You are on page 1of 2491

About the pagination of this eBook

This eBook contains a multi-volume set.

To navigate this eBook by page number, you will need to use the
volume number and the page number, separated by punctuation or a
space.

Refer to the Cumulative Index and match the page reference style
exactly in the Go box at the bottom of the screen.
HORMONES, BRAIN,
AND BEHAVIOR

THIRD EDITION
This page intentionally left blank
HORMONES, BRAIN,
AND BEHAVIOR

THIRD EDITION

EDITORS-IN-CHIEF
Donald W. Pfaff
The Rockefeller University, New York, NY, USA

Marian Joëls
University Medical Center Utrecht, Utrecht, The Netherlands;
and University Medical Center Groningen, Groningen, The Netherlands

VOLUME 1
MAMMALIAN HORMONE-BEHAVIOR SYSTEMS

VOLUME EDITOR
Gabriela González-Mariscal
CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico

AMSTERDAM • BOSTON • HEIDELBERG • LONDON • NEW YORK • OXFORD


PARIS • SAN DIEGO • SAN FRANCISCO • SINGAPORE • SYDNEY • TOKYO
Academic Press is an imprint of Elsevier
Academic Press is an imprint of Elsevier
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
125, London Wall, EC2Y, 5AS
525 B Street, Suite 1800, San Diego, CA 92101-4495, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States

Copyright Ó 2017 Elsevier Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including
photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how
to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the
Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be
noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in
research methods, professional practices, or medical treatment may become necessary.

Practitioners and researchers may always rely on their own experience and knowledge in evaluating and using any information, methods,
compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the
safety of others, including parties for whom they have a professional responsibility.

To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or
damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods,
products, instructions, or ideas contained in the material herein.

Library of Congress Cataloging-in-Publication Data


A catalog record for this book is available from the Library of Congress

British Library Cataloguing-in-Publication Data


A catalogue record for this book is available from the British Library

ISBN: 978-0-12-803592-4

For information on all publications visit our website


at http://store.elsevier.com

Printed and bound in Canada

Publisher: Oliver Walter


Acquisition Editor: Susan Dennis
Content Project Manager: Michael Nicholls
Associate Content Project Manager: Marise Willis
Designer: Greg Harris
EDITORIAL BOARD

EDITORS-IN-CHIEF
Donald W. Pfaff
The Rockefeller University, New York, NY, USA

Marian Joëls
University Medical Center Utrecht, Utrecht, The Netherlands;
and University Medical Center Groningen, Groningen, The Netherlands

VOLUME EDITORS
Gabriela González-Mariscal (Volume 1: Mammalian Hormone-Behavior Systems)
CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico

Jacques Balthazart (Volume 2: Non-Mammalian Hormone-Behavior Systems)


University of Liège, Liège, Belgium

E. Ronald de Kloet (Volume 3: Molecular and Cellular Mechanisms)


Leiden University Medical Center, Leiden, The Netherlands

Stafford Lightman (Volume 4: Clinically Important Hormone Effects on Brain and Behavior)
University of Bristol, Bristol, UK

Anthony P. Auger (Volume 5: Development of Hormone-Behavior Relationships)


University of Wisconsin, Madison, WI, USA

Catherine J. Auger (Volume 5: Development of Hormone-Behavior Relationships)


University of Wisconsin, Madison, WI, USA

v
This page intentionally left blank
CONTRIBUTORS TO VOLUME 1

Raúl Aguilar-Roblero M H Ferkin


Instituto de Fisiología Celular, Ciudad Universitaria, University of Memphis, Memphis, TN, USA
UNAM, México, Mexico
Jeannie M Fiber
Osborne F X Almeida Contract Scientist, Cambridge, MA, USA
Max Planck Institute of Psychiatry, Munich, Germany
Nicole J Gervais
Elvira Arellanes-Licea University of Massachusetts, Amherst, MA, USA
Instituto Potosino de Investigación Científica y
Tecnológica, San Luis Potosí, Mexico Oscar González-Flores
Centro de Investigación en Reproducción Animal,
Wayne G Brake Universidad Autónoma de Tlaxcala-CINVESTAV,
Center for Studies in Behavioral Neurobiology (CSBN), Tlaxcala, México
Concordia University, Montreal, QC, Canada
Gabriela González-Mariscal
Anne Sisto Burt Centro de Investigación en Reproducción Animal,
Facultad de Medicina Veterinaria y Zootecnia, CINVESTAV-Universidad Autónoma de Tlaxcala,
Universidad Nacional Autónoma de México, Mexico Tlaxcala, Mexico
City, Mexico
Erno J Hermans
Matthew P Butler Radboud University Medical Center, Donders Institute
Oregon Health & Science University, Portland, OR, for Brain, Cognition and Behaviour, Radboud University
USA Nijmegen, Nijmegen, The Netherlands
M Caba
A Y Herrera
Centro de Investigaciones Biomédicas, Universidad
University of Southern California, Los Angeles, CA, USA
Veracruzana, Xalapa, Veracruz, México
Kurt L Hoffman
C S Carter
Centro de Investigación en Reproducción Animal,
The Kinsey Institute, Indiana University, Bloomington,
Universidad Autónoma de Tlaxcala-CINVESTAV,
IN, USA
Tlaxcala, México
Annette D de Kloet
Elaine M Hull
College of Medicine, University of Florida, Gainesville,
Florida State University, Tallahassee, FL, USA
FL, USA
Ilia Karatsoreos
J delBarco-Trillo
Washington State University, Pullman, WA, USA
Liverpool John Moores University, Liverpool, UK
Matthieu Keller
José A Delgadillo
Behavioral & Reproductive Physiology, INRA/CNRS/
Centro de Investigación en Reproducción Caprina
University of Tours, Nouzilly, France
(CIRCA), Universidad Autónoma Agraria Antonio
Narro, Torreón, México E B Keverne
University of Cambridge, Cambridge, UK
Mauricio Díaz-Muñoz
Instituto de Neurobiología, Campus UNAM-Juriquilla, Eric G Krause
Querétaro, Mexico College of Pharmacy, University of Florida, Gainesville,
FL, USA

vii
viii Contributors to Volume 1

Lance J Kriegsfeld Ryszard Przewlocki


University of California, Berkeley, CA, USA Institute of Pharmacology, Polish Academy of Sciences,
Krakow, Poland
Agnès Lacreuse
University of Massachusetts, Amherst, MA, USA Gabriela Rodríguez-Manzo
Yazmín Macotela Cinvestav, Mexico City, México
Instituto de Neurobiología, Campus UNAM-Juriquilla, Benno Roozendaal
Querétaro, Mexico Radboud University Medical Center, Donders Institute
A I Melo for Brain, Cognition and Behaviour, Radboud University
Centro de Investigación en Reproducción Animal, Nijmegen, Nijmegen, The Netherlands
CINVESTAV-Universidad Autónoma de Tlaxcala, Rae Silver
Tlaxcala, México Columbia University, New York, NY, USA; Barnard
Sally P Mendoza College, New York, NY, USA; and College of Physicians
University of California, Davis, Davis, CA, USA and Surgeons, New York, NY, USA

Randy J Nelson Cheryl L Sisk


Neuroscience Research Institute, Ohio State University Michigan State University, East Lansing, MI, USA
Wexner Medical Center, Columbus, OH, USA
Tyler J Stevenson
S E Nielsen Institute of Biological and Environmental Sciences,
University of Southern California, Los Angeles, CA, University of Aberdeen, Aberdeen, UK
USA
Jennifer M Swann
Raymond Nowak Lehigh University, Bethlehem, PA, USA
INRA, UMR85 Physiologie de la Reproduction et des
Comportements, Nouzilly, France; CNRS, UMR7247 Brian C Trainor
Physiologie de la Reproduction et des Comportements, University of California, Davis, CA, USA
Nouzilly, France; Université François Rabelais de Tours, Francisco Vázquez-Cuevas
Tours, France; and Institut Français du cheval et de Instituto de Neurobiología, Campus UNAM-Juriquilla,
l’Equitation, Nouzilly, France Querétaro, Mexico
Raúl G Paredes Olivia Vázquez-Martínez
Instituto de Neurobiologia, Universidad Nacional Instituto de Neurobiología, Campus UNAM-Juriquilla,
Autónoma de México, Querétaro, México Querétaro, Mexico
Aras Petrulis Iván Villanueva
Georgia State University, Atlanta, GA, USA Escuela Nacional de Ciencias Biológicas, IPN, México,
Brian J Prendergast Mexico
University of Chicago, Chicago, IL, USA
CONTENTS OF ALL VOLUMES

Editorial Board v
Contributors to Volume 1 vii
Preface of the First Edition xvii
Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease.
An Introduction to the Second Edition xix
Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease.
An Introduction to the Third Edition xxi

VOLUME 1: MAMMALIAN HORMONE-BEHAVIOR SYSTEMS

1.01 Male Sexual Behavior 1


Elaine M Hull and Gabriela Rodríguez-Manzo
1.02 Female Sexual Behavior in Rodents, Lagomorphs, and Goats 59
Oscar González-Flores, Kurt L Hoffman, José A Delgadillo, Matthieu Keller, and Raúl G Paredes
1.03 Parental Behavior 83
G González-Mariscal, M Caba, K L Hoffman, and A I Melo
1.04 The Neurobiology of Social Affiliation and Pair Bonding 117
C S Carter and E B Keverne
1.05 Hormones and the Development and Expression of Aggressive Behavior 145
Brian C Trainor, Cheryl L Sisk, and Randy J Nelson
1.06 Food Intake and Its Control by Signaling Molecules 175
Francisco Vázquez-Cuevas, Raúl Aguilar-Roblero, Elvira Arellanes-Licea, Yazmín Macotela,
Olivia Vázquez-Martínez, Iván Villanueva, and Mauricio Díaz-Muñoz
1.07 Body Fluid Homeostasis 211
Eric G Krause and Annette D de Kloet
1.08 Stress and Opioid Systems 225
Ryszard Przewlocki and Osborne F X Almeida
1.09 Social Stress: Concepts, Assumptions, and Animal Models 261
Sally P Mendoza

ix
x Contents of All Volumes

1.10 Communication by Chemical Signals: Physiological Mechanisms, Ontogeny and Learning,


Function, Evolution, and Cognition 285
M H Ferkin, J delBarco-Trillo, and A Petrulis
1.11 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 329
Aras Petrulis, Jeannie M Fiber, and Jennifer M Swann
1.12 Circadian Regulation of Endocrine Functions 345
Matthew P Butler, Ilia Karatsoreos, Lance J Kriegsfeld, and Rae Silver
1.13 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 371
Tyler J Stevenson, Brian J Prendergast, and Randy J Nelson
1.14 Sex Steroids, Learning and Memory 399
S E Nielsen and A Y Herrera
1.15 Adrenal Stress Hormone Effects on Memory 423
Benno Roozendaal and Erno J Hermans
1.16 Ovarian Hormones and Prefrontal Cortex-Related Cognition 439
Nicole J Gervais, Wayne G Brake, and Agnès Lacreuse
1.17 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 453
Gabriela González-Mariscal, Anne Sisto Burt, and Raymond Nowak

VOLUME 2: NON-MAMMALIAN HORMONE-BEHAVIOR SYSTEMS

Non-mammalian Vertebrates: Fishes


2.01 Neuroendocrinology of Social Behavior in Teleost Fish 3
David Gonçalves, Ana S Félix, and Rui F Oliveira
2.02 Social Regulation of Sex: How the Brain Controls Reproductive Circuits 19
Russell D Fernald
2.03 Socially Controlled Sex Change in Fishes 31
John Godwin and Melissa Lamm
2.04 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish
as Model Systems 47
Ni Y Feng and Andrew H Bass
2.05 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 69
Kent D Dunlap, Ana C Silva, G Troy Smith, and Harold H Zakon

Non-mammalian Vertebrates: Amphibians


2.06 Neuroendocrine Control of Social Behavior in Frogs 101
Walter Wilczynski
2.07 The Vertebrate Brain Stem as the Primary Site of Behavior Command:
Insights from an Amphibian 117
Emma Coddington
2.08 Hormones and Vocal Systems: Insights from Xenopus 131
Erik Zornik and Darcy B Kelley
2.09 Endocrinology of Complex Life Cycles: Amphibians 145
Robert J Denver
Contents of All Volumes xi

Non-mammalian Vertebrates: Reptiles


2.10 Hormones, Brain, and Behavior in Reptiles 171
David Kabelik and David Crews

Non-mammalian Vertebrates: Birds


2.11 Neuroendocrine Regulation of Reproductive Behavior in Birds 217
Gregory F Ball and Jacques Balthazart
2.12 Neural and Hormonal Control of Birdsong 255
Barney A Schlinger and Eliot A Brenowitz
2.13 Rapid Effects of Estrogens on Avian Brain and Social Behavior 291
L Remage-Healey, S A Heimovics, K K Soma, and C A Cornil
2.14 The Function of Behavior as Assessed by Phenotypic Engineering with Testosterone 305
Nicole M Gerlach and Ellen D Ketterson
2.15 Ecophysiological Studies of HormoneeBehavior Relations in Birds: Future Challenges
in a Changing World 321
John C Wingfield

Non-mammalian Invertebrates
2.16 Actions of Developmental Hormones in Adult Social Insects 349
Susan E Fahrbach, Ashton M Trawinski, and Rodrigo A Velarde
2.17 Diverse Functions of Insect Biogenic Amines as Neurotransmitters, Neuromodulators, and
Neurohormones 367
Wendi S Neckameyer and Sandra M Leal
2.18 Regulation of Honeybee Worker (Apis mellifera) Life Histories by Vitellogenin 403
Gyan P Harwood, Kate E Ihle, Heli Salmela (nee Havukainen), and Gro V Amdam
2.19 Endocrine Influences on Insect Societies 421
A R Hamilton, H Shpigler, G Bloch, D E Wheeler, and G E Robinson
2.20 Hormonal Regulation of Behavioral and Phenotypic Plasticity in Bumblebees 453
Abraham Hefetz and Christina M Grozinger
2.21 Stressed Out Insects II. Physiology, Behavior, and Neuroendocrine Circuits Mediating
Stress Responses 465
Erik C Johnson

VOLUME 3: MOLECULAR AND CELLULAR MECHANISMS

3.01 Membrane-Initiated Effects of Estradiol in the Central Nervous System 1


Oline K Rønnekleiv and Martin J Kelly
3.02 Estrogen Regulation of Neurotransmitter and Growth Factor Signaling in the Brain 23
A M Etgen and L M Garcia-Segura
3.03 Genetic and Epigenetic Mechanisms in Neural and Hormonal Controls over Female Reproductive
Behaviors 55
Lee-Ming Kow, A W Lee, Carolyn Klinge, Margaret Warner, Jan-Ake Gustafsson, and D W Pfaff
3.04 Epigenetics of Glucocorticoid Action 83
Karen R Mifsud, Sylvia D Carter, Astrid C E Linthorst, and Johannes M H M Reul
xii Contents of All Volumes

3.05 Androgen Regulation of Neural Circuit Activity: Molecules and Mechanisms 101
Jaroslava Durdiaková and Peter Celec
3.06 Molecular Aspects of Thyroid Hormone-Regulated Behavior 111
Grant W Anderson and Cary N Mariash
3.07 Corticosteroid Actions on Electrical Activity in the Limbic Brain 131
M Joëls, R A Sarabdjitsingh, and H Karst
3.08 Genomic Aspects of Corticosteroid Action in the Brain 149
Lisa T C M van Weert and Onno C Meijer
3.09 Aldosterone Action on Brain and Behavior 159
Harald Murck
3.10 Mechanism of Progesterone Action in the Brain 181
Ignacio Camacho-Arroyo, Valeria Hansberg-Pastor, Edgar Ricardo Vázquez-Martínez, and Marco Cerbón
3.11 Progesterone: Synthesis, Metabolism, Mechanism of Action, and Effects in the Nervous System 215
Michael Schumacher, Xiaoyan Zhu, and Rachida Guennoun
3.12 Neuroactive Steroids and the GABAA Receptor 245
S S Smith and H Shen
3.13 Oxytocin 259
Larry J Young and Hans H Zingg
3.14 Vasopressin: Roles in Modulating Social Behaviors 279
Sarah K Williams Avram and Adi Cymerblit-Sabba
3.15 The Cell Biology of Oxytocin and Vasopressin Cells 305
Jeffrey G Tasker, Daniel L Voisin, and William E Armstrong
3.16 Electrophysiological and Molecular Properties of the Oxytocin- and Vasopressin-Secreting
Systems in Mammals 337
Gary S Bhumbra, Richard E J Dyball, Yoichi Ueta, and Hiroshi Yamashita
3.17 The Gonadotropin-Releasing Hormone and Its Receptor 363
Lothar Jennes, Alfredo Ulloa-Aguirre, Jo Ann Janovick, and P Michael Conn
3.18 Neurophysiology of Gonadotropin-Releasing Hormone Neurons 379
Catherine A Christian
3.19 Action of CRF/Urocortin Peptides 401
Jan M Deussing, Andrey E Ryabinin, Alfredo Zuniga, and Tamas Kozicz
3.20 The Brain Renin Angiotensin System 417
Annette D de Kloet and Eric G Krause

VOLUME 4: CLINICALLY IMPORTANT HORMONE EFFECTS ON BRAIN AND BEHAVIOR

Disorders of Development
4.01 The Gonadal Axis: A Life Perspective 3
Ilpo T Huhtaniemi, Sasha Howard, Leo Dunkel, and Richard A Anderson
4.02 Disorders of Sex Development in Males: Molecular Genetics, Epigenetics, Gender Identity,
and Cognition 59
Yuan-Shan Zhu and Julianne L Imperato-McGinley
Contents of All Volumes xiii

4.03 Genetic Defects of Female Sexual Differentiation 105


Martine Cools, Hedi L Claahsen-van der Grinten, Elfride De Baere, Nina Callens, and Arianne B Dessens
4.04 HypothalamicePituitaryeAdrenal Axis: Congenital Adrenal Hyperplasia 135
Vickie Pasterski and Ieuan Hughes
4.05 Sexual Orientation in Men and Women 151
L J Gooren and W Byne
4.06 Gender Identity Variants 169
Heino F L Meyer-Bahlburg and Wylie C Hembree
4.07 HypothalamicePituitaryeThyroid Axis 179
Russell T Joffe
4.08 Glucocorticoids and Programming of the Fetal Brain 189
Rebecca M Reynolds and Jonathan R Seckl

Disorders in the Adult


4.09 Premenstrual Dysphoric Disorder 197
Henry J Orff and Barbara L Parry
4.10 Mood and Anxiety Disorders in the Context of Fertility Treatments 217
I Fried-Zaig, Y Joskowicz, and M Bloch
4.11 Thyroid Hormone Replacement 229
V Eligar, P N Taylor, O E Okosieme, and C M Dayan
4.12 Glucocorticoid Replacement in Man 241
Georgina M Russell, Kristian Løvås, and Stafford Lightman
4.13 Stress and Anxiety Disorders 251
C V Chen, S A George, and I Liberzon
4.14 Neuroendocrine Aspects of Posttraumatic Stress Disorder 275
Heather N Bader and Rachel Yehuda
4.15 Growth Hormone and Insulin-Like Growth Factor-I: Effects on the Brain 289
Zvi Laron
4.16 Aging and Alzheimer’s Disease 311
Shireen Sindi, Anna Rosenberg, Robert-Paul Juster, and Sonia J Lupien

Specific Disease Entities


4.17 Health-Related Quality of Life and Behavior in Patients with Both Pituitary and Hypothalamic
Diseases 343
Iris Crespo and Susan M Webb
4.18 Eating and Feeding Disorders 355
Madhusmita Misra and Kamryn T Eddy
4.19 Obesity and Appetite: Central Control Mechanisms 369
Tamas L Horvath
4.20 Alcohol Abuse: Endocrine Concomitants 377
P Bortoletto, E W Rosenthal, and E S Ginsburg
4.21 Endocrinological Effects of Drugs of Abuse 403
Brian Reed and Mary Jeanne Kreek
xiv Contents of All Volumes

4.22 Cognitive Dysfunction in Diabetes Mellitus 421


G J Biessels and C M Ryan
4.23 Sex Differences in Autoimmune Diseases 445
R Voskuhl and Taia T Wang

VOLUME 5: DEVELOPMENT OF HORMONE-BEHAVIOR RELATIONSHIPS

Early Life Stages


5.01 Sexual Differentiation of the Brain: A Fresh Look at Mode, Mechanisms, and Meaning 3
Margaret M McCarthy, Geert J De Vries, and Nancy G Forger
5.02 Sex Differences in the Age of Genetics 33
Arthur P Arnold
5.03 Epigenetic Contribution to Sex Differences in Brain and Behavior 49
Anthony P Auger and Margaret M McCarthy
5.04 Environmental Endocrine Disruption of Brain and Behavior 63
H B Patisaul, A C Gore, and D Crews
5.05 Sex Differences in and Hormone Effects on Mammalian Pheromonal Communication 89
James A Cherry and Michael J Baum
5.06 Parental and Early Developmental Stress Impact on Neurodevelopmental and Neuropsychiatric
Disorders 117
B M Nugent, J C Chan, E Jasarevic, and T L Bale
5.07 Early-Life Experiences: Enduring Behavioral, Neurological, and Endocrinological Consequences 133
Russell D Romeo, Akaysha C Tang, and Regina M Sullivan
5.08 Thyroid Hormones and Brain Development 159
Juan Bernal
5.09 Sexual Differentiation of Brain and Behavior in Birds 185
Jacques Balthazart, Arthur P Arnold, and Elizabeth Adkins-Regan
5.10 Sexual Differentiation of Behavior in Nonhuman Primates 225
Kim Wallen
5.11 Gonadal Hormones and Sexual Differentiation of Human Brain and Behavior 247
M Hines
5.12 Sexual Identity and Sexual Orientation 279
Laura Castellanos-Cruz, Ai-Min Bao, and Dick F Swaab

Later Life Stages


5.13 Gonadal Hormonal Influences on the Adolescent Brain and Trajectories of Behavioral
Development 293
Kalynn M Schulz and Cheryl L Sisk
5.14 Neuroendocrine Regulation of Puberty 309
Ei Terasawa and Jon E Levine
5.15 Human Puberty: Physiology, Progression, and Genetic Regulation of Variation in Onset 357
B A Kaminski and M R Palmert
Contents of All Volumes xv

5.16 Hormone Regulation of Neurogenesis Across the Lifespan 373


P Duarte-Guterman, S Yagi, J R Epp, S Brummelte, S R Wainwright, C K Barha,
C Chow, J M Barker, and Liisa A M Galea
5.17 Effects of Sex Steroids on Damaged Neural Systems 411
Donald G Stein
5.18 Effects of Stress Throughout the Lifespan on the Brain and Behavior 443
C Nasca, E Davis, B Bigio, C Sandi, and B S McEwen
5.19 Reproductive Aging of Neuroendocrine Systems 465
Roberta Diaz Brinton

Index 477
This page intentionally left blank
PREFACE OF THE FIRST EDITION

Because of the large number of neuroactive substances discovered by chemical endocrinologists and the
prominent roles of nuclear hormone receptors as ligand-activated transcription factors, scientists studying
hormoneebrainebehavior relations have made discoveries and achieved explanations of behavior that place
their field foremost in neurobiology. The purpose of these volumes is to review the current state of this
knowledge inclusively. That means covering true molecular genetic approaches as well as neuroanatomical,
electrophysiological, zoological, neurochemical, developmental, and behavioral studies. The medical impor-
tance of this work is clear from the last section of this treatise. The editors intend these reviews to be
comprehensive; if there are any gaps in the coverage, a second edition will correct them.

Acknowledgments

Lucy Frank and Carol Oliver, at The Rockefeller University, organized all the volumes. Noelle Gracy and Mica
Haley handled things efficiently at Academic Press. Jasna Markovac at Academic Press presided over all stages of
this project. The editors thank all of these folks for their intelligent and gracious efforts.

Donald W. Pfaff
The Rockefeller University, New York, NY, USA

xvii
This page intentionally left blank
HORMONAL EFFECTS ON SPECIFIC BEHAVIORS, ON GLOBAL
CNS STATES, AND ON HUMAN DISEASE. AN INTRODUCTION
TO THE SECOND EDITION

In 2002, there appeared the first edition of Hormones, Brain, and Behavior, dedicated to three generations of
productive scientists in our field, as it had developed at that point. With five volumes, totaling about 4100
pages, it represented a massive effort by over 200 scientists. The volumes were well received.
At least four developments have taken place since 2002 and justify a second edition. Two of them are
technical: the increasingly sophisticated application of molecular biological techniques and biophysical tech-
niques to the understanding of mechanisms for hormone/behavior relations. Third, we have entered an era of
translational research. Laboratory scientists are under increasing pressure to show how their work might lessen
the burden of disease upon the citizenry. The fourth development is the most subtle one and can be explained as
follows.
Some contemporary approaches to the hormonal controls of various forms of behavior can be distin-
guished sharply from the major theme in neuroscientific research in the past century. I characterize that
classical theme as a hunt for specificity. In the spinal cord, for example, the work of neurophysiologists such as
Sir Charles Sherrington (1906) and Sir John Eccles (1957) sought to explain why a particular reflex response
occurred specifically as a result of one particular stimulus and not another. At the level of the cerebral cortex,
work typified by that of David Hubel and Wiesel (1977) in the visual system and Vernon Mountcastle (1957)
in the somatosensory system described cells whose responses required stimuli of a specific type in a well-
defined receptive field. My own work, explaining mechanisms for lordosis behavior (Pfaff, 1999), fell into that
tradition.
In contrast, new approaches to changes of global states of the central nervous system involve the
regulation of large classes of behavioral responses to large sets of adequate stimuli. Thus, hormonal
influences on mood, exploratory tendencies, arousal, alertness, and attention would not be specific in the
manner displayed by neurons in the visual cortex, but they would be incredibly important for main-
taining mental and physical health. Our field of hormones and behavior increasingly embraces these
concepts.
In order to add new material, discovered since 2002, with special emphasis on clinical research, we had
to adopt the following strategy. We asked authors who had contributed to the first edition to reduce
markedly the length of their treatments of findings already covered in 2002. Therefore, the reader of this
current (second) edition will want to refer to the first edition as well, for the kind of comprehensive review
we intend.
Taking the first and the second editions of Hormones, Brain, and Behavior together will afford the reader
a tremendous number of facts, ranging from molecular genomics of hormone action, through biophysics,
neuroanatomy, and endocrinology to animal behavior and human disease. But this bulky coverage of primary
research findings is not, across the scope of several volumes, intended to be logically systematic. A parallel effort
in the text Principles of Hormone Behavior Relations takes a step in that direction.

References
Eccles, J., 1957. The Physiology of Nerve Cells. Johns Hopkins University Press, Baltimore, MD.
Hubel, D., Wiesel, T., 1977. Functional architecture of macaque monkey visual cortex. Proc. R. Soc. Lond. 198, 1e59.

xix
xx Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease

Mountcastle, V., 1957. Modality and topographic properties of single neurons of cat’s somatic sensory cortex. J. Neurophysiol. 20, 408e434.
Pfaff, D., 1999. Drive: Neural and Molecular Mechanisms of a Motivated Behavior. MIT Press, Cambridge, MA.
Sherrington, C., 1906. The Integrative Action of the Nervous System. Yale University Press, New Haven, CT (Reprinted 1947).

Further Reading
Pfaff, D., Phillips, M.I., Rubin, R.T., 2004. Principles of Hormone/Behavior Relations. Academic Press/Elsevier, San Diego.

Donald W. Pfaff
The Rockefeller University, New York, NY, USA
HORMONAL EFFECTS ON SPECIFIC BEHAVIORS, ON GLOBAL CNS
STATES, AND ON HUMAN DISEASE. AN INTRODUCTION TO THE
THIRD EDITION

In 2002, the first edition of Hormones, Brain, and Behavior appeared, dedicated to three generations of productive
scientists in our field, as it had developed at that point. With five volumes, totaling about 4100 pages, it rep-
resented a massive effort by over 200 scientists. In 2009 the volumes were updated, in the second edition. Both
sets of five volumes were well received.
At least four developments have taken place since 2009 which justify a third edition. Two are technical: the
application of increasingly sophisticated molecular biological techniques and biophysical techniques to the
understanding of mechanisms for hormone/behavior relations. Third, we have entered an era of translational
research. Laboratory scientists are under increasing pressure to show how their work might lessen the burden of
disease upon the citizenry. The fourth development is the most subtle one and can be explained as follows.
Some contemporary approaches to the hormonal controls of various forms of behavior can be distinguished
sharply from the major theme in neuroscientific research in the past century. We characterize that classical theme
as a ‘hunt for specificity.’ In the spinal cord, for example, the work of neurophysiologists such as Sir Charles
Sherrington (1906) and Sir John Eccles (1957) sought to explain why a particular reflex response occurred as
a result of one particular stimulus and not another. At the level of the cerebral cortex, work typified by that of
David Hubel and TorstenWiesel (1977) in the visual system and Vernon Mountcastle (1957) in the somato-
sensory system described cells whose responses required stimuli of a specific type in a well-defined receptive field.
Pfaff’s own work, explaining mechanisms for lordosis behavior (Pfaff, 1999), fell into that tradition.
In contrast, new approaches to changes of global states of the central nervous system involve the regulation of
large classes of behavioral responses to large sets of adequate stimuli. Experimental work on steroid hormone
regulation of responses to stress provide an example of this type of brain research (reviewed, Joels et al., 2013). Thus,
hormonal influences on mood, exploratory tendencies, arousal, alertness, and attention would not be specific in the
manner displayed by neurons in the visual cortex, but they would be incredibly important for maintaining mental
and physical health. Our field of hormones and behavior increasingly embraces these concepts.
In order to add new material, discovered since 2009, with special emphasis on clinical research, we had to
adopt the following strategy. We asked authors who had contributed to the first and second editions to reduce
markedly the length of their treatments of findings already covered in 2009; therefore, the reader of this current
(third) edition may want to refer to the earlier editions as well, for the kind of comprehensive review we intend.
In lieu of these earlier sections, the authors expanded information on more recent concepts and experiments.
Taking all editions of Hormones, Brain, and Behavior together will afford the reader a tremendous number of
facts, ranging from molecular genomics of hormone action, through biophysics, neuroanatomy, and endo-
crinology to animal behavior and human disease. But this bulky coverage of primary research findings is not,
across the scope of several volumes, intended to be logically systematic.
A parallel effort in the text Principles of Hormone Behavior Relations (the second edition of which will appear
soon) takes a step in that direction.

References
Eccles, J., 1957. The Physiology of Nerve Cells. Johns Hopkins University Press, Baltimore, MD.
Hubel, D., Wiesel, T., 1977. Functional architecture of macaque monkey visual cortex. Proc. R. Soc. Lond. 198, 1e59.

xxi
xxii Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease

Joels, M., Pasricha, N., Karst, H., 2013. The interplay between rapid and slow corticosteroid actions in brain. Eur. J. Pharmacol. 719, 44e52.
Mountcastle, V., 1957. Modality and topographic properties of single neurons of cat’s somatic sensory cortex. J. Neurophysiol. 20, 408e434.
Pfaff, D., 1999. Drive: Neural and Molecular Mechanisms of a Motivated Behavior. MIT Press, Cambridge, MA.
Sherrington, C., 1906. The Integrative Action of the Nervous System. Yale University Press, New Haven, CT (Reprinted 1947).

Further Reading
Pfaff, D., Phillips, M.I., Rubin, R.T., 2004. Principles of Hormone/Behavior Relations. Academic Press/Elsevier, San Diego.

Donald W. Pfaff
The Rockefeller University, New York, NY, USA
Marian Joëls
University Medical Center Utrecht, Utrecht, The Netherlands
PERMISSION ACKNOWLEDGMENT

The following material is reproduced with kind permission of Oxford University Press
Figure 1 of Regulation of Honeybee Worker (Apis mellifera) Life Histories by Vitellogenin
www.oup.com

i
1.01 Male Sexual Behavior
Elaine M Hull, Florida State University, Tallahassee, FL, USA
Gabriela Rodrı́guez-Manzo, Cinvestav, Mexico City, México
Ó 2017 Elsevier Inc. All rights reserved.

1.01.1 Patterns of Male Sexual Behavior 2


1.01.1.1 Description of Behavioral Elements 2
1.01.1.1.1 Precopulatory Behaviors 2
1.01.1.1.2 Copulatory Behavior 2
1.01.1.2 Sexual Satiety 3
1.01.1.3 Motivation and Performance 4
1.01.1.4 Sexual Experience 5
1.01.1.5 Sexual Behavior during Puberty and Aging 5
1.01.1.5.1 Puberty 5
1.01.1.5.2 Aging 6
1.01.2 Sexual Reflexes 7
1.01.2.1 Observations during Copulation 7
1.01.2.2 Ex Copula Sexual Reflexes 7
1.01.2.2.1 Spontaneous or Drug-Induced Erections 7
1.01.2.2.2 Noncontact Erections 7
1.01.2.2.3 Reflexive Erections, Anteroflexions, and Seminal Emissions 7
1.01.2.2.4 The Urethrogenital Reflex 8
1.01.2.3 Erection 8
1.01.2.3.1 Anatomy of the Penis and Mechanisms of Erection 8
1.01.2.3.2 Neural Innervation 8
1.01.2.3.3 Cellular Mediators of Erection 9
1.01.2.4 Ejaculation 10
1.01.3 Role of Gonadal Steroids in the Control of Male Sexual Behavior 12
1.01.3.1 Testosterone and Its Metabolites 12
1.01.3.1.1 Time Course of Changes in Copulation Following Castration and T Restoration 12
1.01.3.1.2 Role of T Metabolites in Maintaining and Restoring Copulation 12
1.01.3.1.3 Effects of Castration and Hormone Replacement on Ex Copula Penile Responses 13
1.01.3.2 Steroid Action on Steriod Hormone Receptors 14
1.01.3.3 The Role of Progesterone in Male Sexual Behavior 14
1.01.4 Effects of Systemically and Intraventricularly Injected Drugs 14
1.01.4.1 Dopamine 14
1.01.4.2 Norepinephrine 16
1.01.4.3 Serotonin 16
1.01.4.4 Acetylcholine 18
1.01.4.5 Gamma Aminobutyric Acid 18
1.01.4.6 Glutamate 18
1.01.4.7 Nitric Oxide 18
1.01.4.8 Endocannabinoids 19
1.01.4.9 Endogenous Opioids 19
1.01.4.10 Oxytocin 20
1.01.4.11 Prolactin 20
1.01.4.12 Gonadotropin-Releasing Hormone 21
1.01.4.13 Orexin/Hypocretin 21
1.01.5 Brain Areas and Circuitry Implicated in the Control of Masculine Sexual Behavior 21
1.01.5.1 Sensory Inputs 21
1.01.5.1.1 Olfactory Bulbs 21
1.01.5.1.2 Amygdala 22
1.01.5.1.3 Bed Nucleus of the Stria Terminalis 24
1.01.5.1.4 Central Tegmental Field/Subparafascicular Nucleus of the Thalamus 25
1.01.5.2 Major Integrative Sites 26
1.01.5.2.1 Medial Preoptic Area 26
1.01.5.2.2 Mesocorticolimbic DA Tract 31
1.01.5.2.3 Nigrostriatal DA Tract 33

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00001-8 1


2 Male Sexual Behavior

1.01.5.2.4 Paraventricular Nucleus of the Hypothalamus 33


1.01.5.2.5 Lateral Hypothalamus 35
1.01.5.2.6 Ventromedial Hypothalamus 35
1.01.5.3 Major Motor Outputs 36
1.01.5.3.1 Ventral Premammillary Nucleus 36
1.01.5.3.2 Midbrain Periaqueductal Gray 36
1.01.5.3.3 Nucleus Paragigantocellularis of the Medulla 36
1.01.5.3.4 Other Brain Areas 37
1.01.5.3.5 Spinal Cord 37
1.01.6 Circuitry and Anatomical Interconnections 38
1.01.6.1 Sensory Inputs 38
1.01.6.2 An Ejaculation-Related Circuit 38
1.01.6.3 Efferents from the MPOA 40
1.01.6.4 Sexual Behavior in the Context of Mammalian Social Behavior 40
References 40

1.01.1 Patterns of Male Sexual Behavior not achieve penile insertion and dismounts the female
slowly.
1.01.1.1 Description of Behavioral Elements
Male sexual behavior comprises a complex pattern of genital
and somatomotor responses, elicited, directed, and maintained 1.01.1.1.2.2 Intromissions
by external and internal signals. It includes copulation as well Intromission, the defining event of copulation, refers to
as precopulatory behaviors that allow the male to detect and intravaginal penile insertion. The male rat mounts the
locate a mate, assess her potential mating appropriateness, female, performs pelvic thrusting, and suddenly displays
and stimulate a receptive response. a deeper thrust with a mean duration of 200–300 ms (Beyer
et al., 1981) that coincides with vaginal penetration, fol-
1.01.1.1.1 Precopulatory Behaviors lowed by an abrupt backward dismount and grooming of
Precopulatory behavior may consist simply of anogenital inves- his genitalia. Male hamsters show a similar intromission
tigation or may include highly elaborate behavioral patterns, pattern, but after repeated ejaculations (9–10) a motor intro-
depending on the species. Male and female rodents show an mission pattern called ‘long intromissions’ is displayed,
initial period of mutual anogenital investigation; they also during which penile insertion is longer than during typical
emit ultrasonic vocalizations (USV) of 50 kHz, which have preejaculatory intromissions and than during ejaculation.
been considered mutually arousing (Geyer and Barfield, Long intromissions include intravaginal thrusting, and this
1978; Pomerantz and Clemens, 1981), although recent data motor response precedes sexual satiety (reviewed in Hull
suggest that these USV lack incentive value for both male and and Dominguez, 2007). Male rabbits show rhythmic thrusts
female rats (Snoeren and Ågmo, 2013, 2014). Males engage for several seconds, which may or may not result in intromis-
in urine marking (Meisel and Sachs, 1994), the odor of which sion; however, if intromission does occur, ejaculation begins
possesses incentive value for female rats (Snoeren and Ågmo, almost immediately (Morali et al., 2003).
2014). Receptive females will solicit mating from the male by
characteristic proceptive behaviors, and the male will pursue
and mount her. 1.01.1.1.2.3 Ejaculation
Most mammals achieve the ejaculatory threshold after
multiple intromissions. Ejaculatory behavior in male rats
1.01.1.1.2 Copulatory Behavior starts with an intromission but includes a deeper, longer
Male rodents and rabbits exhibit a highly stereotyped copula- thrust (750–2000 ms) (Beyer et al., 1981) that coincides
tory pattern, shaped by three distinct behavioral motor with seminal ejection. The male then raises his forelegs and
patterns: mount (M), intromission (I), and ejaculation (E). dismounts slowly, then typically grooms himself. Mice
show essentially the same ejaculatory pattern, except that
1.01.1.1.2.1 Mounts during ejaculation, they may freeze before dismounting.
Almost all male mammals mount females dorsally and from Male canids begin ejaculating soon after penile insertion
the rear, posing his forelegs over the female’s back and with and develop a swelling at the base of the penis, which results
his hind feet on the ground. The female may assume in a ‘lock’ of the male to the female (Beach, 1969). Male
a lordosis posture, a reflexive dorsoflexion of the spine, ungulates (Lott, 1981) and rabbits (Morali et al., 2003)
accompanied by deflection of the tail to the side. The male also ejaculate immediately upon intromitting. Rabbits’
then begins anteroposterior pelvic thrusts (19–23 Hz) (Beyer mounts that result in ejaculation are shorter, more regular,
et al., 1981) that induce or intensify the female’s receptive and of higher frequency (Morali et al., 2003). Rhythmic
posture. During a nonintromissive mount, the male does contractions of skeletal and striated perineal muscles usually
Male Sexual Behavior 3

accompany ejaculation and are associated with orgasm in (Dewsbury, 1983), mandarin voles (He et al., 2013), and
humans. rhesus monkeys (Bielert and Goy, 1973).
Copulation to satiety in rats increases dopamine (DA)
1.01.1.1.2.4 Postejaculatory Behavior metabolite levels in the medial preoptic area (MPOA), which
The most patent effect of ejaculation is a period of sexual remain elevated during the first 48-h period of sexual inhibi-
quiescence: the postejaculatory interval (PEI). In rats, refracto- tion (Mas et al., 1995a), while nucleus accumbens (NAc) DA
riness lasts 5–8 min after a first ejaculation and increases with levels also increase during copulation to exhaustion (Fiorino
each successive ejaculation. During the initial 75% of the PEI, et al., 1997). The hypothalamic content of enkephalins is
the male emits 22 kHz USVs and will not copulate in response also increased in sexually satiated rats for at least 48 h after
to any stimulus; for that reason, it is called the absolute refrac- sexual exhaustion (Rodríguez-Manzo et al., 2002a). A reduc-
tory period (Barfield and Geyer, 1975). The remaining 25% of tion in androgen receptor (AR) density within the MPOA,
the PEI, the relative refractory period, is no longer accompa- NAc, and ventromedial hypothalamic nucleus (VMH), but
nied by vocalizations, and its duration can be reduced if the not in the bed nucleus of the stria terminalis (BST), of satiated
male is subjected to nonspecific arousing stimuli, such as rats (Fernández-Guasti et al., 2003) and in the lateral septal
a mild electrical shock (Barfield and Sachs, 1968) or a novel nucleus, medial amygdala (MeA), MPOA, and VMH of satiated
female partner (Zucker and Wade, 1968). After the PEI, male mandarin voles (He et al., 2013) has been detected. Fos-
male rats resume pursuance and mounting of the female. immunoreactivity (Fos-ir, a marker of neural activation) was
Mice resume sexual activity 1–24 h after an ejaculation, increased in regions of the BST, MPOA, and MeA following
depending on the mouse strain, while hamsters have much sexual satiety in male hamsters (Parfitt and Newman, 1998),
shorter PEIs, from a few seconds to 1.5 min (reviewed in while in rats the relevant brain structures were the MeA and
Burns-Cusato et al., 2004; Hull and Dominguez, 2007; Meisel septum (Phillips-Farfán and Fernández-Guasti, 2007). A
and Sachs, 1994). During successive ejaculatory series, the contribution of the nucleus paragigantocellularis (nPGi) in
duration of the PEI increases and the number of satiety can be inferred from the fact that nPGi lesions increased
preejaculatory intromissions decreases (Rodríguez-Manzo the latency to and the number of ejaculations that preceded
and Fernández-Guasti, 1994). sexual satiation (Yells et al., 1992).
The mechanisms involved in the postejaculatory refractory Sexual satiety is regulated by diverse neurotransmitters as
period are poorly understood. In rats, there are data suggesting indicated by drug-induced reversal of the sexual inhibitory
the involvement of serotonergic (McIntosh and Barfield, state. Among them, norepinephrine (NE) (Rodríguez-Manzo,
1984a) and GABAergic (Fernández-Guasti et al., 1986a) trans- 1999b; Rodríguez-Manzo and Fernández-Guasti, 1994,
mission in the induction and maintenance of the PEI, while 1995b), serotonin (5-HT) (Arnone et al., 1995; Fernández-
both dopaminergic (McIntosh and Barfield, 1984b) and norad- Guasti and Rodríguez-Manzo, 1997; Rodríguez-Manzo and
renergic (McIntosh and Barfield, 1984c) transmission are Fernández-Guasti, 1994), endogenous opioids (Garduno-
thought to counteract it, but the evidence is limited. Data on Gutierrez et al., 2013b; Miller and Baum, 1987; Pfaus and
human PEI are scarce and therefore do not contribute to Gorzalka, 1987; Rodríguez-Manzo and Fernández-Guasti,
a significant advance in its understanding (Levin, 2009). 1995a), endocannabinoids (Canseco-Alba and Rodriguez-
Manzo, 2014; Rodriguez-Manzo and Canseco-Alba, 2015b),
glutamate (Rodriguez-Manzo, 2015), and DA (Mas et al.,
1.01.1.2 Sexual Satiety
1995b; Rodríguez-Manzo, 1999b; Guadarrama-Bazante et al.,
When allowed to copulate without restriction, male rats will 2014), but not MPOA gamma-aminobutyric acid (GABA)
show approximately seven ejaculations before reaching sexual (Rodríguez-Manzo et al., 2000), all seem to play a role. Most
satiety (Beach and Jordan, 1956; Rodríguez-Manzo and of the drugs that reverse satiety appear to do so via the NE
Fernández-Guasti 1994). The main feature of this phenom- system (Rodríguez-Manzo and Fernández-Guasti, 1995b),
enon is the instatement of a long-lasting sexual behavior inhi- coupled, in turn, to the DA system, which would be a final
bition which may last up to 3 days, from which males gradually pathway for the reversal (Rodríguez-Manzo, 1999b). Both
recover (Rodriguez-Manzo et al., 2011). Twenty-four hours endogenous opioids (Garduno-Gutierrez et al., 2013a) and
after reaching satiety, male rats may show a complete absence endocannabinoids (Canseco-Alba and Rodriguez-Manzo,
of sexual activity or may execute one ejaculatory series without 2014) are released during copulation to satiety and contribute
recovery (Rodríguez-Manzo and Fernández-Guasti, 1994). to the establishment of the long-lasting sexual inhibitory state.
In rabbits, sexual activity was studied over periods up to The mesolimbic DA system is the target of the actions of the
15 days to determine when the male would become exhausted, released endogenous opioids, through the activation of mu
i.e., fail to copulate with a new female for 24 h (Jiménez et al., opioid receptors within the ventral tegmental area (VTA)
2012). Males copulated to satiety on one day, but would (Garduno-Gutierrez et al., 2013b,c). In male hamsters, the
resume the next day. To attain sexual exhaustion, rabbits copu- cholinergic system in the MPOA was found to play a role in
lated ad libitum during several successive days (the number the development of sexual exhaustion. Infusion of the musca-
varying from 2 to 15). During those tests, the number of rinic receptor agonist oxotremorine delayed, while the musca-
ejaculations decreased from 22 on the first day to 6 on the rinic antagonist scopolamine accelerated, the attainment of
last day, and the number of ‘misses’ (mounts without sexual exhaustion (Floody, 2013).
ejaculation) increased. Thus, rabbits require more days to The so-called Coolidge effect (the renewed sexual respon-
become exhausted than do rats. Sexual satiety has also been siveness of male rats that ceased to copulate after reaching
described in hamsters (Beach and Rabedeau, 1959), mice exhaustion that is produced by changing the stimulus female
4 Male Sexual Behavior

(Fisher, 1962)) interferes with the establishment of the sexual copulate; later, the male is placed into the apparatus alone,
inhibition characteristic of sexual exhaustion, 24 h after satiety; and the number of times he changes levels, presumably in search
neurotoxic lesions of NE neurons did not interfere with of the female, is the measure of motivation (Mendelson and
this effect (Rodríguez-Manzo, 1999a). Bromocriptine, a DA Pfaus, 1989). However, this test also confounds motivation
agonist, was recently found to renew sexual responsiveness of and motor ability. Another test is lever pressing for a secondary
sexually exhausted male rats, to the same sexual mate, immedi- reinforcer that was previously associated with copulation
ately after sexual satiety, mimicking the Coolidge effect but (Everitt, 1990). However, this also confounds motivation with
without changing the female partner (Rojas-Hernandez and motor ability and with the ability to associate the secondary
Juarez, 2015). Together these data suggest that a decreased stimuli with copulation. The X-maze or cross-maze uses the
DA transmission may account for cessation of sexual activity percentage of trials on which the male chooses the
at satiety. compartment containing an estrous female, with which he can
In addition to the inhibition of copulation, other responses copulate, compared to empty compartments or those that
are also modified by sexual exhaustion. Thus, sexually satiated contain other goal objects (Hull et al., 1991; Warner et al.,
rats show a general increase in sensitivity to drug actions, 1991). Motor ability is measured as the running speed to all
including those of yohimbine, 8-hydroxy-2-(di-n propyla- goal boxes and the number of trials on which the male does
mino)tetralin (8-OH-DPAT), naloxone, and desipramine not leave the central start area. Motivation is then scored as
(Martínez-Mota et al., 2005; Rodríguez-Manzo and the percentage of trials on which he chooses the female, out of
Fernández-Guasti, 1994, 1995a). Modifications in the neural all trials on which he leaves the start area. Copulatory
control of copulation appear to occur after sexual satiation in measures can also be assessed. However, drugs may affect
rats. Hence, prior to sexual exhaustion, electrical stimulation sensory processes or memory of cues marking the female’s
of the MPOA (Rodríguez-Manzo et al., 2000), the dorsal region goal box. A similar test uses a runway and a goal compartment
of the VTA (Rodríguez-Manzo and Pellicer, 2007), or the NAc with a one-way door, through which a male can enter
(Rodriguez-Manzo and Pellicer, 2010) markedly facilitated a female’s chamber, copulate, and then leave through a second
sexual behavior, but after sexual satiety the same animals no one-way door to the original start box (Beck et al., 2002).
longer responded to this stimulation. Also, the anxiolytic-like Motivation is measured as time in the female’s compartment,
properties of ejaculation, seen in nonexhausted rats, disap- whereas run latency and duration and the number of
peared in the population of sexually satiated males that can copulatory behaviors reflect both motivation and motor
ejaculate 24 h after satiation (Rodríguez-Manzo et al., 1999). ability. However, drugs on the test day may affect sensory
Thus, copulation to satiation promotes long-lasting changes processes. Therefore, there are no ‘pure’ tests of motivation
in male rats’ physiology. that are free from potential confounds. Without additional
tests of sensory, motor, and learning abilities, it is not possible
to differentiate those factors from sexual motivation.
1.01.1.3 Motivation and Performance
Therefore, reports of effects on sexual motivation should
Motivation for sexual activity may be distinguished from actual include a caveat that those measures of motivation may be
sexual performance. Sexual motivation may be compared to an influenced by changes in sensory, motor, or learning ability.
engine that may drive several goal-directed behaviors, whereas The term sexual arousal has also been discussed as a compo-
hormone-sensitized factors may steer the behavior down nent of motivation, although there is no general agreement
specific paths. However, experimental procedures designed to about its definition. For example, Ågmo (2008) suggested
test sexual motivation may have effects that are confounded that sexual arousal is nothing more than enhanced genital
with sensory, motor, or learning abilities. Therefore, tests that blood flow, while others may regard it as more of a species-
separate effects on motivation from those on motor, sensory, specific first step in a series of appetitive behaviors leading
or learning abilities are preferred. toward copulation (discussed in Heiman and Pfaff, 2011).
Several tests do meet at least some of those criteria. Place pref- Ågmo (2011) later proposed that arousal should “refer to the
erence tests assess whether previous copulation in one compart- general state of activity of the organism and of its nervous sys-
ment leads to greater time spent in that compartment, even tem.and cannot be specific to a particular class of stimuli or
without a stimulus female. An advantage of this test is that little reponses.” Furthermore, he proposed that “the execution of
motor ability is required to show a preference; another advan- copulatory acts leads to increased general arousal.” Similarly,
tage is that any drugs administered during conditioning would Schober et al. (2011) argue that a heightened generalized
be metabolized before the test day. However, it does require CNS arousal energizes sexual arousal and vice versa. They
learning ability for the male to associate the external stimuli propose that a network of ascending excitatory neurons to
with the positive stimuli of copulation. A similar procedure the hypothalamus and basal forebrain promote general arousal
uses naïve males, which are allowed to spend time near either and that descending axons from those areas activate brain stem
an estrous or nonestrous female (Ågmo, 2003; Amstislavskaya areas that control the specific behaviors. They report having
and Popova, 2004). Motor ability to approach the two stimulus successfully bred, over six generations, mice that show high
females is the same; however, sensory processing may be affected versus low general arousal. They then found that males bred
by drugs administered before the test. Another test is the obstruc- for high general arousal had more mounts before vaginal intro-
tion apparatus, in which a male is required to cross a barrier or mission, fewer intromissions before ejaculation, and ejaculated
an electrified grid to access a female. However, this procedure more quickly after the first intromission, which the authors
does confound motivation with motor ability. Another test took as evidence for high sexual arousal. Thus, high general
uses a bilevel apparatus in which a male and female can arousal can increase sexual arousal, which in turn may increase
Male Sexual Behavior 5

sexual motivation and influence copulation. Conversely, sexual behavior, compared to that exhibited by first-time copulators
activity can enhance general motivation. (Can et al., 2007). The MPOA, in particular, seems to be the
target of several sexual experience-induced changes in diverse
neurotransmitter systems. Thus, higher levels of nitric oxide
1.01.1.4 Sexual Experience
synthase (NOS) (Dominguez et al., 2006b), increased expres-
Experience plays an important role in the full development and sion of oxytocin (OT) receptors (Gil et al., 2013), and an
efficiency of sexual behavior. Thus, sexual learning reduces the augmented expression of phosphorylated DARPP-32 (DA-
time to initiate sexual contact and to achieve ejaculation, as and cAMP-regulated phosphoprotein, a downstream marker
well as the amount of stimulation required to ejaculate (Pfeiffer of D1 receptor signaling) were detected in the MPOA of
and Johnston, 1994). Sexual experience also improves copula- sexually experienced rats (McHenry et al., 2012), as well as
tory ability (Domjan 1992; Pfaus et al., 2001; Woodson, 2002) an increased number of AR-positive neurons in the MPOA of
and sharpens the olfactory interest of male rodents in female sexually experienced mice (Swaney et al., 2012), when
sexually related chemosignals (Hayashi and Kimura, 1974; compared with sexually naïve animals. By contrast, mating-
Lydell and Doty, 1972; Nielsen et al., 2013; Pfeiffer and John- induced Fos expression in NOS-containing neurons of this
ston, 1994; Swaney et al., 2007). Some of the learning-induced brain region is higher in sexually naïve rats undergoing their
changes in male sexual ability can be interpreted as experience- first sexual encounter than in sexually experienced males
dependent increases in sexual motivation related to the (Nutsch et al., 2014). Testosterone (T) plasma and
rewarding nature of male sexual activity (Pfaus and Phillips, hippocampal levels are also greater in sexually experienced
1991), since males engage in sexual behavior more rapidly, (Edinger and Frye, 2007) than in sexually naïve males, and
ejaculate more often, and display shorter PEIs (Rodriguez- long-term alterations in glutamate receptor expression in rat
Manzo and Canseco-Alba, 2014). Furthermore, sexual experi- NAc neurons occur as a result of sexual experience (Pitchers
ence diminishes or eliminates the disruptive effect of a novel et al., 2012). Sexual experience increased precontact USVs
environment, seen in sexually naïve males (Pfaus and Wilkins, (50 kHz) (Bialy et al., 2000) and decreased anxiety-like
1995). Increased investigation of female chemosignals has also behavior of male rats (Edinger and Frye, 2007), while
been suggested to result from the increased sexual interest promoting neurogenesis in the hippocampus of sexually
exhibited by sexually experienced male mice (Swaney et al., naïve adult male rats (Leuner et al., 2010).
2007), although a recent study in hamsters revealed that, in In summary, sexual experience induces brain plasticity that
this species, sexual experience did not affect male preference allows males to establish associations that strengthen sexual
for a receptive female (Ballard and Wood, 2007). Interestingly, responding. The rewarding component of male sexual activity
sexually experienced male cats (Rosenblatt and Aronson, appears central to this strengthening, and it can even elicit
1958), mice (Manning and Thompson, 1976), and hamsters sexual arousal in response to nonsexual cues that were paired
(Constantini et al., 2007), but not rats (Bloch and Davidson, with the early sexual experience, such as specific scents or
1968), are less susceptible to the disruptive effects of castration. somatosensory cues (Pfaus et al., 2012). It has been proposed
Sexual experience also attenuates the negative effects of several that natural reinforcers, such as sexual reward, lead to brain
brain lesions on sexual performance, such as bilateral olfactory plasticity as a way to accomplish required behavioral adapta-
bulbectomy in rats (Bermant and Taylor, 1969), ablation of the tions (Olsen, 2011).
vomeronasal organ (VNO) in hamsters (Meredith, 1986) and In spite of this, two specific subpopulations can be found in
rats (Saito and Moltz, 1986), but not those resulting from all strains of rats that do not respond to sexual experience: sexu-
zinc sulfate lesions of the main olfactory epithelium in mice ally sluggish and noncopulating animals. Sexually sluggish rats
(Keller et al., 2006). The effects of lesions in the medial poste- are males that exhibit long latencies to initiate copulation and
rior BST (Claro et al., 1995) on copulation were also greater in to achieve ejaculation which are not reduced by repeated sexual
sexually naïve than in sexually experienced male rats, while in experience (Portillo et al., 2006; Rodriguez-Manzo and
Syrian hamsters this lesion decreased opposite-sex odor prefer- Canseco-Alba, 2014), whereas noncopulating rats are appar-
ence and delayed the whole copulatory sequence in sexually ently normal, otherwise healthy animals that will not copulate
naïve, but not in sexually experienced animals (Been and despite repeated exposure to sexually receptive females (Stefa-
Petrulis, 2010a). Lesion of the sexually dimorphic nucleus nick and Davidson, 1987). The ‘lack’ of experience-driven
(SDN) of the MPOA disrupted copulatory behavior in sexually improvement in copulation of sluggish rats or of induction
inexperienced male rats (de Jonge et al., 1989) but lacked an of sexual behavior display in noncopulating males has no clear
effect in sexually experienced animals (Arendash and Gorski, explanation. However, the endocannabinoid anandamide has
1983). In hamsters, discrete lesions of the MPOA caused severe been found to transiently reduce the ejaculatory threshold of
copulatory deficits only in sexually naïve males and not in sluggish rats (Rodriguez-Manzo and Canseco-Alba, 2015a)
males with sexual experience (Been and Petrulis, 2010b). and to transform noncopulating rats into sexually active males
Enhanced neuronal responses as a result of sexual experience in a long-lasting manner (Canseco-Alba and Rodriguez-Manzo,
have also been reported. Thus, ejaculation activated more cells, 2013).
determined by the number of Fos-ir neurons, within the MPOA
(Lumley and Hull, 1999) and the NAc (Lopez and Ettenberg,
1.01.1.5 Sexual Behavior during Puberty and Aging
2002a) of sexually experienced male rats, compared with sexu-
ally naïve animals. By contrast, in male Japanese quail, sexual 1.01.1.5.1 Puberty
experience decreased the expression of another immediate Puberty is a developmental period during which the hypotha-
early gene, egr-1, in brain areas involved in male sexual lamic–pituitary–gonadal axis is activated and reproductive
6 Male Sexual Behavior

capacity matures (Navarro et al., 2007; Suter et al., 1998). The dimorphic brain regions, such as the SDN of the MPOA and
development of copulatory behavior has been studied in male the MeA (Ahmed et al., 2008). Actually, it has been suggested
rats mainly by testing prepubertal animals with young estrous that structural reorganization at the amygdala might be central
females (Sachs and Meisel, 1979). The data from such studies for the emergence of novel behaviors during adolescence in
established that mounting behavior appears between 40 humans (Scherf et al., 2013). Conversely, in male hamsters
and 50 days of age; intromission, between 44 and 75 days; the number of dendrites and spines in the posterodorsal MeA
and the behavioral pattern of ejaculation, between 48 and (MeApd) decreases during puberty, which coincides with
75 days (reviewed in Meisel and Sachs, 1994). The develop- sexual behavior maturation (Zehr et al., 2006), but in this
ment of copulatory behavior in rats clearly depends on gonadal species pubertally newborn neurons and glia were found to
hormones, since prepubertal castration prevents its appearance integrate into limbic and hypothalamic circuits (Mohr and
(Larsson, 1967), while exogenous T or estrogen (E) hastens its Sisk, 2013).
onset (Södersten et al., 1977). By contrast, in male Syrian
hamsters adult-typical reproductive behavior cannot be acti- 1.01.1.5.2 Aging
vated by gonadal steroids prior to puberty (Sisk et al., 2003). Aging is associated with changes in male sexual function in
In male rats reflexive erections appeared at 40 days of age, humans, monkeys, and rodents. The probability of initiating
penile flips at 44 days, and penile cups at 48 days (Sachs and copulation decreases, and once initiated, the latencies to
Meisel, 1979). mount (ML), intromit (IL), and ejaculate (EL) increase in old
In male rats, T levels begin to rise about day 40, with the so- male rodents and monkeys (Meisel and Sachs, 1994). A rela-
called pubertal T surge occurring around day 50. However, the T tionship between these deficits and lower T levels has not
surge occurs after the onset of mount, intromission, and ejacu- been clearly established. In male rats, the decline in sexual
latory behaviors and the appearance of sexual reflexes (Sachs behavior is accompanied by a decrease in circulating T (Cham-
and Meisel, 1979). Male hamsters begin to copulate after the bers et al., 1991; Smith et al., 1992), but exogenous T only
increase in pubertal T begins, but before the T surge (Romeo partially restores copulatory behaviors (Chambers et al.,
et al., 2002). In this species, the lack of pubertal T impaired T- 1991). Maintenance of T levels by long-term replacement pre-
induced mating in adulthood, produced in hamsters that were vented the decline in intromission frequency (IF) in old rats,
castrated after puberty (Schulz et al., 2004). A series of recent but did not prevent the age-related loss of ejaculatory response
studies in rodents concluded that during puberty sex steroids (Hsu et al., 1986). By contrast, the decrease in sexual activity of
exert a second episode of organizational actions inducing old male rhesus monkeys is not accompanied by a decline in
permanent changes in brain and behavior (Schulz et al., 2009; gonadal hormones or changes in the diurnal pattern of
Sisk and Zehr, 2005). Thus, the effects of pubertal hormones androgen plasma levels, and exogenous T does not increase
on the adolescent brain are important for the maturation of their sexual activity; however, in old long-term castrated
adult social behaviors such as copulation (Schulz and Sisk, monkeys T increased sexual behavior (Phoenix and Chambers,
2006). During adolescence the interpretation of social cues 1986). In men, the decline in T levels during aging has been
must change to enable adult responses. To illustrate this, it correlated with sexual impairment, as in young men with hypo-
has been reported that pheromones contained in female gonadism; however, it is not clear whether T treatment
hamster vaginal secretions (VSs) are rewarding for adult, but improves sexual performance (Moncada, 2006). A direct rela-
not for juvenile male hamsters (Bell et al., 2013a). The tionship between low T levels and decreased libido in aging
rewarding value of VS is T- and DA-dependent; however, T is men has also been difficult to establish (Travison et al.,
not sufficient to activate adult-like mesolimbic Fos expression 2006). However, a role for sexual motivation in male rats’
in response to this female sexual cue, but brain maturation is age-related sexual behavior decline has been inferred from
also required (Bell et al., 2013b). Puberty and adolescence are the fact that treatment with yohimbine, a drug that stimulates
periods of particular vulnerability to stress (Holder and sexual arousal (Viitamaa et al., 2006), improved copulatory
Blaustein, 2014; Schneider, 2013), and perturbations during behavior of old rats and increased mounting in old males
this phase may result in important physiological and with penile anesthesia to levels of untreated young animals
behavioral alterations in adulthood (Romeo, 2005, 2010). (Smith and Davidson, 1990). Interestingly, it was documented
Indeed, chronic consumption of ethanol beginning at puberty that a larger number of hypothalamic cells are recruited in aged
impaired sexual behavior and fertility in adult male rats rats (30–36 months) to elicit a sexual response to chemosen-
(Oliva et al., 2006), and consumption of anabolic androgenic sory stimulation as compared to adult males (12 months),
steroids (AAS), alone or combined, during puberty increased a phenomenon suggested to represent a compensatory mecha-
or decreased sexual and aggressive behaviors in adulthood, nism in the aged animal (Tobiansky et al., 2012).
depending on the drug or drug combination used (Wesson A decline in estrogen receptors (ERs) (Roselli et al., 1993),
and McGinnis, 2006). but not in ARs (Chambers et al., 1991), might underlie the ejac-
Neural changes have been associated with pubertal matura- ulatory deficit of old male rats, since it was found that old intact
tion of sexual behavior. Pubertal maturation of the brain rats had fewer nuclear estrogen receptors (ERn) in the amygdala
includes remodeling of synaptic connections. Accordingly, at than did young animals, with no difference in circulating E
the onset of puberty in male rats, dendritic arborization of levels. Castration reduced ERn in young and old males, but
the sexually dimorphic spinal nucleus of the bulbocavernosus exogenous T restored copulation and ERn in the amygdala
(SNB) increases, followed by a decrease during puberty only in young males; T in old males restored copulation and
(Goldstein and Sengelaub, 1994). Pubertal hormones have ERn only to precastration levels, not to that of young males
been found to modulate the addition of new cells to sexually (Roselli et al., 1993). However, middle-aged (12 months)
Male Sexual Behavior 7

and young (3 months) rats responded to castration and T the absence of a seminal plug in the female’s vagina, in
replacement with similar hypothalamic preoptic upregulation response to the a2-adrenoceptor antagonist yohimbine
of AR and downregulation of ERa densities, suggesting that (Rodríguez-Manzo and Fernández-Guasti, 1995b). Thus, we
middle-aged rat brains retain the capacity to respond to exoge- must be cautious in extrapolating from ex copula tests to copu-
nous T (Wu and Gore, 2010). In men there was an age-related lation or vice versa.
loss of motoneurons in the lumbar spinal cord controlling
penile reflexes (Cruz-Sánchez et al., 1998). In a study in rats,
1.01.2.2 Ex Copula Sexual Reflexes
no age-related decrease in motoneuron number in the spinal
nuclei involved in rats’ sexual reflexes was found, but an age- 1.01.2.2.1 Spontaneous or Drug-Induced Erections
related atrophy of these motoneurons and the muscles they Male rodents sometimes have erections in their home cage or in
innervate was detected (Fargo et al., 2007). Exogenous T a neutral test arena without any obvious sexual stimulus. Such
reversed the decreases in penile muscle weight and motoneu- erections can be increased by certain drugs. These spontaneous
rons’ soma size and dendritic length, suggesting that T might or drug-induced erections usually consist of extension of the
play a role in maintaining neuronal connectivity. engorged glans outside of the penile sheath and are often
Various levels of sexual performance are observed in accompanied by genital grooming. The main advantage of
middle-aged rats (18–19 months), with some able to ejaculate, this model is its simplicity. However, drug-induced erections
others showing only mounts and intromissions, and some can be modified by previous copulation (Sachs et al., 1994).
failing to exhibit any sexual activity. The loss of these specific
components of male sexual behavior has been related with 1.01.2.2.2 Noncontact Erections
an age-related decrease in gonadotropin-releasing hormone Male rodents often have erections in the presence of an inacces-
(GnRH) neurons in hypothalamic and septal regions (Tsai sible receptive female or even the odors of an estrous female
et al., 2014). Incomplete sexual behavior has been associated (Kondo et al., 1999; Sachs, 1997). These erections are similar
with decreased DA and NE, and increased 5-HT, in the NAc to spontaneous or drug-induced erections in rodents and are
(Tsai et al., 2006) and with decreased DA in the MPOA and considered to be a model of psychogenic erection in men.
arcuate nucleus, as well as decreased NE in the MPOA (Chen They are elicited by central, rather than peripheral stimuli.
et al., 2007). Impaired sexual performance, on the other side, However, the primary stimuli to elicit such erections in rodents
was found to be related with decreased DA and NE in the are olfactory, whereas the primary cues for psychogenic erec-
BST (Chen et al., 2008). Aged males showing complete copula- tions in men are visual and auditory; thus, the central pathways
tory behavior had monoamine levels similar to those of young controlling these erections may differ across species. Also, there
sexually active rats. These data suggest that changes in mono- is evidence that different brain areas may regulate noncontact
amine contents in diverse brain regions could play a role in versus in copula erections (Liu et al., 1997b). In addition,
the sexual behavior decline of aged male rats. Recently, taurine noncontact erections may be elicited only in pigmented rats
supplementation to aged rats (20 months) has been found to and not in albinos (Sachs, 1996).
enhance their sexual response by increasing T and nitric oxide
(NO) levels (Yang et al., 2013). 1.01.2.2.3 Reflexive Erections, Anteroflexions, and Seminal
Emissions
Reflexive erections, sometimes referred to as touch-based erec-
1.01.2 Sexual Reflexes tions, can be elicited by manual stimulation of the penis in
numerous species. However, tactile stimulation of the rat penis
1.01.2.1 Observations during Copulation
actually inhibits erection (Hart, 1968). To overcome this diffi-
Although the penis can be observed during copulation in many culty, male rats (Hart, 1968) or mice (Sachs, 1980) can be
species, erections in rodents are usually brief and obscured restrained on their backs and the penile sheath retracted,
from view. Characteristic behaviors are usually used to infer thereby eliciting genital reflexes. Pressure around the base of
the presence of erection, intromission, and ejaculation. The the penis elicits erections of the glans, due to vasodilation in
female’s vagina can be examined for the presence of sperm after the corpus spongiosum, and also anteroflexions (‘flips’), which
a suspected ejaculation, and direct observation of the penis can are elevations of the penis caused by engorgement of the
be made with a slanted mirror under the floor of a clear cage. corpora cavernosa and contraction of the ischiocavernosus
Electrodes may be implanted in the striated perineal muscles muscles. Three gradations of erections have been noted: (1)
to detect electrical activity during copulation (Holmes et al., elongation and rising of the body of the penis; (2) engorge-
1991), and penile pressure has also been measured (Bernabé ment and flaring of the glans; and (3) intense flaring of the
et al., 1999; Giuliano et al., 1994; reviewed in McMurray glans into a cup. The cup deposits the ejaculate around the
et al., 2006). However, these techniques are technically diffi- female’s cervix, where it coagulates to form a copulatory plug.
cult. Therefore, ex copula measures of sexual reflexes have This plug prevents the semen from seeping out of the vagina;
been developed. However, in copula and ex copula erections without it, pregnancy rarely occurs (Sachs, 1982). Ex copula
may differ in their physiological, neurochemical, and seminal emission may also occur. Both reflexive and in copula
hormonal regulation (Sachs, 2000). Furthermore, experimental erections are mediated by parasympathetic vasodilation and
manipulations may affect sexual reflexes without altering the striated penile muscle contractions (Hart, 1968; Holmes
behavioral pattern. For example, male rats with a neurochem- et al., 1991). However, as with noncontact erections, the neural
ical lesion of the NE system repeatedly showed the behavioral mechanisms of erection differ in the two contexts (Sachs,
pattern of ejaculation without seminal ejection, evidenced by 1983).
8 Male Sexual Behavior

1.01.2.2.4 The Urethrogenital Reflex (Gerstenberg et al., 1990). The relative contribution of vascular
A model for both erection and ejaculation has been devel- and muscular factors varies across species, with the rat relying
oped in anesthetized, acutely spinalized male rats (McKenna more on the striated penile muscles, and humans relying
et al., 1991). The urethra is first distended with saline, and more on vascular engorgement (Gerstenberg et al., 1990;
then the pressure is released, resulting in clonic contractions Schmidt and Schmidt, 1993).
of the perineal muscles, rhythmic firing of the cavernous Reflexive erections in the rat are due mostly to steady
nerve, erection, and ejaculation. The simultaneous firing in increases in pressure in the corpora cavernosa (Bernabé et al.,
all the perineal muscles is similar to bursts observed in 1999) and corpus spongiosum (Schmidt et al., 1995). These
human climax (Gerstenberg et al., 1990) and in rats during increases are below systolic pressure, but are interrupted by
ejaculation (Holmes et al., 1991; Miura et al., 2001). 1 s peaks of suprasystolic pressure, elicited by contractions
Synchronized activity in both the pelvic (parasympathetic) of the ischiocavernosus and BS muscles. The bursts occur
and hypogastric (sympathetic) nerves drives the bursts in during glans erections, anteroflexions, and cups. Excision
the cavernous nerve, which are synchronized with somatic (Sachs, 1982) or denervation (Monaghan and Breedlove,
muscle contractions. This reflex is usually evoked only after 1992) of the BS muscle eliminated cups, whereas excision of
spinal transection or lesions of certain brain nuclei, the ischiocavernosus muscle eliminated anteroflexions (Sachs,
evidencing the presence of a tonic descending inhibition 1982). Stimulation of the pelvic and cavernous nerves
(Borgdorff et al., 2008; Carro-Juárez et al., 2003; Gravitt produced plateau increases in penile pressure, whereas
and Marson, 2007; Stafford et al., 2006a,b). (However, see pudendal nerve stimulation elicited suprasystolic increases
Section 1.01.5.2.1.) The urethrogenital (UG) reflex has (Giuliano et al., 1995; Lue et al., 1984). Therefore, erections
been used as a model that provided important information require the coordination of parasympathetically controlled
on the physiological, pharmacological, and neuroanatomical blood flow to the penis and pudendal stimulation of the stri-
control of ejaculation. By using this model the existence of ated perineal muscles.
a spinal pattern generator for ejaculation (SGE) gained
support (Carro-Juárez et al., 2003; McKenna et al., 1991) 1.01.2.3.2 Neural Innervation
(see Section 1.01.2.4). The three major pathways that control penile erection are the
pelvic nerves (primarily parasympathetic and proerectile),
the hypogastric nerves (sympathetic and antierectile), and the
1.01.2.3 Erection
pudendal nerves (somatosensory and motor). The pelvic nerve
1.01.2.3.1 Anatomy of the Penis and Mechanisms of Erection provides the major proerectile innervation (Lue et al., 1995; Tai
An erect penis is needed to deliver sperm into the female’s et al., 1998); it originates in the lumbosacral spinal cord and
reproductive tract. Some mammalian penes are highly vascular, travels via the pelvic plexus (pelvic ganglion) and cavernous
whereas others are more fibroelastic. The vascular penes of nerve to the penile corpora and vasculature. In addition to
humans, monkeys, dogs, cats, and rodents become engorged the parasympathetic fibers, it also carries some sympathetic
due to vascular relaxation, coordinated with striated muscle postganglionic axons (Dail et al., 1986). The SNB and the
contraction. The fibroelastic penes of ungulates, including dorsolateral nuclei of the lumbosacral spinal cord provide
sheep and goats, are extruded by penile muscles and rely very somatomotor control (Schrøder, 1980). Their axons travel in
little on engorgement. There is much variability across species the pudendal nerve, which splits into motor and sensory
in the relative importance of these two factors. branches, both of which also carry sympathetic efferents
The basic structure of the penis is similar across mammalian (McKenna and Nadelhaft, 1986). Stimulation of the ischioca-
species. Most of the penile shaft is occupied by the two corpora vernosus and BS muscles is not sufficient to cause an erection
cavernosa, while the corpus spongiosum surrounds the urethra when the penis is flaccid, but if the penis is erect, stimulation
and enlarges into the glans at the end of the penis. The two dramatically increases its rigidity, and intracavernosal pressure
corpora cavernosa are fused in most species, including humans, increases to suprasystolic levels (Schmidt and Schmidt, 1993).
so that a drug injected anywhere in the structure can diffuse The third major pathway provides mostly antierectile
throughout. Similarly, intracavernous pressure can be moni- sympathetic influence (Diederichs et al., 1991; Giuliano
tored from any site. The corpora cavernosa are large cavernous et al., 1995). Preganglionic axons travel in the lumbar
sinuses (trabeculae) that are supplied by the helicine arteries, splanchnic nerves or the paravertebral sympathetic chain of
which in turn receive blood from the cavernosal artery ganglia (Jänig and McLachlan, 1987). They synapse in the
(reviewed in Anderson, 2011; Hull and Dominguez, 2015). hypogastric plexus or the paravertebral sympathetic chain
The corpora cavernosa are enclosed in a tough capsule (the ganglia, respectively. Postganglionic fibers travel via two major
tunica albuginea), so when they fill with blood, the pressure routes. First, the hypogastric nerve sends input to the pelvic
against the venous outflow traps blood in the penis, which plexus, and then via the cavernous nerve to the penis. Second,
enlarges and becomes rigid. The proximal ends of the corpora paravertebral sympathetic chain axons travel via the pelvic
cavernosa taper into crura (‘legs’) that are attached to the nerve to the pelvic plexus, which then sends output through
ischium (hip bone) and are surrounded by the ischiocaverno- the cavernous nerve to the penis. This route provides most of
sus muscle. The proximal end of the corpus spongiosum the noradrenergic input to the penis (reviewed in Anderson,
enlarges into the urethral bulb and is surrounded by 2011; Giuliano and Rampin, 2000). The penis is maintained
the bulbospongiosus (BS) muscle. Contraction of the in a relaxed, nonerect state by tonic sympathetic input, and
ischiocavernosus muscle increases cavernosal pressure, and injection of noradrenergic antagonists into the corpora caver-
contraction of the BS muscle increases pressure in the glans nosa can elicit erections in men (Brindley, 1986).
Male Sexual Behavior 9

Although the main influence of the sympathetic input is penile response. The authors suggested that the sympathetic
antierectile, a proerectile effect has been observed in anesthe- system may produce vasoconstriction in nonpenile areas,
tized rats (Giuliano et al., 1997; Figure 1). Electrical stimula- thereby diverting blood to the penis and enhancing erection.
tion of the MPOA, a critical brain area for male sexual Therefore, the MPOA can elicit erection by a coordinated acti-
behavior, increased cavernosal pressure. This increase was abol- vation of both the parasympathetic and sympathetic systems.
ished by bilateral section of the pelvic or cavernous nerves,
indicating that the major proerectile effect of MPOA stimula- 1.01.2.3.3 Cellular Mediators of Erection
tion is through the parasympathetic system. However, bilateral Penile erection results from both increases in blood flow into
section of the paravertebral sympathetic chain at the level of L4- the penis, due to relaxation of arterial smooth muscle, and
L5 also significantly decreased the effect of MPOA stimulation, opening sinusoids in the erectile tissue due to relaxation of
as did lesion of sympathetic fibers by the catecholamine neuro- trabecular smooth muscle. Trabecular smooth muscle cells
toxin 6-hydroxydopamine (6-OHDA). Bilateral section of the are linked by gap junctions, which allow current to spread elec-
hypogastric nerves produced a nonsignificant decrease in the trotonically and second messengers to pass from cell to cell.
Therefore, autonomic input can influence the whole network
by innervating relatively few cells (Christ et al., 1999). The
penis is maintained in a flaccid state largely by NE from sympa-
thetic nerves, acting on postsynaptic a1 and, to a lesser extent,
a2 adrenergic receptors (Traish et al., 2000; reviewed in
Andersson, 2011). Drugs that block these receptors can elicit
erection or induce priapism (prolonged erection) (Abber
et al., 1987). Stimulation of a1 and a2 adrenergic receptors acti-
vates phospholipase C and thereby increases intracellular Ca2þ,
which ultimately leads to contraction of arterioles and trabec-
ulae, producing detumescence (Andersson and Stief, 1997;
Christ, 1995; Traish et al., 2000). However, soon after the
initial contraction, Ca2þ levels return to near basal levels, while
the contractile tone remains. The continuing contraction results
from Ca2þ sensitization, mediated by the GTP-binding protein
RhoA and its major effector, Rho-kinase (reviewed in Jin and
Burnett, 2006; Somlyo and Somlyo, 2000; Sopko et al.,
2014). A Rho-kinase antagonist (Y-27632) inhibited
contraction of human or rabbit cavernosal tissue strips that
had been elicited by an a-receptor agonist or electrical
stimulation in vitro (Rees et al., 2001). Y-27632 also
stimulated erection in rats (Dai et al., 2004; Rajasekaran
et al., 2005).
The main mediator of erection is NO, a short-lived gaseous
messenger produced by NOS (reviewed in Burnett, 2006).
Neuronal NOS (nNOS, or NOS I) is present in parasympathetic
nonadrenergic noncholinergic nerves that innervate the erectile
tissue. Endothelial NOS (eNOS, or NOS III) is in the endothe-
lium of erectile tissue. The initial stimulus for erection is NO
produced by nNOS in the parasympathetic nerves. It diffuses
into the smooth muscle cells and activates soluble guanylyl
cyclase, which produces cyclic guanosine monophosphate
(cGMP), which then activates protein kinase G (PKG), and to
a lesser extent protein kinase A (PKA). These enzymes phor-
Figure 1 Diagrammatic representation of peripheral autonomic path- phorylate regulatory proteins, which result in sequestration of
ways potentially involved in erectile response elicited by medial preoptic Ca2þ and the resultant relaxation of smooth muscles and erec-
area (MPOA) stimulation. Sites of neural lesions are represented by tion (reviewed in Burnett, 2006). The activity of cGMP is termi-
hatched bars. No direct projections from MPOA to spinal autonomic nated by phosphodiesterase 5 (PDE5). Several drugs used to
nuclei have been reported. CE, cauda equina; CN, cavernous nerve; HN, treat erectile dysfunction, including sildenafil citrate (Viagra),
hypogastric nerve; L4-L5, fourth and fifth lumbar levels of the para- tadalafil (Cialis), and vardenafil (Levitra), act by inhibiting
vertebral sympathetic chain; L6-S1, sixth lumbar and first sacral level of
PDE5 and thereby prolonging the effects of cGMP.
the spinal cord; MPG, major pelvic ganglion; PN, pelvic nerve; PSC,
Although the initial stimulus for erection is NO from the
paravertebral sympathetic chain; PudN, pudendal nerve; T12-L2, 12th
thoracic to 2nd lumbar level of the spinal cord. Figure is reprinted from parasympathetic nerves, a longer-lasting mediator of erection
Giuliano, F., Bernabé, J., Brown, K., Droupy, S., Benoit, G., Rampin, O., is NO from eNOS in the endothelium of blood vessels and
1997. Erectile response to hypothalamic stimulation in rats: role of sinusoidal spaces. The initial increase in blood flow induces
peripheral nerves. Am. J. Physiol. 273, R1990–R1997, with permission shear stress in those tissues, which activates several enzymes,
from the American Physiological Association. which phosphorylate eNOS at Ser 1177 and sustain the
10 Male Sexual Behavior

production of NO (reviewed in Musicki and Burnett, 2006). in the penis, resulting in greater erectile response to nerve stim-
Phosphorylation at other sites by other enzymes may increase ulation (Escrig et al., 1999). Additional erectile mechanisms
or decrease eNOS activity. However, nNOS can also contribute include activation of melanocortin (MC)4 receptors (Martin
to sustained erection by PKA-induced phosphorylation at Ser et al., 2002). Systemic administration of the MC(4) agonist
1412, which closely resembles the sequence surrounding THIQ increased intracavernous pressure and ex copula erections.
eNOS-Ser 1179 (Hurt et al., 2012). Thus, both neuronal and Intracerebroventricular administration also increased reflexive
endothelial sources of NO contribute to sustained erection. erections. The erections elicited by THIQ were blocked by the
In addition to its direct effects on vasodilation, NO also MC(4) antagonists, agouti-related protein and MPB10, and
inhibits the antierectile RhoA/Rho-kinase pathway (Mills were also attenuated by an OT antagonist (L-368899). Thus,
et al., 2002). Conversely, the RhoA/Rho-kinase pathway may MC(4)-mediated erection may involve OT pathways.
inhibit phosphorylation of eNOS at Ser 1177 and thereby
inhibit erection (Ming et al., 2002). It may also suppress
1.01.2.4 Ejaculation
eNOS gene expression and enzyme activity (Bivalacqua et al.,
2004). Therefore, there is an inverse relationship between the Ejaculation is the physiological process that describes the
proerectile NO/cGMP/PKG pathway and the antierectile expulsion of the semen from the urethra. This process involves
RhoA/Rho-kinase pathway (reviewed in Sopko et al., 2014). several organs including the distal epididymis, the vas deferens,
RhoA activates Rho-associated protein kinase (ROCK), which the seminal vesicles, the prostate, the prostatic urethra, and the
contracts the cavernous sinusoidal and arteriolar smooth bladder neck (Giuliano and Clement, 2005) and consists of
muscle cells, keeping the penile smooth muscle contracted two stages: an emissive phase and an ejective phase (Mitsuya
during the flaccid state of the penis (Kimura et al., 1996). et al., 1976; Newman et al., 1982). The emissive phase involves
The ROCK inhibitor Y-27632, initially studied for antihyper- an autonomic component that elicits synchronized activation
tensive properties, acts independently of NO-mediated of visceral accessory structures such as the prostate and the
relaxation (Chitaley et al., 2001; Wang et al., 2002). tunica albuginea (Exintaris et al., 2006; Shafik et al., 2005),
Inhibition of Rho-kinase not only improves erectile function, while the ejective phase involves rhythmic contraction of peri-
but also decreases penile apoptosis after cavernous nerve neal and pelvic floor striated muscles and the maintenance of
injury in rats (Hannan et al., 2014). Therefore, Rho-kinase/ penile rigidity. During the emissive phase, the bladder neck
ROCK inhibition may provide a new target for treatment of sphincters close, and the seminal vesicles, prostate, vas defer-
erectile dysfunction. ens, and coagulant glands contract, involving parasympathetic
Cavernous nerve damage resulted in increased ROCK and and sympathetic mechanisms that promote deposition of
decreased erectile ability in rats; erectile ability was restored seminal secretions into the prostatic urethra. The expulsive
with Y-27632, which also increased NOS and decreased phase consists of the forceful ejection of semen from the
apoptotic cells (Hannan et al., 2013). Castration also increased urethral meatus, caused by the rhythmic and coordinated
RhoA and Rho-kinase protein levels, but intracavernosal injec- contraction of all genital muscles surrounding the genital tract
tion of Y-27632 increased cavernosal pressure in the castrates, (Carro-Juárez and Rodríguez-Manzo, 2000; Gerstenberg et al.,
as well as in intact and T-replaced rats (Wingard et al., 2003). 1990; Kollberg et al., 1972; Shafik, 1997) that coincides with
Therefore, upregulation of the RhoA/Rho-kinase pathway the rhythmic contraction of muscles from the abdominal
may be one factor in the loss of erectile ability after wall, i.e., cremasteric, iliopsoas, and pubococcygeus muscles
castration. Thus, inhibition of this pathway could provide (Carro-Juárez et al., 2014). Thus, successful ejaculation
a potential treatment for erectile dysfunction. However, the depends on coordinated responses involving autonomic and
decrease in blood pressure that would accompany systemic somatic events. It has recently been proposed that during the
administration could require that these inhibitors be applied emissive phase the sympathetic effect on seminal vesicle’s
locally or that tissue-specific isoforms that target RhoA contraction is dominant in order to squeeze fluid into the
regulatory proteins be developed (Jin and Burnett, 2006). posterior urethra, but that during the ejective phase parasympa-
In addition to NO, there has been recent investigation of the thetic seminal vesicle contractions dominate providing antire-
roles of carbon monoxide (CO) and of hydrogen sulfide (H2S) flux resistance to the pressure generated by BS rhythmic
as gaseous mediators of erection (reviewed in Yetik-Anacak contractions (Hsieh et al., 2014).
et al., 2015). CO is produced by heme oxygenase and, like In copulating animals ejaculation requires the pelvic
NO, activates guanylyl cyclase. H2S is mainly produced by thrusting pattern that results in vaginal penetration and
the smooth muscle in human corpora cavernosa and also seminal deposition, and the genital motor pattern of forceful
targets the guanylyl cyclase pathway. These pathways may seminal ejection (Moralí et al., 2003). Although these patterns
complement and increase NO signaling. These other gaseous can be separately studied (Moralí et al., 2003), only the genital
transmitters may provide additional treatment targets for erec- motor pattern of ejaculation can be observed in experimental
tile dysfunction. models with lesions where penile rigidity, penile movements,
Erection can also be stimulated by activation of adenylyl and ejaculatory expulsions are not disturbed (Carro-Juárez
cyclase by vasoactive intestinal peptide, calcitonin gene- and Rodríguez-Manzo, 2000; Carro-Juárez et al., 2003;
related peptide, and prostaglandin E1 (PGE1) (reviewed in McKenna et al., 1991).
Bivalacqua et al., 2000). The resultant activation of PKA seques- The stimulus that initiates ejaculation is at present
ters intracellular Ca2þ, leading to smooth muscle relaxation unknown; however, it has been proposed that genital sensory
and increased vasodilation. PGE1 also increases production of stimulation that comprises friction of penile skin and intravagi-
NO, and repeated treatments increase both nNOS and eNOS nal pressure, acting against the penis during penetration,
Male Sexual Behavior 11

commences seminal drain into the posterior portion of the (a)


urethra, and when the ejaculatory threshold is reached, due
to the concomitant mechanical stimulation of the urethra by
continuous seminal deposition (de Jong et al., 2006; Sachs
and Barfield, 1976) and penile afferent activation, ejaculation
takes place. The fluids that enter the urethra during emission
elicit single bursts of activity of the BS muscles via the pelvic (b′)
and pudendal nerves, but to elicit rhythmic bursting activity (b)
of perineal muscles during expulsion of seminal fluids, the
concomitant activation of penile afferents (via the pudendal
nerve) is necessary (Tanahashi et al., 2012). The synchronous
contractions of perineal and pelvic floor striated muscles, under
the control of spinal and brain centers are responsible for (c′)
(c)
semen expulsion (Dobberfuhl et al., 2014).
Individual differences in the ejaculatory threshold of rats
were described as part of the biological variability in ejacula-
tory behavior, suggesting the existence of ejaculatory endophe-
notypes in this species, and males have been classified into
(d)
praecox, intermediate, and retarded ejaculators (Pattij et al.,
2005). The physiological control of the ejaculatory threshold
remains one of the biggest riddles in the neurobiology of ejac-
ulatory function. Tonic descending inhibition from the
8-OH-DPAT
nPGi prevents untimely release of the reflex (see Sections
(e)
1.01.2.2.4, 1.01.5.3.3, 1.01.5.3.5, and 1.01.6.2), and supraspi-
nal excitatory influences originate in the MPOA and PVN (see
Sections 1.01.5.2.1 and 1.01.5.2.4).
The neural commands for ejaculation are organized at the Figure 2 Summary figure showing the evidence that the ejaculatory
motor pattern follows the general principles of rhythmic motor patterns
spinal level. It has been demonstrated that a central pattern
produced by a central pattern generator (CPG): (1) a rhythmic muscular
generator, located in the lumbosacral cord, is involved in the
response, the genital motor pattern, is registered during the ejaculatory
control of ejaculation (Figure 2; Carro-Juárez and Rodríguez- event (a); (2) this ejaculatory motor response has similar EMG charac-
Manzo, 2000, 2008; Carro-Juárez et al., 2003). A group of teristics in intact and in spinal urethane-anaesthetized male rats (a and
galanin-containing neurons in this portion of the cord is part b, respectively); (3) deafferentation does not disrupt the expression of
of this ejaculation generator (Truitt and Coolen, 2002), but the ejaculatory motor train (c); (4) a change in the stimulation interval
does not participate in emission in spinally intact male rats does not alter the intrinsic pacing of the ejaculatory-like response (d);
(Staudt et al., 2012). The pattern generator is positioned to and (5) fictive ejaculation can be pharmacologically induced (e). Cali-
relay and integrate all genital sensory and motor signals related bration bar 50 mV, 5 s. Tracing inserts (b0 and c0 ) illustrate two consec-
to ejaculation, and its activation is able to turn on and off all utive ejaculatory motor responses obtained in a 4-min period in a spinal
rat with intact afferents and in a deafferented spinal rat. Figure is adapt-
somatic and autonomic events associated with ejaculation,
ed from Carro-Juárez, M., Cruz, S.L., Rodriguez-Manzo, G., 2003.
including penile erection and movements of the penis driving
Evidence for the involvement of a spinal pattern generator in the control
seminal ejection (Carro-Juárez and Rodríguez-Manzo, 2000, of the genital motor pattern of ejaculation. Brain Res. 975, 222–228,
2003, 2005b, 2006; Carro-Juárez et al., 2003). The SGE can with permission from Elsevier.
modulate genitosensory-induced excitatory and/or inhibitory
mechanisms regulating the facilitation or inhibition of the ejac-
ulatory response at a spinal level (see Carro-Juárez and Rodrí- (Marson and McKenna, 1992), there are data suggesting an
guez-Manzo, 2008, for an extensive review). Thus, repeated intraspinal 5-HT excitatory mechanism, via activation of
activation of ejaculation-related afferent signals modifies the 5-HT1A (Carro-Juárez and Rodríguez-Manzo, 2001; Carro-
activity of the SGE, inducing a short-lasting inhibition that Juárez et al., 2003) and 5-HT2C receptors (Stafford et al.,
could be related to the ejaculation delay phenomenon attained 2006b), in addition to the descending inhibitory influence.
by the ‘pause squeeze method’ as well as long-lasting inhibitory A stimulating role for DA has also been postulated (Peeters
states (Carro-Juárez and Rodríguez-Manzo, 2005b) with char- and Giuliano, 2008; Stafford et al., 2006a), since activation of
acteristics similar to those observed in sexually exhausted DA receptors by apomorphine induces ejaculation-like
animals after repeated ejaculation (Carro-Juárez and Rodrí- responses in anesthetized rats (Stafford and Coote, 2006). Simi-
guez-Manzo, 2000, 2005b). Thus, the SGE can be modulated larly, NO in the lumbar spinal cord participates in the induction
through the activation of spinal sensory feedback mechanisms of a synchronized bursting pattern of sympathetic and somato-
(Carro-Juárez and Rodríguez-Manzo, 2000, 2001, 2005b). motor activity associated with ejaculation (Brack et al., 2007).
Spinal circuits in the lumbosacral spinal cord are modulated The spinal NE system also facilitates ejaculation. Ejaculatory
by supraspinal structures, since the SGE can be activated autonomic and somatic rhythmic patterns, including the
only after lesion of the nPGi or spinal cord transection expulsion of urethral contents and penile erections, are
(Marson and McKenna, 1990). Although the nPGi is thought obtained after increasing NE tone by systemic yohimbine
to exert a 5-HT-mediated tonic inhibition on ejaculation (Carro-Juárez and Rodríguez-Manzo, 2003, 2006). Stimulation
12 Male Sexual Behavior

of a1-adrenoceptors (Carro-Juárez and Rodríguez-Manzo, 2006, 1.01.3.1.1 Time Course of Changes in Copulation Following
2003) also activates the SGE. Moreover, inhibited ejaculation Castration and T Restoration
due to repeated genital mechanical stimulation may be over- Levels of T in plasma decline to unmeasurable levels within
come by blockade of a2-adrenoceptors with yohimbine 24 h after castration (Krey and McGinnis, 1990), but male
(Carro-Juárez and Rodríguez-Manzo, 2003). Cholinergic stimu- rats may continue to copulate for days or weeks, although
lation of the SGE, mediated by the M2, M3, and M4 muscarinic the latency to intromit begins to increase within days. The
receptor subtypes, has also been suggested (Gomez et al., 2005). number of intromissions required to elicit ejaculation actually
Systemic injection of OT in male rats elicits ejaculatory decreases for a few days after castration. Therefore, one function
sequences similar to those obtained by genital-sensory stimula- of T may be to increase the number of intromissions preceding
tion in adult (Carro-Juárez and Rodríguez-Manzo, 2005b) or ejaculation, which would increase the number of sperm in the
neonatal rats (Carro-Juárez and Rodríguez-Manzo, 2005a). ejaculate, facilitate sperm transport, and trigger a progestional
Also gastrin-releasing peptide (Sakamoto, 2011; Takanami state in the female (Toner et al., 1987). There is disagreement
and Sakamoto, 2014) and NMDA receptor-mediated glutama- about the effects of castration on sexual desire and copulation
tergic (Staudt et al., 2011) mechanisms are involved in the excit- in men. Kinsey et al. (1948) used anecdotal reports to conclude
atory modulation of the SGE. Characterization of fast synaptic that castration may not seriously impair sexual function in
responses of the motor neurons controlling BS muscles’ contrac- most men. However, more detailed prospective studies of
tions evidenced the participation of glutamate, GABA, and men who were castrated as ‘treatment’ for sexual offenses found
glycine receptors in their mediation (Best et al., 2013). that half to two-thirds of the men reported a rapid loss of sexual
Endogenous opioids are the only intraspinal inhibitory interest (Heim and Hursch, 1979). The remaining men had
influence on SGE functioning so far described (Carro-Juárez gradual decreases in interest, with 10% still able to copulate
and Rodríguez-Manzo, 2009). Besides its central descending 20 years later. Men who were older at the time of castration
inhibitory role, 5-HT has recently been postulated to modulate were most affected. Imaging studies have reported that visual
the ejaculatory reponse through the activation of peripheral sexual stimuli activated brain areas that control autonomic
receptors. Thus, a facilitative action, mediated by peripheral 5- and neuroendocrine functions and that activation was corre-
HT2 receptors, was recorded after intraurethral application of lated with plasma levels of T (Stoléru et al., 1999). Hypogona-
5-HT (Ishigami et al., 2013), while activation of peripheral 5- dal men showed less activation of some of those areas,
HT1A receptors was reported to inhibit seminal vesicle’s contrac- compared to eugonadal men, and hormone replacement
tion (Hsieh et al., 2011), and selective serotonin reuptake inhib- therapy partially restored the stimulus-activated brain activa-
itors (SSRIs) were found to antagonize sympathetic human tion (Redoute et al., 2005). Testosterone treatment also led to
seminal vesicle muscle contractions in vitro (Birowo et al., a recovery of erectile function in hypogonadal patients, as indi-
2009). Interestingly, chronic treatment with antidepressants cated by nocturnal erections (Foresta et al., 2004).
that modify the ejaculatory response, i.e., desipramine, fluoxe- Five to ten days of exposure to T is necessary to reinstate
tine, and bupropion, also affect the functioning of the SGE copulation in long-term castrated rats (McGinnis et al., 1989;
(Hueletl-Soto et al., 2010, 2012, 2014) (see Section 1.01.6.2). Putnam et al., 2001), and 5–7 weeks are required for hamsters
(Ballard and Wood, 2007). However, in castrated rats T expo-
sure increased firing in the MPOA in response to female odors
1.01.3 Role of Gonadal Steroids in the Control within minutes (Pfaff and Pfaffmann, 1969). Castrated rats
of Male Sexual Behavior started mounting within 35 min after E administration (Cross
and Roselli, 1999), and castrated mice started mounting within
1.01.3.1 Testosterone and Its Metabolites
60 min of T exposure (James and Nyby, 2002). Therefore,
Male sexual behavior depends heavily on T and its metabolites. steroids activate certain brain areas within minutes but require
T is secreted by the Leydig cells of the testes and is carried to its longer-term genomic effects to restore copulation fully.
nontesticular targets in the blood. In all mammals studied, Furthermore, the amount of T affects the degree of recovery.
sexual behavior by adult males is promoted by circulating T Castrated rats that received a high dose of T, which restored
and/or its metabolites, estradiol (E2) and dihydrotestosterone normal levels, had the greatest recovery, compared to those
(DHT). A pubertal increase in T is essential for the increased that received medium and low doses (Harding and Velotta,
sexual activity of maturing males. However, there is interspecies 2011). After 16 days of T exposure, 100% of those that received
variation in the importance of gonadal steroids for sexual the high dose copulated to ejaculation, whereas 80% and 39%
activity. Testicular steroids are essential for mating in most of those receiving the medium and low doses could ejaculate.
rodents, but they play a more modulatory role in humans However, there are individual differences in responsiveness to
(Heim and Hursch, 1979). androgen treatment (Larsson, 1966).
The stimulatory effects of T in adulthood are referred to as
activational effects, as opposed to the organizational effects 1.01.3.1.2 Role of T Metabolites in Maintaining and Restoring
of T during sex differentiation. In adult males T has primarily Copulation
slow, genomically mediated permissive effects that prepare T is the principal hormone produced by the testes and present in
the male to respond to a receptive female. T is usually present systemic circulation. However, T is primarily a prohormone,
in higher concentrations than necessary to activate sexual being converted in target organs either to E2 by aromatase or
behavior, and small fluctuations in levels usually do not affect to DHT by 5a-reductase. There are at least two E receptors,
behavior (reviewed in Ågmo and Ellingsen, 2003). The higher ERa and ERb. Although both T and DHT bind to the
levels are necessary for sperm production in the testes. AR, DHT binds with approximately fivefold greater affinity
Male Sexual Behavior 13

(Wilbert et al., 1983). DHT cannot be aromatized to E2, and there- (Wersinger et al., 1997). Castrated ERaKO males with normal
fore is considered to have only androgenic action. Some target levels of replacement T (Wersinger et al., 1997) or higher
cells may produce both E2 and DHT and have both ERs and ARs. than normal levels of DHT (Ogawa et al., 1998) showed
The relative importance of estrogenic and androgenic stim- increased mounting, but few or no ejaculations. However,
ulation for male sexual behavior is complex and species- treatment with T and a DA agonist restored the ability to ejac-
specific. In castrated male rats E2 is sufficient to reinstate ulate (Wersinger and Rissman, 2000). Aromatase knockout
most aspects of copulation (Cooke et al., 2003). Similarly, (ArKO) mice also showed mating deficits (Bakker et al.,
synthetic androgens that can be aromatized to E2, but not 5a 2002a; Matsumoto et al., 2003). In contrast, bERKO males
reduced to DHT, restored copulation in castrated rats (Moralí mated normally (Ogawa et al., 1999), although the pubertal
et al., 1993) or mice (Ogawa et al., 1996). In contrast, copula- onset of ejaculation was delayed (Temple et al., 2003). There-
tion was not restored by either DHT or the nonaromatizable fore, ERa and aromatase, perhaps together with androgens,
androgen methyltrienolone (R1881) (reviewed in Hull et al., are important for male mouse sexual behavior.
2006). The ability of T to restore copulation in castrated rats However, aromatization of T to E is not required in
was inhibited by aromatase inhibitors (Bonsall et al., 1992; numerous other species, including guinea pigs, hamsters, deer
Roselli et al., 2003; Vagell and McGinnis, 1997) and ER antag- mice, monkeys, and mice (reviewed in Hull et al., 2006).
onists (Beyer et al., 1976). These results support the ‘aromatiza- Although T acting through ARs is most potent, E does contribute
tion hypothesis,’ which states that aromatization of T to E is to the ability of male copulation to ejaculation in macaques,
critical for maintaining or restoring copulation in male rats. though androgens are important for sexual motivation (Barrett
Furthermore, E is also important for sexual behavior in et al., 2006). Also, aromatase inhibitors impaired copulation
macaques. Plasma concentrations of E2 and of the 5-HT in castrated T-treated monkeys (Zumpe et al., 1996). In
precursor tryptophan, but not of T or DHT, were correlated addition, plasma concentrations of E2 or the 5-HT precursor
with male macaques’ ability to copulate to ejaculation, tryptophan, but not of T or DHT, were correlated with male
although androgen concentrations were more important for macaques’ ability to copulate to ejaculation, although sexual
sexual motivation (Barrett et al., 2006). The ER antagonist motivation was more closely related to androgen
(RU 58668) inhibited scent marking and 50 kHz vocalizations concentrations (Barrett et al., 2006). In castrated hamsters the
in male rats, but did not diminish T’s ability to restore copula- aromatizable androgens androstenedione (A) and T restored
tion, although an AR antagonist (hydroxyflutamide) did all sexual behaviors; E promoted anogenital investigation and
inhibit restoration of copulation as well as scent marking and some mounting, but not ejaculation; and DHT was ineffective
vocalizations (Vagell and McGinnis, 1998). Therefore, ARs (Arteaga-Silva et al., 2005). In that study the combination of E
appear to play a role, at least in some experimental conditions, plus DHT was less effective than A or T, suggesting that
in restoring copulation in male rats. In support of this hypoth- aromatization, perhaps local, may be a factor. Although the
esis, E is usually insufficient to fully maintain or restore copu- relative importance of E and A varies across species, males
lation after castration (Kaplan and McGinnis, 1989; Putnam normally have both classes of hormone, which together
et al., 2003, 2005), and antiandrogens also reduce T’s ability promote appetitive and consummatory aspects of mating.
to restore copulation (Vagell and McGinnis, 1998). In another
experiment, male rats were implanted with either E or choles- 1.01.3.1.3 Effects of Castration and Hormone Replacement
terol 35 days after castration and then tested for sexual incen- on Ex Copula Penile Responses
tive motivation every fifth day for 3 weeks (Attila et al., 1.01.3.1.3.1 Animal Studies
2010). E implants failed to restore incentive motivation. In Ex copula reflexes are more sensitive to the presence or absence
other animals, T was administered alone or together with the of gonadal steroids than is copulation. Reflexes are lost more
aromatase inhibitor fadrozole. T did restore behaviors, but rapidly after castration and restored more rapidly by T replace-
fadrozole significantly inhibited those effects. In a third group, ment. Reflexive erections were diminished 24 h after castration
a smaller amount of E was administered with DHT, or E and in spinally transected animals (Hart et al., 1983). In spinally
DHT were administered alone. Only the combination restored intact males the number of cups was decreased by day 4 after
sexual incentive motivation. Therefore, sexual incentive moti- castration, the earliest time tested, anteroflexions were
vation requires simultaneous stimulation of both ARs and decreased by 7 days, and glans erections were decreased by
ERs (see Hull et al., 2006 for a more extensive review). 11 days (Meisel et al., 1984). Reflexive erections were restored
Sexually inexperienced gonad-inact male hamsters showed within 6 h of T replacement in spinally transected males,
a preference for a receptive female versus a stimulus male, which with maximal increases by 24 h (Hart et al., 1983). In spinally
was abolished by castration (Ballard and Wood, 2007). Other intact males, increases were observed 24 h after T replacement,
castrates received a T implant and were tested for 6 weeks. with maximal increases at 48 h (Gray et al., 1980). The longer
Both the loss of sexual preference in castrates and its restoration intervals required in spinally intact males may be due to the
by a T implant occurred after changes in mating behavior. time required to reduce supraspinal inhibition, so that the
Studies of genetically altered mice that lack ERa or ERb have already primed spinal effectors can function. Noncontact erec-
provided new insights into the roles of E in male sexual activity. tions are also lost more rapidly after castration (3 days) and
Gonadally intact males that lacked the ERa (ERa ‘knockout’ restored more rapidly after T replacement (3 days) (Manzo
(ERaKO) mice) mounted, but had fewer intromissions than et al., 1999). Lower levels of T are effective in restoration
wild-type males, and very few ejaculations (Ogawa et al., than are present in gonadally intact males (Davidson et al.,
1997, 1998). These males actually had higher levels of T than 1978), so, as with copulation, plasma T in adult males is higher
wild-type mice, because of decreased negative feedback by E than required for penile reflexes.
14 Male Sexual Behavior

Although copulation in male rats is more dependent on E, protein synthesis inhibitor anisomycin did not interfere with
reflexes rely more heavily on androgenic stimulation. DHT is the ability of T to activate reflexive erections 24 h after T
both necessary and sufficient for maintaining and restoring injection, suggesting that protein synthesis was not necessary
reflexive (Gray et al., 1980; Meisel et al., 1984) or NO-mediated for T’s effect (Sachs and Leipheimer, 1988).
(Lugg et al., 1995, see below) erections in male rats. The DHT Several studies have emphasized the role of local aromatiza-
regimens that maintained or restored ex copula reflexes were not tion of T to E and suggested that E should be considered to be
effective in activating mounting (Gray et al., 1980; Meisel et al., a neuromodulator or neurotransmitter (Balthazart and Ball,
1984). Noncontact erections were also maintained by DHT, 2006; Balthazart et al., 2006; Cornil et al., 2006a; Voigt et al.,
but not by E (Cooke et al., 2003; Manzo et al., 1999). However, 2007). Aromatase is present in presynaptic boutons of several
erections during copulation can be maintained by E. E-treated species, including birds, rats, monkeys, and humans (Hojo
castrates achieved vaginal insertion on as high a percentage of et al., 2004; Naftolin et al., 1996). In mice, systemic injection
intromissions as did control males (O’Hanlon et al., 1981). E of several aromatase inhibitors inhibited copulation in tests
was also as effective as T in maintaining the duration, starting 10 min after the injection; this inhibition was blocked
frequency, and amplitude of EMG bursts in the BS muscles by coadministration of a large dose of E2 (Taziaux et al., 2007).
during intromissions (Holmes and Sachs, 1992). Sachs (1983) The inhibition was not seen in ArKO mice whose behavior was
proposed that E can activate a ‘behavioral cascade,’ organized maintained by daily injections of E2; furthermore, ArKO mice
in the brain that can activate reflexes during copulation, but responded within 15 min to a single dose of E2. Similar inhib-
cannot disinhibit those reflexes ex copula. itory effects of aromatase inhibitors were observed in male
quail (Cornil et al., 2006b). Therefore, locally produced E
1.01.3.1.3.2 Studies on Human Males may facilitate mating. However, questions remain concerning
Levels of T in men with erectile dysfunction are not significantly the behavioral significance of local regulation of aromatase
lower than those in normally functioning men (Becker et al., (see Section 1.01.5.2.1).
2001; Rhoden et al., 2002). However, in aging men with
moderate decreases in T levels, exogenous T did improve erec-
1.01.3.3 The Role of Progesterone in Male Sexual Behavior
tile function and nocturnal penile tumescence (Cavallini et al.,
2004; Schultheiss et al., 2000). Furthermore, T treatment of Progesterone (P), secreted from the testes and adrenals, is
hypogonadal men increased the number of erections in diary a precursor to T. However, there are apparently contradictory
reports (O’Carroll et al., 1985; Salmimies et al., 1982). Further- reports as to whether P facilitates or inhibits male sexual
more, exogenous T in hypogonadal men (which resulted in behavior. P partially restored intromission, but not ejaculation,
normal levels) or in eugonadal men (which produced supra- in castrated rats (Witt et al., 1994, 1995). The P receptor antag-
physiological levels) did increase sexual arousal in response onist RU486 decreased the number of erections elicited by para-
to sexual audiotapes (Alexander et al., 1997). T and DHT doxical sleep deprivation (Andersen and Tufik, 2005). Sexually
were equally effective in stimulating sexual activity in agonadal naïve P receptor knockout (PRKO) mice had fewer mounts than
men, and administration of either an ER antagonist or an aro- did wild-type males, but PRKO mice mated normally on subse-
matase inhibitor did not inhibit sexual function. Thus, andro- quent tests (Phelps et al., 1998). Sexually experienced PRKO
genic stimulation in men appears to facilitate sexual interest males showed more dramatic declines in copulation after castra-
and ability (reviewed in Traish and Guay, 2006). tion than did wild-type males. In contrast to these findings,
which support a facilitative role of P, a later study of PRKO
mice found that they had shorter mount latencies and increased
1.01.3.2 Steroid Action on Steriod Hormone Receptors
likelihood of achieving ejaculation, compared to wild-type mice
ARs and ERs, distributed widely but selectively throughout the (Schneider et al., 2005). In the same report, the P receptor antag-
brain, are thought to mediate most hormonal effects on sexual onist RU486 also decreased intromission latencies and
behavior. These receptors are concentrated in areas that are increased mounts and intromissions. The PRKO mice in that
important for male sexual behavior, including the MPOA study had more ARs in the medial preoptic nucleus (MPN)
(see Simerly, 1995). When bound to the appropriate ligand, and BST, which may have contributed to their better perfor-
classical steroid receptors act as transcription factors to increase mance. The reason for the contradictory results obtained with
synthesis of new proteins. Such actions are consistent with the PRKO mice is not clear. However, in agreement with an inhib-
lengthy time course by which steroids promote male sexual itory role of P, chronic administration of synthetic progestins
behavior (McGinnis and Kahn, 1997; Yahr and Ulibarri, 1987). (medroxyprogesterone acetate and cyproterone acetate) in
However, hormones may also have more rapid effects, men has been used to inhibit deviant sexual behavior (reviewed
mediated by membrane receptors that affect ion channel in Andersen and Tufik, 2006). These compounds block ARs,
activity or second-messenger systems (Kelly et al., 2002; Mer- which may, at least partially, account for their inhibitory effects.
melstein et al., 1996; Shakil et al., 2002; Xiao and Becker,
1998; reviewed in Frye, 2001; in Moore and Evans, 1999; in
Toran-Allerand, 2004). Either E or T in the perfusion medium 1.01.4 Effects of Systemically
rapidly affected firing rates of neurons in MPOA slices; E- and T- and Intraventricularly Injected Drugs
responsive cells were differentially distributed (Silva and
1.01.4.1 Dopamine
Boulant, 1986). In tests of reflexive erections, BS muscle
activity was increased 5 min after T injection, although no The mesolimbic, nigrostriatal, and hypothalamic dopami-
erections were produced at that time; furthermore, the nergic systems are all involved in the integration of male rodent
Male Sexual Behavior 15

sexual response (Hull et al., 1999). DA has long been known to receptor antagonist L-745870 impaired copulatory behavior
facilitate male sexual function. L-DOPA, the precursor of DA, (Sanna et al., 2015). Polymorphism of the dopamine trans-
administered to Parkinsonian patients, increases libido and porter gene (DAT1) was associated with differences in human
sexual potency (Barbeau, 1969; Bowers et al., 1971), and ejaculatory threshold that might be related to premature ejacu-
some DA replacement therapies produce hypersexuality in lation (Santtila et al., 2010).
these patients (Politis et al., 2013). Besides, the nonspecific Male rats can be conditioned to have a same-sex partner
DA agonist apomorphine has been used to treat sexual dysfunc- preference if they undergo cohabitation with other males under
tion (Dula et al., 2000; Giuliano and Allard, 2002; Lal et al., the effects of D2 receptor activation with quinpirole (Triana-Del
1987). Rio et al., 2011), while D1-like receptors mediate ejaculation-
In rats, systemically administered DA agonists facilitate induced conditioned place preference (Dominguez-Salazar
male sexual behavior (reviewed in Bitran and Hull, 1987; et al., 2014), further evidencing the role of DA in the rewarding
Giuliano and Allard, 2001; Hull et al., 2006; Melis and effects of copulation. Sexual arousal has also been shown to be
Argiolas, 1995), induce sexually sluggish males to copulate conditionable in men (Brom et al., 2014), and there is evidence
(Tagliamonte et al., 1974), elicit copulation in sexually for DA mediating the processing of subconsciously perceived
exhausted males (Mas et al., 1995b; Rodríguez-Manzo, sexual stimuli (Oei et al., 2012).
1999b; Guadarrama-Bazante et al., 2014), and partially restore In anesthetized rats, icv and intra-MPOA injection of a D3
copulation in castrates (Malmnäs, 1976; Scaletta and Hull, agonist induced BS rhythmic contractions and ejaculation,
1990). In mice lacking the ERa, which usually show little sexual effects blocked by a D2/D3 (raclopride) and a preferential D3
behavior, apomorphine induces normal copulation (Wersinger antagonist (nafadotride) (Clement et al., 2007a; Kitrey et al.,
and Rissman, 2000). It also elicits penile erections and genital 2007). Thus, supraspinal command of ejaculation appears to
grooming in mice (Rampin et al., 2003). be mediated by D2-like receptors, probably of the D3 subtype,
Systemically administered DA antagonists impair sexual in the MPOA. D1-like receptor agonists increased sexual moti-
behavior in both sexually experienced (Ahlenius and Larsson, vation in rats (Beck et al., 2002) and facilitated copulation in
1990; Pfaus and Phillips, 1989) and naïve rats (Ågmo and DA-deficient mice (Szczypka et al., 1998). Besides, D1-like
Picker, 1990). The negative effects of DA antagonists range receptors mediate the apomorphine-induced reversal of
from increased IL and EL to failure to copulate (reviewed in sexual satiation (Guadarrama-Bazante et al., 2014) and play
Bitran and Hull, 1987; Giuliano and Allard, 2001; Melis and a role in the experience-induced enhancement of male rat
Argiolas, 1995). Haloperidol, a nonspecific DA antagonist, sexual behavior (McHenry et al., 2012). D1-like receptor
inhibits measures of sexual motivation as well as copulation facilitation of male sexual behavior appears to be conserved
itself both after acute (Lopez and Ettenberg, 2000, 2001, across phyla, since it has been detected in lizards (Woolley
2002b) and chronic treatment (Zhang et al., 2011). In human et al., 2001), geckos (Woolley et al., 2004), Japanese quail
males, DA antagonists reduce sexual desire, arousal, and (Balthazart et al., 1997), and European starlings (Schroeder
orgasm (Baldwin and Mayers, 2003). and Riters, 2006).
Differences in male sexual motivation and copulatory The effects of DA agonists are dose-dependent, with low
patterns of two outbred rat lines (Roman high- and low- doses facilitating and high doses inhibiting copulation, the
avoidance rats) have been related to differences in the central latter effect possibly due to the induction of stereotypic
dopaminergic tone, evidenced by a differential sensitivity to behavior (Foreman and Hall, 1987; Szczypka et al., 1998).
apomorphine and haloperidol between strains (Sanna et al., Contradictory effects of D1- and D2-like receptor agonists on
2014). In line with these data, both apomorphine and the ex copula genital reflexes have been reported. On one hand,
D1-like receptor agonist, SKF38393, facilitated sexual behavior a D2 selective agonist was reported to elicit erections, while
display in male rats with a diminished motivational tone due a D1 receptor agonist inhibited them (Zarrindast et al.,
to sexual exhaustion, but not in sexually experienced animals 1992). However, the D2/D3 receptor agonist quinelorane
with an intact sexual motivation (Guadarrama-Bazante et al., decreased the number of reflexive erections in restrained rats
2014). (Bitran et al., 1989b), but promoted penile erection in rhesus
DA receptors are classified into two families; the D1-like monkeys in the presence of sexually receptive females, which
family includes receptors positively coupled to adenylyl cyclase they could see, hear, and smell, but could not contact (Pomer-
and comprises the D1 and D5 receptor subtypes, while the D2- antz, 1991). Other D2/D3 selective receptor agonists elicited
like family consists of D2, D3, and D4 receptor subtypes, which erections in rats in a neutral arena (Ferrari et al., 2002).
are negatively coupled to adenylyl cyclase (Missale et al., 1998). However, a D2/D3 antagonist (eticlopride) actually increased
At the supraspinal level DA has a facilitative effect on ejacula- erections elicited by a presumed-selective D2 agonist or cocaine
tion, mediated especially in the MPOA and PVN by D2-like in rats (Ferrari and Giuliani, 1996b). Cocaine would have
receptors (reviewed in Hull et al., 2004). Systemic activation increased extracellular DA, which would have stimulated
of D2-like receptors with either D3 (7-OH-DPAT) or D2/D3 both D1 and D2 receptors. It is possible that the presumed-
receptor agonists (SND 919 and BHT 920) facilitated ejacula- selective D2 agonist actually stimulated some D1 receptors, as
tion by decreasing both IF and EL in sexually experienced did cocaine, and that inhibition of D2 receptors disinhibited
rats, but not in sexually inactive ones (Ferrari and Giuliani, the D1-like effect. With the use of dopaminergic agonists and
1995, 1996a; Giuliani and Ferrari, 1996). Selective D4 receptor antagonists it was established that the proerectile effects
agonists (PD-168,077 and ABT-724) improved copulatory produced by D2-like receptor agonists, administered either
behavior of sexually potent rats by decreasing MF, EL, and systemically or intra-PVN, are mediated by the D2 receptor
PEI and increasing ejaculation frequency (EF), while the D4 subtype, with D4 receptors playing only a modest role
16 Male Sexual Behavior

(Depoortere et al., 2009; Sanna et al., 2011). DA is released in b1/b2-blockers propanolol and pindolol profoundly inhibited
the MPOA (Hull et al., 1995; reviewed in Hull, 2011) and the sexual behavior, almost abolishing ejaculation (Smith et al.,
NAc (Fiorino et al., 1997) of male rats in response to an estrous 1990, 1996). In sexually vigorous male rats, the b2-adrenocep-
female and during copulation. The DA release patterns during tor agonist clenbuterol reduced mount frequency (MF) and IF
ejaculation in these two brain regions are virtually identical and increased the PEI, but in sexually sluggish animals it
(Pfaus, 2009). improved copulatory behavior by increasing the percentage
of males achieving ejaculation and reducing ML, IL, EL, and
PEI (Benelli et al., 1990). Chronic oral administration of prop-
1.01.4.2 Norepinephrine
anolol to male rabbits impaired sexual behavior, affecting
NE has both facilitative and inhibitory effects on male sexual performance more than arousal aspects (Grotthus et al.,
behavior. Inhibition of NE synthesis impaired copulation, 2007). Besides, an increase in NE transmission produced by
increasing ML, IL, EL, and PEI (McIntosh and Barfield, acute or chronic (14 days) treatment with desipramine, an anti-
1984c). However, lesions of the NE system have rendered depressant that inhibits the NE transporter, reduced the ejacu-
inconsistent results. Electrolytic lesion of the locus coeruleus latory threshold (IF and EL) of sexually vigorous male rats
(LC), which contains the majority of NE cell bodies in the brain (Hueletl-Soto et al., 2010), thus supporting a facilitative role
(Kuhar et al., 1999), inhibited copulation (McIntosh and of NE on copulation.
Barfield, 1984c), but similar lesions of the dorsal NE bundle, The NE system regulates male sexual functions through
which connects the LC with the cerebral cortex and hippo- ascending pathways to the brain and descending pathways to
campus (Kuhar et al., 1999), decreased the PEI and increased the spinal cord (Giuliano and Rampin, 2000), and adrenocep-
the ejaculations in a 1-h test (Clark, 1975, 1980). Central NE tors are found in the brain and spinal cord of animals and
depletion by the neurotoxin DSP-4 increased the PEI (Hansen humans (Roudet et al., 1994; Smith et al., 1995; Wada et al.,
et al., 1982), but in another study it had no effect on copulation 1996). The activity of spinal preganglionic neurons is modu-
(Fernández-Guasti and Rodríguez-Manzo, 1997), neither did lated by NE, and postganglionic sympathetic nerve terminals
the lesion of the dorsal NE bundle with the neurotoxin 6- release NE in the penis, promoting penile detumescence
OHDA (Clark, 1980). DSP-4-induced lesion did not interfere (Giuliano and Rampin, 2004). In contrast to the effects on
with the Coolidge effect, although it reduced the number of copulation, both agonists and antagonists at a2-adrenoceptors
ejaculations prior to satiation and in the ensuing Coolidge inhibit reflexive erections as well as seminal emission in rats.
effect copulatory period (Rodríguez-Manzo, 1999a). However, the effects of the a2-receptor antagonist yohimbine
NE exerts its effects by stimulating a- and b-adrenoceptors. appear to be dose-dependent, with low doses facilitating and
a-adrenoceptors have been shown to modulate sexual arousal higher doses inhibiting the erectile response (reviewed in
(Viitamaa et al., 2006), since a2-adrenoceptor antagonists Meisel and Sachs, 1994). A similar effect is seen on seminal
such as yohimbine, idazoxan, and imiloxan, which enhance emission in dogs (Yonezawa et al., 1991). Activation of a1-
NE transmission by blocking autoreceptors, increased adrenoceptors inhibits reflexive erection in dogs, but stimulates
mounting rates in rats with genital anaesthetization, seminal emission in rats. At the spinal level, it has been shown
augmented copulatory behavior in sexually sluggish and cas- that enhancement of noradrenergic transmission by blockade
trated males (Smith et al., 1987; Tallentire et al., 1996), of a2 autoreceptors with yohimbine and direct a1-adrenoceptor
increased the percentage of sexually naïve rats that copulated stimulation with methoxamine both facilitate male rat ejacula-
to ejaculation (Benelli et al., 1993; Tallentire et al., 1996), tory response (Carro-Juárez and Rodríguez-Manzo, 2006).
reversed the sexual inhibition that follows male rat sexual There is also evidence that the b-adrenergic antagonist
exhaustion in intact rats (Rodriguez-Manzo, 1999b) as well propranolol has a negative impact on rat reflexive erectile and
as exhaustion of the ejaculatory reflex in spinal cord- ejaculatory reflexes (Smith et al., 1995). Together, these data
transected males (Carro-Juárez and Rodríguez-Manzo, 2003). suggest that increased NE activity, either by blockade of a2
Yohimbine produced biphasic effects after its systemic and autoreceptors or by stimulation of a1-adrenoceptors, increases
icv injection; it dose-dependently decreased IL, EL, and PEI in sexual arousal. b-adrenoceptors facilitate copulation, but NE
sexually experienced males (Clark et al., 1984; Sala et al., inhibits reflexive erections, probably at the penis.
1990) and stimulated penile erection through autonomic acti-
vation (Allard and Giuliano, 2001). Conversely, a2-adrenocep-
1.01.4.3 Serotonin
tor agonists (clonidine, guanabenz) dose-dependently
suppressed ejaculation and erection when systemically (Clark 5-HT is generally inhibitory to male sexual behavior (reviewed
and Smith, 1990) and icv administered (Clark, 1991), or in Bitran and Hull, 1987; Hull et al., 2004). A common side
increased ML, IL, and PEI in sexually vigorous male rats and effect of SSRI antidepressants, which increase extracellular
decreased the number of inexperienced males achieving ejacu- 5-HT, is to impair male sexual function in humans and rats.
lation (Benelli et al., 1993). Blockade of a1-adrenoceptors with SSRI’s sexual side effects seem to be best explained by inhibi-
prazosin increased IL, EL, and PEI, while the a1-adrenoceptor tory actions of 5-HT on dopaminergic activity (Bijlsma et al.,
agonist methoxamine had the opposite effect (Clark et al., 2014).
1985; reviewed in Meisel and Sachs, 1994). Contrary to the general inhibitory role of 5-HT in copula-
The influence of b-adrenoceptors on copulation is not clear. tion, stimulation of the 5-HT1A receptor subtype markedly
b-Adrenoceptor antagonists, used for the treatment of cardio- facilitates male rat ejaculation (Ahlenius et al., 1981). Fourteen
vascular diseases (Borchard, 1998), cause sexual dysfunction 5-HT receptor subtypes, belonging to one of seven receptor
in men (Düsing, 2005). In rats, sc and icv injection of the families, have been identified (Hoyer et al., 2002); however,
Male Sexual Behavior 17

only a few of them have been related to male sexual function. satiated rats (Rodríguez-Manzo and Fernández-Guasti 1994;
The inhibitory actions of 5-HT appear to be mediated by 5- Fernández-Guasti and Rodriguez-Manzo, 1997). Other 5-
HT1B receptors in rats and mice (Ahlenius and Larsson, 1998; HT1A agonists such as flesinoxan (Haensel and Slob, 1997),
Hillegaart and Ahlenius, 1998; Rodríguez-Manzo et al., buspirone (Mathes et al., 1990), or ipsapirone (Fernández-
2002b) and by the 5-HT2 receptor in rats (Foreman et al., Guasti et al., 1989) induced a behavioral profile similar to
1989; Klint and Larsson, 1995), while the 5-HT1A receptor that of 8-OH-DPAT, although animals did not ejaculate on
mediates the facilitative actions of this neurotransmitter in their first intromission. That 5-HT1A receptor stimulation yields
rats (Ahlenius et al., 1981). Thus, systemic and intrabrain injec- opposite effects to those produced by 5-HT itself, together with
tion of 5-HT1B agonists inhibit male rat sexual behavior the fact that somatodendritic autoreceptors in 5-HT neurons
(Fernández-Guasti et al., 1989, 1992), and coadministration belong to the 5-HT1A receptor subtype, led to the postulation
of subeffective doses of 5-HT1B agonists and the 5-HT precursor that 8-OH-DPAT acted as an agonist at somatodendritic recep-
5-hydroxytrytophan (5-HTP) synergize to inhibit copulation tors to reduce 5-HT release (Ahlenius et al., 1981). However,
(Fernández-Guasti and Rodríguez-Manzo, 1992). 5-HT1B lesion of raphe nuclei did not alter the 8-OH-DPAT facilitative
receptor antagonists completely block the inhibitory actions actions on copulation, suggesting that they were exerted at
of 5-HTP on copulation, while antagonists to other subtypes postsynaptic sites (Fernández-Guasti and Escalante, 1991). In
do not (Ahlenius and Larsson, 1998). In 5-HT1B receptor addition, facilitative effects of 8-OH-DPAT were obtained after
knockout (KO1B) mice, males become interested earlier in its infusion into the MPOA and the NAc, but not into the dorsal
sexual behavior and are less sensitive to the inhibitory actions raphe, where somatodendritic receptors are found (Fernández-
of 5-HTP than the corresponding wild-type strain. However, Guasti et al., 1992; Matuszewich et al., 1999). The anterior LH
KO1B mice require more stimulation to achieve ejaculation (Riolo et al., 1999) and the MeApd (de Castilhos et al., 2006)
than wild-type males. Pharmacological manipulation in these are two additional brain areas where injection of 8-OH-DPAT
animals revealed that in mice, in contrast to rats, both the facilitates ejaculation.
5-HT1B and the 5-HT1A receptor subtypes contribute to the 8-OH-DPAT’s facilitative effects on copulation require
inhibitory actions of 5-HT (Rodríguez-Manzo et al., 2002b). normal T levels and sexual experience (Rowland and
The inhibitory role ascribed to the 5-HT2 receptor subtype in Houtsmuller, 1998). Both NE and DA systems are involved
male rat sexual behavior is mainly derived from results in 8-OH-DPAT’s facilitative effects, since neurotoxic lesions of
showing that systemic, intraraphe, and intralateral hypothal- the central NE system blocked or attenuated them in sexually
amus (LH) administration of 5-HT2 receptor agonists dose- exhausted and nonexhausted rats, respectively (Fernández-
dependently decreased its expression (Foreman et al., 1989; Guasti and Rodríguez-Manzo, 1997; Rodríguez-Manzo and
Riolo et al., 1999; Watson and Gorzalka, 1991, 1992), while Fernández-Guasti, 1995b). Besides, the D2 antagonist raclopr-
a 5-HT2 antagonist facilitated copulation (Gonzalez et al., ide, but not the 5-HT1A antagonist p-MPPI, decreased 8-OH-
1994). Stimulation of 5-HT2C receptors increased erections DPAT’s facilitative effects in the MPOA (Matuszewich et al.,
and inhibited ejaculation in monkeys (Pomerantz et al., 1999). Intrathecal administration of 8-OH-DPAT at the level
1993) and facilitated erection in anesthetized (Steers and of the lumbosacral spinal cord also reduced the IF and EL
DeGroat, 1989) and conscious rats (Millan et al., 1997). Facil- (Lee et al., 1990).
itative effects of 5-HT2C receptor activation on ejaculation have Chronic SSRI treatment is frequently associated with
also been reported in rats (Yonezawa et al., 2008), and activa- delayed ejaculation. In the case of fluoxetine, this effect seems
tion of spinal 5-HT2C receptors elicited burst pattern responses to be exerted directly at the spinal level (Hueletl-Soto et al.,
and evoked emission associated with ejaculatory-like responses 2012). Acutely, SSRIs inhibit 5-HT reuptake, elevating extracel-
(Stafford et al., 2006b). lular 5-HT levels (Malagie et al., 2000), and usually do not
Brain sites where microinjection of 5-HT has been reported to affect male sexual behavior (de Jong et al., 2005a; Hueletl-
impair male rat sexual behavior are the MPOA (Dominguez and Soto et al., 2012), but when infused into the anterior LH,
Hull, 2010) and the NAc (Fernández-Guasti et al., 1992). they inhibit copulation (Lorrain et al., 1997). Interestingly,
However, accumulation of 5-HT in the MPOA induced by reverse dialysis of 5-HT into this brain region decreases DA
SSRI local injection did not affect sexual behavior, although release in the NAc (Lorrain et al., 1999). After chronic admin-
SSRI injection into the anterior LH did inhibit copulation istration, SSRIs cause desensitization of 5-HT1A receptors (Le
(Lorrain et al., 1997; see Section 1.01.5.2.5). Besides, a tonic Poul et al., 1995).
inhibitory influence on sexual reflexes exerted by brain stem Apparently, this desensitization plays a role, not only in
5-HT descending pathways to the spinal cord has been described SSRIs’ antidepressant properties (Artigas et al., 1996), but
(Marson and McKenna, 1992, 1994a; Marson et al., 1992), while also in the chronic treatment–induced delayed ejaculation
a facilitative action of 5-HT on the expulsion phase of ejacula- (de Jong et al., 2005b). Differences among individual SSRIs
tion, through the activation of peripheral 5-HT2 receptors, was in their ability to desensitize 5-HT1A receptors and to delay ejac-
recently reported in male rats (Ishigami et al., 2013). ulation appear to exist, with paroxetine and fluoxetine being
The facilitative effects on ejaculation resulting from stimula- more potent than citalopram and fluvoxamine in both actions
tion of the 5-HT1A receptor subtype were characterized with the (de Jong et al., 2007).
5-HT1A receptor agonist 8-OH-DPAT, which dramatically The absence of serotonin transporter (SERT) produced
decreased the IF and EL of sexually competent male rats, to lower ejaculation frequencies and prolonged EL in SERT
the point that some rats ejaculated on their first intromission knockout male rats (Chan et al., 2011). Both T and E2 have
(Ahlenius et al., 1981, 1989; Coolen et al., 1997a; Schnur been found to increase SERT and 5-HT2A receptor densities in
et al., 1989). Similar facilitation has been reported in sexually male rats’ brains (Barrett et al., 2006). Stimulation of spinal
18 Male Sexual Behavior

5-HT1A receptors inhibits rat penile erection (Pomerantz et al., sexually naïve animals to inaccessible estrous females
1993; Rehman et al., 1999), but facilitates ejaculation in spinal improves copulatory behavior, and this improvement was
cord-transected rats (Carro-Juárez et al., 2003). The concept of blocked by MK-801 (Powell et al., 2003). There is consider-
5-HT exerting a general inhibitory influence on sexual able evidence for glutamatergic facilitation of both copulation
responses is changing. It has recently been suggested that 5- and genital reflexes following microinjections into the MPOA,
HT is not only involved in the inhibitory control of male sexual PVN, and spinal cord (see Sections 1.01.5.2.1, 1.01.5.2.4, and
behavior, but also in disinhibiting processes to induce sexual 1.01.5.3.5). Thus, glutamate appears to be a major facilitator
behavior (Snoeren et al., 2014). Further, it was proposed that of copulation at both brain and spinal levels. However, the
5-HT released in high concentrations exerts an inhibitory influ- sexual condition of male rats and/or the dose range of gluta-
ence upon male sexual responses, whereas when released at matergic drugs administered appear to influence the role
low concentrations this neurotransmitter contributes to the played by glutamatergic transmission on copulatory behavior.
disinhibition of sexual responses (Rubio-Casillas et al., 2015). Thus, systemic injection of low doses of the NMDA antagonist
ketamine, the AMPA receptor antagonist CNQX, and the
mGluR5 receptor antagonist MPEP all facilitated sexual
1.01.4.4 Acetylcholine
behavior expression of sexually exhausted male rats
There is little information on selective effects of acetylcholine (Rodriguez-Manzo, 2015), characterized by being sexually
(ACh) on copulation in males. Early studies reported suppres- inhibited. Besides, systemic administration of high doses of
sion of male sexual behavior by high doses of nicotine and of MPEP and of the mGluR7 receptor antagonist AMN082 were
physostigmine, an acetylcholinesterase inhibitor, as well as by reported to inhibit sex seeking behavior as well as copulatory
antagonists such as atropine and scopolamine. However, a rela- performance (Li et al., 2013).
tively low dose of nicotine increased EF and reduced IF, EL, and Glutamate is released in the MPOA during copulation
PEI (for review, see Bitran and Hull, 1987), and the muscarinic (Dominguez et al., 2006a), and increases in local extracellular
agonist oxotremorine reduced IF and EL (Ahlenius and glutamate produced by glutamate reuptake inhibitors facili-
Larsson, 1985). Later, physostigmine-induced increases in tated copulation (Dominguez and Hull, 2010). On the other
ACh level and stimulation of muscarinic receptors with pilocar- hand, activation of glutamatergic transmission in the mesocor-
pine increased EF and promoted erections (Maeda et al., 1990; ticolimbic tract has also been associated to male sexual activity
Zarrindast et al., 1994). In hamsters, MPOA cholinergic trans- (Pitchers et al., 2012).
mission seems to play a role in the development of sexual
exhaustion (Floody, 2013) (see Sections 1.01.5.2.1 and
1.01.4.7 Nitric Oxide
1.01.5.3.5).
NO is a soluble gas that acts both as a second messenger and
as a neurotransmitter that has been implicated in male sexual
1.01.4.5 Gamma Aminobutyric Acid
functions that include penile erection (Andersson, 2001) and
GABA is the main inhibitory neurotransmitter in the mamma- brain control of sexual behavior (Bialy et al., 1996). NO is
lian brain (Paul, 1995). Systemic administration of GABAergic produced by the action of NOS, which converts L-arginine
drugs inhibited male rat sexual behavior (Ågmo and Paredes, into citrulline. Systemically administered NOS inhibitors
1985; Paredes et al., 1997). There are reports of inhibitory impair copulation, decreasing the proportion of sexually
effects of GABAergic drugs in the MPOA, PVN, and spinal naïve (Benelli et al., 1995) and experienced (Bialy et al.,
cord (see Sections 1.01.5.2.1, 1.01.5.2.4, and 1.01.5.3.5). In 1996; Hull et al., 1994) male rats that ejaculate, while aug-
male gerbils, a pathway originating in the ventral BST (vBST) menting MF and decreasing IF. NOS inhibitors also decrease
and MPN and projecting to the retrorubral field (RRF) that is the number of noncontact erections (Melis et al., 1998), as
activated during mating was reported to be mostly GABAergic well as reflexive erections, and also increase seminal emis-
(Simmons et al., 2011). An equivalent pathway also important sion (Hull et al., 1994). These effects are consistent with
for mating was described in male rats (Finn and Yahr, 2005). In a facilitative role of NO in parasympathetic function and
addition, cerebrospinal fluid concentrations of GABA increased an inhibitory one in sympathetic activity. A NOS inhibitor
dramatically during the PEI (Qureshi and Södersten, 1986). did not affect sexual motivation, evaluated in a female choice
maze (Hull et al., 1994), although another report found
a diminution evaluated by precoital activity and the
1.01.4.6 Glutamate
percentage of rats mounting (Ratnasooriya et al., 2000).
Excitatory amino acids appear to facilitate copulation and nNOS KO mice have normal penile function, due to
penile erection. Glutamate is the major excitatory neurotrans- a compensatory increase in eNOS (Burnett et al., 1996),
mitter in the CNS. Intraperitoneal injection of low doses of and achieve ejaculation with fewer mounts and intromis-
kainic acid, an agonist of AMPA/kainate receptors, enhanced sions (Kriegsfeld et al., 1999). Ip injection of L-arginine
copulatory behavior in sexually sluggish male rats, but did increased the percentage of sexually naïve rats that copulated
not affect good copulators; however it impaired sexual and improved sexual performance in experienced males,
behavior at higher doses (Drago and Busa, 1997). Systemic whereas icv injection of the NOS inhibitor L-NAME impaired
injection of the NMDA antagonist MK-801 impaired sexual copulatory parameters only in sexually naïve rats (Benelli
behavior in sexually experienced and sexually naïve male et al., 1995), and increases in NO production in the MPOA
rats (Powell et al., 2003), an effect also obtained in the were detected in response to the first sexual experience of
MPOA (Dominguez et al., 2007). Repeated exposure of male rats (Nutsch et al., 2014). NO may control both
Male Sexual Behavior 19

copulation and gential reflexes via actions in the MPOA, the endocannabinoid system exerts a complex influence on
PVN, and spinal cord (see Sections 1.01.5.2.1, 1.01.5.2.4, copulatory responses.
and 1.01.5.3.5). In rodents the expression of NOS in regions
implicated in the control of male sexual behavior is under
1.01.4.9 Endogenous Opioids
the control of gonadal hormones (Panzica et al., 2006).
NO also plays a crucial role in the initiation and mainte- Exogenous opiates such as morphine and heroin are known to
nance of increased intracavernous pressure and penile erec- have negative effects on the sexuality of male addicts, reducing
tion by acting on smooth muscle cells (Toda et al., 2005). sexual interest, impairing genital responses, and blocking ejac-
ulation and orgasm (Pfaus and Gorzalka, 1987). Endogenous
opioids belong to one of three major classes: endorphins,
1.01.4.8 Endocannabinoids
enkephalins, or dynorphins (Mains and Eipper, 1999). These
Endocannabinoids are retrograde messengers acting as neuro- peptides are generated by enzymatic processing from three
modulators in the CNS. They are released from postsynaptic precursor molecules, proopiomelanocortin, proenkephalin,
cell bodies, travel backward across the synaptic cleft, and and prodynorphin (Akil et al., 1984). An inhibitory role of
bind to their receptors mainly located presynaptically at nerve endogenous opioid peptides in the control of male sexual
endings (Wilson and Nicoll, 2002). The main endocannabi- behavior is commonly accepted, based on animal studies
noids are small molecules derived from arachidonic acid: anan- showing that acute and chronically administered morphine,
damide and 2-arachidonoylglycerol. Its neuronal receptor, the heroin, and methadone inhibit copulatory behavior in males
cannabinoid CB1 receptor, is located almost exclusively at of various species (Pfaus and Gorzalka, 1987). Similar results
axon terminals, where its stimulation inhibits neurotransmitter have been obtained after injection of opioid peptides such as
release (Freund et al., 2003). A general inhibitory role on male b-endorphin, morphicepin, and a Met-enkephalin analog
sexual behavior was ascribed to the endocannabinoid system into the MPOA (Hughes et al., 1987; Matuszewich and Dornan,
based on the observation that antagonism of CB1 receptors 1992; Pellegrini-Quarantotti et al., 1978). When infused into
accelerated ejaculation in rats (Gorzalka et al., 2008), while the amygdala, they retard sexual performance (McGregor and
both plant-derived (delta-9-THC) and synthetic (HU 210) Herbert, 1992b). Moreover, it has been reported that sexually
cannabinoid CB1 agonists impaired copulation in mice and inactive male rats have a constitutively increased basal concen-
rats (Shrenker and Barke, 1985; Murphy et al., 1994; Ferrari tration of the endogenous opioid octapeptide Met-Arg(6)-
et al., 2000). Exogenous administration of anandamide, the Gly(7)-Leu(8) in the hypothalamus (Rodríguez-Manzo et al.,
endogenous ligand for CB1 receptors, was also found to impair 2002a), as well as increased expression of proenkephalin (the
male sexual behavior similarly to high doses of exogenous precursor of the endogenous octapeptide) and prodynorphin
agonists (Martínez-González et al., 2004); however, inhibition mRNAs in the PVN (Arletti et al., 1997), compared with sexu-
of anandamide metabolism did not importantly affect male ally active animals.
sexual behavior (Gorzalka et al., 2008). Recently, it was shown Endogenous opioids exert their effects by acting on m, d, and
that low doses of anandamide actually facilitate male sexual k receptor subtypes. The inhibitory actions of these peptides on
behavior of sexually experienced and sexually exhausted rats copulation appear to be mainly mediated by m and d receptors,
(Canseco-Alba and Rodriguez-Manzo, 2014). Facilitative although involvement of k opioid receptors has also been
actions were also obtained after low anandamide doses in sexu- found. Systemic injection of the k opioid receptor agonist
ally sluggish (Rodriguez-Manzo and Canseco-Alba, 2015a), U-50,488H inhibited several sexual parameters, which were
noncopulating (Canseco-Alba and Rodriguez-Manzo, 2013), differentially prevented by intra-VTA, intra-NAc, or intra-
and sexually naïve animals (Rodriguez-Manzo and Canseco- MPOA injections of the k receptor antagonist norbinaltorphi-
Alba, 2015b), leading to the conclusion that anandamide mine (Leyton and Stewart, 1992). The inhibitory role of
exerts dose-based biphasic actions on male rat copulatory endogenous opioids in copulation is also suggested by studies
behavior (Rodriguez-Manzo and Canseco-Alba, 2015b). showing that opioid receptor antagonists (e.g., naloxone and
CB1 receptors are found at glutamatergic terminals in naltrexone) induce sexual behavior expression in sexually inac-
hypothalamic nuclei related to reproductive function (Melis tive rats (Gessa et al., 1979), facilitate the display of mounts
et al., 2006), but also at glutamatergic and GABAergic termi- and intromissions in sexually naïve rats (Pfaus and Wilkins,
nals of the mesolimbic system, where endocannabinoids 1995), reverse the sexual inhibition of sexually satiated rats
modulate the activity of this brain reward circuit (Lupica (Rodríguez-Manzo and Fernández-Guasti, 1995a), decrease
et al., 2004), involved in the motivational aspects of copula- the ejaculatory threshold in sexually competent intact rats,
tion (Kelley and Berridge, 2002). CB1 receptor antagonism and increase the percentage of ejaculating animals (Myers
in the PVN of male rats has also been reported to induce and Baum, 1979). Furthermore, the content of opioid octapep-
penile erection (Melis et al., 2004a; Succu et al., 2006b). In tide was higher in the hypothalamus of sexually inactive males,
this case, it has been hypothesized that CB1 receptor antago- compared to sexually active males (Rodríguez-Manzo et al.,
nists act at GABAergic and glutamatergic neurons to increase 2002a).
oxytocinergic transmission which in turn would promote However, there are inconsistent effects of opioid agonists
penile erection and facilitate ejaculation (Castelli et al., and antagonists on rodent male sexual behavior. Although
2007; Gorzalka et al., 2008). There is also evidence that the a large body of evidence points to the inhibitory nature of
endocannabinoid system interacts with gonadal androgens opioid agonist actions, there are also data suggesting facilitative
by acting on the hypothalamus and the anterior pituitary effects of these peptides. Thus, increasing enkephalin levels by
(Gorzalka and Dang, 2012). Together, these data show that icv administration of an enkephalinase inhibitor decreased IF
20 Male Sexual Behavior

and EL (Ågmo et al., 1994), and intra-VTA injection of produced a degree of VTA m opioid receptor internalization
morphine and dynorphin facilitated male sexual behavior proportional to the amount of ejaculatory activity displayed
and increased DA transmission in the NAc (Mitchell and (Garduno-Gutierrez et al., 2013c).
Stewart, 1990). Opioid facilitation may result from disinhibi- Systemic morphine dose-dependently reduced the propor-
tion of mesolimbic DA neurons by acting at m receptors on tion of animals showing reflexive erections and virtually elim-
VTA GABAergic neurons that exert a tonic inhibitory influence inated seminal emission; naloxone antagonized the effects of
on DA transmission (Balfour et al., 2004). On the other hand, morphine, but the lowest dose, administered alone, also
opioid antagonists also have had inhibitory effects, increasing inhibited erectile response, suggesting that some opioid activity
the duration of the PEI (McConnell et al., 1981; Sachs et al., can facilitate sexual reflexes (Gomez-Marrero et al., 1988).
1981). Thus, a complex picture of potentially excitatory and Stimulation of CNS m opioid receptors prevents penile erection
inhibitory effects of endogenous opioids on male sexual (reviewed in Andersson, 2001), and opioid mechanisms in the
behavior emerges. This complexity appears to be related to spinal cord appear to raise ejaculation threshold, since intra-
several factors, including dose-based, biphasic effects of thecal morphine increased the IF before ejaculation
agonists and antagonists, the brain site where they were admin- (Wiesenfeld-Hallin and Södersten, 1984). Moreover, opioids
istered, the time of the day when rats were tested, and the in the spinal cord have been found to play an inhibitory role
sexual activity level of the animals. Thus, the inhibitory effect in the spinal control of ejaculation (Carro-Juárez and
of b-endorphin is dose dependent (Argiolas, 1999) and is Rodríguez-Manzo, 2009) (see Section 1.01.5.3.5).
believed to occur mainly through actions at the MPOA and
the amygdala. In the former it disrupts copulatory behavior,
1.01.4.10 Oxytocin
while in the latter it disrupts precopulatory exploration
(Bancroft, 1999). Infusion of endomorphin-1 into the MPOA OT is expressed in neurons of the magnocellular PVN and
and the MeA also impaired copulatory behavior, effects that supraoptic nucleus (SON), both of which project to the poste-
were blocked by naloxone (Parra-Gamez et al., 2013). System- rior pituitary, and in parvocellular PVN neurons that project to
ically administered opioid receptor antagonists enhance sexual several brain areas and the lumbosacral spinal cord, where erec-
performance under certain testing conditions; for example, low tile and ejaculatory reflexes are controlled. Either icv or
doses of naloxone facilitated behavior when tested nocturnally, systemic injections of OT facilitated copulation in male rats
but not during the light phase (Van Ree et al., 2000). As to (reduced EL and PEI); conversely, icv injections of an OT antag-
sexual activity level, naloxone in the MPOA facilitates sexual onist impaired or abolished copulation in sexually experienced
performance in poor copulators, but impairs it in average cop- rats (reviewed in Argiolas and Melis, 2005). Facilitative effects
ulators (Van Ree et al., 2000); low ip-administered doses of were also seen in older (20 months) rats that were ‘sexually
both naloxone and naltrexone induce copulation in sexually sluggish’; OT treatment reduced MLs, ILs, ELs, and PEIs and
satiated rats (Rodríguez-Manzo and Fernández-Guasti, increased the number of animals able to resume copulation
1995a), as does naltrexone when directly infused into the after the first ejaculation (Arletti et al., 1990). Intra-MPOA
VTA. However, in sexually experienced males, intra-VTA injection of OT facilitated mating in sexually experienced (Gil
naltrexone impairs copulation (Garduno-Gutierrez et al., et al., 2011) and sexually naïve male rats, and copulation
2013b). This last datum suggests that the motivational status increased OT receptor gene and protein expression in this brain
of the animals rather than the sexual activity level might be region, suggesting a reciprocal interaction between central OT
the defining factor for opioid antagonist effects, since sexually and sexual behavior (Gil et al., 2013). Systemic OT also
exhausted animals were sexually competent males that became reversed the inhibitory effects of chronic fluoxetine, an SSRI,
sexually inhibited as a result of copulation to satiation. Endog- on the ability of male rats to ejaculate (Cantor et al., 1999).
enous opioids acting at the VTA are involved in sexual motiva- SSRI-induced delayed ejaculation was related to
tion (van Furth and van Ree, 1996). desensitization of 5-HT1A receptors on OT neurons (de Jong
Evidence suggests that opioid peptides are released during et al., 2007). Plasma OT is increased during sexual activity in
sexual activity, since the physiological mechanisms of analgesia humans (Carmichael et al., 1987) and is released in the
and reward are concurrently activated during sexual behavior hypothalamus of male rats during mating (Waldherr and
(Szechtman et al., 1981), and both phenomena are blocked Neumann, 2007). OT infusion into the PVN of male rats
by naloxone (Ågmo and Berenfeld, 1990; Forsberg et al., stimulates penile erection (Kita et al., 2006) an effect that is
1987). In addition, increases in Met-enkephalin and the opioid also obtained when OT is injected into brain areas that
octapeptide Met-Arg(6)-Gly(7)-Leu(8) were detected in the receive projections from the PVN, e.g., VTA, hippocampus,
hypothalamus of rats that ejaculated once or copulated to sati- amygdala, and spinal cord (Melis and Argiolas, 2011) (see
ation 24 or 48 h earlier (Rodríguez-Manzo et al., 2002a). Block- Section 1.01.5.2.4).
ing the actions of the endogenous opioids released by multiple
ejaculations during the process of copulation to exhaustion
1.01.4.11 Prolactin
interferes with the establishment of the characteristic sexual
inhibition 24 h later (Garduno-Gutierrez et al., 2013a). Prolactin (PRL) is secreted by the anterior pituitary into the
Mating-induced m opioid receptor internalization, a marker general circulation. This peptide has a well-established role in
for ligand-induced receptor activation, was detected in the lactation; however PRL has been reported to have more than
MPOA (Coolen et al., 2004b) and VTA (Balfour et al., 2004) 300 functions across vertebrates, a majority of which relate to
of rats after one ejaculation. Ejaculation also induced d opioid reproduction (Bancroft, 2005). Hyperprolactinemic patients
receptor internalization in the VTA and repeated ejaculation have low sexual desire often associated with erectile problems
Male Sexual Behavior 21

(Corona et al., 2007). Plasma PRL levels increase markedly neurons project widely to monoaminergic nuclei in the
following ejaculation and orgasm in men (Bancroft, 2005). midbrain and brain stem and to a number of basal forebrain
This postorgasmic PRL increase may act as a feedback control areas, including brain areas relevant for sexual behavior like
of the refractory period following orgasm (Krüger et al., the PVN, the MPOA, and the VTA (Peyron et al., 1998). These
2003). In line with this hypothesis, there is a case report of peptides are primarily known for their ability to regulate
a multiorgasmic healthy man that showed no PRL response feeding and wakefulness; however, they can also enhance
to three consecutive orgasms (Haake et al., 2002). However, male sexual behavior. Their expression is hormonally regu-
male rats engaged in constant sexual activity show a sustained lated, being decreased after castration and restored by E2;
increase in serum PRL during mating (Rojas-Duran et al., a slight increase by DHT was not statistically significant
2015). Animal experiments confirm that chronic exposure to (Muschamp et al., 2007). Both copulation and estrous female
elevated PRL impairs male rat sexual response, increasing IL odors increased Fos expression in orx/hcrt neurons, implying
and EL and reducing reflexive erections; short-term PRL their activation during copulation; in addition, systemic
increase had either no effect or facilitated copulation (Melis administration of an orx/hcrt antagonist (SB334867) delayed
and Argiolas, 1995). the onset of copulation and decreased the number of ejacula-
tions per 30-min test (Muschamp et al., 2007). However,
specific lesion of orexin neurons did not impair sexual
1.01.4.12 Gonadotropin-Releasing Hormone
behavior, but prevented the formation of a conditioned place
GnRH is a peptide hormone produced by the mediobasal preference for copulation (Di Sebastiano et al., 2011) suggest-
hypothalamus, lamina terminalis, and the MPOA that acts on ing a role of this neuropeptide in sexual reward. In line with
the anterior pituitary where it stimulates gonadotropin release. this idea, intra-VTA injection of orexin increased DA release
Earlier studies found that GnRH, administered systemically or in the NAc (Korotkova et al., 2003) as well as dopaminergic
icv, facilitated male sexual behavior in castrated rats main- cell firing (reviewed in Hull, 2011); however, it has been re-
tained with low doses of T, but not in intact animals (Moss ported that icv-administered orexin-A attenuated male sexual
et al., 1975). By contrast, Myers and Baum (1980) found a facil- motivation measured in a preference paradigm (Bai et al.,
itative effect of GnRH only in gonadally intact rats and not in 2009).
castrates replaced with T. A facilitative action of GnRH on
sexual motivation was also established in a mounting test in
rats with penile anesthesia, after its icv infusion (Dorsa and 1.01.5 Brain Areas and Circuitry Implicated
Smith, 1980). GnRH is released naturally when male rodents in the Control of Masculine Sexual Behavior
(mice and hamsters) encounter female vaginal fluid chemosig-
1.01.5.1 Sensory Inputs
nals (Westberry and Meredith, 2003), and GnRH-containing
neurons are activated in male rats by estrous female odors 1.01.5.1.1 Olfactory Bulbs
(Kippin et al., 2003). In sexually naïve hamsters, GnRH Chemosensory signals are transduced in the olfactory mucosa
infused icv restored mating behavior impaired by removal of and VNO and are transmitted to the olfactory bulbs. The
VNOs (Meredith and Fernandez-Fewell, 1994). Systemic importance of chemosensory cues varies across species, but
GnRH facilitates sexual behavior in hyperprolactinemic male these cues are especially important in rodents and other
rats treated with T (Dennison et al., 1996), and exogenous nocturnal species. The olfactory system includes the anatomi-
GnRH restores fertility and sexual activity in men with cally and functionally distinct main and accessory olfactory
hypogonadism associated with low levels of circulating T systems. Volatile odors, processed by the main olfactory
(Mortimer et al., 1974). The loss of GnRH neurons in the system, are transduced by receptors in the nasal mucosa, whose
medial septum and MPOA has been associated with aging- axons project through the cribriform plate to the main olfactory
related diminished mating ability (Tsai et al., 2014). bulb (MOB). Both nonvolatile and volatile species-specific che-
However, adverse effects of GnRH treatment have also been mosensory cues, or pheromones, are detected in the VNO,
reported. Thus, GnRH in old rhesus monkeys increased the PEI located at the base of the nasal cavity and projecting to the
(Phoenix and Chambers, 1990). Chronic administration of accessory olfactory bulb (AOB). Male rats that experienced
a synthetic GnRH analog [(6-D-(2-napthyl)-alanine)GnRH] three ejaculations had a greater density of new cells, and specif-
initially increased IL in intact rats and eliminated sexual ically, new neurons, in the posterior part of the granular cell
behavior 6–8 weeks later, along with a decrease in T (Dorsa layer of the AOB 45 days after copulating (Unda et al., 2016).
et al., 1981). The inhibitory effects probably resulted from No significant differences were found in the MOB. Thus, copu-
the continuous high doses of GnRH, which inhibit LH release lating to three ejaculations can induce lasting changes in the
and gonadal function, whereas endogenous GnRH is released AOB. While vomeronasal cues are important in rodents and
in a pulsatile fashion. Studies in hypogonadal mice bearing other macrosmotic animals, the VNO and AOB are regressed
a deletion of the GnRH-encoding gene suggest that this in humans and may be nonfunctional.
peptide is not essential for male sexual behavior (Gibson
et al., 1997). 1.01.5.1.1.1 Effects of Lesions
Mating by male Syrian hamsters is abolished by olfactory bul-
bectomy. (See Hull et al., 2006 for review of the older research
1.01.4.13 Orexin/Hypocretin
in this area.) However, the relative importance of the main and
The orexin/hypocretin (orx/hcrt) peptides (orexin A and accessory systems is not clear. Removal of the VNO impaired
B/hypocretin 1 and 2) are expressed in the LH; orexinergic copulation in sexually naïve males, but had little effect in
22 Male Sexual Behavior

experienced males (Ballard and Wood, 2007; Meredith, 1986). downstream structures of the vomeronasal pathway
However, vomeronasal nerve cuts, which also damaged part of (Hosokawa and Chiba, 2005; Portillo and Paredes, 2004).
the MOB, produced greater deficits, especially in naïve males The authors suggest that alterations in the pathway from the
(Meredith, 1986). Destruction of receptors in the nasal mucosa VNO render some males unable to process fully the chemosen-
with zinc sulfate produced minimal damage (O’Connell and sory cues of estrous females, resulting in reduced sexual
Meredith, 1984), but deafferentation of both the MOB and motivation.
AOB abolished copulation (Meredith et al., 1980). Although both the main olfactory and vomeronasal systems
Male rats are somewhat less dependent on chemosensory contribute to rodent sexual behavior, male ferrets, which are
input than are hamsters, although bilateral bulbectomy did carnivores, rely only on the main olfactory system (Kelliher
severely compromise mating in some males and increased IL et al., 1998). Female odorants did not elicit Fos-ir in the
and EL in those that did copulate (Edwards et al., 1990, AOB, but did activate clusters of glomeruli in the ventral–
1996; reviewed in Hull et al., 2006). Noncontact erections, eli- caudal MOB (Woodley and Baum, 2004). The ferret MOB
cited by female rat urine, were also eliminated by olfactory bul- has ARs, which have the potential to enhance neuronal
bectomy or zinc sulfate lesions of the nasal mucosa, but not by responses to chemosensory stimuli from the female.
removal of the VNO (Kondo et al., 1999).
Chemosensory stimuli also promote sexual activity of male
1.01.5.1.2 Amygdala
mice (reviewed in Hull et al., 2006). Removal of the VNO
1.01.5.1.2.1 Anatomy
blocked the preference for urinary odors of estrous females
There is controversy about whether ‘the amygdala’ should be
but did not affect olfactory sex discrimination (Pankevich
regarded as a single entity or a collection of nuclei that
et al., 2004). Male mice with a null mutation of the transient
contribute to learning, motivation, and fear (central nucleus
receptor potential 2 (trp2) cation channel, which disrupted
and basolateral division) or to chemosensory processing and
VNO function, had little electrophysiological response to pher-
social behaviors (corticomedial division) (Swanson and
omones, but mated normally with females and also mounted
Petrovich, 1998). Clearly, the corticomedial region serves as
other males (Leypold et al., 2002; Stowers et al., 2002). In
an integration site for chemosensory, somatosensory, and
contrast, mice genetically lacking the cyclic nucleotide-gated
hormonal stimuli and projects to the MPOA and other central
channel alpha 2, necessary for odor-evoked signaling in the
regulatory areas. The AOB projects via the lateral olfactory tract
main olfactory epithelium, failed to mate (Mandiyan et al.,
to the medial and posteromedial nuclei, while the MOB
2005). Thus, in mice, the main olfactory system is more critical
projects diffusely to the anterior and posterolateral cortical
for sexual behavior than the vomeronasal system.
nuclei and to the ventral allocortex (Figure 3). The MeA also
receives somatosensory input from the genitals, relayed via
1.01.5.1.1.2 Activation of c-Fos or Other Immediate-Early Genes
the subparafascicular nucleus (SPF), which also projects
Immediate-early genes are expressed transiently after a stim-
directly to the MPOA. There are abundant ERs and ARs in the
ulus. Their protein products are transcription factors that may
MeA, especially in the posterior subnucleus, as well as in the
affect neural activity or responsiveness to certain stimuli. In
MPOA and other hypothalamic nuclei. The volume and soma
situ hybridization for mRNA from the c-fos gene or immunocy-
size of the MeApd are larger in male than in female rats (Cooke
tochemistry for its protein product Fos is widely used to map
et al., 1999; Hines et al., 1992) and are maintained by stimula-
neural activation in responsive brain areas. Fos was increased
tion of both ARs and ERs (Cooke et al., 2003). Males also have
in the MOB and AOB of male hamsters following either copu-
more neurons and glial cells in that area; the number of glia is
lation or exposure to female odors (Fernandez-Fewell and Mer-
influenced by circulating androgens; neuron number appears
edith. 1994; Fiber et al., 1993; Kollack-Walker and Newman,
to be ‘organized’ before adulthood (Morris et al., 2008).
1997; Swann, 1997). Furthermore, female odors elicited Fos
expression in the olfactory bulb even in castrates, although
the castrates were unable to copulate (Swann, 1997). Therefore, 1.01.5.1.2.2 Effects of Lesions
castration may result in loss of signal in downstream areas Lesions of the central nucleus and basolateral division have
without altering detection of female cues. In rats and mice, little effect on copulation, although they do impair condi-
too, either copulation or exposure to female odors elicited tioned responses during fear and reinforcement (reviewed in
Fos-ir in the MOB, AOB, and downstream structures (Baum Everitt, 1990). In contrast, corticomedial lesions do impair
and Everitt, 1992; Coolen et al., 1997b; Halem et al., 1999; copulation, with the severity dependent on the specific location
Robertson et al., 1991), with mating being more effective and the species. Anterior MeA lesions abolish copulation in
than odor alone (Kelliher et al., 1999). In male mice, Fos-ir eli- male hamsters, while more posterior lesions impair, but do
cited by female odors was localized to the rostral portion of the not eliminate mating (Lehman et al., 1980). A more recent
AOB and was dependent on an intact VNO (Dudley and Moss, study reported that lesions of the MeApd in male hamsters
1999; Matsuoka et al., 1999). However, intranasal zinc sulfate decreased attraction to female odors, but anterior MeA lesions
in mice, intended to disrupt the main olfactory epithelium, led to prolonged investigation of both male and female odors
blocked the Fos response to female urine in both the MOB (Maras and Petrulis, 2006). The authors suggested that the
and AOB (Martel and Baum, 2007). In rats, exposure to female MeApd may increase arousal toward relevant social stimuli,
bedding elicited Fos-ir in the AOB of all males, regardless of whereas the anterior MeA may filter stimuli and direct the
whether they were sexually experienced, noncopulators, or arousal toward appropriate social cues. In rats (de Jonge
sexually naïve (Portillo and Paredes, 2004); however, only in et al., 1992; Dominguez et al., 2001; Kondo, 1992; McGregor
sexually experienced copulators was Fos expressed in all and Herbert, 1992a) and gerbils (Heeb and Yahr, 2000), MeA
Male Sexual Behavior 23

Figure 3 Diagram of the ventral surface of the hamster brain. Shaded areas indicate brain regions that receive efferent projections of the olfactory
system, via the main olfactory bulb (left), and of the vomeronasal system, via the accessory olfactory bulb (right). ACo, anterior cortical nucleus of
the amygdala; AOB, accessory olfactory bulb; AON, anterior olfactory nucleus; BNST, bed nucleus of the stria terminalis; EC, entorhinal cortex; HR,
hippocampal rudiment; Me, medial nucleus of the amygdala; MeA, medial nucleus of the amygdala, anterior division; MeP, medial nucleus of the
amygdala, posterior division; MOB, main olfactory bulb; nAOT, nucleus of the accessory olfactory tract; nLOT, nucleus of the lateral olfactory tract;
OLF EPITH, olfactory epithelium; OTu, olfactory tubercle; PC, piriform cortex; PLCo, posterolateral cortical nucleus of the amygdala; PMCo, poster-
omedial cortical nucleus of the amygdala; VNO, vomeronasal organ. Figure is reprinted from Wood, R.I., Newman, S.W., 1995. Hormonal influence
on neurons of the mating behavior pathway in male hamsters. In: Micevych, P.E., Hammer Jr., R.P. (Eds.), Neurobiological Effects of Sex Steroid
Hormones. Cambridge University Press, New York, pp. 3–39 (Chapter 1) with permission from Cambridge University Press.

lesions delay and slow copulation and increase the intromis- nuclei of the amygdala of male rats (Baum and Everitt, 1992;
sions required to elicit ejaculation. Coolen et al., 1996; Veening and Coolen, 1998), with the
Radiofrequency lesions of the posterior MeA of male rats largest number of Fos-ir cells in the MeA, especially the poste-
eliminated noncontact erections and decreased preference for rior subnucleus, and fewer in the anterior and posteromedial
an estrous female, but did not affect copulation (Kondo and cortical nuclei (Kollack and Newman, 1992). However, expo-
Sachs, 2002; Kondo et al., 1998). A subregion of the MeApd sure to an inaccessible estrous female rat elicited about as
has been linked to sexual satiety. Lesions of the MeApd in much Fos-ir in arginine vasopressin-containing neurons in
male hamsters delayed sexual satiety (Parfitt et al., 1996) (see the MeApd as did copulation in male rats (Hari Dass and
Section 1.01.5.1.2.3). Vyas, 2014). Perhaps those neurons are more attuned to chem-
ical stimuli than to copulation. Mating also elicited Fos-ir in
1.01.5.1.2.3 Activation of c-Fos or Other Immediate-Early Genes amygdaloid nuclei of hamsters (Kollack and Newman, 1992,
or Other Measures of Activity 1997), gerbils (Heeb and Yahr, 1996), prairie voles (Wang
Chemosensory stimuli can induce Fos-ir in the corticomedial et al., 1997), and musk shrews (Gill et al., 1998). In gerbils
amygdala in rats (Bressler and Baum, 1996; Coolen et al., the medial part of the MeApd was activated (Fos-ir) by sex-
1997b; Kelliher et al., 1999), hamsters (Fernandez-Fewell and related odors, whereas the lateral portion was activated only
Meredith, 1994; Fiber et al., 1993; Kollack and Newman, following ejaculation (Heeb and Yahr, 1996). Similarly, in
1992), and gerbils (Heeb and Yahr, 1996). Increasing amounts hamsters Fos-ir in the MeApd was correlated with the onset
of sexual behavior elicited increasing amounts of Fos-ir in of satiety (Parfitt and Newman, 1998). Fos-ir in the MeA, but
24 Male Sexual Behavior

not in the MPOA, of male rats was positively correlated with for both aromatase and AR in the MPOA. The authors did
the length of the PEI (Lumley and Hull, 1999), suggesting not suggest an explanation for the apparent inverse
that some neurons in that area may contribute to postejacula- relationship between aromatase (and AR in the MPOA) and
tory quiescence. In rats (Gréco et al., 1998b) and hamsters copulatory ability.
(Wood and Newman, 1993), AR-containing cells in the
posterior MeA are activated by mating; however, castration 1.01.5.1.2.5 Microinjections of Drugs
did not reduce mating-induced Fos-ir in the MeA of rats Microinjection of the serotonin 5-HT1A agonist 8-OH-DPAT
(Baum and Wersinger, 1993) or odor-induced Fos-ir in the into the MeApd speeded the onset of copulation and decreased
MeA of hamsters (Swann, 1997). Similarly, mice lacking the the PEI in male rats (de Castilhos et al., 2006). Therefore, the
gene for aromatase had similar response to odors as in wild- MeApd is one site at which 5-HT1A agonists facilitate copula-
type mice (Aste et al., 2003). tion. Angiotensin microinjections into the MeA impaired copu-
Men and women underwent functional magnetic resonance lation in male rats; however, it is not clear whether increased
imaging (fMRI) while watching sexual or nonsexual pictures thirst may have interfered with sexual behavior (Breigeiron
(Cyders et al., 2016). Only men showed increased brain activity et al., 2002).
in response to the sexual stimuli. Areas activated included the
bilateral amygdala, anterior cingulate cortex/medial prefrontal 1.01.5.1.2.6 Amygdaloid Efferents
cortex, anterior insula/lateral orbitofrontal cortex, and occipital A major output of the MeA is to the MPOA. Unilateral lesions
regions. of the MPOA impaired, but did not abolish copulation in male
rats; however, contralateral lesions of the MeA and MPOA
1.01.5.1.2.4 Effects of Hormone Implants or Manipulations severely disrupted sexual behavior (Kondo and Arai, 1995).
Affecting Hormone Receptors or Enzymes Similar results were reported for gerbils (Heeb and Yahr,
In castrated male rats, bilateral T implants in the MeA delayed 2000). Chemical stimulation of the MeA increased DA release
the loss of mating and of noncontact erections (Berendsen and in the MPOA (Dominguez and Hull, 2001), and microinjec-
Broekkamp, 1987), and E implants stimulated mounting, but tion of the DA agonist apomorphine into the MPOA restored
not ejaculation (Huddleston et al., 2003). Similarly, in cas- copulation that had been abolished by large excitotoxic lesions
trated hamsters either T (Wood, 1996) or E (Coolen and of the MeA (Dominguez et al., 2001). Basal extracellular DA in
Wood, 1999; Wood and Coolen, 1997), but not DHT the MPOA was not affected by small radiofrequency MeA
(Wood, 1996), implants in the MeA restored copulation. lesions, but the DA release in response to a female and during
However, DHT implants were successful in male rats treated copulation was eliminated by the lesions, which also impaired
with subthreshold systemic injections of E (Baum et al., but did not abolish copulation (Dominguez et al., 2001).
1982). Either an ER antagonist in male hamsters (Wood and Indeed, chemical stimulation of the MeA elicited DA release
Williams, 2001), or an aromatase (Huddleston et al., 2006) in the MPOA of a magnitude similar to that observed during
or AR (McGinnis et al., 1996) antagonist in male rats impaired, copulation (Dominguez and Hull, 2001). Thus, it appears
but did not block, copulation, suggesting that both androgenic that one result of MeA activity is to increase DA in the MPOA
and estrogenic stimulation contributes to mating in male rats in anticipation of and during mating. There are no dopami-
and hamsters. Furthermore, MeA implants of E2 conjugated nergic neurons in the MeA of rats; therefore, efferents from
to bovine serum albumin (which prevents E2 from crossing the MeA may activate, either directly or indirectly, dopami-
the membrane) were ineffective in maintaining mating nergic cell bodies or terminals in the MPOA and thereby facil-
behavior (Huddleston et al., 2006), suggesting an intracellular itate mating.
mechanism of action. Because both hormonal and chemosen- Chronic isolation of normally monogamous prairie voles
sory stimuli are critical for mating in male hamsters and are resulted in accelerated copulation when males were paired
processed in the MeA, their interactions were probed. Unilat- with a female (Perry et al., 2016). Pups had been reared with
eral olfactory bulbectomy, either ipsi- or contralateral to a T same-sex siblings until adulthood, when they were isolated
implant in the posterior MeA of castrated hamsters, rendered for 4 weeks. Males had increased ERa in the MeApd but
the implant ineffective (Wood and Coolen, 1997), suggesting decreased tyrosine hysroxylase (TH), which is the rate-
that bilateral chemosensory input is essential for T in this limiting enzyme for catecholamine production. As noted
nucleus to stimulate sexual behavior. However, with similar T above, the MeA projects to the MPOA; however, it lacks
implants in both the MPOA and BST, only ipsilateral bulbec- DOPA decarboxylase, which would produce DA; therefore,
tomy blocked the facilitation by T (Wood and Newman, those neurons could release either L-DOPA or other trace
1995c). Thus, communication between groups of neurons amines into the MPOA. Trace amine receptors have been
that process hormonal and chemosensory stimuli is critical shown to decrease DA release (Lindemann et al., 2008), which
for male hamster sexual behavior. is important for copulation. Therefore, a reduction in trace
The P receptor antagonist RU-486, administered to male hormone release in the MPOA could promote mating before
rat pups during the first postnatal week, resulted in increased pair bond formation could occur, which would facilitate polyg-
AR-ir in the MeA and several other areas, including BST, amous strategies.
MPOA, and ventromedial nucleus (Forbes-Lorman et al.,
2014). Copulation was also enhanced by the RU-486 1.01.5.1.3 Bed Nucleus of the Stria Terminalis
treatment. In contrast, Antaramian et al. (2015) reported 1.01.5.1.3.1 Anatomy
that sexually sluggish male rats actually had increased Output from the MeA travels either directly to the MPOA or
mRNA for aromatase in the amygdala and increased mRNA synapses in the BST, which then relays the information to the
Male Sexual Behavior 25

MPOA and other sites. The posteromedial division of the BST is Subregions of the tegmentum have been referred to as the
especially relevant to the control of male sexual behavior central tegmental field (CTF, e.g., Baum and Everitt, 1992;
(Alheid et al., 1995). As with the posterior MeA, the posterome- Gréco et al., 1999; Simerly and Swanson, 1986) or dorsolateral
dial BST has abundant steroid hormone receptors (Li et al., tegmentum (DLT, e.g., Edwards and Einhorn, 1986; Giordano
1993; Simerly et al., 1990; Wood and Newman, 1993). et al., 1998; Maillard and Edwards, 1991). The CTF/DLT is
dorsal to the lateral half of the substantia nigra and includes
1.01.5.1.3.2 Effects of Lesions the SPF, part of the zona incerta, the peripeduncular nucleus,
In male rats and hamsters the more dorsal part of the BST the mesencephalic reticular nucleus, and the anterior pretectal
appears to contribute more to preparation for mating than to nucleus. The medial parvocellular division of the subparafascic-
copulation per se (reviewed in Hull et al., 2006). In rats, lesions ular nucleus (SPFp) relays somatosensory input from the geni-
decreased the number of noncontact erections and slowed the tals to the MPOA and MeA. It is considered to be part of an
rate of copulation, but did not affect the intromission ratio, ejaculation circuit (see Section 1.01.6).
suggesting that in copula erections were unaffected; in contrast,
lesions of the MPOA had little effect on noncontact erections 1.01.5.1.4.2 Effects of Lesions
but severely impaired copulation (Liu et al., 1997b). Finn Bilateral neurotoxic lesions of the CTF decreased the percent-
and Yahr (2005) suggested that previous studies that showed ages of male rats that could mount, intromit, and ejaculate
little effect on copulation targeted the dorsal part of the BST. (Romero-Carbente et al., 2007). Partner preference and sexual
In contrast, they made excitotoxic lesions of the vBST, which incentive motivation were not affected. Even misplaced lesions
projects to the retrorubral field (A8) of the midbrain and other in adjacent areas impaired behavior, suggesting that the effects
downstream sites. vBST lesions in rats resulted in severe copu- were not specific to the CTF. In tests 3 weeks after the lesion,
latory deficits, although damage to the adjacent posterodorsal copulation had returned to normal. Earlier studies reported
preoptic nucleus (PdPN) may have contributed to the impair- more severe deficits (Brackett and Edwards, 1984; Giordano
ment (Finn and Yahr, 2005). The authors suggested that the et al., 1998), and contralateral lesions of the MPOA and CTF
vBST in rats may be the counterpart to the gerbil sexually impaired both copulation and pursuit and sniffing of the
dimorphic area (SDA) of the preoptic area, which is critical female (Edwards and Einhorn, 1986). Combined ipsilateral
for male gerbil sexual behavior. In gerbils contralateral lesions lesions of the CTF and MeA abolished mating-induced Fos-ir
of the caudal medial BST and the SDA severely impaired copu- in the MPOA, whereas single lesions of each did not (Baum
lation, but contralateral lesions of the MeA and SDA produced and Everitt, 1992). Bilateral SPFp lesions in gerbils did not
less of a deficit, suggesting that the caudal medial BST does affect copulation, suggesting that the Fos-ir elicited in that
more than simply transmit information from the MeA to the nucleus following ejaculation resulted from sensory input,
SDA (Sayag et al., 1994). rather than motor control of ejaculation (Heeb and Yahr,
2000). However, in male rats the SPFp has been shown to
1.01.5.1.3.3 Activation of c-Fos or Other Immediate-Early Genes receive projections from lumbar spinothalamic neurons
In male rats, hamsters, and gerbils, copulation, or to a lesser (Coolen et al., 2003), which are essential for ejaculation (Truitt
extent, exposure to female odors elicited Fos-ir in the BST, espe- and Coolen, 2002) and express ejaculation-related Fos-ir
cially in the posterior and medial subdivisions (Coolen et al., (Truitt et al., 2003).
1996; Fernandez-Fewell and Meredith, 1994; Heeb and Yahr,
1996; Kelliher et al., 1999; Kollack-Walker and Newman, 1.01.5.1.4.3 Activation of c-Fos or Other Immediate-Early Genes
1995). In quail, both visual and physical contact increased Fos-ir was selectively increased in either the CTF or SPFp after
Fos-ir in the BST (Iyilikci et al., 2014). In contrast, mating ejaculation in rats (Baum and Everitt, 1992; Coolen et al.,
decreased Fos-ir in male macaques (Michael et al., 1999), 1997a,b, 1996; Wersinger et al., 1993), gerbils (Heeb and
and female odors did not affect Fos-ir in male ferrets (Kelliher Yahr, 1996), hamsters (Kollack-Walker and Newman, 1997),
et al., 1998). and musk shrews (Gill et al., 1998). Fos-ir was not increased
after chemosensory investigation, mounts, or intromissions.
1.01.5.1.3.4 Hormonal Effects In men, ejaculation stimulated blood flow in the SPFp, as
There are abundant steroid receptors in the posteromedial BST, measured with positron emission tomography (Holstege
extending from the stria terminalis to the MPN (Li et al., 1993; et al., 2003). Also, electrical stimulation of the CTF facilitated
Simerly, 1995; Simerly et al., 1990; Wood and Newman, mating in rats (Shimura and Shimokochi, 1991).
1993). Castration decreased, and T restored, immunoreactivity
to CCK in the encapsulated portion of the BST (Simerly and 1.01.5.1.4.4 Presence of Steroid Receptors
Swanson, 1987). Similar effects were seen in SDAs of the SPFp neurons contain ARs (Gréco et al., 1996), and many AR-ir
MeA and MPN. neurons that project to the MPOA express ejaculation-induced
Fos-ir (Gréco et al., 1998b).
1.01.5.1.4 Central Tegmental Field/Subparafascicular Nucleus
of the Thalamus 1.01.5.1.4.5 Connections
1.01.5.1.4.1 Anatomy The medial SPFp contains many galanin-ir fibers, and neurons
There are reciprocal connections between the midbrain containing mating-induced Fos-ir in the medial SPFp are
tegmentum and the MPOA, MeA, and anterior hypothalamus surrounded by galanin-ir fibers (Veening and Coolen, 1998).
(Coolen et al., 1998, 2003; Gréco et al., 1999, 1998b; Murphy Neurons in laminae 7 and 10 of the lumbosacral spinal
et al., 1999a; Simerly, 1995; Simerly and Swanson, 1988). cord project to the medial SPFp (Coolen et al., 2003;
26 Male Sexual Behavior

Gréco et al., 1999; Truitt et al., 2003), which also connects recip- ejaculation (Heeb and Yahr, 2000). Unilateral lesions of the
rocally with forebrain nuclei that control copulation (Coolen medial SDA in gerbils, together with lesions of the contralateral
et al., 2003). Therefore, galanin may convey ejaculation-related lateral SDA, impaired copulation as effectively as bilateral
somatosensory input through the SPFp to higher brain areas. lesions of each (Yahr and Gregory, 1993).
Large MPOA lesions disrupt the initiation of copulation as
well as its performance, suggesting that it contributes to sexual
1.01.5.2 Major Integrative Sites
motivation. However, male rats, cats, dogs, and monkeys
1.01.5.2.1 Medial Preoptic Area showed interest in sexual behavior even after MPOA lesions
1.01.5.2.1.1 Anatomy abolished their ability to copulate (reviewed in Everitt, 1990;
The MPOA is a critical integrative site for male sexual behavior Hull and Dominguez, 2015). Similarly, noncontact erections
in all vertebrate species tested. This is remarkable, in that a wide were not affected by lesions that abolished copulation (Liu
range of sensory stimuli elicit mating, and diverse motor et al., 1997b). This apparent dissociation of copulatory perfor-
patterns comprise the species-specific behaviors. Every sensory mance and sexual motivation following MPOA lesions led
modality sends indirect input to the MPOA, and reciprocal Everitt (1990) to suggest that the MPOA controls only copula-
connections allow the MPOA to modify the processing of tory performance and not sexual motivation.
sensory input (Simerly and Swanson, 1986). Furthermore, However, MPOA lesions have decreased sexual motivation
steroid receptors in the MPOA and its major afferents can in several contexts, including female partner preference in rats
bias input to favor sexually relevant stimuli. T can influence (Edwards et al., 1996; Paredes et al., 1998) and ferrets (Kindon
the neuroanatomy of MPOA neurons. Gonadally intact and et al., 1996; Paredes and Baum, 1995), pursuit of a female by
T-treated castrates had more dendritic spines and greater male rats (Paredes et al., 1993), or precopulatory behaviors
spine density than did castrates without T (Garelick and in marmosets (Lloyd and Dixson, 1988). Therefore, MPOA
Swann, 2014). However, dendrite length was not increased. lesions do not eliminate sexual motivation, but they clearly
Therefore, existing dendrites produce more spines that are diminish it. The evolutionary conservation of MPOA influence
available for synapses, but do not extend their length. on both sexual performance and motivation is seen in studies
Efferents from the MPOA to hypothalamic, midbrain, and of Japanese quail and starlings, in which small MPOA lesions
brain stem nuclei regulate somatomotor or autonomic decreased time spent in front of a window through which the
patterns and motivational states (reviewed in Simerly and male could see a female (Balthazart et al., 1998) or reduced
Swanson, 1988; Yahr, 1995). singing and gathering of nesting materials (Riters and Ball,
The MPOA comprises heavily interconnected subnuclei 1999).
with various functions, connections (Maragos et al., 1989; Sim-
erly et al., 1986; Wang and Swann, 2014), and neurotrans-
mitter content (Simerly et al., 1986). A medial periventricular 1.01.5.2.1.3 Effects of Electrical or Chemical Stimulation
zone regulates neuroendocrine function, and a medial zone, Because MPOA lesions impair male sexual behavior, it was ex-
including MPN and PdPN, controls male sexual behavior and pected that stimulation would facilitate it, and, indeed,
maternal behavior. The magnocellular medial preoptic nucleus numerous early studies of rats, guinea pigs, and opossums
in male Syrian hamsters receives input from chemosensory showed such facilitative effects (reviewed in Hull and Domi-
pathways and steroid receptor-containing areas and sends its nguez, 2015). Decreases in mounts and intromissions
efferents to several brain stem nuclei that regulate aspects of preceding ejaculation and shortened ejaculation latency and
male sexual behavior, including the periaqueductal gray PEIs were reported. One such study found facilitation of copu-
(PAG), deep mesencephalic nucleus, retrorubral field, VTA, lation following electrical stimulation of the MPOA of male
and lateral paragigantocellular nucleus (Wang and Swann, rats; however, such stimulation did not restore copulation
2014). Other targets are steroid-containing nuclei in the 24 h after they had copulated to sexual satiety (Rodríguez-
septum, preoptic area, and hypothalamus. Wang and Swann Manzo et al., 2000). Furthermore, combining electrical stimu-
suggest that its lack of efferents to chemosensory processing lation with subthreshold doses of the DA agonist apomorphine
areas implies that it integrates the chemosensory and hormonal or the a2-adrenoceptor antagonist yohimbine was also ineffec-
signals and then relays that information to brain stem areas tive. Higher doses of both drugs had previously restored sexual
that control motor behavior. behavior in satiated males (Rodríguez-Manzo, 1999b). There-
fore, sexual satiety and postejaculatory quiescence may be
1.01.5.2.1.2 Effects of Lesions based on different neural mechanisms.
Large lesions of the MPOA abolish copulation in numerous Repeated electrical stimulation of the MPOA in previously
species ranging from monkeys to fish (reviewed in Hull and noncopulating male rats resulted in ‘kindling’ (increased after-
Dominguez, 2015). Similar effects are obtained with electro- discharges leading to a seizure) and the ability of most males to
lytic lesions, which destroy both cell bodies and axons passing copulate on subsequent stimulation-free tests (Paredes et al.,
through, and axon-sparing neurotoxic lesions; thus neurons in 1990). Males that could initially copulate without stimulation
the MPOA are critical for activating copulation. Generally, were not further facilitated. In another study, kindlinglike stim-
more severe deficits were found with more caudal lesions ulation that did not produce a seizure nevertheless facilitated
that included portions of the anterior hypothalamus. Smaller copulation for at least 8 months in previously noncopulating
lesions have produced less severe deficits. In gerbils small rats (Portillo et al., 2003). Even in T-treated female rats,
lesions of the PdPN, which is activated selectively by ejacula- MPOA kindling resulted in male-like sexual behavior
tion (Heeb and Yahr, 1996), decreased mounting and delayed (Dominguez-Salazar et al., 2003).
Male Sexual Behavior 27

Either electrical or glutamatergic stimulation of the MPOA copulation in almost all males (Butera and Czaja, 1989). While
of anesthetized rats increased intracavernous pressure E is the primary steroid for activating sexual behavior in male
(Giuliano et al., 1996; Sato et al., 2001) and elicited the UG rats, androgen in the MPOA does contribute to their sexual
reflex, even without urethral stimulation (Marson and behavior. The antiandrogen flutamide, administered into the
McKenna, 1994b). The effects of MPOA stimulation on intraca- anterior MPOA, impaired copulation, but not partner preference,
vernous pressure were enhanced by intrathecal injection of an whereas flutamide in the posterior MPOA impaired motivation,
NO donor, a cGMP analog, and sildenafil, and were inhibited but not copulatory performance (McGinnis et al., 2002).
by the NOS inhibitor L-NAME (Sato et al., 2001). Axons from Gonadally intact males that did not copulate when tested
the MPOA do not project directly to the lumbosacral spinal repeatedly received either E or T implants in the MPOA
cord, where erection and ejaculatory reflexes are controlled; (Antonio-Cabrera and Paredes, 2014). All noncopulating males
thus, efferents from the MPOA must stimulate downstream receiving T ejaculated consistently from week 11 after the
sites that then regulate the reflexes. However, the MPOA is implant until the end of testing at 5 months. All subjects
not necessary for genital reflexes, since MPOA lesions have implanted with E ejaculated from week 16 until the end of
minimal effect on noncontact erections (Liu et al., 1997b), testing. The noncopulating rats had been shown to have normal
reflexive erections (Szechtman et al., 1978), or spontaneous T and E plasma concentrations, but did have reduced
seminal emission (Ågmo et al., 1977). aromatase in the MPOA. Therefore, providing additional T or
E directly into the MPOA was sufficient to stimulate copulation.
1.01.5.2.1.4 Effects of Direct Application of Steroids or Steroid
Antagonists 1.01.5.2.1.5 Effects of Direct Application of Drugs Affecting
Numerous studies have shown facilitative effects of steroid Specific Neurotransmitters
implants in the MPOA of castrated rats, ferrets, birds, and Drugs administered into the MPOA influence a number of
lizards (reviewed in Hull and Dominguez, 2015). However, behavioral measures. Most of these effects resemble those of
these implants have not resulted in complete copulatory the same drug administered systemically, though there are
behavior, as other central and peripheral targets also require some site-specific differences.
hormonal priming. In castrated male rabbits, T implants
restored territorial marking (chin rubbing), but not copulation 1.01.5.2.1.5.1 Dopamine. Neurons of the periventricular DA
(Melo et al., 2008). Aromatization of T to E in the MPOA is system lie along the third ventricle and send branching axons
important for T’s facilitative effects. The aromatase inhibitor laterally into the MPOA and anterior hypothalamus (Moore
fadrozole, microinjected into the MPOA of gonadally intact and Lookingland, 1995). The classic D1/D2 agonist apomor-
male rats, inhibited copulation; only the MPOA contained phine, microinjected into the MPOA, facilitated copulation in
numerous unoccupied ERs as a result of the inhibitor, suggest- both intact and castrated male rats and increased the number
ing that the inhibitor had not spread to other sites (Clancy of ex copula reflexes; conversely, the D1/D2 DA antagonist cis-
et al., 1995). Conversely, implantation of E into the MPOA flupenthixol impaired copulation, reflexes, and sexual motiva-
of intact males treated systemically with fadrozole increased tion, measured as choice of the female in an X-maze (reviewed
MF, IF, and EF, compared to males with systemic fadrozole in Hull and Dominguez, 2015). There was both spatial and
but no E in the MPOA (Clancy et al., 2000). Therefore, E in behavioral specificity in the drug effects. MPOA injections of
the MPOA, together with androgens throughout the body, a different antagonist, haloperidol, decreased the number of
could partially restore male sexual behavior. The importance anticipatory level changes in search of a female in a bilevel
of aromatization for male sexual behavior is also seen in quail, apparatus and also impaired copulation (Pfaus and Phillips,
in which the effects of T implants in castrates correlated with 1991). Similarly, 6-OHDA lesions of the MPOA impaired
the induction of aromatase immunoreactivity (ARO-ir) in the copulation for at least 4 h after the injection, although
caudal POM (Balthazart and Ball, 2007). Similarly, lesions in increased synthesis in the remaining neurons restored both
the caudal POM of quail produced the most marked DA levels and sexual behavior within 24 h (Bazzett et al.,
copulatory deficits (Balthazart et al., 1998), and there was the 1992). The effect was specific to DA; levels of NE and
highest Fos-ir in response to copulation in the caudal POM epinephrine were not affected. In addition, apomorphine in
(reviewed in Balthazart and Ball, 2007). On the other hand, the MPOA fully restored copulatory ability of males whose
anterior POM lesions were associated with deficits in large excitotoxic lesions of the amygdala had severely
appetitive measures (Balthazart et al., 1998). Sexual impaired their ability to copulate (Dominguez et al., 2001).
preference in rams may also be affected by aromatization in D1- and D2-like receptors in the MPOA appear to have
the MPOA. Rams that preferred to mate with other males had different roles in the regulation of copulation. Stimulation of
lower levels of serum T and E, as well as decreased aromatase D1-like receptors facilitated copulation and reflexive erections,
activity in the MPOA, than did those that preferred to mate whereas stimulation of D2-like receptors produced dose-
with females (Resko et al., 1999). Also, sexually high- dependent effects (reviewed in Hull and Dominguez, 2015).
performing rams with ewes had more ERs and more A low dose of the D3/D2 agonist quinelorane decreased the
occupied ERs in the MPOA than did low-performing rams latency to the first ex copula reflex without affecting the numbers
(Alexander et al., 1993). of reflexes, suggesting a disinhibition of reflexes; however,
In contrast to E, DHT in the MPOA of castrated male rats is a high dose of quinelorane, or of the D1 antagonist SCH-
relatively ineffective, unless administered with subthreshold 23390, decreased erections but increased sympathetically
systemic E or DHT (reviewed in Hull and Dominguez, 2015). mediated seminal emissions (Bazzett et al., 1991). The D1
However, such implants in castrated guinea pigs restored agonist dihydroxyphenyl-tetrahydrothienopyridine (THP)
28 Male Sexual Behavior

increased reflexive erections and facilitated copulation but effects of 8-OH-DPAT, but the 5-HT1A antagonist did not, sug-
inhibited ex copula seminal emissions (Hull et al., 1995; gesting that elevation of extracellular DA levels resulted in stim-
Markowski et al., 1994). Experiments using low and high doses ulation of D2 receptors in the MPOA, which mediated much of
of apomorphine with D1- and D2-like antagonists also support the facilitation (Matuszewich et al., 1999). Microinjection of an
the suggestion that stimulation of D1-like receptors promotes SSRI (alaproclate) into the MPOA did not significantly affect
erections, while intense stimulation of D2-like receptors facili- behavior, although it did inhibit copulation when microin-
tates ejaculation (Hull et al., 1992). Similarly, in tests of copu- jected into the anterior lateral hypothalamus (LHA) (Lorrain
lation the D1-like agonist increased copulatory efficiency et al., 1997).
(Markowski et al., 1994), and the D1-like antagonist increased
intromission latency but decreased the threshold for ejacula- 1.01.5.2.1.5.3 Gamma-Aminobutyric Acid. High concentrations
tion (Hull et al., 1989). The antagonist also decreased sexual of GABA have been reported in the MPOA of male rat brains
motivation (choice of the female in an X-maze) (Moses and (Andersson, 2001) as well as in mating-activated neurons of
Hull, 1999). Similarly to the D1-like antagonist, a high dose this brain area and of the MeA in male gerbils (Simmons and
of the D3/D2 agonist quinelorane delayed the start of Yahr, 2003). Enhancement of GABAergic transmission in the
copulation, but decreased ejaculation threshold (Hull et al., MPOA by blocking GABA degradation or stimulating GABAA
1989; Moses et al., 1995). It is not clear whether the dose- receptors with muscimol reduced the proportion of copulating
dependent effects of quinelorane are mediated by different rats (Fernández-Guasti et al., 1986a). Inhibition of GABA
subtypes of D2-like receptors or by different populations of synthesis or blockade of GABAA receptors within the MPOA
neurons that are under different levels of tonic inhibition. had a potent stimulatory effect on male rat sexual behavior,
However, the data consistently support the hypothesis that expressed by a marked reduction of the PEI (Fernández-
stimulation of D1-like receptors increases parasympathetically Guasti et al., 1986a) and the virtual elimination of the USVs
mediated erections and the early phase of copulation, that accompany the absolute refractory period (Fernández-
whereas stimulation of D2-like receptors shifts the autonomic Guasti et al., 1986b). However, bicuculline did not restore
balance to favor sympathetically mediated ejaculation. This copulation in satiated male rats, suggesting that sexual satiety
hypothesis is supported by a report that the preferential D3 is regulated by mechanisms different from those that control
agonist 7-OH-DPAT, microinjected into the MPOA, elicited the PEI (Rodríguez-Manzo et al., 2000).
ejaculation-related events in anesthetized male rats (Kitrey
et al., 2007). In addition, the facilitative role of D1-like 1.01.5.2.1.5.4 Opioids. The lowest dose of the m agonist
receptors was supported by a demonstration that a D1 morphine and of the k agonist dynorphin (1–13), microin-
antagonist (SCH-23390), microinjected into the MPOA jected into the MPOA, facilitated copulation, but the highest
before each of seven exposures of a male rat to an estrous dose of morphine led to the failure to resume mating after
female placed over his cage, blocked the facilitative effects of the second ejaculation (Band and Hull, 1990). The m agonist
those exposures on a drug-free copulation test (McHenry morphiceptin increased the latency to copulate, but did not
et al., 2012). In that same report, other animals received affect copulatory performance, motivation, or locomotion
acute and/or chronic mating experience; those with both (Matuszewich et al., 1995). However, b-endorphin delayed
acute and chronic mating showed increased phosphorylation the start of mating and inhibited its performance (Holmes
of DA- and cAMP-regulated phosphoprotein (DARPP-32) in et al., 1997; van Furth et al., 1995). In support of facilitative
the MPOA, compared with all other groups. DARPP-32 is effects of low levels of opioids in the MPOA, the antagonist
a downstream marker of D1 receptor signaling and is naloxone in the MPOA prevented the induction of sexual rein-
a substrate of cAMP-dependent protein kinase (PKA). forcement (Ågmo and Paredes, 1988). A review by Paredes
Therefore, D1 receptors in the MPOA appear to contribute to (2014) summarizes both indirect evidence of opioid activity
experience-induced enhancement of male sexual behavior, during copulation and conditioned place preference (CPP)
perhaps through a PKA-regulated mechanism. studies that directly test the importance of opioids in the
In male European starlings microinjections of a D1 agonist MPOA for sexual reward in both males and females.
into the MPOA elicited sexually motivated, but not aggression-
motivated, song (Riters et al., 2014). However, there was an 1.01.5.2.1.5.5 Norepinephrine. Microinjection of NE into the
inverted U curve, with either very low or very high doses elicit- MPOA facilitated both sexual arousal and copulation, whereas
ing little singing. The effect of the D1 agonist was blocked by the a-adrenergic antagonist phenoxybenzamine and the
the D1 antagonist SCH-23390. Therefore, an optimal level of b-adrenergic antagonist propranolol inhibited mating (Mallick
D1 receptor stimulation is needed for sexually motivated vocal et al., 1996). Stimulating autoreceptors with the a2-agonist
production. clonidine, which would decrease NE levels in the MPOA,
impaired copulation, and inhibiting autoreceptors with the
1.01.5.2.1.5.2 Serotonin. Microinjection of large doses of a2-antagonist yohimbine blocked the inhibitory effects of
5-HT into the MPOA inhibited copulation (Fernández-Guasti systemic clonidine (Clark, 1991). Therefore, NE in the MPOA
et al., 1992; Verma et al., 1989), and a 5-HT1B agonist delayed may facilitate copulation.
ejaculation (Fernández-Guasti et al., 1992). As with systemic
injections of 5-HT1A agonists, reverse-dialysis of 8-OH-DPAT 1.01.5.2.1.5.6 Nitric Oxide. Reverse dialysis of the NO
into the MPOA facilitated copulation (Matuszewich et al., precursor L-arginine into the MPOA increased, and the NOS
1999) and increased both DA and 5-HT levels (Lorrain et al., inhibitor L-NMMA decreased the rate of mounting by male
1998). The D2 antagonist raclopride partially blocked the rats (Sato et al., 1998). L-NMMA also decreased the number
Male Sexual Behavior 29

of sexually naïve males that copulated and increased the 1.01.5.2.1.5.8 Orx/Hcrt, OT, and Other Neurotransmitters. Orx/
number of ex copula seminal emissions (Moses and Hull, hcrt is best known for its regulation of arousal and feeding.
1999; Moses et al., 1995). The NOS inhibitor L-NAME also pre- However, microinjections of Orx/hcrt into the MPOA of male
vented copulation in sexually naïve male rats and decreased rats also facilitated mating (Gulia et al., 2003). Microinjection
numbers of intromissions and ejaculations in experienced of OT into the MPOA also facilitated copulation, whereas
males (Lagoda et al., 2004). L-NAME microinjections before microinjection of an OT antagonist impaired copulation (Gil
each of seven exposures to an inaccessible estrous female et al., 2011). Furthermore, sexual experience increased both
blocked the facilitative effects of those exposures, as seen in OT receptor mRNA and protein levels in the MPOA (Gil
saline-injected males, compared to males not exposed to et al., 2013). Thus, a neuropeptide known for facilitating
a female (Lagoda et al., 2004). parental behavior, mating, and social trust also acts in the
NOS in the MPOA is hormonally regulated. In both rats MPOA to facilitate mating in male rats. In addition, there is
(Du and Hull, 1999; Putnam et al., 2005) and hamsters evidence for facilitative effects of ACh (Hull et al., 1988a,b)
(Hadeishi and Wood, 1996), castration decreased NOS-ir in and PGE2 (Clemens and Gladue, 1977) in the MPOA
the MPN. T replacement in castrated rats for 2, 5, or 10 days (reviewed in Hull et al., 2006).
increased NOS-ir and improved copulation; greater NOS-ir
density was correlated with shorter mount latencies (Sato
1.01.5.2.1.6 Electrophysiological Recordings
et al., 2005). There was also less NOS-ir in ERaKO mice,
Some neurons in the MPOA increased their firing rates only
compared to both wild-type mice and those lacking the AR
before male rats began to mate, and others increased their firing
due to the testicular feminization mutation (tfm)
only during copulation (Shimura et al., 1994). MPOA neurons
(Scordalakes et al., 2002). Therefore, ERs are more likely than
in monkeys also responded differentially during lever pressing
ARs to mediate the hormonal upregulation of NOS. nNOS is
to gain access to a female and during copulation; activity
colocalized with both ERa and AR in the MPOA of rats (Sato
decreased markedly after ejaculation (Oomura et al., 1988).
et al., 2005), mice (Scordalakes et al., 2002), and hamsters
In MPOA slices from male quail, bath applications of DA
(Hadeishi and Wood, 1996). Furthermore, improvement in
inhibited 52–80% of cells, but excited 10–25% (Cornil et al.,
copulation following T replacement correlated with density
2002). The inhibitory and excitatory effects were mediated by
of NOS-ir in the MPOA, but not the number of NOS-positive
a2 and a1 adrenoreceptors, respectively. Therefore, in the quail
neurons (Sato et al., 2005). Reverse dialysis of an NO donor
MPOA DA affects neural activity via cross-talk with NE recep-
(sodium nitroprusside) into the MPOA of castrated rats that
tors. In voltage-clamp recordings from dissociated neurons
were maintained on systemic DHT (to maintain genital and
from the MPOA of male rats, with presynaptic nerve endings
sensory structures) fully restored copulation in half the
intact, 5-HT inhibited both GABAergic miniature inhibitory
animals (Sato et al., 2007). NO’s effects are at least partially
postsynaptic currents (mIPSCs) and glutamatergic miniature
mediated by cGMP. A guanylyl cyclase inhibitor decreased
excitatory postsynaptic currents (mEPSCs) (Lee et al., 2008).
the ability of the NO donor to improve copulation and
The inhibition of mIPSCs was mediated by 5-HT1A receptors,
increased the MPOA DA release in gonadally intact male rats
whereas the inhibition of mEPSCs was mediated by 5-HT1B
(Sato and Hull, 2006). In contrast to the reports of
receptors. The authors suggest that stimulation of adenylyl
facilitative effects of hormones in mice, rats, and hamsters,
cyclase, and the consequent activation of PKA, increase mIPSCs
there is one report of increased NOS-ir after castration of rats
and mEPSCs and counteract the effects of 5-HT. Therefore, even
and a reduction following T or DHT replacement (Singh
though 5-HT did not increase in the MPOA at the time of ejac-
et al., 2000). The reason for the seeming contradiction may
ulation (Lorrain et al., 1997; see Section 1.01.5.2.5.3), and an
lie in the more posterior location examined by Singh et al. or
SSRI microinjected into the MPOA did not significantly impair
their use of an antibody that labeled very few cells. In
behavior (Lorrain et al., 1997; see above), 5-HT may influence
summary, NO in the MPOA facilitates copulation, and NOS
sexual behavior by reducing the effects of GABAergic and gluta-
is upregulated by T, probably via aromatization to E, in the
matergic transmission.
MPN, but not in more posterior areas.

1.01.5.2.1.5.7 Glutamate. Microinjection of glutamate into 1.01.5.2.1.7 Chemical Changes Detected by Microdialysis or from
the MPOA of anesthetized male rats elicited erectile Tissue Punches
responses (Giuliano et al., 1996) and also the UG reflex There is a close relationship between extracellular DA levels in
without genital stimulation (Marson and McKenna, the MPOA and male rat sexual behavior. DA levels rose when
1994b). Conversely, copulation was inhibited by microinjec- an inaccessible female was introduced and remained high or
tions of the NMDA antagonist MK-801 into the MPOA of increased further during copulation (Hull et al., 1995; Sato
sexually naïve or experienced males (Dominguez et al., et al., 1995). The recent presence of T was permissive for
2007). Nearly all neurons in the MPOA that expressed Fos both copulation and DA release, with two-thirds of 1-week
after mating also contained the NR1 subunit of NMDA recep- castrates able to copulate and to show a DA response to the
tors, and mating increased phosphorylation of NR1 in the female, while no 2-week castrate could copulate or show
MPOA, thereby activating the receptor. In addition, blocking a DA response (Hull et al., 1995). There was both anatomical
NMDA receptors decreased mating-induced Fos-ir and and behavioral specificity for the DA response (Hull et al.,
mating-induced phosphorylation of NMDA receptors. There- 1995, 1993). The fact that DA increased before copulation
fore, glutamate in the MPOA facilitates several measures of began suggests that the increase was not caused by copulation,
sexual behavior, at least in part through NMDA receptors. but was likely associated with sexual motivation. Not only is
30 Male Sexual Behavior

the DA response to the female lost after castration, but basal MPOA and arcuate nucleus, decreased serum PRL, and facili-
extracellular levels are also lower than in gonadally intact tated copulation (Yeh et al., 2008). Similarly, tissue levels of
males; however, intracellular levels were actually higher than DA in the MPOA and arcuate were higher in middle-aged rats
in intact males (Du et al., 1998). Therefore, synthesis and that could copulate to ejaculation, compared to middle-aged
storage of DA in the MPOA was at least as great in castrates males that could not copulate or that could mount and
as in intact males; the deficiency in castrates was in their ability intromit, but not ejaculate (Chen et al., 2007). Tissue levels of
to release their abundant stores. Restoration of T for 2, 5, or NE were not different among the groups.
10 days resulted in increasing sexual behavior and DA response Chemosensory stimuli processed by the olfactory bulbs
to the female (Putnam et al., 2001). No 2-day T-treated castrate provide the major signal relayed by the amygdala to the
could copulate or show a DA response; eight of nine 5-day MPOA of rodents. In male hamsters with sham bulbectomies,
T-treated castrates copulated and showed a DA response, with or with unilateral bulbectomy contralateral to the MPOA micro-
five of the eight able to ejaculate. All of the 10-day T-treated dialysis probe, presentation of an estrous female increased DA
castrates copulated and all showed the DA response. There levels, and animals copulated to ejaculation (Triemstra et al.,
were numerous correlations between DA levels and 2005). Males with ipsilateral bulbectomy also copulated to ejac-
copulatory measures. Therefore, both the loss of copulation ulation, but there was no female-stimulated DA response. Bilat-
following castration and its restoration by T are closely eral bulbectomy abolished both copulation and the DA
associated with the female-elicited DA response in the response to the female. Threfore, chemosensory stimuli are
MPOA. E and DHT were differentially effective in restoring essential for the MPOA DA response in male hamsters.
basal and female-stimulated DA release in long-term castrates Glutamate is also released in the MPOA of male rats during
(Putnam et al., 2003). E2 resulted in high basal DA levels but copulation (Dominguez et al., 2006a). Extracellular glutamate
no increase in response to a female; E2-treated castrates increased 170% during copulation and 300% in the 2-min
intromitted, but did not show an ejaculation pattern. DHT sample during which the male ejaculated. Glutamate fell
was no more effective than the oil vehicle in restoring dramatically after ejaculation, and the decrease was highly
copulation, basal, and female-stimulated DA levels. However, correlated with the length of the PEI. Reverse dialysis of a gluta-
the combination of E2 and DHT fully restored both basal and mate reuptake inhibitor increased levels to 280%, increased
female-stimulated DA levels, as well as copulation. In the number of ejaculations, and decreased the latencies to ejac-
contrast to the positive correlations between extracellular DA ulate and to resume copulation after the PEI. In a later study,
and behavior, tissue (stored) DA levels were negatively reverse dialysis of 5-HT into the MPOA decreased glutamate
correlated with mating, suggesting that tissue DA levels were levels, decreased the number of ejaculations, and increased
high because it could not be released (Putnam et al., 2005). ejaculation latencies and PEIs (Dominguez and Hull, 2010).
A similar relation between apparent DA release and ability to The 5-HT metabolite 5-hydroxyindole acetic acid (5-HIAA)
copulate was reported in male hamsters (Schulz et al., 2003). was increased in the POA of male rats following ejaculation
Adult males had an increase in the major metabolite DOPAC (Fumero et al., 1994; Mas et al., 1995a), and 5-HT was
in punches from the MPOA in response to VSs from female increased in POA tissue punches after ejaculation (Mas et al.,
hamsters. The increase in DOPAC is taken as evidence that 1987). The authors suggested that increased 5-HT may have
DA was released, transported back into the axon terminal, contributed to the PEI. However, a more recent microdialysis
and metabolized. Juveniles showed no such increase and also study that measured 5-HT itself found that 5-HT in the
were unable to copulate. MPOA and the more lateral POA remained constant during
DA release in the MPOA is regulated by NO. Both basal copulation, ejaculation, and the PEI (Lorrain et al., 1997).
(Lorrain and Hull, 1993) and copulation-induced (Lorrain However, 5-HT did increase in the LHA in the sample during
et al., 1996) extracellular DA levels were dependent on NOS which the male ejaculated. The 5-HIAA increase in the earlier
activity. As noted above, T and E positively regulate NOS-ir in studies may have diffused from the LHA to the adjacent POA.
the MPOA. Thus, gonadal steroids may maintain DA levels in Thus, 5-HT in the POA may not contribute significantly to
the MPOA by upregulating NOS. the PEI. However, see below for a microdialysis study showing
A major stimulus for the DA response to a female is input that reverse dialysis of 5-HT into the MPOA decreases gluta-
from the MeA. Small radiofreqency lesions of the MeA impaired mate release and number of ejaculations and increases ejacula-
copulation and abolished the MPOA DA response to the female tion latency and PEI (Dominguez and Hull, 2010). Two
but did not affect basal DA levels in the MPOA (Dominguez resolutions to this apparent contradiction may be that 5-HT
et al., 2001). Thus, as with E2 restoration of copulation in levels during reverse dialysis are higher than during copulation
castrates (Putnam et al., 2003), basal DA levels in the MPOA and the PEI or that a small increase in 5-HT during the PEI was
were sufficient for suboptimal mating ability, but an additional not detectable by microdialysis.
increase in response to a female is required for optimal copula-
tion. As noted above, microinjections of apomorphine into the 1.01.5.2.1.8 Activation of c-Fos or Other Measures of Neural
MPOA completely restored copulation in males with large exci- Activity
totoxic lesions of the amygdala (Dominguez et al., 2001), Copulation increases Fos expression in the MPOA of male
providing additional evidence that a major way in which the rats (Baum and Everitt, 1992; Robertson et al., 1991; Veening
MeA facilitates sexual behavior is by increasing extracellular and Coolen, 1998), gerbils (Heeb and Yahr, 1996), hamsters
DA in the MPOA in response to a female. In agreement with (Fernandez-Fewell and Meredith, 1994; Kollack and
a facilitative effect of DA, chronic systemic administration of Newman, 1992; Kollack-Walker and Newman, 1995), and
an extract of Ginkgo biloba increased tissue DA levels in the mice (Halem et al., 1999). Sexual experience may enhance
Male Sexual Behavior 31

future responsiveness to sexual stimuli; there were more Fos- expressing neurons correlated with levels of sexual experience
ir neurons in the MPN of sexually experienced male rats after and sexual activity (Nutsch et al., 2016).
one ejaculation than in previously naïve males, even though Some B6D2F1 mice are able to copulate after castration
the naïve males required more intromissions to trigger (McInnis et al., 2016). The ability to copulate without T is
the ejaculation (Lumley and Hull, 1999). Similarly, passed from father to son and may be related to increased
chemosensory cues in sexually experienced male hamsters production of amyloid precursor protein, tau, and synaptophy-
stimulated greater Fos expression than in sexually naïve sin in the MPOA. The mechanisms by which these proteins may
males (Fewell and Meredith, 2002). Fos expression was affect mating behavior are not clear.
also increased in the MPN and BST of Japanese quail by
both visual and physical contact with females (Iyilikci 1.01.5.2.1.9 Effects of Intracerebral Grafts
et al., 2014). Injection of fetal MPOA neurons into the MPOA of aging male
Copulation-induced Fos-ir in the MPOA can be elicited by rats improved their ability to copulate between 21 and 45 days
input from either the MeA or the CTF (Baum and Everitt, after implantation; the benefits lasted until the end of the exper-
1992), and Fos-ir neurons in the MPOA project to the iment, 4.5 months later (Hung et al., 1997). Serum T was also
PAG (Struthers, 2001) and to the BST (Maejima et al., 2015). increased. Implants from other brain areas were ineffective.
Sexual behavior was also associated with an increase in
2-deoxyglucose incorporation in the MPOA of birds (Dermon 1.01.5.2.1.10 Activation of Neurotransmitter Receptors
et al., 1999) and increased cytochrome oxidase activity in lizards Endocytosis of m opioid receptors occurred within 30 min after
(Sakata and Crews, 2004). copulation and lasted for at least 6 h (Coolen et al., 2004b).
There is disagreement as to whether chemosensory cues are The opioid antagonist naloxone prevented receptor internaliza-
sufficient to induce Fos expression in the MPOA. There have tion. Microinjection of a m agonist elicited similar internaliza-
been positive reports of such induction in rats (Bressler and tion, and mating elicited Fos-ir in m receptor-containing
Baum, 1996; Kelliher et al., 1999), hamsters (Fiber et al., neurons. However, naloxone did not prevent the mating-
1993; Kollack-Walker and Newman, 1997), macaques induced Fos expression in m receptor-containing neurons, sug-
(Michael et al., 1999), gerbils (Heeb and Yahr, 1996), and gesting that the mating-induced Fos response did not result
ArKO mice (Aste et al., 2003). It is not clear whether the from stimulation of m receptors.
main or accessory olfactory system mediates the Fos responses
to chemosensory cues (Fewell and Meredith, 2002; Swann 1.01.5.2.1.11 Summary of MPOA Functional Roles
et al., 2001). Similarly, fMRI indicated pheromone-induced The MPOA receives indirect sensory input from all senses and
arousal of the MPOA in marmosets (Ferris et al., 2001), and has reciprocal connections with the sources of afferent input,
temperature fluctuations in the MPOA of rats occurred in allowing hormone-concentrating neurons to bias the processing
response to odor cues (Kiyatkin and Mitchum, 2003). Fos of sexually relevant input. MPOA efferents to other hypotha-
was also elicited in the MPOA of male hamsters by electrical lamic, midbrain, and brain stem nuclei regulate somatomotor
stimulation of the VNO (Meredith and Fewell, 2001). patterns and genital reflexes and contribute to sexual motivation.
However, there have also been negative reports of Fos induc-
tion in rats (Baum and Everitt, 1992; Coolen et al., 1997b), 1.01.5.2.2 Mesocorticolimbic DA Tract
mice (Halem et al., 1999), hamsters (Fernandez-Fewell and 1.01.5.2.2.1 Anatomy
Meredith, 1994), and ferrets (Kelliher et al., 1998). Ejaculation The mesocorticolimbic circuit comprises DA cell bodies in
selectively activated the MPN of hamsters (Kollack-Walker the VTA ascending to the NAc and the medial prefrontal
and Newman, 1997) and rats (Coolen et al., 1997b), and the cortex (mPFC) and plays a central role in motivated behav-
PdPN of gerbils (Heeb and Yahr, 1996). iors (Alcaro et al., 2007). Electrical stimulation studies
The neurochemical identity of Fos-ir neurons has been have implicated this system in positive rewarding states
addressed. In gerbils half of the Fos-ir neurons in the medial (Wise and Rompre, 1989) and appetitive-motivated behav-
SDA and PdPN were GABAergic; almost one-fourth of the iors (Berridge and Robinson, 1998). Sexual behavior is
neurons in the medial SDA were glutamatergic, but there a rewarding and reinforcing behavior that activates the mes-
were no glutamatergic cells in the PdPN (Simmons and Yahr, ocorticolimbic circuit (Balfour et al., 2006). Hence, DA efflux
2003). In rats galanin-containing cells are selectively activated is increased in the NAc in response to the presence of an
by ejaculation (Bakker et al., 2002b). Ejaculation-induced estrous female behind a barrier, as well as during copulation
Fos-ir in the MPOA of male rats was decreased by administra- (Damsma et al., 1992; Fiorino et al., 1997; Pfaus et al., 1990;
tion of a D1 antagonist (Lumley and Hull, 1999), suggesting Robinson et al., 2002). The VTA and the NAc are activated
that stimulation of D1 receptors mediated at least some of during sexual arousal (Guevara et al., 2008). Recording of
the copulation-induced Fos-ir. Copulation increased Fos-ir in male rat NAc neuronal activity during sexual behavior
AR-containing neurons in the MPOA of male rats (Greco, showed that many NAc shell neurons exhibited firing rate
1998a), but not in aromatase-containing neurons in male changes during sexual activity that were related to sexual
quail (Foidart et al., 1999). Copulation also increased behavior performance, its reward-related processing, and
expression of Fos in NOS-containing neurons in the MPOA the sexual behavior inhibition that follows ejaculation. These
of male rats (Nutsch et al., 2014). As mentioned above, NO data suggest that the NAc shell is importantly involved in
is important for MPOA DA release and copulation. Previous encoding distinct aspects of male sexual behavior
sexual experience also increased the coexpression of Fos and (Matsumoto et al., 2012). In the VTA, sexual behavior acti-
D2-like receptors in the MPOA, and numbers of D2- and Fos- vates both DA and non-DA neurons. VTA activation appears
32 Male Sexual Behavior

to be mediated by endogenous opioids released during 1.01.5.2.2.2 Effects of Lesions or Electrical Stimulation
sexual activity, which inhibit VTA GABAergic interneurons, Lesion of the mPFC disrupted male rat sexual behavior, partic-
thereby releasing DA cells from tonic inhibition (Balfour ularly its initiation (Ågmo et al., 1995). VTA lesion increased
et al., 2004). Actually, ejaculation induced m and d opioid the duration of the PEI (Brackett et al., 1986), while NAc
receptor internalization (an indicator of ligand-induced lesions impaired both copulation and noncontact erections,
receptor activation) in neurons of the VTA, and a correlation suggesting that the NAc plays an excitatory role in the regula-
between the amount of ejaculatory activity displayed and the tion of sexual arousal (Kippin et al., 2004). Electrical stimula-
degree of in vivo m opioid receptor internalization could be tion of the VTA facilitated copulation (Markowski and Hull,
established (Garduno-Gutierrez et al., 2013c). The mPFC 1995); however, the facilitative effect is restricted to its dorsal
receives DA inputs from the VTA and sends projections portion, and stimulation of its ventral portion inhibited mating
back to this structure (Tzschentke, 2000). Projections from (Rodríguez-Manzo and Pellicer, 2007). Electrical stimulation
the mPFC to the VTA are largely glutamatergic; thus, it is of the NAc also facilitated sexual behavior display in sexually
possible that mPFC inputs contribute to excitation of VTA experienced rats (Figure 4), but lacked an effect when the
neurons via glutamate release during copulation (Balfour same animals were sexually exhausted (Rodriguez-Manzo and
et al., 2006). Efferent projections from the mPFC also contact Pellicer, 2010).
other brain areas involved in sexual behavior and motiva-
tion, including the NAc shell and core, the MPOA, BST, 1.01.5.2.2.3 Activation of c-Fos or Receptors
and SPF. Fos expression is induced in the NAc and VTA of male rats in
Functional neuroimaging studies in humans showed that response to mating (Robertson et al., 1991; Balfour et al.,
the NAc is likely to participate in the motivational component 2004). Female odors and conditioned nonsexual odors paired
of male sexual arousal and is activated during erection evoked with mating-stimulated Fos expression in NAc (Kippin et al.,
by visual erotic stimulation (Redoute et al., 2000), while gray 2003). Moreover, sexual experience enhanced estrous female-
matter atrophy of the NAc was related to psychogenic erectile induced NAc Fos-ir (Lopez and Ettenberg, 2002a). mPFC is
dysfunction in men (Cera et al., 2012). also activated during sexual behavior and sends projections to

Figure 4 Percentage of sexually experienced male rats achieving one to four successive ejaculations (EJAC) within a 30-min period when electri-
cally stimulated with high- (100 Hz) or low-frequency (20 Hz) in the nucleus accumbens (dashed columns) or caudate-putamen nucleus (double
dashed columns). Fisher exact probability test, þ, p ¼ 0.054; *, p < 0.05; **, p < 0.01; ***, p < 0.001. Ventral striatum stimulation facilitated, while
dorsal striatum stimulation inhibited, copulation, regardless of the stimulation frequency used. Reproduced from Rodriguez-Manzo, G., Pellicer, F.,
2010. Electrical stimulation of dorsal and ventral striatum differentially alters the copulatory behavior of male rats. Behav. Neurosci. 124, 686–694,
adapted with permission from APA.
Male Sexual Behavior 33

sex-activated neurons in the VTA, as determined by Fos-ir (Balfour orexin-A and orexin-B was reported to increase DA release in
et al., 2006). Mating increased Fos-ir in the dopaminergic neurons the NAc (Narita et al., 2006) and, at specific doses, to augment
of the VTA that were apposed to orexin fibers, suggesting that dopaminergic cell firing (Muschamp et al., 2007). Using rat
orexin promotes sexual behavior by activating the mesocortico- midbrain slices, it was demonstrated that orexin-B enhances
limbic circuit (Hull, 2011). Sexual behavior was shown to cause glutamatergic transmission in the VTA (Borgland et al., 2008).
DFosB (a stable transcription factor of the Fos family related to
enhanced motivation and reward induced by drugs of abuse) 1.01.5.2.3 Nigrostriatal DA Tract
accumulation in the NAc. Overexpression of DFosB in this brain The nigrostriatal pathway originates in the zona compacta of
region facilitated sexual behavior in naïve male rats which the substantia nigra and sends its projections to the caudate
behaved more like experienced animals, but had no effect in and putamen nuclei of the striatum. The nigrostriatal DA
already experienced males (Wallace et al., 2008) DFosB accumu- system has been implicated in the control of procedural
lation was also found to occur in the mPFC and VTA as a result of aspects of movement and motivated behaviors, as it reaches
sexual experience (Pitchers et al., 2010). Besides, sexual activity dorsal areas of the basal ganglia (Alcaro et al., 2007). Mating
causes long-term changes in glutamate NMDA and AMPA induces an increase in DA concentrations both in ventral and
receptor expression and function in the NAc (Pitchers et al., dorsal striatum, but DA is released in the dorsal striatum only
2012). after the male begins to copulate, suggesting that this release
reflects motor activation, rather than motivational aspects of
1.01.5.2.2.4 Microinjections of Drugs copulation (Damsma et al., 1992). Electrical stimulation of
Microinjection of D1- and D2-like receptor antagonists into the the dorsal striatum (caudate putamen) had an inhibitory
NAc decreased sexual motivation (Pfaus and Phillips, 1991). influence on copulation of sexually competent male rats, con-
Apomorphine, a nonselective DA agonist, microinjected into sisting of slowing its display and decreasing the ejaculatory
the VTA, delayed the onset of copulation and slowed its rate, frequency, in contrast to ventral striatum stimulation, which
presumably by stimulating autoreceptors and thereby decreasing accelerated copulation (Rodriguez-Manzo and Pellicer,
dopaminergic activity (Hull et al., 1990). Conversely, the nonse- 2010; Figure 4). Bilateral lesions of the substantia nigra also
lective DA antagonist cis-flupenthixol reduced IL for those rats slowed the rate of copulation and decreased ejaculatory
that copulated, presumably by blocking autoreceptors, but also capacity (Brackett et al., 1986). Copulation did not induce
decreased the proportion of copulating rats, perhaps due to depo- Fos-ir in the dorsal striatum, in contrast to mesolimbic areas
larization block. These effects appeared to be due to motoric slow- such as the NAc, VTA, and mPFC (Coolen et al., 1996);
ing, not a decrease in specifically sexual motivation or genital however, DFosB accumulated in the caudate putamen, as
reflexes (Hull et al., 1991). D1 agonist injection into the NAc well as in the NAc and VTA, as a result of sexual experience
augmented the percentage of sexually naïve male rats copulating (Pitchers et al., 2010). Apomorphine injected into the stria-
during their first sexual encounter (Bialy et al., 2010). Infusion of tum did not affect copulation in male rats (Hull et al.,
5-HT into the NAc impairs copulation in rats, while a facilitative 1986), and bilateral haloperidol infusions only increased EF
effect was obtained after 8-OH-DPAT (Fernández-Guasti et al., (Pfaus and Phillips, 1991).
1992). Intra-VTA microinjection of morphine and dynorphin It is important to call attention to the fact that anatomical
facilitated male sexual behavior and increased DA transmission and neurochemical studies suggest that nigrostriatal and meso-
in the NAc (Mitchell and Stewart, 1990), both effects probably limbic dopaminergic systems are involved in the organization
resulting from m opioid receptor-mediated release from of many if not all motivated behaviors including sexual
GABAergic tonic inhibition (Balfour et al., 2004). Conversely, behavior (Alcaro et al., 2007). No clear boundaries can be
the opioid receptor antagonist naltrexone microinjected into established between these two systems, since VTA and substan-
the VTA inhibited ejaculation in sexually experienced rats, but tia nigra dopaminergic neurons partly overlap in their projec-
facilitated sexual behavior expression in sexually satiated males. tion fields (Nauta et al., 1978), while copulation-related cues
This result suggests that endogenous opioids within the VTA and mating simultaneously activate nigrostriatal and mesolim-
take part in the maintenance of the sexual inhibition that follows bic circuits (Robinson et al., 2002). Thus, motor, motivational,
sexual exhaustion, evidencing the participation of the mesocorti- and reward functions of these two dopaminergic systems func-
colimbic system in this phenomenon. The reversal of sexual tion in a unified way (Nicola, 2007; Robbins and Everitt, 2007)
exhaustion was due to blockade of m opioid receptors, since and might in the future be integrated in a single dopaminergic
intra-VTA infusion of the d opioid receptor antagonist naltrindole circuit.
was ineffective (Garduno-Gutierrez et al., 2013b).
It was shown that direct administration of phosphodies- 1.01.5.2.4 Paraventricular Nucleus of the Hypothalamus
terase type 5 inhibitors in the caudal VTA facilitates noncontact 1.01.5.2.4.1 Anatomy
penile erections and increases DA levels in the NAc, suggesting The PVN is an important integrative site for endocrine and
that the increase in sexual arousal induced by these drugs autonomic functions. It comprises a parvocellular division,
involves the mesolimbic circuit (Sanna et al., 2009). Similarly, which projects to several brain areas and the spinal cord, and
OT injected into the VTA and other brain regions induces a magnocellular division, which releases OT and vasopressin
penile erection (Melis and Argiolas, 2011). In particular, penile from the posterior pituitary (reviewed in Swanson and
erection induced by OT infusion into the hippocampus is Sawchenko, 1980). Axons that project to the spinal cord release
mediated by an increase in glutamate levels in the VTA, which OT, vasopressin, somatostatin, DA, and other, undetermined
in turn increase DA levels within the mesocorticolimbic system transmitters. Direct projections from the PVN to the motoneu-
(NAc and mPFC) (Succu et al., 2011). Intra-VTA injection of rons of the dorsomedial nucleus of the lumbosacral cord,
34 Male Sexual Behavior

which innervate the levator ani and BS muscles mediating erec- stimulated by direct infusion of OT into the parvocellular
tion and ejaculation, have also been identified (Dobberfuhl division of the PVN (Kita et al., 2006). D2-like receptors are
et al., 2014). Periventricular DA neurons and brain stem NE coexpressed by OT neurons of the PVN, and it has been
and 5-HT nuclei provide input to the parvocellular PVN. The suggested that the OT neurons projecting to the spinal cord
PVN integrates afferent input of multiple sources as well as respond to DA to induce penile erection (Baskerville et al.,
efferent autonomic output, the latter with the participation of 2009). Besides, PVN apomorphine acts via OT projections to
both glutamate and GABA interneurons that play a role in the VTA to increase DA release in the NAc and elicit erections
controlling the excitability of magnocellular and parvocellular (Melis et al., 2007).
output neurons (Ferguson et al., 2008).
OT is present in neurons whose cell bodies are located in the 1.01.5.2.4.3.2 Nitric Oxide. Microinjection of NO donors or
PVN and project to the neurohypophysis as well as in neurons a high dose of the NO precursor L-arginine into the PVN eli-
that project from the PVN to extrahypothalamic brain regions. cited erections, whereas the NOS inhibitor L-NAME decreased
The PVN is the most sensitive brain area to OT -induced penile noncontact erections and impaired mating (Melis et al.,
erection in male rats. Actually, all other brain areas in which 1998). Reverse dialysis of another NOS inhibitor, L-NMMA,
local OT injection was reported to induce penile erection decreased reflexive erections, and L-arginine increased erec-
receive oxytocinergic projections from the PVN. DA, glutamate, tions; however, neither drug affected copulation (Sato et al.,
and NO can facilitate penile erection and copulation by stimu- 1999). Those authors reported that similar administration
lating OTergic neurons of the PVN, while GABA, opioid of L-NMMA into the MPOA did impair copulation. Thus,
peptides, and endocannabinoids inhibit those neurons NO-related drugs have consistently affected noncontact and
(Argiolas and Melis, 2013). reflexive erections, but have had inconsistent effects on
mating.
1.01.5.2.4.2 Effects of Electrolytic or Cell Body Lesions
Excitotoxic lesions of the parvocellular PVN decreased noncon- 1.01.5.2.4.3.3 Amino Acids. Microinjections of the glutamate
tact erections without affecting copulation (Liu et al., 1997a). agonist NMDA elicited erections, an effect blocked by an
Similar lesions decreased the volume of semen ejaculated NMDA antagonist and a NOS inhibitor in the PVN (Melis
and decreased the OT-ir innervation of the lumbosacral et al., 1997) and also by an OT antagonist injected icv, but
spinal cord, but again did not affect mating (Ackerman et al., not into the PVN (Melis et al., 2000b). The NMDA antagonist
1997). Larger lesions of both the parvo- and magnocellular impaired both noncontact erections and copulation when
divisions inhibited both reflexive and noncontact erections administered alone (Melis et al., 2005). NMDA in the PVN
and led to some impairment of mating (Liu et al., 1997a). also increased intracavernosal pressure in awake and anesthe-
However, electrolytic lesions actually speeded the first tized male rats (Chen and Chang, 2003; Zahran et al., 2000).
reflexive erection (Monaghan et al., 1993). Thus, the PVN Conversely, a GABAA, but not a GABAB, agonist in the PVN
contributes to both noncontact and reflexive erections and inhibited erections elicited by apomorphine, OT, or NMDA
seminal emission, but does not consistently affect copulation (Melis et al., 2000a).
or reflexive erections. Lateral parvocellular PVN lesions
destroyed neurophysin-ir axons to the sexually dimorphic 1.01.5.2.4.3.4 Other Transmitters. Morphine microinjections
SNB (Wagner and Clemens, 1993). Neurophysin is a marker before introduction of an estrous female prevented both
for OT and vasopressin; thus, the PVN is the source of OT noncontact erections and the rise in NO that occurred in
projections to the SNB that promote erection, seminal control rats (see below) (Melis et al., 1999b). Hexarelin analog
emission, and ejaculation. peptides, which were originally known for their ability to
induce growth hormone release, activate specific receptors on
1.01.5.2.4.3 Effects of Direct Applications of Drugs Affecting OT neurons in the PVN to admit calcium and thereby stimulate
Specific Transmitters NOS in the OT neurons and elicit erections (reviewed in
1.01.5.2.4.3.1 DA and OT. Microinjections of mixed and selec- Argiolas and Melis, 2005). A cannabinoid CB1 antagonist,
tive D2-like agonists or of OT elicited drug-induced erections microinjected into the PVN, elicited erections, an effect reduced
and increased reflexive erections and seminal emissions by CB1 agonists, an NMDA antagonist, and a NOS inhibitor in
(reviewed in Argiolas and Melis, 2005). The mixed DA agonist the PVN, and by an icv OT antagonist (Melis et al., 2004b). The
apomorphine also increased intracavernous pressure in anes- CB1 receptors in the PVN are mostly on GABAergic terminals,
thetized rats (Allard et al., 2002). The D2-like receptor has which in turn impinge on both OT and glutamatergic neurons
more recently been hypothesized to be the D4 receptor, which (Castelli et al., 2007). The authors note that one would expect
apparently opens N-type calcium channels; the calcium then inhibition of the inhibitory CB1 receptors to increase GABA
activates NOS in the OT neurons (Melis et al., 2005, and release; however, they suggest that stimulation of CB1 receptors
references therein). The noncontact erections elicited by DA may inhibit GABA reuptake in the PVN; therefore, blocking CB1
agonists were inhibited by an OT antagonist administered receptors would increase GABA reuptake and disinhibit gluta-
icv, but not into the PVN, suggesting that DA excites neurons matergic and OT neurons. Thus, the mechanism of CB1 antag-
that release OT elsewhere (Melis et al., 1999a). OT released onists’ effects is not clear. Pro-VGF-derived peptides are
in the PVN itself apparently acts via OT receptors on OT cleavage products of VGF, the product of the vgf gene, which
neurons by increasing calcium influx, thereby stimulating is selectively induced by nerve growth factor (see references
NOS, which acts intracellularly to stimulate OT neurons in Argiolas and Melis, 2005). They may activate OT neurons
(reviewed in Argiolas and Melis, 2005). Penile erection is in the PVN and thereby facilitate erections; their effects are
Male Sexual Behavior 35

inhibited by both NOS and OT antagonists (reviewed in sexual behavior deterioration (Kobayashi et al., 2015).
Argiolas and Melis, 2005). Sexually competent male rats had more OT mRNA and less
mRNA for proenkephalin and prodynorphin in the PVN than
1.01.5.2.4.4 Chemical Changes Detected by Microdialysis did impotent males (Arletti et al., 1997). Furthermore, NOS
DA levels increased during noncontact erections and, to and OT were colocalized (Yamada et al., 1996).
a greater extent, copulation (Melis et al., 2003, 2004b). Both In addition to projections to the hippocampus, lumbosacral
noncontact erections and copulation were accompanied by spinal cord, and other brain areas, there is a direct projection to
an increase in NO, inferred from increases in NO2 and NO3 the nPGi, where terminals form close appositions to seroto-
(Melis et al., 1998). In the same study, L-NAME in the PVN nergic neurons that project to the lumbosacral cord and inhibit
inhibited both the increase in NO2 and noncontact erections; genital reflexes (Bancila et al., 2002).
however, PVN injections of hemoglobin, an NO scavenger,
blocked the increase in NO2, but not noncontact erections, sug- 1.01.5.2.5 Lateral Hypothalamus
gesting that NO works only intracellularly in the PVN. Dopa- 1.01.5.2.5.1 Anatomy
mine agonists of the D2-like receptor family injected into the The LH is reciprocally connected with brain regions that
PVN increase NO production, apparently with the D2 receptor process emotional information. It may control the autonomic
subtype playing a major role in this effect (Sanna et al., 2012). nervous system via connections to the nucleus of the solitary
Morphine microinjections into the PVN inhibited the tract and parabrachial nucleus, endocrine secretion via those
copulation-induced increase in NO and copulation (Melis to the PVN, and emotional responses via connections to PAG
et al., 1999b). Reverse dialysis of L-arginine increased both and midbrain extrapyramidal area. Activation of this system
reflexive erections and the NO increase, and L-NMMA inhibited leads to both motor arousal and emotional/cognitive arousal
both (Sato et al., 1999). Apomorphine or a D2 agonist, but not (Ikemoto, 2007).
a D1 agonist, increased NO production and erections (Melis
et al., 1996), as did NMDA (Melis et al., 1997). Omega cono- 1.01.5.2.5.2 Effects of Electrical Stimulation and Lesions
toxin, an inhibitor of N-type calcium channels, prevented the LH electrical stimulation has rewarding effects and has induced
elicitation of erections and NO production by apomorphine stimulation-bound copulation in male rats (Huston, 1971),
and OT; NO donors overcame the need for calcium channel while males with lesions in the anterior LH intromitted repeat-
activation (Succu et al., 1998). Microinjection of a CB1 edly, but few copulated to ejaculation (Kippin et al., 2004).
antagonist increased extracellular glutamate (Succu et al., During exposure to an inaccessible receptive female, anterior
2006b) and NO production (Melis et al., 2006), whereas LH-lesioned males had increased noncontact erections. Thus,
microinjection of morphine decreased both erections elicited the anterior LH may inhibit sexual arousal but facilitate
by a CB1 antagonist and the increases in glutamate and NO ejaculation. On the other hand, orexin cell-specific lesion in
production (Succu et al., 2006a). the LH did not affect sexual behavior expression, but
prevented formation of a CPP for sexual behavior, suggesting
1.01.5.2.4.5 Immunocytochemistry that orexin is not essential for male sexual behavior display,
30% of lateral parvocellular neurons in the PVN contain ER-ir, but is important for copulation-related reward processing
and almost half of them project to the lumbar spinal cord (Di Sebastiano et al., 2011).
(Wagner et al., 1993). Therefore, some hormonal effects on
the SNB may be indirect, via steroid-sensitive afferents. 1.01.5.2.5.3 Microinjections, Microdialysis, and Fos-ir
Ejaculation induced greater Fos expression in the parvocel- Microinjection of an SSRI into the anterior LH delayed the onset
lular and magnocellular PVN than did intromission, especially of copulation and increased the EL (Lorrain et al., 1997), while
in the caudal lateral parvocellular PVN, with one-third of the reverse dialysis of 5-HT decreased basal and female-elicited DA
Fos-ir neurons containing OT (Witt and Insel, 1994). Neurons release in the NAc (Lorrain et al., 1999). In addition, anterior
in that area project to the brain stem and lumbar spinal cord. LH 5-HT levels were increased at the time of ejaculation, coin-
Furthermore, female odor elicited Fos-ir in OT-containing cident with the copulatory refractory period (Lorrain et al.,
neurons in the parvocellular PVN of sexually experienced, but 1997). The LH contains orx/hcrt neurons, which appear to
not naïve male rats; direct exposure to an anesthetized play a critical role in arousal and reward (Harris and Aston-
estrous female elicited Fos-ir in both groups, compared to Jones, 2006) and are activated following copulation
nonexposed controls (Nishitani et al., 2004). However, in (Muschamp et al., 2007). It has been proposed that 5-HT in
gerbils neither mating nor exposure to a previous mating the anterior LH might inhibit male sexual behavior by inhibit-
arena elicited Fos-ir in the PVN (Heeb and Yahr, 1996). The ing orx/hcrt neurons, which would eliminate their facilitative
PVN has been implicated in the deterioration of male sexual influence on VTA DA cell firing (Muschamp et al., 2007).
behavior produced by the alarm pheromone, since the Measurement of brain perfusion in healthy men during
copulatory decline is accompanied by an increase in Fos sexual activity with the aid of fMRI showed increased activity-
expression in the parvocellular division and a decrease in the related blood flow in the LH that correlated with penile erec-
magnocellular division of the PVN. Naloxone pretreatment tion (Georgiadis et al., 2010).
blocked sexual behavior decline and magnocellular PVN Fos
expression changes. The authors suggest that pheromone 1.01.5.2.6 Ventromedial Hypothalamus
detection by the olfactory system activates the parvocellular 1.01.5.2.6.1 Anatomy
PVN, which in turn activates opioidergic neurons that Although the VMH is primarily known for its role in female
suppress magnocellular PVN activation, thus contributing to receptive behavior, it may also influence male sexual behavior.
36 Male Sexual Behavior

It has numerous ERs and ARs (Simerly et al., 1990; Wood et al., represent a unique site for the integration of sexual odorant
1992) and receives both genitosensory (Coolen et al., 2003) cues with leptin-mediated signals of energy balance to
and chemosensory (Canteras et al., 1995; Coolen and Wood, regulate reproduction (Leshan et al., 2009).
1998; Gomez and Newman, 1992) information. Copulation
to satiety reduces AR density in the VMH of male rats and 1.01.5.3.2 Midbrain Periaqueductal Gray
mandarin voles (Fernández-Guasti et al., 2003; He et al., 2013). The ventrolateral portion of the PAG (vlPAG) contains neurons
that receive input from the MPOA (Murphy and Hoffman,
1.01.5.2.6.2 Effects of Lesions and Hormonal Manipulations 2001) and project to the nPGi. These neurons also receive input
Electrolytic microlesions in the dorsomedial VMH impaired the from the lumbosacral spinal cord (Normandin and Murphy,
ability of T implants to restore USVs and scent marking, but 2008). Sexual behavior induces extensive FOS expression in
had relatively little effect on mating (Harding and McGinnis, the PAG; however, very little FOS is observed in the PAG output
2005). T implants in VMH failed to restore copulation or neurons to the nPGi, suggesting that these neurons may be
USVs in rats (Harding and McGinnis, 2003) or mice (Nyby inhibited during mating. Since the nPGi exerts a descending
et al., 1992), but did restore partner preference in rats (Harding inhibitory influence onto the spinal cord, this pathway must
and McGinnis, 2003) and increased urine marking in mice be disinhibited to allow genital reflexes to occur. It has been
(Nyby et al., 1992). Hydroxyflutamide (AR antagonist) in the speculated that the PAG output to the nPGi might be the source
VMH of castrated, T-replaced rats inhibited both sexual of disinhibition to the spinal cord during mating (Normandin
motivation (Harding and McGinnis, 2004) and mating and Murphy, 2008). Neurons of different regions of the PAG
(Harding and McGinnis, 2004; McGinnis et al., 1996). Thus, express 5-HT, including those projecting from the vlPAG to
ARs in the VMH may contribute to male sexual behavior. the nPGi; this serotonergic input might be relevant for the
modulation of the tonic inhibition of spinal cord sexual
1.01.5.2.6.3 Expression of c-Fos reflexes from the nPGi (Bago et al., 2002). Supporting this
Copulation induced Fos-ir in the VMH of both rats (Coolen idea, serotonergic lesion of the vlPAG decreases ejaculation
et al., 1996) and gerbils (Heeb and Yahr, 1996). Chemosensory latency and increases the mean number of ejaculations
signals induced Fos-ir in gerbils (Heeb and Yahr, 1996), but exhibited by male rats (Normandin and Murphy, 2011). Elec-
not in rats (Bressler and Baum, 1996). Pelvic nerve transection trolytic lesions of the PAG, including the vlPAG, blocked elici-
did not affect ejaculation-induced Fos-ir in rats, suggesting that tation of the UG reflex by MPOA electrical stimulation
other sensory inputs, such as the pudendal nerve or chemosen- (Marson, 2004), but unilateral lesions of vlPAG and contralat-
sory input, contribute to mating-induced Fos-ir (Wersinger eral SDA of gerbils did not affect copulation (Finn and Yahr,
et al., 1993). Copulation did not increase Fos-ir in the VMH 1994).
of musk shrews (Gill et al., 1998), hamsters (Kollack-Walker There are numerous ERs and ARs in the caudal two-thirds of
and Newman, 1997), mice (Halem et al., 1999), or ferrets the PAG (Murphy et al., 1999b), with afferents from the MPOA
(Kelliher et al., 1998; Lambert et al., 1992) and actually in close apposition to ER- and AR-ir neurons, some of which
decreased Fos-ir in macaques (Michael et al., 1999). The project to nPGi (Murphy and Hoffman, 2001). AR and ERa
ventrolateral portion of the VMH contains neuronal subpopu- distribution within the caudal PAG of male rats was dense,
lations that are activated at the initiation of male–female with the majority of these receptors localized in the lateral/
encounters in mice and become progressively inhibited as the ventrolateral PAG (Loyd and Murphy, 2008). Thus,
encounter proceeded to active copulation, intermingled with hormones can modulate transmission from the MPOA to the
neurons activated by intermale aggression (Lin et al., 2011). nPGi, via the PAG. The PAG of monkeys projects to
a premotor area of the medulla, the nucleus retroambiguous
(Vanderhorst et al., 2000).
1.01.5.3 Major Motor Outputs
1.01.5.3.1 Ventral Premammillary Nucleus 1.01.5.3.3 Nucleus Paragigantocellularis of the Medulla
The MeA, the BST, and the MPN are the major sources of inputs In rats, the nPGi of the rostroventrolateral medulla is consid-
to the ventral premammillary nucleus (PMv), while the ventral ered the site of origin of the descending inhibitory influence
subiculum, the arcuate nucleus, and the ventrolateral subdivi- to the spinal cord that controls genital reflexes (Marson and
sion of the VMH provide moderate-to-high-density afferents McKenna, 1990). The nPGi receives direct projections from
to this nucleus (Cavalcante et al., 2014). The PMv contains the PVN, MPOA, and PAG and indirect projections from the
ARs in rats (Canteras et al., 1992; Simerly et al., 1990; Yokosuka MPOA through a relay in the PAG. Direct efferents from the
and Hayashi, 1996; Yokosuka et al., 1997) and hamsters MPOA are activated during mating, while indirect projections
(Wood and Newman, 1995a). Mating increased Fos-ir in are not (Normandin and Murphy, 2008). The nPGi projects
AR-containing neurons in rats (Gréco et al., 1998b). Fos-ir directly to pudendal motoneurons in the lumbosacral spinal
was elicited in the PMv of male mice by female-soiled cord involved in sexual refelexes (Hermann et al., 2003;
bedding (Yokosuka et al., 1999). The PMv contains Marson et al., 1992) (see Section 1.01.6.2).
numerous neurons expressing receptors for leptin (LepRb),
the adipocyte-derived hormone signaling energy status and 1.01.5.3.3.1 Effects of Lesions
contributing thereby to the metabolic control of Spinal nuclei that control genital reflexes are under tonic
reproduction. LepRb neurons in the PMv of male mice are inhibitory control from the nPGi; spinal transection releases
activated by female-soiled bedding odors and directly fictive ejaculation (Carro-Juárez and Rodríguez-Manzo,
innervate hypothalamic GnRH neurons, suggesting that they 2008) and increases the number or intensity of erections
Male Sexual Behavior 37

(reviewed in Hull et al., 2006). Bilateral nPGi lesions antierectile pathway with a concomitant increase in the
increased the proportion of sexually naïve male rats that activity of the proerectile parasympathetic sacral and
ejaculated on their first exposure to an estrous female and pudendal pathways (for review see Giuliano and Rampin,
reduced the stimulation and time needed by sexually experi- 2004). The neural commands controlling ejaculation are
enced male rats to achieve ejaculation. This lesion also also organized at the spinal level, and a central pattern gener-
increased the latency and number of ejaculations displayed ator, located at the lumbosacral-cord level, is involved in the
before reaching sexual exhaustion (Yells et al., 1992). Similar relay and integration of the genital sensory and motor signals
lesions decreased the latency and increased the numbers of related to ejaculation (Carro-Juárez and Rodríguez-Manzo,
reflexive erections (Holmes et al., 2002; Marson et al., 1992) 2008; Carro-Juárez et al., 2003). A group of lumbar spinotha-
and allowed the UG reflex to be elicited without spinal tran- lamic neurons, located in lamina VII and X of L3 and L4
section (Marson and McKenna, 1990). segments in this portion of the cord, which express galanin,
cholecystokinin, and neurokinin receptors, form part of this
1.01.5.3.3.2 Effects of Electrical Stimulation ejaculation generator (Truitt and Coolen, 2002). Selective
Electrical stimulation of the nPGi elicited field potentials in the lesion of these neurons disrupted bulbocavernosous muscle
lumbosacral spinal cord near the SNB (Tanaka and Arnold, bursting characteristic of ejaculation in anesthetized, spinal-
1993). nPGi stimulation specifically activated sympathetic ized male rats, but did not affect emission or erectile function
fibers in the pudendal nerve (Johnson and Hubscher, 2000), in spinally intact rats (Staudt et al., 2012).
and activation of pudendal neurons by electrical stimulation A system of gastrin-releasing peptide (GRP) neurons in the
of the dorsal nerve of the penis was inhibited by stimulation upper lumbar spinal cord (L3-L4) that innervates lower lumbar
of nPGi (Johnson and Hubscher, 1998). (L5-L6) and sacral S1 regions controlling erection and ejacula-
tion has been described in male rats. This system is the first
1.01.5.3.3.3 Immunocytochemistry neural pathway known to restore ejaculation in castrated
Most nPGi neurons that project to the lumbosacral spinal cord male rats (Sakamoto et al., 2008). It was recently demonstrated
contain 5-HT (Marson and McKenna, 1992). The main 5-HT that the perinatal androgen surge is essential for the develop-
source to the nPGi comes from the vlPAG neurons that project ment of this GRP system, which is sexually dimorphic (Oti
directly to the nPGi (Normandin and Murphy, 2011). The et al., 2016).
inhibitory effects of systemic SSRIs on ejaculation are reversed In humans, innervation of anatomical structures involved
(Clement et al., 2007b) or prevented (Yells et al., 1994) by in ejaculation is organized similarly to rats, but the spinal
nPGi lesions. Serotonergic receptors of the 5-HT1A (Thor cord segments harboring the sympathetic, parasympathetic,
et al., 1990), 5-HT1C (Hoffman and Mezey, 1989), 5-HT2A, 5- and somatic centers are slightly different due to differences in
HT2C, and 5-HT3 (Fonseca et al., 2001) subtypes are expressed metamerization between species. Thus, the parasympathetic
in this brain structure. Interestingly, the inhibitory effects of centers that innervate the prostate and seminal vesicles that
systemic SSRIs on ejaculation are reversed or prevented by secrete seminal fluid are located in the L6-S1 segments of rats
nPGi lesions, leading to the suggestion that SSRIs might inhibit but in the S2-S4 segments of men; the sympathetic centers
male rats’ genital reflexes, not only at the spinal-cord level innervating smooth muscles of the tract, including epydidimus,
(Marson and McKenna, 1992), but this could also involve vas deferens, seminal vesicles, prostate, prostatic urethra, and
the nPGi via vlPAG 5-HT input (Normandin and Murphy, the bladder neck, are located in T13-L2 rat segments and
2011). between T12 and L2 in humans, while the location of the
somatic centers that innervate the pelvic striated muscles is
1.01.5.3.4 Other Brain Areas between L6 and S1 in rats, and in the ventral horn of S2-S4
Several additional brain areas have been implicated in the regu- segments in humans (Onuf’s nucleus). Studies about ejacula-
lation of male sexual behavior, although there is little informa- tion in spinal cord-injured men support the existence of an
tion about their specific contributions. These include the lateral SGE in humans that appears to be located at L3-L5 spinal
septum (Kondo et al., 1993), hippocampus (Chen et al., 1992), segments (Chehensse et al., 2013).
caudal zona incerta (Edwards and Isaacs, 1991; Maillard and Spinal cord transection releases sexual reflexes, indicating
Edwards, 1991), and cortical regions in men: inferior temporal that supraspinal centers exert a descending inhibitory influence
cortex, right insula, right inferior frontal cortex, and left anterior (McKenna et al., 1991), probably mediated by 5-HT (Marson
cingulate cortex (Stoleru et al., 1999). and McKenna, 1994a). Intrathecal administration of 5-HT
abolished the UG reflex (Marson and McKenna, 1992), and
1.01.5.3.5 Spinal Cord chronic treatment with the SSRI fluoxetine diminished the
The spinal cord contains the autonomic and somatic nuclei response capacity of the ejaculation generator (Hueletl-Soto
controlling erection and ejaculation. The sympathetic and et al., 2012); however 5-HT1A (Carro-Juárez and Rodríguez-
parasympathetic tones involved in the control of these Manzo, 2001; Carro-Juárez et al., 2003) and 5-HT2C receptor
reflexes are under the influence of sensory stimuli from the stimulation (Stafford et al., 2006b) facilitated the UG reflex,
genitalia and are integrated at the spinal-cord level, where suggesting that 5-HT exerts a dual influence on male sexual
supraspinal information also converges. The spinal cord behavior at the spinal level.
contains thoracolumbar sympathetic, sacral parasympathetic, Cerebrospinal fluid levels of GABA, glutamate, and aspar-
and sacral pudendal motoneurons anatomically linked with tate markedly increased after ejaculation, with the increase in
the penis. Erection is likely to occur when the spinal cord GABA concentrations almost fivefold that of excitatory amino
reduces the activity of the thoracolumbar sympathetic acids (Qureshi and Södersten, 1986). Lumbosacral injection
38 Male Sexual Behavior

of the GABAB agonist baclofen inhibited ex copula reflexive 1.01.6 Circuitry and Anatomical Interconnections
penile erection (Bitran et al., 1989a), while that of a competitive
NMDA or AMPA-kainate receptor antagonist depressed both Organisms in natural settings weigh, prioritize, and integrate
dorsal penile nerve (DPN)-stimulated erection in multiple internal and external signals in order to choose and
anesthetized rats and reflexive erection in conscious animals perform various motivated behaviors, including mating. Here
(Rampin et al., 2004). Also, NMDA receptor-mediated we summarize several functional circuits that analyze and inte-
glutamatergic mechanisms are involved in the excitatory grate sensory and hormonal stimuli and execute behavioral
modulation of the SGE (Staudt et al., 2011). Thus, GABA outcomes.
may inhibit, and glutamate may facilitate, sexual reflexes at
the spinal level. The DA agonist apomorphine, injected
1.01.6.1 Sensory Inputs
intrathecally at the lumbosacral level of conscious rats,
impaired reflexive erection, but evoked erection in normal Rodents depend heavily on chemosensory signals for mating.
and spinal animals (Andersson, 2001) and induced The MeA, BNST, and MPOA are critical nodes in this circuit,
ejaculation-like responses in anesthetized rats (Stafford and and each contains numerous steroid receptors (Rasia-Filho
Coote, 2006). However, chronic treatment with bupropion, et al., 1991; Wood and Newman, 1995b). T or E, but not
a dual reuptake inhibitor increasing DA and NA availability, DHT, in the MeA or MPOA enhances mating (Wood, 1996)
disrupted the functioning of the SGE (Hueletl-Soto et al., and responsiveness to odor cues (Sipos and Nyby, 1998;
2014). Wood and Newman, 1995b). Hormones may permit or
Reflexive erections are inhibited by both a1- and a2-adreno- enhance release of transmitters, promote the production or
ceptor agonists. a2-antagonists have dose-dependent effects, placement of postsynaptic receptors, and increase growth and
with low doses facilitating, and higher doses inhibiting, branching of neurons.
reflexive erections in rats (reviewed in Meisel and Sachs, The major source of afferent input from the penis is the
1994) and seminal emission in dogs (Yonezawa et al., 1991). dorsal nerve of the penis (DNP, a branch of the pudendal nerve),
The spinal NE system facilitates ejaculation, since increasing which carries signals from the penile skin, prepuce, and glans;
NE tone by systemic yohimbine and stimulation of a1- or a2- input from deeper structures is carried by the cavernous nerve
adrenoceptors (Carro-Juárez and Rodríguez-Manzo, 2003, (Steers, 2000). Afferents end mostly in medial portions of the
2006) all activate the SGE. dorsal horn and the medial central gray matter of the lumbosa-
The ACh agonist muscarine, intrathecally administered, cral cord (reviewed in Hull et al., 2002, 2006). Electrical
facilitated ejaculation, while intraspinal injection of different stimulation of the DNP elicited responses in the nPGi, PVN,
muscarinic receptor antagonists decreased the percentage of MPOA, and cortex (Hubscher and Johnson, 1996; Yanagimoto
rats that ejaculated and increased the EL in those ejaculating et al., 1996). Mating elicits Fos-ir in the CTF of rats (Baum and
(Durán et al., 2000; Gomez-Martinez and Cueva-Rolon, Everitt, 1992) and SPFp of rats (Coolen et al., 1996, 1997a,b),
2009). Intrathecal injection of a NOS inhibitor abolished, gerbils (Heeb and Yahr, 1996), and hamsters (Kollack-Walker
and an NO donor enhanced apomorphine-evoked bursting and Newman, 1997); these areas all project to the MPOA.
activity in the vas deferens nerve that is associated with ejacula- Axons from the L3 and L4 spinal segments terminate adjacent
tory responses (Brack et al., 2007). to SPFp neurons that project to the MPOA or BSTpm
Opioids exert an inhibitory influence on the functioning of (Wells and Coolen, 2001). Electrical stimulation of the DNP
the SGE, since morphine blocks the expression of the in men elicited activity in the midline of somatosensory cortex
rhythmic motor pattern that accompanies ejaculation in (Gerstenberg et al., 1991; Guerit and Opsomer, 1991).
spinal rats, and this effect is prevented by naloxone pretreat-
ment. By contrast, naloxone facilitated the SGE activity and
1.01.6.2 An Ejaculation-Related Circuit
delayed the inhibition of the ejaculatory response resulting
from repeated urethral stimulation. So far, endogenous As mentioned above, ejaculation is controlled by a central
opioids represent the only intraspinal inhibitory influence pattern generator, located in the lumbosacral spinal cord, named
on SGE functioning described (Carro-Juárez and Rodríguez- the SGE (Carro-Juárez et al., 2003), coordinating genital inputs
Manzo, 2009). and sympathetic, parasympathetic, and motor outputs to trigger
Electrical microstimulation of the SGE in rapid ejaculating ejaculation (Carro-Juárez and Rodríguez-Manzo, 2008). The
rats revealed that the expulsion phase in this subpopulation activity the SGE depends on cerebral descending facilitatory
is accelerated, supporting the notion that the reduced ejacula- and inhibitory influences. A group of spinothalamic neurons
tory latency in these rats is related to an altered SGE functioning located in the lumbar spinal cord (LSt cells) form part of the ejac-
(Borgdorff et al., 2009). ulation generator and project to the SPFp (Truitt and Coolen,
These data show that the effects of neurotransmitters in the 2002). Ejaculation, but not intromissions without ejaculation,
spinal cord may vary from those exerted in the brain or periph- elicited small areas of Fos-ir in MeApd, BSTpm, PdPN, and
eral nervous system. Thus, opposite results may be obtained SPFp of rats (for review see Coolen, 2005; Coolen et al.,
with the same drug on a given sexual reflex, depending on 2004a), hamsters (Fernandez-Fewell and Meredith, 1994;
the animal model used (i.e., in copula vs ex copula responses, Kollack-Walker and Newman, 1997; Parfitt and Newman,
conscious vs anesthetized rats, spinally intact vs spinally trans- 1998), and gerbils (Heeb and Yahr, 1996, 2001; Simmons and
ected animals). To integrate this knowledge to understand the Yahr, 2002, 2003; Figure 5). These regions, activated specifically
spinal cord-mediated sexual functioning of an intact copu- following ejaculation, are part of a broader ejaculation-related
lating animal is one of today’s challenges. network that includes the MPOA, the PVN, and the nPGi.
Male Sexual Behavior 39

AOB
Chemosensory inputs

vl SPFp
st st
sm ot
fx
v3 fx
aq
v3 PD

fr ml

MPN
MPN

Rostral MeApd
Caudal
BNSTpm BNSTpm LSSC

Visceral and somatosensory inputs

Figure 5 Schematic overview of Fos immunoreactivity in medial preoptic nucleus (MPN), posteriomedial part of the bed nucleus of the stria termi-
nalis (BNSTpm), posterodorsal preoptic nucleus (PD), posterodorsal part of the medial amygdala (MeApd), parvocellular division of the sub-
parafascicular nucleus (SPFp), and accessory olfactory bulbs (AOB). Areas where Fos is induced following chemosensory cues or chemosensory
investigation are illustrated by diagonal stripes from upper left to lower right. Areas where Fos is induced only following ejaculation are illustrated in
gray. Areas where Fos is induced following all consummatory elements of sexual behavior are illustrated by diagonal stripes from lower left to upper
right. LSSC, lumbosacral spinal cord; v3, third ventricle; fx, fornix; vl; lateral ventricle; st, stria terminalis; s, stria medularis; ot, optic tract; aq, aque-
duct; fr, fasciculus retroflexus; ml, medial lemniscus. Figure is reprinted from Coolen, L.M., Allard, J., Truitt, W.A., McKenna, K.E., 2004. Central
regulation of ejaculation. Physiol. Behav. 83, 203–215.

The MPOA and PVN appear to mediate excitatory, and the nPGi, However, lesions that included the SPFp and zona incerta of
inhibitory, influences on the spinal control of ejaculation. The rats almost eliminated ejaculation (Maillard and Edwards,
MPOA is a key structure for ejaculation, but it does not directly 1991). The BSTpm and MePD contribute to sexual satiety of
project to the spinal cord. However, neurons in each of the hamsters, as Fos-ir increased only after multiple ejaculations
ejaculation-activated regions have reciprocal connections with (Parfitt and Newman, 1998), and small lesions increased the
the MPOA (Coolen et al., 1998; Gréco et al., 1998a; Heeb and number of ejaculations before satiety (Parfitt et al., 1996). LH
Yahr, 2001), and the MPOA projects to the PVN and nPGi. lesions in rats disrupt ejaculation, but not intromissions or
The parvocellular region of the PVN sends neural projections mounts (Kippin et al., 2004). 5-HT is released in the anterior
to the spinal cord, including the lumbosacral segments LH at the time of ejaculation (Lorrain et al., 1997); however,
(Hallbeck and Blomqvist, 1999), to the vlPAG (Luiten et al., it is not clear whether this release is related to triggering ejacu-
1985), and the nPGi (Kobayashi et al., 2013). This PVN descend- lation or promoting the quiescence of the PEI. However, brain-
ing pathway is considered to exert, directly or indirectly, a facili- descending pathways originating in the LH, caudal raphe
tatory influence on ejaculation (Veening and Coolen, 2014). The nuclei, and the ventrolateral division of the gigantocellular
MPOA projects also to the nPGi, the primary source of descend- nucleus were recently described which directly project onto
ing inhibition on the SGE, both directly (Normandin and spinal neurons, including the LSt cells (Facchinetti et al., 2014).
Murphy, 2008) and through a PAG relay (Murphy and Hoffman, In men, positron emission tomography showed increases in
2001; Figure 6). It was recently postulated that the 5-HT regional cerebral blood flow during ejaculation in the meso-
pathway that runs from the vlPAG to the nPGi may act as a gating diencephalic region, which includes the SPFp, as well as the
mechanism for descending inhibition of genital reflexes cerebellum, lateral putamen, claustrum, and several cortical
(Normandin and Murphy, 2011). regions (Holstege et al., 2003). No increases were seen in the
The MeApd and SPFp of rats and gerbils contribute to acti- MPOA or BNST, in agreement with a study in male macaques
vation of the MPOA, as unilateral lesions decreased mating- (Michael et al., 1999). Neuroimaging techniques also showed
induced Fos-ir there (Baum and Everitt, 1992; Heeb and that the major effect of ejaculation in male human brain is
Yahr, 2000). It is not clear whether the Fos-ir neurons received not the activation of certain regions, but deactivation of regions
sensory input or activated motor patterns of ejaculation. involved in emotional processing. Activation following ejacula-
In gerbils, PdPN or MeApd lesions decreased mounting and tion was detected in the right posterior insula, the dorsal poste-
delayed ejaculation; thus, these areas contribute to mating, but rior temporal lobe, the central midbrain, the medial globus
are not essential for ejaculation (Heeb and Yahr, 2000). pallidus, and major parts of the cerebellum, while deactivation
40 Male Sexual Behavior

Ejaculatory circuit

BNSTpm MeApd

mPOA PVN
SPFp

PAG

nPGi

SGE

Figure 6 Brain control of ejaculation. Scheme illustrating the principal brain regions that form part of the ejaculatory circuit influencing the
functioning of the spinal ejaculation generator. BNSTpm, posteriomedial part of the bed nucleus of the stria terminalis; MeApd, posterodorsal part of
the medial amygdala; mPOA, hypothalamic medial preoptic area; nPGi, nucleus paragigantocellularis; PAG, periaqueductal gray; PVN, hypothalamic
paraventricular nucleus; SGE, spinal generator for ejaculation; SPFp, parvocellular division of the subparafascicular nucleus (described in
Section 1.01.6.2).

was identified in the ventral part of the temporal lobe and these brain areas form a reciprocally interconnected circuit
extended regions of the parietal, medial orbitofrontal, and that serves all mammalian social behaviors. Most of the areas,
temporal cortices (Holstege and Huynh, 2011). except the midbrain, are richly endowed with steroid receptors,
and all influence more than one behavior. Therefore, perinatal,
1.01.6.3 Efferents from the MPOA adolescent, and adult hormones can provide a bias toward
sexually dimorphic responses to social stimuli. It is not clear
Efferents from the MPOA target the periventricular and medial whether the same neurons within a structure contribute to
zones of the hypothalamus; the LH; midbrain motivation/ more than one behavior, or whether neurons specific for one
somatomotor regions, including the VTA and pedunculopon- behavior mingle with those for other behaviors. However, there
tine nucleus; midbrain and brain stem areas that project to may be common themes that underlie the various social behav-
the spinal cord, including the PAG, certain raphe nuclei, iors and the neural mechanisms that control them.
and nPGi; the BNST; and part of the septal area (Simerly and
Swanson, 1988; reviewed in Hull et al., 2006). These connec-
tions are mostly reciprocal, allowing ‘downstream’ sites to
influence the input that they receive. They provide multiple
References
ways to influence autonomic, motivational, and somatomotor
Abber, J.C., Lue, T.F., Luo, J.A., Juenemann, K.P., Tanagho, E.A., 1987. Priapism
patterns. Output to the nPGi, either directly or via the PAG, induced by chlorpromazine and trazodone: mechanism of action. J. Urol. 137,
may disinhibit erections and the UG reflex. However, it seems 1039–1042.
unlikely that mere disinhibition could elicit the reflexes that Ackerman, A.E., Lange, G.M., Clemens, L.G., 1997. Effects of paraventricular lesions
can be evoked by MPOA stimulation (Giuliano et al., 1996, on sex behavior and seminal emissions in male rats. Physiol. Behav. 63, 49–53.
Ågmo, A., 2003. Lack of opioid or dopaminergic effects on unconditioned sexual
1997; Marson and McKenna, 1994b). Such excitatory inputs incentive motivation in male rats. Behav. Neurosci. 117, 55–68.
may include parasympathetic efferents, via the pelvic and Ågmo, A., 2008. On the concept of sexual arousal: a simpler alternative. Horm. Behav.
cavernous nerves, and sympathetic outflow, via the paraverte- 53, 312–314.
bral sympathetic chain and possibly the hypogastric nerve Ågmo, A., 2011. On the intricate relationship between sexual motivation and arousal.
Horm. Behav. 59, 681–688.
(Giuliano et al., 1997). A number of projection sites contain
Ågmo, A., Berenfeld, R., 1990. Reinforcing properties of ejaculation in the male rat:
abundant steroid hormone receptors, allowing hormones to role of opioids and dopamine. Behav. Neurosci. 104, 177–182.
bias sexually relevant cues and responses. Ågmo, A., Ellingsen, E., 2003. Relevance of non-human animal studies to the
understanding of human sexuality. Scand. J. Psychol. 44, 293–301.
Ågmo, A., Paredes, R., 1985. GABAergic drugs and sexual behavior in the male rat.
1.01.6.4 Sexual Behavior in the Context of Mammalian Social Eur. J. Pharmacol. 112, 371–378.
Behavior Ågmo, A., Paredes, R., 1988. Opioids and sexual behavior in the male rat. Pharmacol.
Biochem. Behav. 30, 1021–1034.
The brain areas that regulate male sexual behavior also influ- Ågmo, A., Picker, Z., 1990. Catecholamines and the initiation of sexual behavior in
ence other social behaviors. Newman (1999) suggested that male rats without sexual experience. Pharmacol. Biochem. Behav. 35, 327–334.
Male Sexual Behavior 41

Ågmo, A., Gomez, M., Irazabal, Y., 1994. Enkephalinase inhibition facilitates sexual Argiolas, A., Melis, M.R., 2013. Neuropeptides and central control of sexual behaviour
behavior in the male rat but does not produce conditioned place preference. from the past to the present: a review. Prog. Neurobiol. 108, 80–81.
Pharmacol. Biochem. Behav. 47, 771–778. Arletti, R., Benelli, A., Bertolini, A., 1990. Sexual behavior of aging male rats is
Ågmo, A., Soulairac, M.L., Soulairac, A., 1977. Preoptic lesions, sexual behavior, and stimulated by oxytocin. Eur. J. Pharmacol. 179, 377–381.
spontaneous ejaculation in the rat. Scand. J. Psychol. 18, 345–347. Arletti, R., Calza, L., Giardino, L., Benelli, A., Cavazutti, E., Bertolini, A., 1997. Sexual
Ågmo, A., Villalpando, A., Picker, Z., Fernández, H., 1995. Lesions of the medial impotence is associated with a reduced production of oxytocin and with an
prefrontal cortex and sexual behavior in the male rat. Brain Res. 696, increased production of opioid peptides in the paraventricular nucleus of male rats.
177–186. Neurosci. Lett. 233, 65–68.
Ahlenius, S., Larsson, K., 1985. Central muscarinic receptors and male rat sexual Arnone, M., Baroni, M., Gai, J., Guzzi, U., Desclaux, M.F., Keane, P.E., LeFur, G.,
behavior: facilitation by oxotremorine but not arecoline or pilocarpine in meths- Soubrié, P., 1995. Effect of SR 59026A, a new 5-HT(1A) receptor agonist, on
copolamine pretreated animals. Psychopharmacology (Berlin) 87, 127–129. sexual activity in male rats. Behav. Pharmacol. 6, 276–282.
Ahlenius, S., Larsson, K., 1990. Effects of selective D1 and D2 antagonists on male rat Arteaga-Silva, M., Marquez-Villanueva, Y., Martinez-Garcia, R., Hernandez-
sexual behavior. Experientia 46, 1026–1028. Gonzalez, M., Bonilla-Jaime, H., Retana-Marquez, S., 2005. Effects of hormonal
Ahlenius, S., Larsson, K., 1998. Evidence for the involvement of 5-HT1B receptors in replacement with androgens and estrogens on male sexual behavior and plasma
the inhibition of male rat sexual behavior produced by 5-HTP. Psychopharmacology levels of these steroids in gonadectomized golden hamsters (Mesocricetus aur-
(Berlin) 137, 374–382. atus). Physiol. Behav. 85, 571–580.
Ahlenius, S., Larsson, K., Arvidsson, L.E., 1989. Effects of stereoselective 5-HT1A Artigas, F., Romero, L., de Montigny, C., Blier, P., 1996. Acceleration of the effect of
agonists on male rat sexual behavior. Pharmacol. Biochem. Behav. 33, selected antidepressant drugs in major depression by 5-HT1A antagonists. Trends
691–695. Neurosci. 19, 378–383.
Ahlenius, S., Larsson, K., Svensson, L., Hjorth, S., Carlsson, A., Lindberg, P., Aste, N., Honda, S., Harada, N., 2003. Forebrain Fos responses to reproductively related
Wikström, H., Sanchez, D., Arvidsson, L.E., Hacksell, U., Nilsson, J.L., 1981. chemosensory cues in aromatase knockout mice. Brain Res. Bull. 60, 191–200.
Effects of a new type of 5-HT receptor agonist on male rat sexual behavior. Attila, M., Oksala, R., Agmo, A., 2010. Sexual incentive motivation in male rats
Pharmacol. Biochem. Behav. 15, 785–792. requires both androgens and estrogens. Horm. Behav. 58, 341–351.
Ahmed, E.I., Zehr, J.L., Schulz, K.M., Lorenz, B.H., DonCarlos, L.L., Sisk, C.L., 2008. Bago, M., Marson, L., Dean, C., 2002. Serotonergic projections to the rostroventro-
Pubertal hormones modulate the addition of new cells to sexually dimorphic brain lateral medulla from midbrain and raphe nuclei. Brain Res. Mol. Brain Res. 945,
regions. Nat. Neurosci. 11, 995–997. 249–258.
Akil, H., Watson, S.J., Young, E., Lewis, M.E., Khachaturian, H., Walker, J.M., 1984. Bai, Y.J., Li, Y.H., Zheng, X.G., Han, J., Yang, X.Y., Sui, N., 2009. Orexin A attenuates
Endogenous opioids: biology and function. Annu. Rev. Neurosci. 7, 223–255. unconditioned sexual motivation in male rats. Pharmacol. Biochem. Behav. 91,
Alcaro, A., Huber, R., Panksepp, J., 2007. Behavioral functions of the mesolimbic 581–589.
dopaminergic system: an affective neuroethological perspective. Brain Res. Rev. Bakker, J., Honda, S., Harada, N., Balthazart, J., 2002a. Sexual partner preference requires
56, 283–321. a functional aromatase (cyp19) gene in male mice. Horm. Behav. 42, 158–171.
Alexander, B.M., Perkins, A., Van Kirk, E.A., Moss, G.E., Fitzgerald, J.A., 1993. Bakker, J., Woodley, S.K., Kelliher, K.R., Baum, M.J., 2002b. Sexually dimorphic
Hypothalamic and hypophyseal receptors for estradiol in high and low sexually activation of galanin neurones in the ferret’s dorsomedial preoptic area/anterior
performing rams. Horm. Behav. 27, 296–307. hypothalamus after mating. J. Neuroendocrinol. 14, 116–125.
Alexander, G.M., Swerdloff, R.S., Wang, C., Davidson, T., McDonald, V., Steiner, B., Baldwin, D., Mayers, A., 2003. Sexual side-effects of antidepressant and antipsychotic
Hines, M., 1997. Androgen-behavior correlations in hypogonadal men and eugo- drugs. Adv. Psychiatr. Treat. 9, 202–210.
nadal men: I. Mood and response to auditory sexual stimuli. Horm. Behav. 31, Balfour, M.E., Yu, L., Coolen, L.M., 2004. Sexual behavior and sex-associated envi-
110–119. ronmental cues activate the mesolimbic system in male rats. Neuro-
Alheid, G.F., de Olmos, J.S., Beltramino, C.A., 1995. Extended amygdala and basal psychopharmacology 29, 718–730.
forebrain. In: Paxinos, G.E. (Ed.), The Rat Nervous System. Academic Press, San Balfour, M.E., Brown, J.L., Yu, L., Coolen, L.M., 2006. Potential contributions of
Diego, pp. 495–578 (Chapter 22). efferents from medial prefrontal cortex to neural activation following sexual
Allard, J., Giuliano, F., 2001. Central nervous system agents in the treatment of behavior in the male rat. Neuroscience 137, 1259–1276.
erectile dysfunction: how do they work? Curr. Urol. Rep. 2, 488–494. Ballard, C.L., Wood, R.I., 2007. Partner preference in male hamsters: steroids, sexual
Allard, J., Bernabe, J., Derdinger, F., Alexandre, L., McKenna, K., Giuliano, F., 2002. experience and chemosensory cues. Physiol. Behav. 91, 1–8.
Selegiline enhances erectile activity induced by dopamine injection in the para- Balthazart, J., Ball, G.F., 2006. Is brain estradiol a hormone or a neurotransmitter?
ventricular nucleus of the hypothalamus in anesthetized rats. Int. J. Impot. Res. 14, Trends Neurosci. 29, 241–249.
518–522. Balthazart, J., Ball, G.F., 2007. Topography in the preoptic region: differential regu-
Amstislavskaya, T.G., Popova, N.K., 2004. Female-induced sexual arousal in male lation of appetitive and consummatory male sexual behaviors. Front. Neuro-
mice and rats: behavioral and testosterone response. Horm. Behav. 46, 544–550. endocrinol. 28, 161–178.
Andersen, M.L., Tufik, S., 2005. Effects of progesterone blockade over cocaine- Balthazart, J., Absil, P., Gerard, M., Appeltants, D., Ball, G.F., 1998. Appetitive and
induced genital reflexes of paradoxical sleep deprived male rats. Horm. Behav. consummatory male sexual behavior in Japanese quail are differentially regulated
47, 477–484. by subregions of the preoptic medial nucleus. J. Neurosci. 18, 6512–6527.
Andersen, M.L., Tufik, S., 2006. Does male sexual behavior require progesterone? Balthazart, J., Castagna, C., Ball, G.F., 1997. Differential effects of D1 and D2
Brain Res. Rev. 51, 136–143. dopamine-receptor agonists and antagonists on appetitive and consummatory
Andersson, K.E., 2001. Pharmacology of penile erection. Pharmacol. Rev. 53, aspects of male sexual behavior in Japanese quail. Physiol. Behav. 62, 571–580.
417–450. Balthazart, J., Cornil, C.A., Taziaux, M., Charlier, T.D., Baillien, M., Ball, G.F., 2006.
Andersson, K.E., 2011. Mechanisms of penile erection and basis for pharmacological Rapid changes in production and behavioral action of estrogens. Neuroscience
treatment of erectile dysfunction. Pharmacol. Rev. 63, 811–859. 138, 783–791.
Andersson, K.E., Stief, C.G., 1997. Neurotransmission and the contraction and Bancila, M., Giuliano, F., Rampin, O., Mailly, P., Brisorgueil, M.J., Calas, A., Verge, D.,
relaxation of penile erectile tissues. World J. Urol. 15, 14–20. 2002. Evidence for a direct projection from the paraventricular nucleus of the
Antaramian, A., Gonzalez-Gallardo, A., Garcia-Ugalde, C., Portillo, W., Paredes, R.G., hypothalamus to putative serotoninergic neurons of the nucleus paragigantocellularis
2015. Steroid receptors and aromatase gene expression in different brain areas of involved in the control of erection in rats. Eur. J. Neurosci. 16, 1240–1248.
copulating and sexually sluggish male rats. J. Sex. Med. 12, 2267–2275. Bancroft, J., 1999. Central inhibition of sexual response in the male: a theoretical
Antonio-Cabrera, E., Paredes, R.G., 2014. Testosterone or oestradiol implants in the perspective. Neurosci. Biobehav. Rev. 23, 763–784.
medial preoptic area induce mating in noncopulating male rats. Bancroft, J., 2005. The endocrinology of sexual arousal. J. Endocrinol. 186, 411–427.
J. Neuroendocrinology 26, 448–458. Band, L.C., Hull, E.M., 1990. Morphine and dynorphin (1-13) microinjected into the
Arendash, G.W., Gorski, R.A., 1983. Effects of discrete lesions of the sexually medial preoptic area and nucleus accumbens: effects on sexual behavior in male
dimorphic nucleus of the preoptic area or other medial preoptic regions on the rats. Brain Res. 524, 77–84.
sexual behavior of male rats. Brain Res. Bull. 10, 147–154. Barbeau, A., 1969. L-DOPA therapy in Parkinson’s disease: a critical review of nine
Argiolas, A., 1999. Neuropeptides and sexual behavior. Neurosci. Biobehav. Rev. 23, years’ experience. Can. Med. Assoc. J. 101, 791–800.
1127–1142. Barfield, R.J., Geyer, L.A., 1975. The ultrasonic postejaculatory vocalization and
Argiolas, A., Melis, M.R., 2005. Central control of penile erection: role of the para- the postejaculatory refractory period of the male rat. J. Comp. Physiol. Psychol.
ventricular nucleus of the hypothalamus. Prog. Neurobiol. 76, 1–21. 88, 723–734.
42 Male Sexual Behavior

Barfield, R.J., Sachs, B.D., 1968. Sexual behavior: stimulation by painful electrical Bialy, M., Beck, J., Abramczyk, P., Trzebski, A., Przybylski, J., 1996. Sexual behavior
shock to skin in male rats. Science 161, 392–393. in male rats after nitric oxide synthesis inhibition. Physiol. Behav. 60, 139–143.
Barrett, G.M., Bardi, M., Guillen, A.K., Mori, A., Shimizu, K., 2006. Regulation of Bialy, M., Rydz, M., Kaczmarek, L., 2000. Precontact 50-kHz vocalizations in male rats
sexual behaviour in male macaques by sex steroid modulation of the serotonergic during acquisition of sexual experience. Behav. Neurosci. 114, 983–990.
system. Exp. Physiol. 91, 445–456. Bialy, M., Kalata, U., Nikolaev-Diak, A., Nikolaev, E., 2010. D1 receptors involved in the
Baskerville, T.A., Allard, J., Wayman, C., Douglas, A.J., 2009. Dopamine-oxytocin acquisition of sexual experience in male rats. Behav. Brain Res. 206, 166–176.
interactions in penile erection. Eur. J. Neurosci. 30, 2151–2164. Bielert, C.F., Goy, R.W., 1973. Sexual behavior of male rhesus: effects of repeated
Baum, M.J., Everitt, B.J., 1992. Increased expression of c-fos in the medial preoptic ejaculation and partner’s cycle stage. Horm. Behav. 4, 109–122.
area after mating in male rats: role of afferent inputs from the medial amygdala and Bijlsma, E.Y., Chan, J.S., Olivier, B., Veening, J.G., Millan, M.J., Waldinger, M.D.,
midbrain central tegmental field. Neuroscience 50, 627–646. Oosting, R.S., 2014. Sexual side effects of serotonergic antidepressants: mediated
Baum, M.J., Wersinger, S.R., 1993. Equivalent levels of mating-induced neural c-fos by inhibition of serotonin on central dopamine release? Pharmacol. Biochem.
immunoreactivity in castrated male rats given androgen, estrogen, or no steroid Behav. 121, 88–101.
replacement. Biol. Reprod. 48, 1341–1347. Birowo, P., Uckert, S., Kedia, G.T., et al., 2009. Evaluating the role of the seroto-
Baum, M.J., Tobet, S.A., Starr, M.S., Bradshaw, W.G., 1982. Implantation of dihy- ninergic system in the control of human seminal vesicle smooth muscle-an in vitro
drotestosterone propionate into the lateral septum or medial amygdala facilitates approach. J. Sex. Med. 6, 2672–2679.
copulation in castrated male rats given estradiol systemically. Horm. Behav. 16, Bitran, D., Hull, E.M., 1987. Pharmacological analysis of male rat sexual behavior.
208–223. Neurosci. Biobehav. Rev. 11, 365–389.
Bazzett, T.J., Eaton, R.C., Thompson, J.T., Markowski, V.P., Lumley, L.A., Hull, E.M., Bitran, D., Miller, S.A., McQuade, D.B., Leipheier, R.E., Sachs, B.D., 1989a. Inhibition
1991. Dose dependent D2 effects on genital reflexes after MPOA injections of of sexual reflexes by lumbosacral injection of a GABAB agonist in the male rat.
quinelorane and apomorphine. Life Sci. 48, 2309–2315. Pharmacol. Biochem. Behav. 31, 657–666.
Bazzett, T.J., Lumley, L., Bitran, D., Markowski, V., Warner, R., Hull, E., 1992. Male Bitran, D., Thompson, J.T., Hull, E.M., Sachs, B.D., 1989b. Quinelorane (LY 163502),
rat copulation following 6-OHDA lesions of the medial preoptic area: resistance to a D2 dopamine receptor agonist, facilitates seminal emission but inhibits penile
repeated administration and rapid behavioral recovery. Brain Res. 580, 164–170. erection in the rat. Pharmacol. Biochem. Behav. 34, 453–458.
Beach, F.A., 1969. Locks and beagles. Am. Psychol. 24, 971–989. Bivalacqua, T.J., Champion, H.C., Hellstrom, W.J.G., Kadowitz, P.J., 2000. Pharma-
Beach, F.A., Jordan, L., 1956. Sexual exhaustion and recovery in the male rat. Q. J. cotherapy for erectile dysfunction. Trends Pharmacol. Sci. 21, 484–489.
Exp. Psychol. 8, 121–133. Bivalacqua, T.J., Champion, H.C., Usta, M.F., Cellek, S., Chitaley, K., Webb, R.C.,
Beach, F.A., Rabedeau, R.G., 1959. Sexual exhaustion and recovery in the male Lewis, R.L., Mills, T.M., Hellstrom, W.J., Kadowitz, P.J., 2004. RhoA/Rho kinase
hamster. J. Comp. Physiol. Psychol. 52, 56–66. suppresses endothelial nitric oxide synthase in the penis: a mechanism for diabetes-
Beck, J., Bialy, M., Kostowski, W., 2002. Effects of D(1) receptor agonist SKF 38393 associated erectile dysfunction. Proc. Natl. Acad. Sci. U.S.A. 101, 9121–9126.
on male rat sexual behavior and postcopulatory departure in the goal compartment- Bloch, G.J., Davidson, J.M., 1968. Effects of adrenalectomy and experience on
runaway paradigm. Physiol. Behav. 76, 91–97. postcastration sex behavior in the male rat. Physiol. Behav. 3, 461–465.
Becker, A.J., Uckert, S., Stief, C.G., Scheller, F., Knapp, W.H., Hartmann, U., Bonsall, R.W., Clancy, A.N., Michael, R.P., 1992. Effects of the nonsteroidal aro-
Jonas, U., 2001. Cavernous and systemic testosterone plasma levels during matase inhibitor, fadrozole, on sexual behavior in male rats. Horm. Behav. 26,
different penile conditions in healthy males and patients with erectile dysfunction. 240–254.
Urology 58, 435–440. Borchard, U., 1998. Pharmacological properties of beta-adrenoceptor blocking drugs.
Been, L.E., Petrulis, A., 2010a. Lesions of the posterior bed nucleus of the stria J. Clin. Basic Cardiol. 1, 5–9.
terminalis eliminate opposite-sex odor preference and delay copulation in male Borgdorff, A.J., Bernabe, J., Denys, P., Alexandre, L., Giuliano, F., 2008. Ejaculation elicited
syrian hamsters: role of odor volatility and sexual experience. Eur. J. Neurosci. 32, by microstimulation of lumbar spinothalamic neurons. Eur. Urol. 54, 449–456.
483–493. Borgdorff, A.J., Rossler, A.S., Clement, P., Bernabe, J., Alexandre, L., Giuliano, F.,
Been, L.E., Petrulis, A., 2010b. The role of the medial preoptic area in appetitive and 2009. Differences in the spinal command of ejaculation in rapid ejaculating rats.
consummatory reproductive behaviors depends on sexual experience and odor J. Sex. Med. 6, 2197–2205.
volatility in male syrian hamsters. Neuroscience 170, 1120–1132. Borgland, S.L., Storm, E., Bonci, A., 2008. Orexin B/hypocretin 2 increases gluta-
Bell, M.R., De Lorme, K.C., Figueira, R.J., Kashy, D.A., Sisk, C.L., 2013a. Adolescent matergic transmission to ventral tegmental area neurons. Eur. J. Neurosci. 28,
gain in positive valence of a socially relevant stimulus: engagement of the mes- 1545–1556.
ocorticolimbic reward circuitry. Eur. J. Neurosci. 37, 457–468. Bowers Jr., M.B., Van Woert, M., Davis, L., 1971. Sexual behavior during L-dopa
Bell, M.R., Meerts, S.H., Sisk, C.L., 2013b. Adolescent brain maturation is necessary treatment for Parkinsonism. Am. J. Psychiatry 127, 1691–1693.
for adult-typical mesocorticolimbic responses to a rewarding social cue. Dev. Brack, K.E., Watkins, N., Pyner, S., Coote, J.H., 2007. A physiological role for nitric
Neurobiol. 73, 856–869. oxide in the centrally mediated sympathetic and somatomotor ejaculatory response
Benelli, A., Arletti, R., Basaglia, R., Bertolini, A., 1993. Male sexual behavior: further in anesthetized male wistar rats. Neuroscience 150, 487–497.
studies on the role of alpha 2-adrenoceptors. Pharmacol. Res. 28, 35–45. Brackett, N.L., Edwards, D.A., 1984. Medial preoptic connections with the midbrain
Benelli, A., Bertolini, A., Poggioli, R., Cavazutti, E., Calza, L., Giardino, L., Arletti, R., tegmentum are essential for male sexual behavior. Physiol. Behav. 32, 79–84.
1995. Nitric oxide is involved in male sexual behavior of rats. Eur. J. Pharmacol. Brackett, N.L., Iuvone, P.M., Edwards, D.A., 1986. Midbrain lesions, dopamine and
294, 505–510. male sexual behavior. Behav. Brain Res. 20, 231–240.
Benelli, A., Zanoli, P., Bertolini, A., 1990. Effect of clenbuterol on sexual behavior in Breigeiron, M.K., Morris, M., Lucion, A.B., Sanvito, G.L., 2002. Effects of angiotensin II
male rats. Physiol. Behav. 47, 373–376. microinjected into medial amygdala on male sexual behavior in rats. Horm. Behav.
Berendsen, H.H., Broekkamp, C.L., 1987. Drug-induced penile erections in rats, 41, 267–274.
indications of serotoninlB receptor mediation. Eur. J. Pharmacol. 135, 279–287. Bressler, S.C., Baum, M.J., 1996. Sex comparison of neuronal Fos immunoreactivity in
Bermant, G., Taylor, L., 1969. Interactive effects of experience and olfactory bulb the rat vomeronasal projection circuit after chemosensory stimulation. Neurosci-
lesions in male rat copulation. Physiol. Behav. 4, 13–17. ence 71, 1063–1072.
Bernabé, J., Rampin, O., Sachs, B.D., Giuliano, F., 1999. Intracavernous pressure Brindley, G.S., 1986. Pilot experiments on the actions of drugs injected into the human
during erection in rats: an integrative approach based on telemetric recording. Am. corpus cavernosum penis. Br. J. Pharmacol. 87, 495–500.
J. Physiol. 276, R441–R449. Brom, M., Both, S., Laan, E., Everaerd, W., Spinhoven, P., 2014. The role of
Berridge, K.C., Robinson, T.E., 1998. What is the role of dopamine in reward: hedonic conditioning, learning and dopamine in sexual behavior: a narrative review of
impact, reward learning, or incentive salience? Brain Res. Rev. 28, 309–369. animal and human studies. Neurosci. Biobehav. Rev. 38, 38–59.
Best, T.K., Marson, L., Thor, K.B., Burgard, E.C., 2013. Synaptic activation of bul- Burnett, A.L., 2006. Nitric oxide in the penis-science and therapeutic implications from
bospongiosus motoneurons via dorsal gray commissural inputs. J. Neurophysiol. erectile dysfunction to priapism. J. Sex. Med. 3, 578–582.
109, 58–67. Burnett, A.L., Nelson, R.J., Calvin, D.C., Liu, J.X., Demas, G.E., Klein, S.L.,
Beyer, C., Contreras, J.L., Moralí, G., Larsson, K., 1981. Effects of castration and sex Kriegsfeld, L.J., Dawson, T.M., Snyder, S.H., 1996. Nitric oxide-dependent
steroid treatment on the motor copulatory pattern of the rat. Physiol. Behav. 27, penile erection in mice lacking neuronal nitric oxide synthase. Mol. Med. 2,
727–730. 288–296.
Beyer, C., Moralí, G., Naftolin, F., Larsson, K., Perez-Palacios, G., 1976. Effect of Burns-Cusato, M., Scordalakes, E.M., Rissman, E.R., 2004. Of mice and missing
some antiestrogens and aromatase inhibitors on androgen induced sexual behavior data: what we know (and need to learn) about male sexual behavior. Physiol.
in castrated male rats. Horm. Behav. 7, 353–363. Behav. 83, 217–232.
Male Sexual Behavior 43

Butera, P.C., Czaja, J.A., 1989. Effects of intracranial implants of dihydrotestosterone Chehensse, C., Bahrami, S., Denys, P., Clement, P., Bernabe, J., Giuliano, F., 2013.
on the reproductive physiology and behavior of male guinea pigs. Horm. Behav. 23, The spinal control of ejaculation revisited: a systematic review and meta-analysis
424–431. of anejaculation in spinal cord injured patients. Hum. Reprod. Update 19,
Can, A., Domjan, M., Delville, Y., 2007. Sexual experience modulates neuronal activity 507–526.
in male Japanese quail. Horm. Behav. 52, 590–599. Chen, J.C., Tsai, H.W., Yeh, K.Y., Tai, M.Y., Tsai, Y.F., 2007. Male sexual behavior and
Canseco-Alba, A., Rodriguez-Manzo, G., 2013. Anandamide transforms noncopulating catecholamine levels in the medial preoptic area and arcuate nucleus in middle-
rats into sexually active animals. J. Sex. Med. 10, 686–693. aged rats. Brain Res. 1184, 186–192.
Canseco-Alba, A., Rodriguez-Manzo, G., 2014. Low anandamide doses facilitate male Chen, J.C., Tsai, H.W., Yeh, K.Y., Tai, M.Y., Tsai, Y.F., 2008. Correlation of cate-
rat sexual behaviour through the activation of CB1 receptors. Psychopharmacology cholamine levels in the bed nucleus of the stria terminalis and reduced sexual
(Berlin) 231, 4071–4080. behavior in middle-aged male rats. J. Gerontol. A Biol. Sci. Med. Sci. 63,
Canteras, N.S., Simerly, R.B., Swanson, L.W., 1992. Connections of the posterior 678–682.
nucleus of the amygdala. J. Comp. Neurol. 324, 143–179. Chen, K.K., Chang, L.S., 2003. Effect of excitatory amino acid receptor agonists on
Canteras, N.S., Simerly, R.B., Swanson, L.W., 1995. Organization of projections from penile erection after administration into paraventricular nucleus of hypothalamus in
the medial nucleus of the amygdala: a PHAL study in the rat (published erratum the rat. Urology 62, 575–580.
appears in J. Comp. Neurol. 1996, 369, 328–330). J. Comp. Neurol. 360, Chen, K.K., Chan, J.Y.H., Chang, L.S., Chen, M.T., Chan, S.H.H., 1992. Elicitation of
213–245. penile erection following activation of the hippocampal formation in the rat. Neu-
Cantor, J.M., Binik, Y.M., Pfaus, J.G., 1999. Chronic fluoxetine inhibits sexual behavior rosci. Lett. 141, 218–222.
in the male rat: reversal with oxytocin. Psychopharmacology (Berlin) 144, Chitaley, K., Wingard, C.J., Clinton Webb, R., Branam, H., Stopper, V.S., Lewis, R.W.,
355–362. Mills, T.M., 2001. Antagonism of Rho-kinase stimulates rat penile erection via
Carmichael, M.S., Humbert, R., Dixen, J., Palmisano, G., Greenleaf, W., a nitric oxide-independent pathway. Nat. Med. 7, 119–122.
Davidson, J.M., 1987. Plasma oxytocin increases in the human sexual response. Christ, G.J., 1995. The penis as a vascular organ: the importance of corporal smooth
J. Clin. Endocrinol. Metab. 64, 27–31. muscle tone in the control of erection. Urol. Clin. N. Am. 22, 727–745.
Carro-Juárez, M., Rodríguez-Manzo, G., 2000. Sensory and motor aspects of the coital Christ, G.J., Wang, H.Z., Venkateswarlu, K., Zhao, W., Day, N.S., 1999. Ion channels
reflex in the spinal male rat. Behav. Brain Res. 108, 97–103. and gap junctions: their role in erectile physiology, dysfunction, and future therapy.
Carro-Juárez, M., Rodríguez-Manzo, G., 2001. Exhaustion of the coital reflex in spinal Mol. Urol. 3, 61–73.
male rats is reversed by the serotonergic agonist 8-OH-DPAT. Behav. Brain Res. Clancy, A.N., Zumpe, D., Michael, R.P., 1995. Intracerebral infusion of an aromatase
118, 161–168. inhibitor, sexual behavior and brain estrogen receptor-like immunoreactivity in
Carro-Juárez, M., Rodríguez-Manzo, G., 2003. Yohimbine reverses the exhaustion of intact male rats. Neuroendocrinology 61, 98–111.
the coital reflex in spinal male rats. Behav. Brain Res. 141, 43. Clancy, A.N., Zumpe, D., Michael, R.P., 2000. Estrogen in the medial preoptic area of
Carro-Juárez, M., Rodríguez-Manzo, G., 2005a. Evidence for the presence and male rats facilitates copulatory behavior. Horm. Behav. 38, 86–93.
functioning of the spinal generator for ejaculation in neonatal male rats. Int. J. Clark, J.T., 1991. Suppression of copulatory behavior in male rats following central
Impot. Res. 17, 270–276. administration of clonidine. Neuropharmacology 30, 373–382.
Carro-Juárez, M., Rodríguez-Manzo, G., 2005b. Role of the genital sensory information Clark, J.T., Smith, E.R., 1990. Clonidine suppresses copulatory behavior and erectile
in the control of the functioning of the spinal generator for ejaculation. Int. J. Impot. reflexes in male rats: lack of effect of naloxone pretreatment. Neuroendocrinology
Res. 17, 114–120. 51, 357–364.
Carro-Juárez, M., Rodriguez-Manzo, G., 2006. Alpha-adrenergic agents modulate the Clark, J.T., Smith, E.R., Davidson, J.M., 1984. Enhancement of sexual motivation in
activity of the spinal pattern generator for ejaculation. Int. J. Impot. Res. 18, male rats by yohimbine. Science 225, 847–849.
32–38. Clark, J.T., Smith, E.R., Davidson, J.M., 1985. Evidence for the modulation of sexual
Carro-Juárez, M., Rodríguez-Manzo, G., 2008. The spinal pattern generator for behavior by a-adrenoceptors in male rats. Neuroendocrinology 41, 36–43.
ejaculation. Brain Res. Rev. 58, 106–120. Clark, T.K., 1975. Sexual inhibition is reduced by rostral midbrain lesions in the male
Carro-Juárez, M., Rodríguez-Manzo, G., 2009. Participation of endogenous opioids in rat. Science 190, 169–171.
the inhibition of the spinal generator for ejaculation in rats. J. Sex. Med. 6, Clark, T.K., 1980. Male rat sexual behavior compared after 6-OHDA and electrolytic
3045–3055. lesions in the dorsal NA bundle region of the midbrain. Brain Res. 202,
Carro-Juárez, M., Cruz, S.L., Rodriguez-Manzo, G., 2003. Evidence for the involvement 429–443.
of a spinal pattern generator in the control of the genital motor pattern of ejacu- Claro, F., Segovia, S., Guilamón, A., Del Abril, A., 1995. Lesions in the medial posterior
lation. Brain Res. 975, 222–228. region of the BST impair sexual behavior in sexually experienced and inexperienced
Carro-Juárez, M., Rodriguez-Manzo, G., de Lourdes Rodriguez Pena, M., Franco, M.A., male rats. Brain Res. Bull. 36, 1–10.
2014. Rhythmic motor patterns accompanying ejaculation in spinal cord- Clemens, L.G., Gladue, B.A., 1977. Effect of prostaglandin E2 on masculine sexual
transected male rats. Int. J. Impot. Res. 26, 191–195. behaviour in the rat. J. Endocrinol. 75, 383–389.
Castelli, M.P., Piras, A.P., Melis, T., Succu, S., Sanna, F., Melis, M.R., Collu, S., Clement, P., Bernabe, J., Denys, P., Alexandre, L., Giuliano, F., 2007a. Ejaculation
Grazia Ennas, M., Diaz, G., Mackie, K., Argiolas, A., 2007. Cannabinoid CB 1 induced by i.c.v. injection of the preferential dopamine D(3) receptor agonist 7-
receptors in the paraventricular nucleus and central control of penile erection: hydroxy-2-(di-N-propylamino)tetralin in anesthetized rats. Neuroscience 145,
immunocytochemistry, autoradiography and behavioral studies. Neuroscience 605–610.
147, 197–206. Clement, P., Bernabe, J., Gengo, P., Denys, P., Laurin, M., Alexandre, L., Giuliano, F.,
de Castilhos, J., Marcuzzo, S., Forti, C.D., Frey, R.M., Stein, D., Achaval, M., Rasia- 2007b. Supraspinal site of action for the inhibition of ejaculatory reflex by
Filho, A.A., 2006. Further studies on the rat posterodorsal medial amygdala: dapoxetine. Eur. Urol. 51, 825–832.
dendritic spine density and effect of 8-OH-DPAT microinjection on male sexual Constantini, R.M., Park, J.H., Beery, A.K., Paul, M.J., Ko, J.J., Zucker, I., 2007. Post-
behavior. Brain Res. Bull. 69, 131–139. castration retention of reproductive behavior and olfactory preferences in male
Cavalcante, J.C., Bittencourt, J.C., Elias, C.F., 2014. Distribution of the neuronal inputs Siberian hamsters: role of prior experience. Horm. Behav. 51, 149–155.
to the ventral premammillary nucleus of male and female rats. Brain Res. 1582, Cooke, B.M., Breedlove, S.M., Jordan, C., 2003. Both estrogen receptors and
77–90. androgen receptors contribute to testosterone-induced changes in the morphology
Cavallini, G., Caracciolo, S., Vitali, G., Modenini, F., Biagiotti, G., 2004. Carnitine of the medial amygdala and sexual arousal in male rats. Horm. Behav. 43,
versus androgen administration in the treatment of sexual dysfunction, depressed 335–346.
mood, and fatigue associated with male aging. Urology 63, 641–646. Cooke, B.M., Tabibnia, G., Breedlove, S.M., 1999. A brain sexual dimorphism
Cera, N., Delli Pizzi, S., Di Pierro, E.D., et al., 2012. Macrostructural alterations of controlled by adult circulating androgens. Proc. Natl. Acad. Sci. U.S.A. 96,
subcortical grey matter in psychogenic erectile dysfunction. PLoS One 7, 7538–7540.
e39118. Coolen, L.M., 2005. Neural control of ejaculation. J. Comp. Neurol. 493, 39–45.
Chambers, K.C., Thornton, J.E., Roselli, C.E., 1991. Age-related deficits in brain Coolen, L.M., Wood, R.I., 1998. Bidirectional collections of the medial amygdaloid
androgen binding and metabolism, testosterone and sexual behavior of male rats. nucleus in the Syrian hamster brain: simultaneous anterograde and retrograde tract
Neurobiol. Aging 12, 123–130. tracing. J. Comp. Neurol. 399, 189–209.
Chan, J.S., Snoeren, E.M., Cuppen, E., Waldinger, M.D., Olivier, B., Oosting, R.S., Coolen, L.M., Wood, R.I., 1999. Testosterone stimulation of the medial preoptic area
2011. The serotonin transporter plays an important role in male sexual behavior: and medial amygdala in the control of male hamster sexual behavior: redundancy
a study in serotonin transporter knockout rats. J. Sex. Med. 8, 97–108. without amplification. Behav. Brain Res. 98, 143–153.
44 Male Sexual Behavior

Coolen, L.M., Allard, J., Truitt, W.A., McKenna, K.E., 2004a. Central regulation of Dobberfuhl, A.D., Oti, T., Sakamoto, H., Marson, L., 2014. Identification of CNS
ejaculation. Physiol. Behav. 83, 203–215. neurons innervating the levator ani and ventral bulbospongiosus muscles in male
Coolen, L.M., Fitzgerald, M.E., Yu, L., Lehman, M.N., 2004b. Activation of m opioid rats. J. Sex. Med. 11, 664–677.
receptors in the medial preoptic area following copulation in male rats. Neuro- Dominguez, J.M., Hull, E.M., 2001. Stimulation of the medial amygdala enhances
science 124, 11–21. medial preoptic dopamine release: implications for male rat sexual behavior. Brain
Coolen, L.M., Olivier, B., Peters, H.J., Veening, J.G., 1997a. Demonstration of Res. 917, 225–229.
ejaculation-induced neural activity in the male rat brain using 5-HT1A agonist 8-OH- Dominguez, J.M., Gil, M., Hull, E.M., 2006a. Preoptic glutamate facilitates male sexual
DPAT. Physiol. Behav. 62, 881–891. behavior. J. Neurosci. 26, 1699–1703.
Coolen, L.M., Peters, H.J., Veening, J.G., 1996. Fos immunoreactivity in the rat brain Dominguez, J.M., Hull, E.M., 2010. Serotonin impairs copulation and attenuates
following consummatory elements of sexual behavior: a sex comparison. Brain Res. ejaculation-induced glutamate activity in the medial preoptic area. Behav. Neurosci.
738, 67–82. 124, 554–557.
Coolen, L.M., Peters, H.J., Veening, J.G., 1997b. Distribution of Fos immunoreactivity Dominguez, J.M., Balfour, M.E., Lee, H.S., Brown, J.L., Davis, B.A., Coolen, L.M.,
following mating versus anogenital investigation in the male rat brain. Neuroscience 2007. Mating activates NMDA receptors in the medial preoptic area of male rats.
77, 1151–1161. Behav. Neurosci. 121, 1023–1031.
Coolen, L.M., Peters, H.J., Veening, J.G., 1998. Anatomical interrelationships of the Dominguez, J.M., Brann, J.H., Gil, M., Hull, E.M., 2006b. Sexual experience increases
medial preoptic area and other brain regions activated following male sexual nitric oxide synthase in the medial preoptic area of male rats. Behav. Neurosci.
behavior: a combined c-fos and tract-tracing study. J. Comp. Neurol. 397, 120, 1389–1394.
421–435. Dominguez, J., Riolo, J.V., Xu, Z., Hull, E.M., 2001. Regulation by the medial
Coolen, L.M., Veening, J.G., Petersen, D.W., Shipley, M.T., 2003. Parvocellular amygdala of copulation and medial preoptic dopamine release. J. Neurosci. 21,
subparafascicular thalamic nucleus in the rat: anatomical and functional 349–355.
compartmentalization. J. Comp. Neurol. 463, 117–131. Dominguez-Salazar, E., Portillo, W., Velazquez-Moctezuma, J., Paredes, R.G., 2003.
Cornil, C.A., Ball, G.F., Balthazart, J., 2006a. Functional significance of the rapid Facilitation of male-like coital behavior in female rats by kindling. Behav. Brain Res.
regulation of brain estrogen action: where do the estrogens come from? Brain Res. 140, 57–64.
1126, 2–26. Dominguez-Salazar, E., Naser, H.F., Velazquez-Moctezuma, J., 2014. D1-like antag-
Cornil, C.A., Balthazart, J., Motte, P., Massotte, L., Seutin, V., 2002. Dopamine onist blocks conditioned place preference induced by ejaculation in male rats.
activates noradrenergic receptors in the preoptic area. J. Neurosci. 22, Behav. Brain Res. 269, 15–19.
9320–9330. Domjan, M., 1992. Adult learning and mate choice: possibilities and experimental
Cornil, C.A., Taziaux, M., Baillien, M., Ball, G.F., Balthazart, J., 2006b. Rapid effects of evidence. Am. Zool. 32, 48–61.
aromatase inhibition on male reproductive behaviors in Japanese quail. Horm. Dorsa, D.M., Smith, E.R., 1980. Facilitation of mounting behavior in male rats by
Behav. 49, 45–67. intracranial injections of luteinizing hormone-releasing hormone. Regul. Pept. 1,
Corona, G., Mannucci, E., Fisher, A.D., Lotti, F., Ricca, V., Balercia, G., Petrone, L., 147–155.
Forti, G., Maggi, M., 2007. Effect of hyperprolactinemia in male patients consulting Dorsa, D.M., Smith, E.R., Davidson, J.M., 1981. Endocrine and behavioral effects of
for sexual dysfunction. J. Sex. Med. 4, 1485–1493. continuous exposure of male rats to a potent luteinizing hormone-releasing
Cross, E., Roselli, C.E., 1999. 17beta-estradiol rapidly facilitates chemoinvestigation hormone (LHRH) agonist: evidence for central nervous system actions of LHRH.
and mounting in castrated male rats. Am. J. Physiol. 276 (5 Pt 2), R1346–R1350. Endocrinology 109, 729–735.
Cruz-Sánchez, F.F., Moral, A., Tolosa, E., de Belleroche, J., Rossi, M.L., 1998. Drago, F., Busa, L., 1997. Dose-dependent effects of acute kainic acid treatment on
Evaluation of neuronal loss, astrocytosis and abnormalities of cytoskeletal sexual behavior of male rats. Brain Res. 778, 135–139.
components of large motor neurons in the human anterior horn in aging. J. Neural Du, J., Hull, E.M., 1999. Effects of testosterone on neuronal nitric oxide synthase and
Transm. 105, 689–701. tyrosine hydroxylase. Brain Res. 836, 90–98.
Cyders, M.A., Dzemidzic, M., Eiler, W.J., Kareken, D.A., 2016. An fMRI study of Du, J., Lorrain, D.S., Hull, E.M., 1998. Castration decreases extracellular, but
responses to sexual stimuli as a function of gender and sensation seeking: increases intracellular, dopamine in medial preoptic area of male rats. Brain Res.
a preliminary analysis. J. Sex Res. 1–7. 782, 11–17.
Dai, Y., Chitaly, K., Webb, R.C., Lewis, R.W., Mills, T.M., 2004. Topical application of Dudley, C.A., Moss, R.L., 1999. Activation of an anatomically distinct subpopulation of
a Rho-kinase inhibitor in rats causes penile erection. Int. J. Impot. Res. 16, accessory olfactory bulb neurons by chemosensory stimulation. Neuroscience 91,
294–298. 1549–1556.
Dail, W.G., Minorsky, N., Moll, M.A., Manzanares, K., 1986. The hypogastric nerve Dula, E., Keating, W., Siami, P.F., Edmonds, A., O’Neil, J., Buttler, S., 2000. Efficacy
pathway to penile erectile tissue: histochemical evidence supporting a vasodilator and safety of fixed-dose and dose-optimization regimens of sublingual apomor-
role. J. Auton. Nerv. Syst. 15, 341–349. phine versus placebo in men with erectile dysfunction. The Apomorphine Study
Damsma, G., Pfaus, J.G., Wenkstern, D., Phillips, A.G., Fibiger, H.C., 1992. Sexual Group. Urology 56, 130–135.
behavior increases dopamine transmission in the nucleus accumbens and striatum Durán, I., Gil, L., Cueva-Rolón, R., 2000. Masculine copulatory behavior is facilitated
of male rats: comparison with novelty and locomotion. Behav. Neurosci. 106, by intrathecally administered muscarine. Exp. Brain Res. 134, 490–496.
181–191. Düsing, R., 2005. Sexual dysfunction in male patients with hypertension: influence of
Davidson, J.M., Stefanick, M.L., Sachs, B.D., Smith, E.R., 1978. Role of androgen in antihypertensive drugs. Drugs 65, 773–786.
sexual reflexes of the male rat. Physiol. Behav. 21, 141–146. Edinger, K.L., Frye, C.A., 2007. Sexual experience of male rats influences anxiety-like
Depoortere, R., Bardin, L., Rodrigues, M., Abrial, E., Aliaga, M., Newman-Tancredi, A., behavior and androgen levels. Physiol. Behav. 92, 443–453.
2009. Penile erection and yawning induced by dopamine D2-like receptor agonists Edwards, D.A., Einhorn, L.C., 1986. Preoptic and midbrain control of sexual motiva-
in rats: influence of strain and contribution of dopamine D2, but not D3 and D4 tion. Physiol. Behav. 37, 329–335.
receptors. Behav. Pharmacol. 20, 303–311. Edwards, D.A., Isaacs, S., 1991. Zona incerta lesions: effects on copulation, partner-
Dennison, E., Bain, P.A., Bartke, A., Meliska, C.J., 1996. Systemically admin- preference and other socio-sexual behaviors. Behav. Brain Res. 44, 145–150.
istered gonadotrophin-releasing hormone enhances copulatory behavior in Edwards, D.A., Griffis, K.T., Tardival, C., 1990. Olfactory bulb removal: effects on
castrated, testosterone-treated hyperprolactinemic male rats. Int. J. Androl. sexual behavior and partner-preference in male rats. Physiol. Behav. 48,
19, 253–259. 447–450.
Dermon, C.R., Stamatakis, A., Tlemcani, O., Balthazart, J., 1999. Performance of Edwards, D.A., Walter, B., Liang, P., 1996. Hypothalamic and olfactory control of
appetitive or consummatory components of male sexual behavior is mediated by sexual behavior and partner preference in male rats. Physiol. Behav. 60,
different brain areas: a 2-deoxyglucose autoradiographic study. Neuroscience 94, 1347–1354.
1261–1277. Escrig, A., Marin, R., Mas, M., 1999. Repeated PGE1 treatment enhances nitric oxide
Dewsbury, D.A., 1983. Recovery from sexual satiety in deer mice (Peromyscus and erection responses to nerve stimulation in the rat penis by upregulating
maniculatus bairdi). J. Comp. Psychol. 97, 34–42. constitutive NOS isoforms. J. Urol. 162, 2205–2210.
Diederichs, W., Stief, C.G., Benard, F., Bosch, R., Lue, T.F., Tanagho, E.A., 1991. The Everitt, B.J., 1990. Sexual motivation: a neural and behavioral analysis of the
sympathetic role as an antagonist of erection. Urol. Res. 19, 123–126. mechanisms underlying appetitive and copulatory responses of male rats. Neu-
Di Sebastiano, A.R., Wilson-Perez, H.E., Lehman, M.N., Coolen, L.M., 2011. Lesions rosci. Biobehav. Rev. 14, 217–232.
of orexin neurons block conditioned place preference for sexual behavior in male Exintaris, B., Nguyen, D.T., Dey, A., Lang, R.J., 2006. Spontaneous electrical activity
rats. Horm. Behav. 59, 1–8. in the prostate. Auton. Neurosci. 126–127, 371–379.
Male Sexual Behavior 45

Facchinetti, P., Giuliano, F., Laurin, M., Bernabe, J., Clement, P., 2014. Direct brain Foidart, A., Meddle, S.L., Balthazart, J., 1999. Mating-induced Fos and aromatase are
projections onto the spinal generator of ejaculation in the rat. Neuroscience 272, not co-localized in the preoptic area. Neuroreport 10, 907–912.
207–216. Fonseca, M.I., Ni, Y.G., Dunning, D.D., Miledi, R., 2001. Distribution of serotonin 2A,
Fargo, K.N., Iwema, C.L., Clark-Phelps, M.C., Sengelaub, D.R., 2007. Exogenous 2C and 3 receptor mRNA in spinal cord and medulla oblongata. Brain Res. Mol.
testosterone reverses age-related atrophy in a spinal neuromuscular system. Horm. Brain Res. 89, 11–19.
Behav. 51, 20–30. Forbes-Lorman, R., Auger, A.P., Auger, C.J., 2014. Neonatal RU-486 (mifepristone)
Ferguson, A.V., Latchford, K.J., Samson, W.K., 2008. The paraventricular nucleus of exposure increases androgen receptor immunoreactivity and sexual behavior in
the hypothalamus – a potential target for integrative treatment of autonomic male rats. Brain Res. 1543, 143–150.
dysfunction. Expert Opin. Ther. Targets 12, 717–727. Foreman, M.M., Hall, J.L., 1987. Effects of D2-dopaminergic receptor stimulation on
Fernandez-Fewell, G.D., Meredith, M., 1994. C-fos expression in vomeronasal path- male rat sexual behavior. J. Neural Transm. 68, 153–170.
ways of mated or pheromone-stimulated male golden hamsters: contribution from Foreman, M.M., Hall, J.L., Love, R.L., 1989. The role of the 5-HT2 receptor in the
vomeronasal sensory input and expression related to mating performance. regulation of sexual performance of male rats. Life Sci. 45, 1263–1270.
J. Neurosci. 14, 3643–3654. Foresta, C., Caretta, N., Rossato, M., Garolla, A., Ferlin, A., 2004. Role of androgens in
Fernández-Guasti, A., Escalante, A.L., 1991. Role of presynaptic serotonergic erectile function. J. Urol. 171, 2358–2362 quiz 2435.
receptors o the mechanism of action of 5-HT1A and 5-HT1B agonists on masculine Forsberg, G., Wiesenfeld-Hallin, Z., Eneroth, R., Södersten, P., 1987. Sexual behavior
sexual behavior: physiological and pharmacological implications. J. Neural Transm. induces naloxone-reversible hypoalgesia in male rats. Neurosci. Lett. 81, 151–154.
85, 95–107. Freund, T.F., Katona, I., Piomelli, D., 2003. Role of endogenous cannabinoids in
Fernández-Guasti, A., Rodríguez-Manzo, G., 1992. Further evidence showing that synaptic signaling. Physiol. Rev. 83, 1017–1066.
the inhibitory actions of serotonin on rat masculine sexual behavior is mediated Frye, C.A., 2001. The role of neurosteroids and non-genomic effects of progestins and
after the stimulation of 5-HT1B receptors. Pharmacol. Biochem. Behav. 42, androgens in mediating sexual receptivity of rodents. Brain Res. Rev. 37, 201–222.
529–533. Fumero, B., Fernandez-Vera, J.R., Gonzalez-Mora, J.L., Mas, M., 1994. Changes in
Fernández-Guasti, A., Rodríguez-Manzo, G., 1997. 8-OH-DPAT and male rat sexual monoamine turnover in forebrain areas associated with masculine sexual behavior:
behavior: partial blockade by noradrenergic lesion and sexual exhaustion. Phar- a microdialysis study. Brain Res. 662, 233–239.
macol. Biochem. Behav. 56, 11–116. van Furth, W.R., van Ernst, M.G., van Ree, J.M., 1995. Opioids and sexual behavior of
Fernández-Guasti, A., Escalante, A.L., Agmo, A., 1989. Inhibitory action of various 5- male rats: involvement of the medial preoptic area. Behav. Neurosci. 109,
HT1B receptor agonists on rat masculine sexual behavior. Pharmacol. Biochem. 123–134.
Behav. 34, 811–816. van Furth, W.R., van Ree, J.M., 1996. Sexual motivation: involvement of endogenous
Fernández-Guasti, A., Escalante, A.L., Ahlenius, S., Hillegaart, V., Larsson, K., 1992. opioids in the ventral tegmental area. Brain Res. 729, 20–28.
Stimulation of 5-HT1A and 5-HT1B receptors in brain regions and its effects on male Garduno-Gutierrez, R., Guadarrama-Bazante, L., Leon-Olea, M., Rodriguez-Manzo, G.,
rat sexual behavior. Eur. J. Pharmacol. 210, 121–129. 2013a. Endogenous opioids mediate the sexual inhibition but not the drug
Fernández-Guasti, A., Larsson, K., Beyer, C., 1986a. GABAergic control of masculine hypersensitivity induced by sexual satiation in male rats. Behav. Neurosci. 127,
sexual behavior. Pharmacol. Biochem. Behav. 24, 1065–1070. 458–464.
Fernández-Guasti, A., Larsson, K., Vega-Sanabria, J., 1986b. Depression of post- Garduno-Gutierrez, R., Leon-Olea, M., Rodriguez-Manzo, G., 2013b. The meso-
ejaculatory ultrasonic vocalization by (þ) bicuculline. Behav. Brain Res. 19, 35–39. limbic system participates in the naltrexone-induced reversal of sexual
Fernández-Guasti, A., Swaab, D., Rodríguez-Manzo, G., 2003. Sexual behavior exhaustion: opposite effects of intra-VTA naltrexone administration on copulation
reduces hypothalamic androgen receptor immunoreactivity. Psychoneur- of sexually experienced and sexually exhausted male rats. Behav. Brain Res.
oendocrinology 28, 501–512. 256, 64–71.
Ferrari, F., Giuliani, D., 1995. Sexual attraction and copulation in male rats: effects of Garduno-Gutierrez, R., Leon-Olea, M., Rodriguez-Manzo, G., 2013c. Different amounts
the dopamine agonist SND 919. Pharmacol. Biochem. Behav. 50, 29–34. of ejaculatory activity, a natural rewarding behavior, induce differential mu and
Ferrari, F., Giuliani, D., 1996a. Behavioral effects induced by the dopamine D3 agonist delta opioid receptor internalization in the rat’s ventral tegmental area. Brain Res.
7-OH-DPAT in sexually-active and -inactive male rats. Neuropharmacology 35, 1541, 22–32.
279–284. Garelick, T., Swann, J., 2014. Testosterone regulates the density of dendritic spines in
Ferrari, F., Giuliani, D., 1996b. Influence of eticlopride on cocaine- and DA D2 the male preoptic area. Horm. Behav. 65, 249–253.
agonist-induced behavioral effects in rats. Pharmacol. Biochem. Behav. 53, Georgiadis, J.R., Farrell, M.J., Boessen, R., et al., 2010. Dynamic subcortical blood
525–530. flow during male sexual activity with ecological validity: a perfusion fMRI study.
Ferrari, F., Ottani, A., Giuliani, D., 2000. Inhibitory effects of the cannabinoid agonist Neuroimage 50, 208–216.
HU 210 on rat sexual behavior. Physiol. Behav. 69, 547–554. Gerstenberg, T.C., Levin, R.J., Wagner, G., 1990. Erection and ejaculation in man.
Ferrari, F., Ottani, A., Giuliani, D., 2002. Influence of sildenafil on central dopamine- Assesment of the electromyographic activity of the bulbocavernosus and ischio-
mediated behavior in male rats. Life Sci. 70, 1501–1508. cavernosus muscles. Br. J. Urol. 65, 395–402.
Ferris, C.F., Snowdon, C.T., King, J.A., Duong, T.Q., Ziegler, T.E., Ugurbil, K., Gerstenberg, T.C., Nordling, J., Hald, T., 1991. Evoked potentials from the lower
Ludwig, R., Schultz-Darken, N.J., Wu, Z., Olson, D.P., Sullivan Jr., J.M., urinary tract. II. The spino-cortical neuraxis. A methodological study. Scand. J. Urol.
Tannenbaum, P.L., Vaughan, J.T., 2001. Functional imaging of brain activity in Nephrol. Suppl. 138, 41–46.
conscious monkeys responding to sexually arousing cues. Neuroreport 12, Gessa, G.L., Paglietti, E., Pellegrini-Quarantotti, B., 1979. Induction of copulatory
2231–2236. behavior in sexually inactive rats by naloxone. Science 204, 203–205.
Fewell, G.D., Meredith, M., 2002. Experience facilitates vomeronasal and olfactory Geyer, L.A., Barfield, R.J., 1978. Influence of gonadal hormones and sexual behavior
influence on Fos expression in medial preoptic area during pheromone exposure or on ultrasonic vocalization in rats: I. Treatment of females. J. Comp. Physiol.
mating in male hamsters. Brain Res. 941, 91–106. Psychol. 92, 438–446.
Fiber, J.M., Adages, P., Swain, J.M., 1993. Pheromones induce c-fos in limbic areas Gibson, M.J., Wu, T.J., Miller, G.M., Silverman, A.J., 1997. What nature’s knockout
regulating male hamster mating behavior. Neuroreport 4, 871–874. teaches us about GnRH activity: hypogonadal mice and neuronal grafts. Horm.
Finn, P.D., Yahr, P., 1994. Projection of the sexually dimorphic area of the gerbil Behav. 31, 212–220.
hypothalamus to the retrorubral field is essential for male sexual behavior: role of Gil, M., Bhatt, R., Picotte, K.B., Hull, E.M., 2011. Oxytocin in the medial preoptic area
A8 and other cells. Behav. Neurosci. 108, 362–378. facilitates male sexual behavior in the rat. Horm. Behav. 59, 435–443.
Finn, P.D., Yahr, P., 2005. Projection from the ventral bed nucleus of the stria ter- Gil, M., Bhatt, R., Picotte, K.B., Hull, E.M., 2013. Sexual experience increases oxytocin
minalis to the retrorubral field in rats and the effects of cells in these areas on receptor gene expression and protein in the medial preoptic area of the male rat.
mating in male rats versus gerbils. Horm. Behav. 47, 123–138. Psychoneuroendocrinology 38, 1688–1697.
Fiorino, D.F., Coury, A., Phillips, A.G., 1997. Dynamic changes in nucleus accumbens Gill, C.J., Wersinger, S.R., Veney, S.L., Rissman, E.F., 1998. Induction of c-fos-like
dopamine efflux during the Coolidge effect in male rats. J. Neurosci. 17, immunoreactivity in musk shrews after mating. Brain Res. 811, 21–28.
4849–4855. Giordano, M., Guemes, M., Lopez-Arias, V., Paredes, R.G., 1998. Socio-sexual
Fisher, A.E., 1962. Effects of stimulus variation on sexual satiation in the male rat. behavior in male rats after lesions of the dorsolateral tegmentum. Physiol.
J. Comp. Physiol. Psychol. 55, 614–620. Behav. 65, 89–94.
Floody, O.R., 2013. Oxotremorine delays and scopolamine accelerates sexual Giuliani, D., Ferrari, F., 1996. Differential behavioral response to dopamine D2 agonists
exhaustion when applied to the preoptic area in male hamsters. Pharmacol. Bio- by sexually naïve, sexually active, and sexually inactive male rats. Behav. Neurosci.
chem. Behav. 110, 75–88. 110, 802–808.
46 Male Sexual Behavior

Giuliano, F., Allard, J., 2001. Dopamine and sexual function. Int. J. Impot. Res. 13 Guevara, M.A., Martinez-Pelayo, M., Arteaga Silva, M., Bonilla-Jaime, H., Hernandez-
(Suppl. 3), S18–S28. Gonzalez, M., 2008. Electrophysiological correlates of the mesoaccumbens system
Giuliano, F., Allard, J., 2002. Apomorphine SL (Uprima): preclinical and clinical during male rat sexual behaviour. Physiol. Behav. 95, 545–552.
experiences learned the first central nervous system-acting ED drug. Int. J. Impot. Gulia, K.K., Mallick, H.N., Kumar, V.M., 2003. Orexin A (hypocretin-1) application at
Res. 14 (Suppl. 1), S53–S56. the medial preoptic area potentiates male sexual behavior in rats. Neuroscience
Giuliano, F., Rampin, O., 2000. Central neural regulation of penile erection. Neurosci. 116, 921–923.
Biobehav. Rev. 24, 517–533. Haake, P., Exton, M.S., Haverkamp, J., Krämer, M., Leygraf, N., Hartmann, U.,
Giuliano, F., Rampin, O., 2004. Neural control of erection. Physiol. Behav. 83, Schedlowski, M., Krüger, T.H., 2002. Absence of orgasm-induced prolactin secretion
189–201. in a healthy multi-orgasmic male subject. Int. J. Impot. Res. 14, 133–135.
Giuliano, F., Clement, P., 2005. Physiology of ejaculation: emphasis on serotonergic Hadeishi, Y., Wood, R.I., 1996. Nitric oxide synthase in mating behavior circuitry of
control. Eur. Urol. 48, 408–417. male Syrian hamster brain. J. Neurobiol. 30, 480–492.
Giuliano, F., Bernabé, J., Brown, K., Droupy, S., Benoit, G., Rampin, O., 1997. Erectile Haensel, S.M., Slob, A., 1997. Flesinoxan: a prosexual drug for male rats. Eur. J.
response to hypothalamic stimulation in rats: role of peripheral nerves. Am. J. Pharmacol. 330, 1–9.
Physiol. 273, R1990–R1997. Halem, H.A., Cherry, J.A., Baum, M.J., 1999. Vomeronasal neuroepithelium and
Giuliano, F., Bernabé, J., Rampin, O., Courtois, F., Benoit, G., Rousseau, J.P., 1994. forebrain Fos responses to male pheromones in male and female mice.
Telemetric monitoring of intracavernous pressure in freely moving rats d;uring J. Neurobiol. 39, 249–263.
copulation. J. Urol. 152, 1271–1274. Hallbeck, M., Blomqvist, A., 1999. Spinal cord-projecting vasopressinergic neurons in
Giuliano, F., Rampin, O., Bernabé, J., Rousseau, J.P., 1995. Neural control of penile the rat paraventricular hypothalamus. J. Comp. Neurol. 411, 201–211.
erection in the rat. J. Auton. Nerv. Syst. 55, 36–44. Hannan, J.L., Albersen, M., Kutlu, O., et al., 2013. Inhibition of Rho-kinase improves
Giuliano, F., Rampin, O., Brown, K., Courtois, F., Benoit, G., Jardin, A., 1996. erectile function, increases nitric oxide signaling and decreases penile apoptosis in
Stimulation of the media1 preoptic area of the hypothalamus in the rat elicits a rat model of cavernous nerve injury. J. Urol. 189, 1155–1161.
increases in intracavernous pressure. Neurosci. Lett. 209, 1–4. Hannan, J.L., Kutlu, O., Stopak, B.L., et al., 2014. Valproic acid prevents penile
Goldstein, L.A., Sengelaub, D.R., 1994. Differential effects of dihydrotestosterone and fibrosis and erectile dysfunction in cavernous nerve-injured rats. J. Sex. Med. 11,
estrogen on the development of motoneuron morphology in a sexually dimorphic rat 1442–1451.
spinal nucleus. J. Neurobiol. 25, 878–892. Hansen, S., Köhler, C., Ross, S.B., 1982. On the role of the dorsal mesencephalic
Gomez, D.M., Newman, S.W., 1992. Differential projections of the anterior and tegmentum in the control of masculine sexual behavior in the rat: effects of
posterior regions of the media1 amygdaloid nucleus in the Syrian hamster. electrolytic lesions, ibotenic acid and DSP4. Brain Res. 240, 311–320.
J. Comp. Neurol. 317, 195–218. Harding, S.M., McGinnis, M.Y., 2003. Effects of testosterone in the VMN on copulation,
Gomez, L.E., Flores, R., Cano, P., Durán, I., Cueva-Rolón, R., 2005. Cholinergic partner preference, and vocalizations in male rats. Horm. Behav. 43, 327–335.
modulation of the urethro genital reflex in spinal cord-transected rats. Pharmacol. Harding, S.M., McGinnis, M.Y., 2004. Androgen receptor blockade in the MPOA or
Biochem. Behav. 81, 100–113. VMN: effects on male sociosexual behaviors. Physiol. Behav. 81, 671–680.
Gomez-Marrero, J., Feria, M., Mas, M., 1988. Stimulation of opioid receptors Harding, S.M., McGinnis, M.Y., 2005. Microlesions of the ventromedial nucleus of the
suppresses penile erectile reflexes and seminal emission in rats. Pharmacol. hypothalamus: effects on sociosexual behaviors in male rats. Behav. Neurosci.
Biochem. Behav. 31, 393–396. 119, 1227–1234.
Gomez-Martinez, L.E., Cueva-Rolon, R., 2009. Muscarinic receptor antagonism at the Harding, S.M., Velotta, J.P., 2011. Comparing the relative amount of testosterone
spinal cord level causes inhibitory effects on male rat sexual behavior. Behav. Brain required to restore sexual arousal, motivation, and performance in male rats. Horm.
Res. 203, 247–255. Behav. 59, 666–673.
Gonzalez, M.I., Farabollini, F., Albonetti, E., Wilson, C.A., 1994. Interactions between Hari Dass, S.A., Vyas, A., 2014. Copulation or sensory cues from the female augment
5-hydroxytryptamine (5-HT) and testosterone in the control of sexual and nonsexual Fos expression in arginine vasopressin neurons of the posterodorsal medial
behavior in male and female rats. Pharmacol. Biochem. Behav. 47, 591–601. amygdala of male rats. Front. Zool. 11, 42.
Gorzalka, B.B., Dang, S.S., 2012. Minireview: endocannabinoids and gonadal Harris, G.C., Aston-Jones, G., 2006. Arousal and reward: a dichotomy in orexin
hormones: bidirectional interactions in physiology and behavior. Endocrinology 153, function. Trends Neurosci. 29, 571–577.
1016–1024. Hart, B.L., 1968. Sexual reflexes and mating behavior in the male rat. J. Comp.
Gorzalka, B.B., Morrish, A.C., Hill, M.N., 2008. Endocannabinoid modulation of male Physiol. Psychol. 65, 453–460.
rat sexual behavior. Psychopharmacology (Berlin) 198, 479–486. Hart, B.L., Wallach, S.J.R., Melese-D’Hospital, P.Y., 1983. Differences in respon-
Gravitt, K., Marson, L., 2007. Effect of the destruction of cells containing the serotonin siveness to testosterone of penile reflexes and copulatory behavior of male rats.
reuptake transporter on urethrogenital reflexes. J. Sex. Med. 4, 322–330 Horm. Behav. 17, 274–283.
discussion 330–321. Hayashi, S., Kimura, T., 1974. Sex-attractant emitted by female mice. Physiol. Behav.
Gray, G.D., Smith, E.R., Davidson, J.M., 1980. Hormonal regulation of penile erection 13, 563–567.
in castrated male rats. Physiol. Behav. 24, 463–468. He, F., Yu, P., Wu, R., 2013. Relationship between sexual satiety and motivation, brain
Gréco, B., Edwards, D.A., Michael, R.P., Clancy, A.N., 1996. Androgen receptor androgen receptors and testosterone in male mandarin voles. Behav. Brain Res.
immunoreactivity and mating-induced Fos expression in the forebrain and midbrain 250, 257–263.
structures in the male rat. Neuroscience 75, 161–171. Heeb, M.M., Yahr, P., 1996. c-fos immunoreactivity in the sexually dimorphic area of
Gréco, B., Edwards, D.A., Michael, R.P., Clancy, A.N., 1998a. Androgen receptors and the hypothalamus and related brain regions of male gerbils after exposure to sex-
estrogen receptors are colocalized in male rat hypothalamic and limbic neurons related stimuli or performance of specific sexual behaviors. Neuroscience 72,
that express Fos immunoreactivity induced by mating. Neuroendocrinology 67, 1049–1071.
18–28. Heeb, M.M., Yahr, P., 2000. Cell-body lesions of the posterodorsal preoptic nucleus or
Gréco, B., Edwards, D.A., Michael, R.P., Zumpe, D., Clancy, A.N., 1999. Coloc- posterodorsal medial amygdala, but not the parvicellular subparafascicular thal-
alization of androgen receptors and mating-induced FOS immunoreactivity in amus, disrupt mating in male gerbils. Physiol. Behav. 68, 317–331.
neurons that project to the central tegmental field in male rats. J. Comp. Neurol. Heeb, M.M., Yahr, P., 2001. Anatomical and functional connections among cell groups in
408, 220–236. the gerbil brain that are activated with ejaculation. J. Comp. Neurol. 439, 248–258.
Gréco, B., Edwards, D.A., Zumpe, D., Clancy, A.N., 1998b. Androgen receptor and Heim, N., Hursch, C.J., 1979. Castration for sex offenders: treatment or punishment?
mating-induced Fos immunoreactivity are co-localized in limbic and midbrain A review and critique of recent European literature. Arch. Sex. Behav. 8, 281–304.
neurons that project to the male rat medial preoptic area. Brain Res. 781, 15–24. Heiman, J.R., Pfaff, D., 2011. Sexual arousal and related concepts: an introduction.
Grotthus, B., Piasecki, T., Piesniewska, M., Marszalik, P., Kwiatkowska, J., Skrzypiec- Horm. Behav. 59, 613–615.
Spring, M., Szelag, A., 2007. The influence of prolonged b-blockers treatment on Hermann, G.E., Holmes, G.M., Rogers, R.C., Beattie, M.S., Bresnahan, J.C., 2003.
male rabbit’s sexual behavior and penile microcirculation. Int. J. Impot. Res. 19, 49–54. Descending spinal projections from the rostral gigantocellular reticular nuclei
Guadarrama-Bazante, I.L., Canseco-Alba, A., Rodriguez-Manzo, G., 2014. Dopamine complex. J. Comp. Neurol. 455, 210–221.
receptors play distinct roles in sexual behavior expression of rats with a different Hillegaart, V., Ahlenius, S., 1998. Facilitation and inhibition of male rat ejacu-
sexual motivational tone. Behav. Pharmacol. 25, 684–694. latory behavior by the respective 5-HT 1A and 5-HT1B receptor agonists 8-OH-
Guerit, J.M., Opsomer, R.J., 1991. Bit-mapped imaging of somatosensory evoked DPAT and anpirtoline, as evidenced by use of the corresponding new and
potentials after stimulation of the posterior tibial nerves and dorsal nerve of the selective receptor antagonists NAD-299 and NAS-181. Br. J. Pharmacol.
penis/clitoris. Electroencephalogr. Clin. Neurophysiol. 80, 228–237. 125, 1733–1743.
Male Sexual Behavior 47

Hines, M., Allen, L.S., Gorski, R.A., 1992. Sex differences in subregions of the medial Hull, E.M., Bitran, D., Pehek, E.A., Warner, R.K., Band, L.C., Holmes, G.M., 1986.
nucleus of the amygdala and the bed nucleus of the stria terminalis of the rat. Brain Dopaminergic control of male sex behavior in rats: effects of an intracerebrally-
Res. 579, 321–326. infused agonist. Brain Res. 370, 73–81.
Hoffman, B.J., Mezey, E., 1989. Distribution of serotonin 5-HT1C receptor mRNA in Hull, E.M., Du, J., Lorrain, D.S., Matuszewich, L., 1995. Extracellular dopamine in the
adult rat brain. FEBS Lett. 247, 453–462. medial preoptic area: implications for sexual motivation and hormonal control of
Hojo, Y., Hattori, T., Furukawa, A., Suzuki, K., Ishii, H., Mukai, H., Morrison, J.H., copulation. J. Neurosci. 15, 7465–7471.
Janssen, W.J.M., Kominami, S., Harada, N., Kimoto, T., Kawato, S., 2004. Hull, E.M., Eaton, R.C., Markowski, V.P., Moses, J., Lumley, L.A., Loucks, J.A., 1992.
Adult male rat hippocampus synthesizes estradiol from pregnenolone by Opposite influence of medial preoptic D1 and D2 receptors on genital reflexes:
cytochromes P45017 and P450 aromatase localized in neurons. PNAS 101, implications for copulation. Life Sci. 51, 1705–1713.
865–870. Hull, E.M., Eaton, R.C., Moses, J., Lorrain, D., 1993. Copulation increases dopamine
Holder, M.K., Blaustein, J.D., 2014. Puberty and adolescence as a time of vulnerability activity in the medial preoptic area of male rats. Life Sci. 52, 935–940.
to stressors that alter neurobehavioral processes. Front. Neuroendocrinol. 35, Hull, E.M., Lorrain, D.S., Du, J., Matuszewich, L., Lumley, L.A., Putnam, S.K.,
89–110. Moses, J., 1999. Hormone-neurotransmitter interactions in the control of sexual
Holmes, G.M., Sachs, B.D., 1992. Erectile function and bulbospongiosus EMG activity behavior. Behav. Brain Res. 105, 105–116.
in -estrogen-maintained castrated rats vary with behavioral context. Horm. Behav. Hull, E.M., Lumley, L.A., Matuszewich, J., Dominguez, J., Moses, J., Lorrain, D.S.,
26, 406–419. 1994. The roles of nitric oxide in sexual function of male rats. Neuropharmacology
Holmes, G.M., Chapple, W.D., Leipheimer, R.E., Sachs, B.D., 1991. Electromyographic 33, 1499–1504.
analysis of male rat perineal muscles during copulation and reflexive erections. Hull, E.M., Meisel, R.L., Sachs, B.D., 2002. Male sexual behavior. In: Pfaff, D.W.,
Physiol. Behav. 49, 1235–1246. Arnold, A.P., Etgen, A.M., Fahrbach, S.E., Rubin, R.T. (Eds.), Hormones, Brain and
Holmes, G.M., Hermann, G.E., Rogers, R.C., Bresnahan, J.C., Beattie, M.S., 2002. Behavior, vol. 1. Academic Press, San Diego, pp. 3–137 (Chapter 1).
Dissociation of the effects of nucleus raphe obscurus or rostral ventrolateral Hull, E.M., Muschamp, J.W., Sato, S., 2004. Dopamine and serotonin: influences on
medulla lesions on eliminatory and sexual reflexes. Physiol. Behav. 75, 49–55. male sexual behavior. Physiol. Behav. 83, 291–307.
Holmes, G.M., Rogers, R.C., Bresnahan, J.C., Beattie, M.S., 1997. Differential effects Hull, E.M., Pehek, E.A., Bitran, D., Holmes, G.M., Warner, R.K., Band, L.C., Bazzett, T.,
of intrathecal thyrotropin-releasing hormone (TRH) on perineal reflexes in male rats. Clemens, L.G., 1988b. Brain localizations of cholinergic influence on male sex
Physiol. Behav. 61, 57–63. behavior in rats: agonists. Pharmacol. Biochem. Behav. 31, 175–178.
Holstege, G., Huynh, H.K., 2011. Brain circuits for mating behavior in cats and brain Hull, E.M., Warner, R.K., Bazzett, T.J., Eaton, R.C., Thompson, J.T., Scaletta, L.L.,
activations and de-activations during sexual stimulation and ejaculation and orgasm 1989. D2/D1 ratio in the medial preoptic area affects copulation of male rats.
in humans. Horm. Behav. 59, 702–707. J. Pharmacol. Exp. Ther. 251, 422–427.
Holstege, G., Georgiadis, J.R., Paans, A.M., Meiners, L.C., van der Graaf, F.H., Hull, E.M., Weber, M.S., Eaton, R.C., Dua, R., Markowski, V.P., Lumley, L.A.,
Reinders, A.A., 2003. Brain activation during human male ejaculation. J. Neurosci. Moses, J., 1991. Dopamine receptors in the ventral tegmental area affect
23, 9185–9193. motor, but not motivational or reflexive, components of copulation in male rats.
Hosokawa, N., Chiba, A., 2005. Effects of sexual experience on conspecific odor Brain Res. 554, 72–76.
preference and estrous odor-induced activation of the vomeronasal projection Hull, E.M., Wood, R.I., McKenna, K., 2006. The neurobiology of male sexual behavior.
pathway and the nucleus accumbens in male rats. Brain Res. 1066, 101–108. In: Neill, J.D. (Ed.), Knobil and Neill’s Physiology of Reproduction, third ed., vol. 2.
Hoyer, D., Hannon, J.P., Martin, G.R., 2002. Molecular, pharmacological and func- Academic Press, pp. 1729–1824 (Chapter 33).
tional diversity of 5-HT receptors. Pharmacol. Biochem. Behav. 71, 533–554. Hung, S.H., Pi, W.P., Tsai, Y.F., Peng, M.T., 1997. Restoration of sexual behavior in
Hsu, H.K., Hsu, C., Yu, J.Y., Peng, M.T., 1986. Effects of long-term testosterone aged male rats by intracerebral grafts of fetal preoptic area neurons. J. Formos.
replacement on copulatory activity in old male rats. Gerontology 32, 10–17. Med. Assoc. 96, 812–818.
Hubscher, C.H., Johnson, R.D., 1996. Responses of medullary reticular formation Hurt, K.J., Sezen, S.F., Lagoda, G.F., Musicki, B., Rameau, G.A., Snyder, S.H.,
neurons to input from the male genitalia. J. Neurophysiol. 76, 2474–2482. Burnett, A.L., 2012. Cyclic amp-dependent phosphorylation of neuronal nitric oxide
Huddleston, G.G., Michael, R.P., Zumpe, D., Clancy, A.N., 2003. Estradiol in the male synthase mediates penile erection. Proc. Natl. Acad. Sci. U.S.A. 109, 16624–
rat amygdala facilitates mounting but not ejaculation. Physiol. Behav. 79, 16629.
239–246. Huston, J.P., 1971. Relationship between motivating and rewarding stimulation of the
Huddleston, G.G., Paisley, J.C., Clancy, A.N., 2006. Effects of estrogen in the male rat lateral hypothalamus. Physiol. Behav. 6, 711–716.
medial amygdala: infusion of an aromatase inhibitor lowers mating and bovine Hsieh, J.T., Kuo, Y.C., Chang, H.C., Liu, S.P., Chen, J.H., Tsai, V.F., 2014. The role of
serum albumin-conjugated estradiol implants do not promote mating. Neuroen- sympathetic and parasympathetic nerve systems on the smooth muscle of rat
docrinology 83, 106–116. seminal vesicles – experimental results and speculation for physiological impli-
Hueletl-Soto, M.E., Carro-Juarez, M., Rodriguez-Manzo, G., 2010. DMI-induced sexual cation on ejaculation. Andrology 2, 59–64.
effects in male rats: analysis of DMI’s acute and chronic actions on copulatory Hsieh, J.T., Liu, S.P., Chang, H.C., Tsai, V.F., Chien, C.T., Yu, H.J., Ho, C.H., 2011.
behavior and on the genital motor pattern of ejaculation. Pharmacol. Biochem. The activation of peripheral 5-HT1A receptors can inhibit seminal vesicle
Behav. 94, 423–430. contraction: an in vivo animal study. Urology 78, 376–379.
Hueletl-Soto, M.E., Carro-Juarez, M., Rodriguez-Manzo, G., 2012. Fluoxetine chronic Ikemoto, S., 2007. Dopamine reward circuitry: two projection systems from the ventral
treatment inhibits male rat sexual behavior by affecting both copulatory behavior midbrain to the nucleus accumbens-olfactory tubercle complex. Brain Res. Rev.
and the genital motor pattern of ejaculation. J. Sex. Med. 9, 1015–1026. 56, 27–78.
Hueletl-Soto, M.E., Carro-Juarez, M., Rodriguez-Manzo, G., 2014. Effects of bupropion Ishigami, T., Yoshioka, K., Karicheti, V., Marson, L., 2013. A role for peripheral 5-HT2
on the ejaculatory response of male rats. Int. J. Impot. Res. 26, 205–212. receptors in serotonin-induced facilitation of the expulsion phase of ejaculation in
Hughes, A.M., Everitt, B.J., Herbert, J., 1987. Selective effects of b-endorphin male rats. J. Sex. Med. 10, 2688–2702.
infused into the hypothalamus, preoptic area and bed nucleus of the stria Iyilikci, O., Baxter, S., Balthazart, J., Ball, G.F., 2014. Fos expression in mono-
terminalis on the sexual and ingestive behavior of male rats. Neuroscience 23, aminergic cell groups in response to sociosexual interactions in male and female
1063–1073. japanese quail. Behav. Neurosci. 128, 48–60.
Hull, E.M., 2011. Sex, drugs and gluttony: how the brain controls motivated behaviors. James, P.J., Nyby, J.G., 2002. Testosterone rapidly affects the expression of copu-
Physiol. Behav. 104, 173–177. latory behavior in house mice (Mus musculus). Physiol. Behav. 75, 287–294.
Hull, E.M., Dominguez, J.M., 2007. Sexual behavior in male rodents. Horm. Behav. 52, Jänig, W., McLachlan, E.M., 1987. Organization of lumbar spinal outflow to distal
45–55. colon and pelvic organs. Physiol. Rev. 67, 1332–1404.
Hull, E.M., Dominguez, J.M., 2015. The neurobiology of male sexual behavior. In: Jiménez, P., Serrano-Meneses, M.A., Cuamatzi, E., González-Mariscal, G., 2012.
Knobil, E., Neill (Editors in Chief), J., Plant (Section Editor), T. (Eds.), Physiology of Analysis of copulatory parameters in male rabbits across successive tests leading
Reproduction, fourth ed. Elsevier Press,. to sexual exhaustion. World Rabbit Sci. 20, 13–23.
Hull, E.M., Bazzett, T.J., Warner, R.K., Eaton, R.C., Thompson, J.T., 1990. Dopamine Jin, L., Burnett, A.L., 2006. RhoA/Rho-kinase in erectile tissue: mechanisms of
receptors in the ventral tegmental area modulate male sexual behavior in rats. disease and therapeutic insights. Clin. Sci. 110, 153–165.
Brain Res. 512, 1–6. Johnson, R.D., Hubscher, C.H., 1998. Brainstem microstimulation differentially inhibits
Hull, E.M., Bitran, D., Pehek, E.A., Holmes, G.M., Warner, R.K., Band, L.C., pudendal motoneuron reflex inputs. Neuroreport 9, 341–345.
Clemens, L.G., 1988a. Brain localization of cholinergic influence on male sex Johnson, R.D., Hubscher, C.H., 2000. Brainstem microstimulation activates sympa-
behavior in rats: antagonists. Pharmacol. Biochem. Behav. 31, 169–174. thetic fibers in pudendal nerve motor branch. Neuroreport 11, 379–382.
48 Male Sexual Behavior

de Jong, T.R., Pattij, T., Veening, J.G., Waldinger, M.D., Cools, A.R., Olivier, B., Kollack, S.S., Newman, S.W., 1992. Mating behavior induces selective expression of
2005a. Effects of chronic selective serotonin reuptake inhibitors on 8-OH-DPAT- Fos protein within the chemosensory pathways of the male Syrian hamster brain.
induced facilitation of ejaculation in rats: comparison of fluvoxamine and Neurosci. Lett. 143, 223–228.
paroxetine. Psychopharmacology (Berlin) 179, 509–515. Kollack-Walker, S., Newman, S.W., 1995. Mating and agonistic behavior produce
de Jong, T.R., Pattij, T., Veening, J.G., Dederen, P.J., Waldinger, M.D., different patterns of Fos immunolabeling in the male Syrian hamster brain.
Cools, A.R., Olivier, B., 2005b. Citalopram combined with WAY 100635 Neuroscience 66, 721–736.
inhibits ejaculation and ejaculation-related Fos immunoreactivity. Eur. J. Kollack-Walker, S., Newman, S.W., 1997. Mating-induced expression of c-fos in the
Pharmacol. 509, 49–59. male Syrian hamster brain: role of experience, pheromones, and ejaculations.
de Jong, T.R., Veening, J.G., Olivier, B., Waldinger, M.D., 2007. Oxytocin involvement J. Neurobiol. 32, 481–501.
in SSRI-induced delayed ejaculation: a review of animal studies. J. Sex. Med. 4, Kollberg, S., Petersen, I., Stener, I., 1972. Preliminary results of an electromyographic
14–28. study of ejaculation. Acta Chir. Scand. 123, 478–483.
de Jong, T.R., Veening, J.G., Waldinger, M.D., Cools, A.R., Olivier, B., 2006. Serotonin Kondo, Y., 1992. Lesions of the medial amygdala produce severe impairment of copulatory
and the neurobiology of the ejaculatory threshold. Neurosci. Biobehav. Rev. 30, behavior in sexually inexperienced male rats. Physiol. Behav. 51, 939–943.
893–907. Kondo, Y., Arai, Y., 1995. Functional association between the medial amygdala and the
de Jonge, F.H., Louwerse, A.L., Ooms, M.P., Evers, P., Endert, E., van de Poll, N.E., medial preoptic area in regulation of mating behavior in the male rat. Physiol.
1989. Lesions of the SDN-POA inhibit sexual behavior of male Wistar rats. Brain Behav. 57, 69–73.
Res. Bull. 23, 483–492. Kondo, Y., Sachs, B.D., 2002. Disparate effects of small medial amygdala lesions
de Jonge, F.H., Oldenburger, W.P., Louwerse, A.L., Van de Poll, N.E., 1992. Changes on noncontact erection, copulation, and partner preference. Physiol. Behav. 76,
in male copulatory behavior after sexual exciting stimuli: effects of medial amygdala 443–447.
lesions. Physiol. Behav. 52, 327–332. Kondo, Y., Sachs, B.D., Sakuma, Y., 1998. Importance of the medial amygdala in rat
Kaplan, M.E., McGinnis, M.Y., 1989. Effects of ATD on male sexual behavior and penile erection evoked by remote stimuli from estrous females. Behav. Brain Res.
androgen receptor binding: a reexamination of the aromatization hypothesis. Horm. 91, 215–221.
Behav. 23, 10–26. Kondo, Y., Tomihara, K., Sakuma, Y., 1999. Sensory requirements for noncontact
Keller, M., Douhard, Q., Baum, M.J., Bakker, J., 2006. Sexual experience does not penile erection in the rat. Behav. Neurosci. 113, 1062–1070.
compensate for the disruptive effects of zinc sulfate-lesioning of the main Kondo, Y., Yamanouchi, K., Arai, Y., 1993. P-chlorophenylalanine facilitates copulatory
olfactory epithelium on sexual behavior in male mice. Chem. Senses 31, behavior in septal lesioned but not in preoptic lesioned male rats.
753–762. J. Neuroendocrinol. 5, 629–633.
Kelley, A.E., Berridge, K.C., 2002. The neuroscience of natural rewards: relevance to Korotkova, T.M., Sergeeva, O.A., Eriksson, K.S., Haas, H.L., Brown, R.E., 2003.
addictive drugs. J. Neurosci. 22, 3306–3311. Excitation of ventral tegmental area dopaminergic and nondopaminergic neurons by
Kelliher, K.R., Chang, Y.-M., Wersinger, S.R., Baum, M.J., 1998. Sex difference and orexins/hypocretins. J. Neurosci. 23, 7–11.
testosterone modulation of pheromone-induced neuronal Fos in the ferret’s main Krey, L.C., McGinnis, M.Y., 1990. Time-courses of the appearance/disappearance of
olfactory bulb and hypothalamus. Biol. Reprod. 59, 1454–1463. nuclear androgen þ receptor complexes in the brain and adenohypophysis
Kelliher, K.R., Liu, Y.C., Baum, M.J., Sachs, B.D., 1999. Neuronal Fos activation in following testosterone administration/withdrawal to castrated male rats: relation-
olfactory bulb and forebrain of male rats having erections in the presence of ships with gonadotropin secretion. J. Steroid. Biochem. 35, 403–408.
inaccessible estrous females. Neuroscience 92, 1025–1033. Kriegsfeld, L.J., Demas, G.E., Huang, P.L., Burnett, A.L., Nelson, R.J., 1999. Ejacu-
Kelly, M.J., Qiu, J., Wagner, E.J., Ronnekleiv, O.K., 2002. Rapid effects of estrogen on latory abnormalities in mice lacking the gene for endothelial nitric oxide synthase
G protein-coupled receptor activation of potassium channels in the central nervous (eNOS /). Physiol. Behav. 67, 561–566.
system (CNS). J. Steroid. Biochem. Mol. Biol. 83, 187–193. Krüger, T.H., Haake, P., Haverkamp, J., Krämer, M., Exton, M.S., Saller, B.,
Kimura, K., Ito, M., Amano, M., Chihara, K., Fukata, Y., Nakafuku, M., Yamamori, B., Leygraf, N., Hartmann, U., Schedlowski, M., 2003. Effects of acute prolactin
Feng, J., Nakano, T., Okawa, K., Iwamatsu, A., Kaibuchi, K., 1996. Regulation of manipulation on sexual drive and function in males. J. Endocrinol. 179, 357–365.
myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase). Science Kuhar, M.J., Couceyro, P.R., Lambert, P.D., 1999. Catecholamines. In: Siegel, G.J.,
273, 245–248. Agranoff, B.W., Albers, R.W., Fisher, S.K., Uhler, M.D. (Eds.), Basic Neurochem-
Kindon, H.A., Baum, M.J., Paredes, R.J., 1996. Medial preoptic/anterior hypothalamic istry, sixth ed. Lippincott-Raven, Philadelphia, pp. 243–262.
lesions induce a female-typical profile of sexual partner preference in male ferrets. Lagoda, G., Muschamp, J.W., Vigdorchik, A., Hull, E.M., 2004. A nitric oxide synthesis
Horm. Behav. 30, 514–527. inhibitor in the medial preoptic area inhibits copulation and stimulus sensitization in
Kinsey, A.C., Pomeroy, W.B., Martin, C.E., 1948. Sexual Behavior in the Human Male. male rats. Behav. Neurosci. 118, 1317–1323.
W. B. Saunders Co, Philadelphia. Lal, S., Laryea, E., Thavundayil, J.X., Nair, N.P., Negrete, J., Ackman, D., Blundell, P.,
Kippin, T.E., Cain, S.W., Pfaus, J.G., 2003. Estrous odors and sexually conditioned Gardiner, R.J., 1987. Apomorphine-induced penile tumescence in impotent
neutral odors activate separate neural pathways in the male rat. Neuroscience 117, patients: preliminary findings. Prog. Neuropsychopharmacol. Biol. Psychiatry 11,
971–979. 235–242.
Kippin, T.E., Sotiropoulos, V., Badih, J., Pfaus, J.G., 2004. Opposing roles of the Lambert, G.M., Rubin, B.S., Baum, M.J., 1992. Sex difference in the effect of mating
nucleus accumbens and anterior lateral hypothalamic area in the control of sexual on c-fos expression in luteinizing hormone-releasing hormone neurons of the ferret
behavior in the male rat. Eur. J. Neurosci. 19, 698–704. forebrain. Endocrinology 131, 1473–1480.
Kita, I., Yoshida, Y., Nishino, S., 2006. An activation of parvocellular oxytocinergic Larsson, K., 1966. Individual differences in reactivity to androgen in male rats. Physiol.
neurons in the paraventricular nucleus in oxytocin-induced yawning and penile Behav. 1, 255–258.
erection. Neurosci. Res. 54, 269–275. Larsson, K., 1967. Testicular hormone and developmental changes in mating behavior
Kitrey, N.D., Clement, P., Bernabe, J., Alexandre, L., Giuliano, F., 2007. of the male rat. J. Comp. Physiol. Psychol. 63, 223–230.
Microinjection of the preferential dopamine receptor D(3) agonist 7-hydroxy- Le Poul, E., Laaris, N., Doucet, E., Laporte, A.M., Hamon, M., Lanfumey, L., 1995. Early
N,N-di-n-propylaminotetralin hydrobromide into the hypothalamic medial desensitization of somato-dendritic 5-HT1A autoreceptors in rats treated with fluox-
preoptic area induced ejaculation in anesthetized rats. Neuroscience 149, etine or paroxetine. Naunyn Schmiedebergs Arch. Pharmacol. 352, 141–148.
636–641. Lee, J.J., Hahm, E.T., Lee, C.H., Cho, Y.W., 2008. Serotonergic modulation of
Kiyatkin, E.A., Mitchum Jr., R.D., 2003. Fluctuations in brain temperature during GABAergic and glutamatergic synaptic transmission in mechanically isolated rat
sexual interaction in male rats: an approach for evaluating neural activity underlying medial preoptic area neurons. Neuropsychopharmacology 33, 340–352.
motivated behavior. Neuroscience 119, 1169–1183. Lee, R.L., Smith, E.R., Mas, M., Davidson, J.M., 1990. Effects of intrathecal
Klint, T., Larsson, K., 1995. Clozapine acts as a 5-HT2 antagonist by attenuating DOI- administration of 8-OH-DPAT on genital reflexes and mating behavior in male rats.
induced inhibition of male rat sexual behavior. Psychopharmacology (Berlin) 119, Physiol. Behav. 47, 665–669.
291–294. Lehman, M.N., Winans, S.S., Powers, J.B., 1980. Medial nucleus of the amygdala
Kobayashi, T., Kiyokawa, Y., Arata, S., Takeuchi, Y., Mori, Y., 2013. C-fos expression mediates chemosensory control of male hamster sexual behavior. Science 210,
during the modulation of sexual behavior by an alarm pheromone. Behav. Brain 557–560.
Res. 237, 230–237. Leshan, R.L., Louis, G.W., Jo, Y.H., Rhodes, C.J., Munzberg, H., Myers Jr., M.G.,
Kobayashi, T., Kiyokawa, Y., Takeuchi, Y., Mori, Y., 2015. Neural correlates underlying 2009. Direct innervation of GnRH neurons by metabolic- and sexual odorant-
naloxone-induced amelioration of sexual behavior deterioration due to an alarm sensing leptin receptor neurons in the hypothalamic ventral premammillary
pheromone. Front. Neurosci. 9, 52. nucleus. J. Neurosci. 29, 3138–3147.
Male Sexual Behavior 49

Leuner, B., Glasper, E.R., Gould, E., 2010. Sexual experience promotes adult neu- Lumley, L.A., Hull, E.M., 1999. Effects of a D-1 antagonist and of sexual experience on
rogenesis in the hippocampus despite an initial elevation in stress hormones. PLoS copulation-induced Fos-like immunoreactivity in the medial preoptic nucleus. Brain
One 5, e11597. Res. 829, 55–68.
Levin, R.J., 2009. Revisiting post-ejaculation refractory time-what we know and what Lupica, C.R., Riegel, A.C., Hoffman, A.F., 2004. Marijuana and cannabinoid regulation
we do not know in males and in females. J. Sex. Med. 6, 2376–2389. of brain reward circuits. Br. J. Pharmacol. 143, 227–234.
Leypold, B.G., Yu, C.R., Leinders-Zufall, T., Kim, M.M., Zufall, F., Axel, R., 2002. Lydell, K., Doty, R.L., 1972. Male rat of odor preferences for female urine as
Altered sexual and social behaviors in trp2 mutant mice. Proc. Natl. Acad. Sci. a function of sexual experience, urine age, and urine source. Horm. Behav. 3,
U.S.A. 99, 6376–6381. 205–212.
Leyton, M., Stewart, J., 1992. The stimulation of central k opiod receptors decreased Maeda, N., Matsuoka, N., Yamaguchi, I., 1990. Septohippocampal cholinergic pathway
male sexual behavior and locomotor activity. Brain Res. 594, 56–74. and penile erections induced by dopaminergic and cholinergic stimulants. Brain
Li, H.Y., Blaustein, J.D., De Vries, G.J., Wade, G.N., 1993. Estrogen-receptor Res. 537, 163–168.
immunoreactivity in hamster brain: preoptic area, hypothalamus and amygdala. Maejima, S., Ohishi, N., Yamaguchi, S., Tsukahara, S., 2015. A neural connection
Brain Res. 631, 304–312. between the central part of the medial preoptic nucleus and the bed nucleus of the
Li, X., Higley, A., Song, R., Xi, Z.X., 2013. Effects of metabotropic glutamate receptor stria terminalis to regulate sexual behavior in male rats. Neurosci. Lett. 606, 66–71.
ligands on male sexual behavior in rats. Neuropharmacology 66, 373–381. Maillard, C.A., Edwards, D.A., 1991. Excitotoxin lesions of the zone incerta/lateral
Lin, D., Boyle, M.P., Dollar, P., Lee, H., Lein, E.S., Perona, P., Anderson, D.J., 2011. tegementum continuum: effects on male sexual behavior in rats. Behav. Brain Res.
Functional identification of an aggression locus in the mouse hypothalamus. Nature 46, 143–149.
470, 221–226. Mains, R.E., Eipper, B.A., 1999. Peptides. In: Siegel, G.J., Agranoff, B.W.,
Lindemann, L., Meyer, C.A., Jeanneau, K., et al., 2008. Trace amine-associated Albers, R.W., Fisher, S.K., Uhler, M.D. (Eds.), Basic Neurochemistry, sixth ed.
receptor 1 modulates dopaminergic activity. J. Pharmacol. Exp. Ther. 324, Lippincott-Raven, Philadelphia, pp. 363–382.
948–956. Malagie, I., Deslandes, A., Gardier, A.M., 2000. Effects of acute and chronic tianeptine
Liu, Y.C., Salamone, J.D., Sachs, B.D., 1997a. Impaired sexual response after lesions administration on serotonin outflow in rats: comparison with paroxetine by using
of the paraventricular nucleus of the hypothalamus in male rats. Behav. Neurosci. in vivo microdialysis. Eur. J. Pharmacol. 403, 55–65.
111, 1361–1367. Mallick, H., Manchanda, S.K., Kumar, V.H., 1996. b-Adrenergic modulation of male
Liu, Y.C., Salamone, J.D., Sachs, B.D., 1997b. Lesions in medial preoptic area and sexual behavior elicited from the medial preoptic area in rats. Behav. Brain Res. 74,
bed nucleus of stria terminalis: differential effects on copulatory behavior and 181–187.
noncontact erection in male rats. J. Neurosci. 17, 5245–5253. Malmnäs, C.O., 1976. The significance of dopamine, versus other catecholamines, for
Lloyd, S.A., Dixson, A.F., 1988. Effects of hypothalamic lesions upon the sexual and L-dopa induced facilitation of sexual behavior in the castrated male rat. Pharmacol.
social behaviour of the male common marmoset (Callithrix jacchus). Brain Res. Biochem. Behav. 4, 521–526.
463, 317–329. Mandiyan, V.S., Coats, J.K., Shah, N.M., 2005. Deficits in sexual and aggressive
Lopez, H.H., Ettenberg, A., 2000. Haloperidol challenge during copulation prevents behaviors in Cnga2 mutant mice. Nat. Neurosci. 8, 1660–1662.
subsequent increase in male sexual motivation. Pharmacol. Biochem. Behav. 67, Manning, A., Thompson, M.L., 1976. Postcastration retention of sexual behavior in the
387–393. male BDF1 mouse: the role of experience. Anim. Behav. 24, 523–533.
Lopez, H.H., Ettenberg, A., 2001. Dopamine antagonism attenuates the unconditioned Manzo, J., Cruz, M.R., Hernandez, M.E., Pacheco, P., Sachs, B.D., 1999. Regulation
incentive value of estrous female cues. Pharmacol. Biochem. Behav. 68, of noncontact erection in rats by gonadal steroids. Horm. Behav. 35, 264–270.
411–416. Maragos, W.F., Newman, S.W., Lehman, M.N., Powers, J.B., 1989. Neurons of origin
Lopez, H.H., Ettenberg, A., 2002a. Exposure to female rats produces differences in c- and fiber trajectory of amygdalofugal projections to the medial preoptic area in
fos induction between sexually-naïve and experienced male rats. Brain Res. 947, Syrian hamsters. J. Comp. Neurol. 280, 59–71.
57–66. Maras, P.M., Petrulis, A., 2006. Chemosensory and steroid-responsive regions of the
Lopez, H.H., Ettenberg, A., 2002b. Sexually conditioned incentives: attenuation of medial amygdala regulate distinct aspects of opposite-sex odor preference in male
motivational impact during dopamine receptor antagonism. Pharmacol. Biochem. Syrian hamsters. Eur. J. Neurosci. 24, 3541–3552.
Behav. 72, 65–72. Markowski, V.P., Hull, E.M., 1995. Cholecystokinin modulates mesolimbic
Lorrain, D.S., Hull, E.M., 1993. Nitric oxide increases dopamine and serotonin release dopaminergic influences on male rat copulatory behavior. Brain Res. 699,
in the medial preoptic area. Neuroreport 5, 87–89. 266–274.
Lorrain, D.S., Matuszewich, L., Friedman, R.D., Hull, E.M., 1997. Extracellular sero- Markowski, V.P., Eaton, R.C., Lumey, L.A., Moses, J., Hull, E.M., 1994. A D1 agonist
tonin in the lateral hypothalamic area is increased during the postejaculatory in the MPOA facilitates copulation in male rats. Pharmacol. Biochem. Behav. 47,
interval and impairs copulation in rats. J. Neurosci. 17, 9361–9366. 483–486.
Lorrain, D.S., Matuszewich, L., Howard, R.V., Du, J., Hull, E.M., 1996. Nitric oxide Marson, L., 2004. Lesions of the periaqueductal gray block the medial preoptic area-
promotes medial preoptic dopamine release during male rat copulation. Neuro- induced activation of the urethrogenital reflex in male rats. Neurosci. Lett. 367,
report 8, 31–34. 278–282.
Lorrain, D.S., Matuszewich, L., Hull, E.M., 1998. 8-OH-DPAT influences extracellular Marson, L., McKenna, K.E., 1990. The identification of a brainstem site controlling
levels of serotonin and dopamine in the medial preoptic area of male rats. Brain spinal sexual reflexes in male rats. Brain Res. 515, 303–308.
Res. 790, 217–223. Marson, L., McKenna, K.E., 1992. A role for 5-hydroxytryptamine in descending
Lorrain, D.S., Riolo, J.V., Matuszewich, L., Hull, E.M., 1999. Lateral hypothalamic inhibition of spinal sexual reflexes. Exp. Brain Res. 88, 313–320.
serotonin inhibits nucleus accumbens dopamine: implications for sexual satiety. Marson, L., McKenna, K.E., 1994a. Serotonergic neurotoxic lesions facilitate male
J. Neurosci. 19, 7648–7652. sexual reflexes. Pharmacol. Biochem. Behav. 47, 883–888.
Lott, D.F., 1981. Sexual behavior and intersexual strategies in American bison. Marson, L., McKenna, K.E., 1994b. Stimulation of the hypothalamus initiates the
Z. Tierpsychol. 56, 97–114. urethrogenital reflex in male rats. Brain Res. 638, 103–108.
Loyd, D.R., Murphy, A.Z., 2008. Androgen and estrogen (alpha) receptor localization Marson, L., List, M.S., McKenna, K.E., 1992. Lesions of the nucleus para-
on periaqueductal gray neurons projecting to the rostral ventromedial medulla in gigantocellularis alter ex- copula penile reflexes. Brain Res. 592, 187–192.
the male and female rat. J. Chem. Neuroanat. 36, 216–226. Martel, K.L., Baum, M.J., 2007. Sexually dimorphic activation of the accessory, but not
Lue, T.F., Gleason, C.A., Brock, G.B., Carroll, P.R., Tanagho, E.A., 1995. Intraoperative the main, olfactory bulb in mice by urinary volatiles. Eur. J. Neurosci. 26, 463–475.
electrostimulation of the cavernous nerve: technique, results and limitations. Martin, W.J., McGowan, E., Cashen, D.E., Gantert, L.T., Drisko, J.E., Hom, G.J.,
J. Urol. 154, 1426–1428. Nargund, R., Sebhat, I., Howard, A.D., Van der Ploeg, L.H., MacIntyre, D.E., 2002.
Lue, T.F., Takamura, T., Umraiya, M., Schmidt, R.A., Tanagho, E.A., 1984. Activation of melanocortin MC(4) receptors increases erectile activity in rats ex
Hemodynamics of canine corpora cavernosa during erection. Urology 24, copula. Eur. J. Pharmacol. 454, 71–79.
347–352. Martínez-González, D., Bonilla-Jaime, H., Morales-Otal, A., Henriksen, S.J., Velázquez-
Lugg, J.A., Rajfer, J., Gonzalez-Cadavid, N.F., 1995. Dihydrotestosterone is the active Moctezuma, J., Prospéro-García, O., 2004. Oleamide and anandamide effects on
androgen in the maintenance of nitric oxide-mediated penile erection in the rat. food intake and sexual behavior of rats. Neurosci. Lett. 364, 1–6.
Endocrinology 136, 1495–1501. Martínez-Mota, L., López-Rubalcava, C., Rodríguez-Manzo, G., 2005. Ejaculation
Luiten, P.G., ter Horst, G.J., Karst, H., Steffens, A.B., 1985. The course of para- induces long-lasting behavioural changes in male rats in the forced swimming test:
ventricular hypothalamic efferents to autonomic structures in medulla and spinal evidence for an increased sensitivity to the antidepressant desipramine. Brain Res.
cord. Brain Res. 329, 374–378. Bull. 65, 323–329.
50 Male Sexual Behavior

Mas, M., Fumero, B., Fernandez-Vera, J.R., Gonzáez-Mora, J.L., 1995a. Neuro- Meisel, R.L., Sachs, B.D., 1994. The physiology of male sexual behavior. In: Knobil, E.,
chemical correlates of sexual exhaustion and recovery as assessed by in vivo Neill, J. (Eds.), Physiology of Reproduction, second ed. Raven Press, New York,
microdyalisis. Brain Res. 675, 13–19. pp. 3–105 (Chapter 1).
Mas, M., Fumero, B., Perez-Rodriguez, I., 1995b. Induction of mating behavior by Meisel, R.L., O’Hanlon, J.K., Sachs, B.D., 1984. Differential maintenance of penile
apomorphine in sexually sated rats. Eur. J. Pharmacol. 280, 331–334. responses and copulatory behavior by gonadal hormones in castrated male rats.
Mas, M., Rodriguez del Castillo, A., Guerra, M., Davidson, J.M., Battaner, E., 1987. Horm. Behav. 18, 56–64.
Neurochemical correlates of male sexual behavior. Physiol. Behav. 41, Melis, M.R., Argiolas, A., 1995. Dopamine and sexual behavior. Neurosci. Biobehav.
341–345. Rev. 19, 19–38.
Mathes, C.W., Smith, E.R., Popa, B.R., Davidson, J.M., 1990. Effects of intrathecal Melis, M.R., Argiolas, A., 2011. Central control of penile erection: a re-visitation of the
and systemic administration of buspirone on genital reflexes and mating behavior in role of oxytocin and its interaction with dopamine and glutamic acid in male rats.
male rats. Pharmacol. Biochem. Behav. 36, 63–68. Neurosci. Biobehav. Rev. 35, 939–955.
Matsumoto, J., Urakawa, S., Hori, E., de Araujo, M.F., Sakuma, Y., Ono, T., Nishijo, H., Melis, M.R., Melis, T., Cocco, C., Succu, S., Sanna, F., Pillolla, G., Boi, A., Ferri, G.L.,
2012. Neuronal responses in the nucleus accumbens shell during sexual behavior Argiolas, A., 2007. Oxytocin injected into the ventral tegmental area induces penile
in male rats. J. Neurosci. 32, 1672–1686. erection and increases extracellular dopamine in the nucleus accumbens and
Matsumoto, T., Honda, S., Harada, N., 2003. Neurological effects of aromatase paraventricular nucleus of the hypothalamus of male rats. Eur. J. Neurosci. 26,
deficiency in the mouse. J. Steroid. Biochem. Mol. Biol. 86, 357–365. 1026–1035.
Matsuoka, M., Yokosuka, M., Mori, Y., Ichikawa, M., 1999. Specific expression pattern Melis, M.R., Spano, M.S., Succu, S., Argiolas, A., 1999a. The oxytocin antagonist
of Fos in the accessory olfactory bulb of male mice after exposure to soiled bedding d(CH 2)5Tyr(Me)2-Orn8-vasotocin reduces non-contact penile erections in male rats.
of females. Neurosci. Res. 35, 189–195. Neurosci. Lett. 265, 171–174.
Matuszewich, L., Dornan, W.A., 1992. Bilateral injections of a selective m-receptor Melis, M.R., Spano, M.S., Succu, S., Argiolas, A., 2000a. Activation of gamma-
agonist (morphiceptin) into the medial preoptic nucleus produces a marked delay in aminobutyric acid(A) receptors in the paraventricular nucleus of the hypothal-
the initiation of sexual behavior in the male rat. Psychopharmacology (Berlin) 106, amus reduces apomorphine-, N-methyl-D-aspartic acid- and oxytocin-induced
391–396. penile erection and yawning in male rats. Neurosci. Lett. 281, 127–130.
Matuszewich, L., Lorrain, D.S., Trujillo, R., Dominguez, J., Putnam, S.K., Hull, E.M., Melis, M.R., Spano, M.S., Succu, S., Locatelli, V., Torsello, A., Muller, E.E.,
1999. Partial antagonism of 8-OH-DPAT’s effects on male rat sexual behavior with Deghenghi, R., Argiolas, A., 2000b. EP 60761- and EP 50885- induced penile
a D2, but not a 5-HT1A, antagonist. Brain Res. 820, 55–62. erection: structure-activity studies and comparison with apomorphine, oxytocin and
Matuszewich, L., Ormsby, J.L., Moses, J., Lorrain, D.S., Hull, E.M., 1995. Effects of N-methyl-D-aspartic acid. Int. J. Impot. Res. 12, 255–262.
morphiceptin in the medial preoptic area on male sexual behavior. Psychophar- Melis, M.R., Succu, S., Argiolas, A., 1996. Dopamine agonists increase nitric oxide
macology (Berlin) 122, 330–335. production in the paraventricular nucleus of the hypothalamus: correlation with
McConnell, S., Baum, M., Badger, T., 1981. Lack of correlation between naloxone- penile erection and yawning. Eur. J. Neurosci. 8, 2056–2063.
induced changes in sexual behavior and serum LH in male rats. Horm. Behav. Melis, M.R., Succu, S., Iannucci, U., Argiolas, A., 1997. N-methyl-D-aspartic acid-
15, 16–35. induced penile erection and yawning: role of hypothalamic paraventricular nitric
McGinnis, M.Y., Kahn, D.F., 1997. Inhibition of male sexual behavior by intracranial oxide. Eur. J. Pharmacol. 328, 115–123.
implants of the protein synthesis inhibitor anisomycin into the medial preoptic area Melis, M.R., Succu, S., Mascia, M.S., Argiolas, A., 2004a. Antagonism of cannabinoid
of the rat. Horm. Behav. 31, 15–23. CB1 receptors in the paraventricular nucleus of male rats induces penile erection.
McGinnis, M.Y., Mirth, M.C., Zebrowski, A.F., Dreifuss, R.M., 1989. Critical exposure Neurosci. Lett. 359, 17–20.
time for androgen activation of male sexual behavior in rats. Physiol. Behav. 46, Melis, M.R., Succu, S., Mascia, M.S., Argiolas, A., 2005. PD-168077, a selective
159–165. dopamine D4 receptor agonist, induces penile erection when injected into the
McGinnis, M.Y., Montana, R.C., Lumia, A.R., 2002. Effects of hydroxyflutamide in the paraventricular nucleus of male rats. Neurosci. Lett. 379, 59–62.
medial preoptic area or lateral septum on reproductive behaviors in male rats. Brain Melis, M.R., Succu, S., Mascia, M.S., Cortis, L., Argiolas, A., 2003. Extra-cellular
Res. Bull. 59, 227–234. dopamine increases in the paraventricular nucleus of the hypothalamus: correla-
McGinnis, M.Y., Williams, G.W., Lumia, A.R., 1996. Inhibition of male sex behavior by tion with penile erection and yawning. Eur. J. Neurosci. 17, 1266–1272.
androgen receptor blockade in preoptic area or hypothalamus, but not amygdala or Melis, M.R., Succu, S., Mascia, M.S., Cortis, L., Argiolas, A., 2004b. Extracellular
septum. Physiol. Behav. 60, 783–789. excitatory amino acids increase in the paraventricular nucleus of male rats during
McGregor, A., Herbert, J., 1992a. Differential effects of excitotoxic basolateral and sexual activity: main role of N-methyl-D-aspartic acid receptors in erectile function.
corticomedial lesions of the amygdala on the behavioural and endocrine responses Eur. J. Neurosci. 19, 2569–2575.
to either sexual or aggression-promoting stimuli in the male rat. Brain Res. 574, Melis, M.R., Succu, S., Mascia, M.S., Sanna, F., Melis, T., Castelli, M.P., Argiolas, A.,
9–20. 2006. The cannabinoid receptor antagonist SR-141716A induces penile erection
McGregor, A., Herbert, J., 1992b. Specific effects of b-endorphin infusions in male rats: involvement of paraventricular glutamic acid and nitric oxide.
into the amygdala on sexual behavior in the male rat. Neuroscience 46, Neuropharmacology 50, 219–228.
165–172. Melis, M.R., Succu, S., Mauri, A., Argiolas, A., 1998. Nitric oxide production is
McHenry, J.A., Bell, G.A., Parrish, B.P., Hull, E.M., 2012. Dopamine D1 receptors and increased in the paraventricular nucleus of the hypothalamus of male rats during
phosphorylation of dopamine- and cyclic AMP-regulated phosphoprotein-32 in the non-contact penile erections and copulation. Eur. J. Neurosci. 10, 1968–1974.
medial preoptic area are involved in experience-induced enhancement of male Melis, M.R., Succu, S., Spano, M.S., Argiolas, A., 1999b. Morphine injected into the
sexual behavior in rats. Behav. Neurosci. 126, 523–529. paraventricular nucleus of the hypothalamus prevents noncontact penile erections
McInnis, C.M., Bonthuis, P.J., Rissman, E.F., Park, J.H., 2016. Inheritance of steroid- and impairs copulation involvement of nitric oxide. Eur. J. Neurosci. 11,
independent male sexual behavior in male offspring of B6D2F1 mice. Horm. Behav. 1857–1864.
80, 132–138. Melo, A.I., Chirino, R., Jiménez, A., Cuamatzi, E., Beyer, C., González-Mariscal, G.,
McIntosh, T.K., Barfield, R.J., 1984a. Brain monoaminergic control of male repro- 2008. Effect of forebrain implants of testosterone or estradiol on scent-marking
ductive behavior. I. Serotonin and the post-ejaculatory refractory period. Behav. and sexual behavior in male and female rabbits. Horm. Behav. 54, 676–683.
Brain Res. 12, 255–265. Mendelson, S.D., Pfaus, J.G., 1989. Level searching: a new assay of sexual motivation
McIntosh, T.K., Barfield, R.J., 1984b. Brain monoaminergic control of male repro- in the male rat. Physiol. Behav. 45, 337–341.
ductive behavior. II. Dopamine and the post-ejaculatory refractory period. Behav. Meredith, M., 1986. Vomeronasal organ removal before sexual experience impairs
Brain Res. 12, 267–273. male hamster mating behavior. Physiol. Behav. 36, 737–743.
McIntosh, T.K., Barfield, R.J., 1984c. Brain monoaminergic control of male repro- Meredith, M., Fernandez-Fewell, G., 1994. Vomeronasal system, LHRH, and sex
ductive behavior. III. Norepinephrine and the post-ejaculatory refractory period. behavior. Psychoneuroendocrinology 19, 657–672.
Behav. Brain Res. 12, 275–281. Meredith, M., Fewell, G., 2001. Vomeronasal organ: electrical stimulation activates Fos
McKenna, K.E., Chung, S.K., McVary, K.T., 1991. A model for the study of sexual in mating pathways and in GnRH neurons. Brain Res. 922, 87–94.
function in anesthetized male and female rats. Am. J. Physiol. 261, R1276–R1285. Meredith, M., Marques, D.M., O’Connell, R.J., Stem, F.L., 1980. Vomeronasal pump:
McKenna, K.E., Nadelhaft, I., 1986. The organization of the pudenda1 nerve in the significance for male hamster sexual behavior. Science 207, 1224–1226.
male and female rat. J. Comp. Neurol. 248, 532–549. Mermelstein, P.G., Becker, J.B., Surmeier, D.J., 1996. Estradiol reduces calcium
McMurray, G., Casey, J.H., Naylor, A.M., 2006. Animal models in urological disease currents in rat neostriatal neurons via a membrane receptor. J. Neurosci. 16,
and sexual dysfunction. Br. J. Pharmacol. 147 (Suppl. 2), S62–S79. 595–604.
Male Sexual Behavior 51

Michael, R.P., Clancy, A.N., Zumpe, D., 1999. Effects of mating on c-fos expression in Muschamp, J.W., Dominguez, J.M., Sato, S.M., Shen, R.Y., Hull, E.M., 2007. A role
the brains of male macaques. Physiol. Behav. 66, 591–597. for hypocretin (orexin) in male sexual behavior. J. Neurosci. 27, 2837–2845.
Millan, M.J., Peglion, J.L., Lavielle, G., Perrin-Monneyron, S., 1997. 5-HT2C receptors Musicki, B., Burnett, A.L., 2006. eNOS function and dysfunction in the penis. Exp. Biol.
mediate penile erections in rats, actions of novel and selective agonists and Med. (Maywood) 231, 154–165.
antagonists. Eur. J. Pharmacol. 325, 9–12. Myers, B., Baum, M.J., 1979. Facilitation by opiate antagonists of sexual performance
Miller, R.L., Baum, M.J., 1987. Naloxone inhibits mating and conditioned place in the male rat. Pharmacol. Biochem. Behav. 10, 615–618.
preference for an estrous female in male rats soon after castration. Pharmacol. Myers, B., Baum, M.J., 1980. Facilitation of copulatory performance in male rats by
Biochem. Behav. 26, 781–789. naloxone: effects of hypophysectomy, 17 alpha-estradiol, and luteinizing hormone
Mills, T.M., Chitaley, K., Lewis, R.W., Webb, R.C., 2002. Nitric oxide inhibits RhoA/ releasing hormone. Pharmacol. Biochem. Behav. 12, 365–370.
Rho-kinase signaling to cause penile erection. Eur. J. Pharmacol. 439, 173–174. Naftolin, F., Horvath, T.L., Jakab, R.L., Leranth, C., Harada, N., Balthazart, J., 1996.
Ming, X.F., Viswambharan, H., Barandier, C., Ruffieux, J., Kaibuchi, K., Rusconi, S., Aromatase immunoreactivity in axon terminals of the vertebrate brain. An immu-
Yang, Z., 2002. Rho GTPase/Rho kinase negatively regulates endothelial nitric nocytochemical study on quail, rat, monkey and human tissues. Neuroendocri-
oxide synthase phosphorylation through the inhibition of protein kinase B/Akt in nology 63, 149–155.
human endothelial cells. Mol. Cell. Biol. 22, 8467–8477. Narita, M., Nagumo, Y., Hashimoto, S., et al., 2006. Direct involvement of orexinergic
Missale, C., Nash, S.R., Robinson, S.W., Jaber, M., Caron, M.G., 1998. Dopamine systems in the activation of the mesolimbic dopamine pathway and related
receptors: from structure to function. Physiol. Rev. 78, 189–225. behaviors induced by morphine. J. Neurosci. 26, 398–405.
Mitchell, J.B., Stewart, J., 1990. Facilitation of sexual behaviors in the male rat associated Nauta, W.J., Smith, G.P., Faull, R.L., Domesick, V.B., 1978. Efferent connections and
with intra-VTA injections of opiates. Pharmacol. Biochem. Behav. 35, 643–650. nigral afferents of the nucleus accumbens septi in the rat. Neuroscience 3,
Mitsuya, H., Asai, J., Suyama, K., Ushida, T., Hosoe, K., 1976. Application of x-ray 385–401.
cinematography in urology: I. Mechanism of ejaculation. J. Urol. 83, 86–92. Navarro, V.M., Castellano, J.M., Garcia-Galiano, D., Tena-Sempere, M., 2007.
Miura, T., Kondo, Y., Akimoto, M., Sakuma, Y., 2001. Electromyography of male rat Neuroendocrine factors in the initiation of puberty: the emergent role of kisspeptin.
perineal musculature during copulatory behavior. Urol. Int. 67, 240–245. Rev. Endocr. Metab. Disord. 8, 11–20.
Mohr, M.A., Sisk, C.L., 2013. Pubertally born neurons and glia are functionally inte- Newman, H., Reiss, H., Northurp, J.D., 1982. Physical basis of emission, ejaculation,
grated into limbic and hypothalamic circuits of the male syrian hamster. Proc. Natl. and orgasm in the male. Urology 19, 341–350.
Acad. Sci. U.S.A. 110, 4792–4797. Newman, S.W., 1999. The medial extended amygdala in male reproductive behavior:
Monaghan, E.P., Breedlove, S.M., 1992. The role of the bulbocavernosus in penile a node in the mammalian social behavior network. Ann. N.Y. Acad. Sci. 877,
reflex behavior in rats. Brain Res. 587, 178–180. 242–257.
Monaghan, E.P., Arjomand, J., Breedlove, S.M., 1993. Brain lesions affect penile Nicola, S.M., 2007. The nucleus accumbens as part of a basal ganglia action selection
reflexes. Horm. Behav. 27, 122–131. circuit. Psychopharmacology (Berlin) 191, 521–550.
Moncada, I., 2006. Testosterone and men’s quality of life. Aging Male 9, 189–193. Nielsen, B.L., Jerome, N., Saint-Albin, A., Rampin, O., Maurin, Y., 2013. Behavioural
Moore, F.L., Evans, S.J., 1999. Steroid hormones use non-genomic mechanisms to control response of sexually naive and experienced male rats to the smell of 6-methyl-5-
brain functions and behaviors: a review of evidence. Brain Behav. Evol. 54, 41–50. hepten-2-one and female rat faeces. Physiol. Behav. 120, 150–155.
Moore, K.E., Lookingland, K.J., 1995. Dopaminergic neuronal systems in the hypo- Nishitani, S., Moriya, T., Kondo, Y., Sakuma, Y., Shinohara, K., 2004. Induction of Fos
thalamus. In: Bloom, F.E., Kupfer, D.J. (Eds.), Psychopharmacology: The Fourth immunoreactivity in oxytocin neurons in the paraventricular nucleus after female
Generation of Progress. Raven Press, New York, pp. 245–246 (Chapter 22). odor exposure in male rats: effects of sexual experience. Cell. Mol. Neurobiol. 24,
Moralí, G., Lemus, A.E., Munguia, R., Arteaga, M., Perez-Palacios, G., Sundaram, K., 283–291.
Kumar, N., Bardin, C.W., 1993. Induction of male sexual behavior in the rat by 7 Normandin, J.J., Murphy, A.Z., 2008. Nucleus paragigantocellularis afferents in male
alpha-methyl-19-nortestosterone, an androgen that does not undergo 5 alpha- and female rats: organization, gonadal steroid receptor expression, and activation
reduction. Biol. Reprod. 49, 577–581. during sexual behavior. J. Comp. Neurol. 508, 771–794.
Moralí, G., Pía, M.A., Contreras, J.L., Arteaga, M., González-Vidal, M.D., Beyer, C., Normandin, J.J., Murphy, A.Z., 2011. Serotonergic lesions of the periaqueductal
2003. Detailed analysis of the male copulatory motor pattern in mammals: gray, a primary source of serotonin to the nucleus paragigantocellularis, facilitate
hormonal bases. Scand. J. Psychol. 44, 279–288. sexual behavior in male rats. Pharmacol. Biochem. Behav. 98, 369–375.
Morris, J.A., Jordan, C.L., Breedlove, M., 2008. Sexual dimorphism in neuronal Nutsch, V.L., Will, R.G., Hattori, T., Tobiansky, D.J., Dominguez, J.M., 2014. Sexual
number of the posterodorsal medial amygdala is independent of circulating experience influences mating-induced activity in nitric oxide synthase-containing
androgens and regional volume in adult rats. J. Comp. Neurol. 506, 851–859. neurons in the medial preoptic area. Neurosci. Lett. 579, 92–96.
Mortimer, C.H., McNeilly, A.S., Fisher, R.A., Murray, M.A., Besser, G.M., 1974. Nutsch, V.L., Will, R.G., Robison, C.L., Martz, J.R., Tobiansky, D.J., Dominguez, J.M.,
Gonadotrophin-releasing hormone therapy in hypogonadal males with hypothalamic 2016. Colocalization of mating-induced Fos and D2-like dopamine receptors in the
or pituitary dysfunction. Br. Med. J. 4, 617–621. medial preoptic area: influence of sexual experience. Front. Behav. Neurosci.
Mos, J., Mollet, I., Tolboom, J.T., Waldinger, M.D., Olivier, B., 1999. A comparison of 10, 75.
the effects of different serotonin reuptake blockers on sexual behavior of the male Nyby, J., Matochik, J.A., Barfield, R.J., 1992. Intracranial androgenic and estrogenic
rat. Eur. Neuropsychopharmacol. 9, 123–135. stimulation of male-typical behaviors in house mice (Mus domesticus). Horm.
Moses, J., Hull, E.M., 1999. A nitric oxide synthesis inhibitor administered into the Behav. 26, 24–45.
medial preoptic area increases seminal emissions in an ex copula reflex test. O’Carroll, R., Shapiro, C., Bancroft, J., 1985. Androgens, behaviour and nocturnal
Pharmacol. Biochem. Behav. 63, 345–348. erection in hypogonadal men: the effects of varying the replacement dose. Clin.
Moses, J., Loucks, J.A., Watson, H.L., Matuszewich, L., Hull, E.M., 1995. Dopami- Endocrinol. (Oxf.) 23, 527–538.
nergic drugs in the medial preoptic area and nucleus accumbens: effects on motor O’Connell, R.J., Meredith, M., 1984. Effects of volatile and nonvolatile chemical signals
activity, sexual motivation, and sexual performance. Pharmacol. Biochem. Behav. on male sex behaviors mediated by the main and accessory olfactory systems.
51, 681–686. Behav. Neurosci. 98, 1083–1093.
Moss, R.L., McCann, S.M., Dudley, C.A., 1975. Releasing hormones and sexual Oei, N.Y., Rombouts, S.A., Soeter, R.P., van Gerven, J.M., Both, S., 2012. Dopamine
behavior. Prog. Brain Res. 42, 37–46. modulates reward system activity during subconscious processing of sexual stimuli.
Murphy, A.Z., Hoffman, G.E., 2001. Distribution of gonadal steroid receptor-containing Neuropsychopharmacology 37, 1729–1737.
neurons in the preoptic-periaqueductal gray-brainstem pathway: a potential circuit Ogawa, S., Chan, J., Chester, A.E., Gustafsson, J.A., Korach, K.S., Pfaff, D.W., 1999.
for the initiation of male sexual behavior. J. Comp. Neurol. 438, 191–212. Survival of reproductive behaviors in estrogen receptor beta gene-deficient
Murphy, A.Z., Rizvi, T.A., Ennis, M., Shipley, M.T., 1999a. The organization of (betaERKO) male and female mice. Proc. Natl. Acad. Sci. U.S.A. 96, 12887–12892.
preoptic-medullary circuits in the male rat, evidence for interconnectivity of neural Ogawa, S., Lubahn, D.B., Korach, K.S., Pfaff, D.W., 1997. Behavioral effects of
structures involved in reproductive behavior, antinociception and cardiovascular estrogen receptor gene disruption in male mice. Proc. Natl. Acad. Sci. U.S.A. 94,
regulation. Neuroscience 91, 1103–1116. 1476–1481.
Murphy, A.Z., Shupnik, M.A., Hoffman, G.E., 1999b. Androgen and estrogen (alpha) Ogawa, S., Robbins, A., Kumar, N., Pfaff, D.W., Sundaram, K., Bardin, C.W., 1996.
receptor distribution in the periaqueductal gray of the male rat. Horm. Behav. 36, Effects of testosterone and 7 alpha-methyl-19-nortestosterone (MENT) on sexual and
98–108. aggressive behaviors in two inbred strains of male mice. Horm. Behav. 30, 74–84.
Murphy, L.L., Gher, J., Steger, R.W., Bartke, A., 1994. Effects of delta 9- Ogawa, S., Washburn, T.F., Taylor, J., Lubahn, D.B., Korach, K.S., Pfaff, D.W., 1998.
tetrahydrocannabinol on copulatory behavior and neuroendocrine responses of Modification of testosterone-dependent behaviors by estrogen receptor-alpha gene
male rats to female conspecifics. Pharmacol. Biochem. Behav. 48, 1011–1017. disruption in male mice. Endocrinology 139, 5058–5069.
52 Male Sexual Behavior

O’Hanlon, J.K., Meisel, R.L., Sachs, B.D., 1981. Estradiol maintains castrated male Pfaus, J.G., Phillips, A.G., 1991. Role of dopamine in anticipatory and consummatory
rats’ sexual reflexes in copula, but not ex copula. Behav. Neural Biol. 32, aspects of sexual-behavior in the male-rat. Behav. Neurosci. 105, 727–743.
269–273. Pfaus, J.G., Wilkins, M.F., 1995. A novel environment disrupts copulation in sexually
Oliva, S.U., Messias, A.G., Silva, D.A., Pereira, O.C., Gerardin, D.C., Kempinas, W.G., naïve but not experienced male rats: reversal with naloxone. Physiol. Behav. 57,
2006. Impairment of adult male reproductive function in rats exposed to ethanol 1045–1049.
since puberty. Reprod. Toxicol. 22, 599–605. Pfaus, J.G., Damsma, G., Nomikos, G.G., Wenkstern, D.G., Blaha, C.D., Phillips, A.G.,
Olsen, C.M., 2011. Natural rewards, neuroplasticity, and non-drug addictions. Fibiger, H.C., 1990. Sexual behavior enhances central dopaminergic transmission
Neuropharmacology 61, 1109–1122. in the rat. Brain Res. 530, 345–348.
Oomura, Y., Aou, S., Koyama, Y., Yoshimatsu, H., 1988. Central control of sexual Pfaus, J.G., Kippin, T.E., Centeno, S., 2001. Conditioning and sexual behavior:
behavior. Brain Res. Bull. 20, 863–870. a review. Horm. Behav. 402, 291–321.
Oti, T., Takanami, K., Katayama, N., Edey, T., Satoh, K., Sakamoto, T., Sakamoto, H., Pfaus, J.G., Kippin, T.E., Coria-Avila, G.A., Gelez, H., Afonso, V.M., Ismail, N.,
2016. Perinatal testosterone exposure is critical for the development of the male- Parada, M., 2012. Who, what, where, when (and maybe even why)? How the
specific sexually dimorphic gastrin-releasing peptide system in the lumbosacral experience of sexual reward connects sexual desire, preference, and performance.
spinal cord that mediates erection and ejaculation. Biol. Sex Differ. 7, 4. Arch. Sex. Behav. 41, 31–62.
Pankevich, D.E., Baum, M.J., Cherry, J.A., 2004. Olfactory sex discrimination Pfeiffer, C.A., Johnston, R.E., 1994. Hormonal and behavioral responses of male
persists, whereas the preference for urinary odorants from estrous females hamsters to females and female odors: roles of olfaction, the vomeronasal system,
disappears in male mice after vomeronasal organ removal. J. Neurosci. 24, and sexual experience. Physiol. Behav. 55, 129–138.
9451–9457. Phelps, S.M., Lydon, J.P., O’Malley, B.W., Crews, D., 1998. Regulation of male sexual
Panzica, G.C., Viglietti-Panzica, C., Sica, M., Gotti, S., Martini, M., Pinos, H., behavior by progesterone receptor, sexual experience, and androgen. Horm. Behav.
Carrillo, B., Collado, P., 2006. Effects of gonadal hormones on central nitric oxide 34, 294–302.
producing systems. Neuroscience 13, 8987–8995. Phillips-Farfán, B.V., Fernández-Guasti, A., 2007. c-Fos expression related to sexual
Paredes, R.G., 2014. Opioids and sexual reward. Pharmacol. Biochem. Behav. 121, satiety in the male rat forebrain. Physiol. Behav. 91, 609–619.
124–131. Phoenix, C.H., Chambers, K.C., 1986. Threshold for behavioral response to testos-
Paredes, R.G., Baum, M.J., 1995. Altered sexual partner preference in male ferrets terone in old castrated male rhesus monkeys. Biol. Reprod. 35, 918–926.
given excitotoxic lesions of the preoptic area/anterior hypothalamus. J. Neurosci. Phoenix, C.H., Chambers, K.C., 1990. Sexual performance of old and young male
15, 6619–6630. rhesus macaques following treatment with GnRH. Physiol. Behav. 47, 513–517.
Paredes, R.G., Haller, A.E., Manero, M.C., Alvaradom, R., Ågmo, A., 1990. Medial Pitchers, K.K., Frohmader, K.S., Vialou, V., Mouzon, E., Nestler, E.J., Lehman, M.N.,
preoptic area kindling induces sexual behavior in sexually inactive male rats. Brain Coolen, L.M., 2010. DeltaFosB in the nucleus accumbens is critical for reinforcing
Res. 515, 20–26. effects of sexual reward. Genes Brain Behav. 9, 831–840.
Paredes, R.G., Highland, L., Karam, P., 1993. Socio-sexual behavior in male rats after Pitchers, K.K., Schmid, S., Di Sebastiano, A.R., Wang, X., Laviolette, S.R.,
lesions of the medial preoptic area: evidence for reduced sexual motivation. Brain Lehman, M.N., Coolen, L.M., 2012. Natural reward experience alters AMPA and
Res. 618, 271–276. NMDA receptor distribution and function in the nucleus accumbens. PLoS One 7,
Paredes, R.G., Karam, P., Highland, L., Ågmo, A., 1997. GABAergic drugs and socio- e34700.
sexual behavior. Pharmacol. Biochem. Behav. 58, 291–298. Politis, M., Loane, C., Wu, K., et al., 2013. Neural response to visual sexual cues in
Paredes, R.G., Tzschentke, T., Nakach, N., 1998. Lesions of the medial preoptic area/ dopamine treatment-linked hypersexuality in Parkinson’s disease. Brain 136, 400–411.
anterior hypothalamus (MPOA/AH) modify partner preference in male rats. Brain Pomerantz, S.M., 1991. Quinelorane (LY163502), a D2 dopamine receptor agonist,
Res. 813, 81–83. acts centrally to facilitate penile erections of male rhesus monkeys. Pharmacol.
Parfitt, D.B., Newman, S.W., 1998. Fos-immunoreactivity within the extended Biochem. Behav. 39, 123–128.
amygdala is correlated with the onset of sexual satiety. Horm. Behav. 34, 17–29. Pomerantz, S.M., Clemens, L.G., 1981. Ultrasonic vocalizations in male deer mice
Parfitt, D.B., Coolen, L.M., Newman, S.W., Wood, R.I., 1996. Lesions of the posterior (Peromyscus maniculatus bairdi), their role in male sexual behavior. Physiol.
medial nucleus of the amygdala delay sexual satiety. Soc. Neurosci. Abstr. Behav. 27, 869–872.
22, 155. Pomerantz, S.M., Hepner, B.C., Wertz, J.M., 1993. 5-HT1A and 5-HT1C/1D receptor
Parra-Gamez, L., Garcia-Hidalgo, A., Paredes, R.G., 2013. Infusion of endomorphin-1 agonists produce reciprocal effects on male sexual behavior of rhesus monkeys.
(EM-1) in the MPOA and the Me modulate sexual and socio-sexual behavior in the Eur. J. Pharmacol. 243, 227–234.
male rat. Brain Res. 1517, 36–43. Portillo, W., Paredes, R.G., 2004. Sexual incentive motivation, olfactory preference,
Pattij, T., de Jong, T.R., Uitterdijk, A., Waldinger, M.D., Veening, J.G., Cools, A.R., and activation of the vomeronasal projection pathway by sexually relevant cues in
van der Graaf, P.H., Olivier, B., 2005. Individual differences in male rat ejac- non-copulating and naive male rats. Horm. Behav. 46, 330–340.
ulatory behavior: searching for models to study ejaculation disorders. Eur. J. Portillo, W., Basanez, E., Paredes, R.G., 2003. Permanent changes in sexual behavior
Neurosci. 22, 724–734. induced by medial preoptic area kindling-like stimulation. Brain Res. 96, 10–14.
Paul, S.M., 1995. GABA and glycine. In: Bloom, F.E., Kupfer, D.J. (Eds.), Psycho- Portillo, W., Diaz, N.F., Retana-Marquez, S., Paredes, R.G., 2006. Olfactory, partner
pharmacology: The Fourth Generation of Progress. Raven Press, New York, preference and Fos expression in the vomeronasal projection pathway of sexually
pp. 87–94. sluggish male rats. Physiol. Behav. 88, 389–397.
Peeters, M., Giuliano, F., 2008. Central neurophysiology and dopaminergic control of Powell, W.S., Dominguez, J.M., Hull, E.M., 2003. An NMDA antagonist impairs
ejaculation. Neurosci. Biobehav. Rev. 32, 438–453. copulation and the experience induced enhancement of male sexual behavior in the
Pellegrini-Quarantotti, B., Corda, M., Paglietti, E., Biggio, G., Gessa, G.L., 1978. rat. Behav. Neurosci. 117, 69–75.
Inhibition of copulatory behavior in male rats by D-Ala2-Met-Enkephalinamide. Life Putnam, S.K., Du, J., Hull, E.M., 2001. Testosterone restoration of copulation and
Sci. 23, 673–678. medial preoptic dopamine release in castrated male rats: 2-, 5-, and 10-day
Perry, A.N., Carter, C.S., Cushing, B.S., 2016. Chronic social isolation enhances treatments. Horm. Behav. 39, 216–224.
reproduction in the monogamous prairie vole (Microtus ochrogaster). Psycho- Putnam, S.K., Sato, S., Hull, E.M., 2003. Hormonal maintenance of copulation in
neuroendocrinology 68, 20–28. castrates: association with extracellular dopamine in MPOA. Horm. Behav. 44,
Peyron, C., Tighe, D.K., van den Pol, A.N., de Lecea, L., Heller, H.C., Sutcliffe, J.G., 419–426.
Kilduff, T.S., 1998. Neurons containing hypocretin (orexin) project to multiple Putnam, S.K., Sato, S., Riolo, J.V., Hull, E.M., 2005. Effects of testosterone metab-
neuronal systems. J. Neurosci. 18, 9996–10015. olites on copulation, medial preoptic dopamine, and NOS-immunoreactivity in
Pfaff, D.W., Pfaffmann, C., 1969. Olfactory and hormonal influences on the basal castrated male rats. Horm. Behav. 47, 513–522.
forebrain of the male rat. Brain Res. 15, 137–156. Qureshi, G.A., Södersten, P., 1986. Sexual activity alters the concentration of amino
Pfaus, J.G., 1996. Frank A. Beach awarddhomologies of of animal and human sexual acids in the cerebrospinal fluid of male rats. Neurosci. Lett. 70, 374–378.
behaviors. Horm. Behav. 30, 187–200. Rajasekaran, M., White, S., Baquir, A., Wilkes, N., 2005. Rho-kinase inhibition improves
Pfaus, J.G., 2009. Pathways of sexual desire. J. Sex. Med. 6, 1506–1533. erectile function in aging male Brown-Norway rats. J. Androl. 26, 182–188.
Pfaus, J.G., Gorzalka, B.B., 1987. Opioids and sexual behavior. Neurosci. Biobehav. Rampin, O., Jerome, N., Suaudeau, C., 2003. Proerectile effects of apomorphine in
Rev. 11, 1–34. mice. Life Sci. 72, 2329–2336.
Pfaus, J.G., Phillips, A.G., 1989. Differential effects of dopamine receptor antag- Rampin, O., Monnerie, R., Jerome, N., McKenna, K., Maurin, Y., 2004. Spinal control
onists on the sexual behavior of male rats. Psychopharmacology (Berlin) 98, of erection by glutamate in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 286,
363–368. R710–R718.
Male Sexual Behavior 53

Rasia-Filho, A.A., Peres, T.M., Cubilla-Gutierrez, F.H., Lucion, A.B., 1991. Effect of Rodríguez-Manzo, G., Asai, M., Fernández-Guasti, A., 2002a. Evidence for changes in
estradiol implanted in the corticomedial amygdala on the sexual behavior of cas- brain enkephalin contents associated to male rat sexual activity. Behav. Brain Res.
trated male rats. Braz. J. Med. Biol. Res. 24, 1041–1049. 131, 47–55.
Ratnasooriya, W.D., Dharmasiri, M.G., Wadsworth, R.M., 2000. Reduction in libido and Rodriguez-Manzo, G., Guadarrama-Bazante, I.L., Morales-Calderon, A., 2011.
fertility of male rats by administration of the nitric oxide (NO) synthase inhibitor N- Recovery from sexual exhaustion-induced copulatory inhibition and drug hyper-
nitro-L-arginine methylesther. Int. J. Androl. 23, 187–191. sensitivity follow a same time course: two expressions of a same process? Behav.
Redoute, J., Stoleru, S., Gregoire, M.C., et al., 2000. Brain processing of visual sexual Brain Res. 217, 253–260.
stimuli in human males. Hum. Brain Mapp. 11, 162–177. Rodríguez-Manzo, G., López-Rubalcava, C., Fernández-Guasti, A., 1999. Anxiolytic-like
Redoute, J., Stoleru, S., Pugeat, M., et al., 2005. Brain processing of visual sexual effect of ejaculation under various sexual behavior conditions in the male rat.
stimuli in treated and untreated hypogonadal patients. Psychoneuroendocrinology Physiol. Behav. 67, 651–657.
30, 461–482. Rodríguez-Manzo, G., López-Rubalcava, C., Hen, R., Fernández-Guasti, A., 2002b.
Rees, R.W., Ralph, D.J., Royle, M., Moncada, S., Cellek, S., 2001. Y-27632, an Participation of 5-HT1B receptors in the inhibitory actions of serotonin on masculine
inhibitor of Rho-kinase, antagonizes noradrenergic contractions in the rabbit and sexual behavior of mice: pharmacological analysis in 5-HT1B receptor knockout
human corpus cavernosum. Br. J. Pharmacol. 133, 455–458. mice. Br. J. Pharmacol. 136, 1127–1134.
Rehman, J., Kaynan, A., Christ, G., Valcic, M., Maayani, S., Melman, A., 1999. Rodríguez-Manzo, G., Pellicer, F., Larsson, K., Fernández-Guasti, A., 2000. Stimulation
Modification of sexual behavior of Long-Evans male rats by drugs acting on the 5- of the medical preoptic area facilitates sexual behavior but does not reverse sexual
HT1A receptor. Brain Res. 821, 414–425. satiation. Behav. Neurosci. 114, 553–560.
Resko, J.A., Perkins, A., Roselli, C.E., Stellflug, J.N., Stormshak, F.K., 1999. Sexual Rojas-Duran, F., Pascual-Mathey, L.I., Serrano, K., Aranda-Abreu, G.E., Manzo, J.,
behaviour of rams: male orientation and its endocrine correlates. J. Reprod. Fertil. Soto-Cid, A.H., Hernandez, M.E., 2015. Correlation of prolactin levels and PRL-
Suppl. 54, 259–269. receptor expression with stat and MAPK cell signaling in the prostate of long-
Rhoden, E.L., Teloken, C., Mafessoni, R., Souto, C.A., 2002. Is there any relation term sexually active rats. Physiol. Behav. 138, 188–192.
between serum levels of total testosterone and the severity of erectile dysfunction? Rojas-Hernandez, J., Juarez, J., 2015. Copulation is reactivated by bromocriptine in
Int. J. Impot. Res. 14, 167–171. male rats after reaching sexual satiety with a same sexual mate. Physiol. Behav.
Riolo, J.V., Friedman, R.D., Hull, E.M., 1999. Microinjection of Selective 5-HT2 and 5- 151, 551–556.
HT1A Agonists and Antagonists: Effects on Male Rat Copulatory Behavior. 29th Romeo, R.D., 2005. Neuroendocrine and behavioral development during puberty:
Annual Meeting of the SFN, Miami Beach, Florida. a tale of two axes. Vitam. Horm. 71, 1–25.
Riters, L.V., Ball, G.F., 1999. Lesions to the medial preoptic area affect singing in the Romeo, R.D., 2010. Adolescence: a central event in shaping stress reactivity. Dev.
male European starling (Sturnus vulgaris). Horm. Behav. 36, 276–286. Psychobiol. 52, 244–253.
Riters, L.V., Pawlisch, B.A., Kelm-Nelson, C.A., Stevenson, S.A., 2014. Inverted-U Romeo, R.D., Richardson, H.N., Sisk, C.L., 2002. Puberty and the maturation of the
shaped effects of D1 dopamine receptor stimulation in the medial preoptic male brain and sexual behavior: recasting a behavioral potential. Neurosci. Bio-
nucleus on sexually motivated song in male European starlings. Eur. J. Neurosci. behav. Rev. 26, 381–391.
39, 650–662. Romero-Carbente, J.C., Hurtazo, E.A., Paredes, R.G., 2007. Central tegmental field
Robbins, T.W., Everitt, B.J., 2007. A role for mesencephalic dopamine in activation: and sexual behavior in the male rat: effects of neurotoxic lesions. Neuroscience
commentary on Berridge (2006). Psychopharmacology (Berlin) 191, 433–437. 148, 867–875.
Robertson, G.S., Pfaus, J.G., Atkinson, L.J., Matsumura, H., Phillips, A.G., Roselli, C.E., Cross, E., Poonyagariyagorn, H.K., Stadelman, H.L., 2003. Role of
Fibiger, H.C., 1991. Sexual behavior increases c-Fos expression in the forebrain of aromatization in anticipatory and consummatory aspects of sexual behavior in male
the male rat. Brain Res. 564, 352–357. rats. Horm. Behav. 44, 146–151.
Robinson, D.L., Heien, M.L., Wightman, R.M., 2002. Frequency of dopamine Roselli, C.E., Thornton, J.E., Chambers, K.C., 1993. Age-related deficits in brain
concentration transients increases in dorsal and ventral striatum of male rats estrogen receptors and sexual behavior of male rats. Behav. Neurosci. 107,
during introduction of conspecifics. J. Neurosci. 22, 10477–10486. 202–209.
Rodríguez-Manzo, G., 1999a. Blockade of the establishment of the sexual inhibition Rosenblatt, J.S., Aronson, L.R., 1958. The decline of sexual behavior in male cats after
resulting from sexual exhaustion by the Coolidge effect. Behav. Brain Res. 100, castration with special reference to the role of prior sexual experience. Behaviour
245–254. 12, 285–338.
Rodríguez-Manzo, G., 1999b. Yohimbine interacts with the dopaminergic system to Roudet, C., Mouchet, P., Feuerstein, C., Savasta, M., 1994. Normal distribution of alpha
reverse sexual satiation: further evidence for a role of sexual motivation in sexual 2-adrenoceptors in the rat spinal cord and its modification after noradrenergic dener-
exhaustion. Eur. J. Pharmacol. 372, 1–8. vation: a quantitative autoradiographic study. J. Neurosci. Res. 39, 319–329.
Rodriguez-Manzo, G., 2015. Glutamatergic transmission is involved in the long lasting Rowland, D.L., Houtsmuller, E.J., 1998. 8-OH-DPAT interacts with sexual experience
sexual inhibition of sexually exhausted male rats. Pharmacol. Biochem. Behav. and testosterone to affect ejaculatory response in rats. Pharmacol. Biochem.
131, 64–70. Behav. 60, 143–149.
Rodriguez-Manzo, G., Canseco-Alba, A., 2014. A role for learning and memory in the Rubio-Casillas, A., Rodriguez-Quintero, C.M., Rodriguez-Manzo, G., Fernandez-
expression of an innate behavior: the case of copulatory behavior. In: Meneses, A. Guasti, A., 2015. Unraveling the modulatory actions of serotonin on male rat
(Ed.), Identification of Neural Markers Accompanying Memory. Elsevier Press, sexual responses. Neurosci. Biobehav. Rev. 55, 234–246.
pp. 135–147. Sachs, B.D., 1980. Sexual reflexes of spinal male house mice. Physiol. Behav. 24,
Rodriguez-Manzo, G., Canseco-Alba, A., 2015a. Anandamide reduces the ejaculatory 489–492.
threshold of sexually sluggish male rats: possible relevance for human lifelong Sachs, B.D., 1982. Role of striated penile muscles in penile reflexes, copulation, and
delayed ejaculation disorder. J. Sex. Med. 12, 1128–1135. induction of pregnancy in the rat. J. Reprod. Fertil. 66, 433–443.
Rodriguez-Manzo, G., Canseco-Alba, A., 2015b. Biphasic effects of anandamide on Sachs, B.D., 1983. Potency and fertility: hormonal and mechanical causes
behavioural responses: emphasis on copulatory behaviour. Behav. Pharmacol. 26, and effects of penile actions in rats. In: Balthazart, J., Pröve, E., Gilles, R.
607–615. (Eds.), Hormones and Behaviour in Higher Vertebrates. Springer-Verlag, Berlin,
Rodríguez-Manzo, G., Fernández-Guasti, A., 1994. Reversal of sexual exhaustion by pp. 86–110 (Chapter 7).
serotonergic and noradrenergic agents. Behav. Brain Res. 62, 127–134. Sachs, B.D., 1996. Penile erection in response to remote cues from females: albino
Rodríguez-Manzo, G., Fernández-Guasti, A., 1995a. Opioid antagonists and the sexual rats severely impaired relative to pigmented strains. Physiol. Behav. 60, 803–808.
satiation phenomenon. Psychopharmacology (Berlin) 122, 131–136. Sachs, B.D., 1997. Erection evoked in male rats by airborne scent from estrous
Rodríguez-Manzo, G., Fernández-Guasti, A., 1995b. Participation of the central females. Physiol. Behav. 62, 921–924.
noradrenergic system in the reestablishment of copulatory behavior of sexually Sachs, B.D., 2000. Contextual approaches to the physiology and classification of
exhausted rats by yohimbine, naloxone and 8-OH-DPAT. Brain Res. Bull. 38, erectile function, erectile dysfunction, and sexual arousal. Neurosci. Biobehav. Rev.
399–404. 24, 541–560.
Rodríguez-Manzo, G., Pellicer, F., 2007. Electrical stimulation of the ventral tegmental Sachs, B.D., Barfield, R., 1976. Functional analysis of masculine copulatory behavior in
area exerts opposite effects on male rat sexual behavior expression depending on the rat. Adv. Stud. Behav. 7, 91–154.
the stimulated sub region. Behav. Brain Res. 179, 310–313. Sachs, B.D., Leipheimer, R.E., 1988. Rapid effect of testosterone on striated muscle
Rodriguez-Manzo, G., Pellicer, F., 2010. Electrical stimulation of dorsal and ventral activity in rats. Neuroendocrinology 48, 453–458.
striatum differentially alters the copulatory behavior of male rats. Behav. Neurosci. Sachs, B.D., Meisel, R.L., 1979. Pubertal development of penile reflexes and copu-
124, 686–694. lation in male rats. Psychoneuroendocrinology 4, 287–296.
54 Male Sexual Behavior

Sachs, B.D., Akasofu, K., McEldowney, S., 1994. Effects of copulation on Schmidt, M.H., Schmidt, H.S., 1993. The ischiocavernosus and bulbospongiosus
apomorphine-induced erection in rats. Pharmacol. Biochem. Behav. 48, 423–428. muscles in mammalian penile rigidity. Sleep 16, 171–183.
Sachs, B., Valcourt, R., Flagg, H., 1981. Copulatory behavior and sexual reflexes of Schmidt, M.H., Valatx, J.L., Sakai, K., Debilly, G., Jouvet, M., 1995. Corpus spon-
male rats treated with naloxone. Pharmacol. Biochem. Behav. 14, 251–253. giosum penis pressure and perineal muscle activity during reflexive erections in the
Saito, T.R., Moltz, H., 1986. Copulatory behavior of sexually naive and sexually rat. Am. J. Physiol. 269, R904–R913.
experienced male rats following removal of the vomeronasal organ. Physiol. Behav. Schneider, M., 2013. Adolescence as a vulnerable period to alter rodent behavior. Cell
37, 507–510. Tissue Res. 354, 99–106.
Sakamoto, H., 2011. Gastrin-releasing peptide system in the spinal cord mediates Schneider, J.S., Burgess, C., Sleiter, N.C., DonCarlos, L.L., Lydon, J.P., O’Malley, B.,
masculine sexual function. Anat. Sci. Int. 86, 19–29. Levine, J.E., 2005. Enhanced sexual behaviors and androgen receptor immuno-
Sakamoto, H., Matsuda, K., Zuloaga, D.G., et al., 2008. Sexually dimorphic gastrin reactivity in the male progesterone receptor knockout mouse. Endocrinology 146,
releasing peptide system in the spinal cord controls male reproductive functions. 4340–4348.
Nat. Neurosci. 11, 634–636. Schnur, S.L., Smith, E.R., Lee, R.L., Mas, M., Davidson, J.M., 1989. A component
Sakata, J.T., Crews, D., 2004. Cytochrome oxidase activity in the preoptic area analysis of the effects of DPAT on male rat sexual behavior. Physiol. Behav. 45,
correlates with differences in sexual behavior of intact and castrated male leopard 897–901.
geckos (Eublepharis macularius). Behav. Neurosci. 118, 857–862. Schober, J., Weil, Z., Pfaff, D., 2011. How generalized CNS arousal strengthens sexual
Sala, M., Braida, D., Leone, M.P., Calcaterra, P., Monti, S., Gori, E., 1990. Central arousal (and vice versa). Horm. Behav. 59, 689–695.
effect of yohimbine on sexual behavior in the rat. Physiol. Behav. 47, 165–173. Schrøder, H.D., 1980. Organization of the motoneurons innervating the pelvic muscles
Salmimies, P., Kockott, G., Pirke, K.M., Vogt, H.J., Schill, W.B., 1982. Effects of of the male rat. J. Comp. Neurol. 192, 567–587.
testosterone replacement on sexual behavior in hypogonadal men. Arch. Sex. Schroeder, M.B., Riters, L.V., 2006. Pharmacological manipulations of dopamine and
Behav. 11, 345–353. opioids have differential effects on sexually motivated song in male European
Sanna, F., Contini, A., Melis, M.R., Argiolas, A., 2015. Role of dopamine D4 receptors starlings. Physiol. Behav. 88, 575–584.
in copulatory behavior: studies with selective D4 agonists and antagonists in male Schultheiss, D., Hiltl, D.M., Meschi, M.R., Machtens, S.A., Truss, M.C., Stief, C.G.,
rats. Pharmacol. Biochem. Behav. 137, 110–118. Jonas, U., 2000. Pilot study of the transdermal application of testosterone gel to
Sanna, F., Piludu, M.A., Corda, M.G., Argiolas, A., Giorgi, O., Melis, M.R., 2014. the penile skin for the treatment of hypogonadotropic men with erectile dysfunc-
Dopamine is involved in the different patterns of copulatory behaviour of Roman tion. World J. Urol. 18, 431–435.
high and low avoidance rats: studies with apomorphine and haloperidol. Phar- Schulz, K.M., Sisk, C.L., 2006. Pubertal hormones, the adolescent brain, and the
macol. Biochem. Behav. 124, 211–219. maturation of social behaviors: lessons from the Syrian hamster. Mol. Cell.
Sanna, F., Succu, S., Boi, A., Melis, M.R., Argiolas, A., 2009. Phosphodiesterase type Endocrinol. 254–255, 120–126.
5 inhibitors facilitate noncontact erections in male rats: site of action in the brain Schulz, K.M., Molenda-Figueira, H.A., Sisk, C.L., 2009. Back to the future: the
and mechanism of action. J. Sex. Med. 6, 2680–2689. organizational-activational hypothesis adapted to puberty and adolescence. Horm.
Sanna, F., Succu, S., Hubner, H., Gmeiner, P., Argiolas, A., Melis, M.R., 2011. Behav. 55, 597–604.
Dopamine D2-like receptor agonists induce penile erection in male rats: differential Schulz, K.M., Richardson, H.N., Romeo, R.D., Morris, J.A., Lookingland, K.J.,
role of D2, D3 and D4 receptors in the paraventricular nucleus of the hypothalamus. Sisk, C.L., 2003. Medial preoptic area dopaminergic responses to female phero-
Behav. Brain Res. 225, 169–176. mones develop during puberty in the male Syrian hamster. Brain Res. 988,
Sanna, F., Succu, S., Melis, M.R., Argiolas, A., 2012. Dopamine agonist-induced 139–145.
penile erection and yawning: differential role of D(2)-like receptor subtypes and Schulz, K.M., Richardson, H.N., Zehr, J.L., Osetek, A.J., Menard, T.A., Sisk, C.L.,
correlation with nitric oxide production in the paraventricular nucleus of the 2004. Gonadal hormones masculinize and defeminize reproductive behaviors
hypothalamus of male rats. Behav. Brain Res. 230, 355–364. during puberty in the male Syrian hamster. Horm. Behav. 45, 242–249.
Santtila, P., Jern, P., Westberg, L., Walum, H., Pedersen, C.T., Eriksson, E., Scordalakes, E.M., Shetty, S.J., Rissman, E.F., 2002. Roles of estrogen receptor alpha
Kenneth Sandnabba, N., 2010. The dopamine transporter gene (DAT1) poly- and androgen receptor in the regulation of neuronal nitric oxide synthase. J. Comp.
morphism is associated with premature ejaculation. J. Sex. Med. 7, Neurol. 453, 336–344.
1538–1546. Shafik, A., 1997. Pelvic floor muscles and sphincters during erection and ejaculation.
Sato, S., Braham, C.S., Putnam, S.K., Hull, E.M., 2005. Neuronal nitric oxide synthase Arch. Androl. 39, 71–78.
and gonadal steroid interaction in the MPOA of male rats: co-localization and Shafik, A., Shafik, A.A., Shafik, I., El-Sibai, O., 2005. Urethral sphincters response to
testosterone-induced restoration of copulation and nNOS-immunoreactivity. Brain cavernosus muscles stimulation with identification of cavernoso-urethral reflex.
Res. 1043, 205–213. Arch. Androl. 51, 335–343.
Sato, S.M., Hull, E.M., 2006. The nitric oxide-guanosine 3ʹ, 5ʹ- cyclic monophosphate Shakil, T., Hoque, A.N., Husain, M., Belsham, D.D., 2002. Differential regulation
pathway regulates dopamine efflux in the medial preoptic area and copulation in of gonadotropin-releasing hormone secretion and gene expression by
male rats. Neuroscience 139, 417–428. androgen: membrane versus nuclear receptor activation. Mol. Endocrinol. 16,
Sato, S.M., Wersinger, S.R., Hull, E.M., 2007. The effect of nitric oxide-cGMP pathway 2592–2602.
stimulation on dopamine in the medial preoptic area and copulation in DHT- treated Shimura, T., Shimokochi, M., 1991. Modification of male rat copulatory behavior by
castrated male rats. Horm. Behav. 52, 177–182. lateral midbrain stimulation. Physiol. Behav. 50, 989–994.
Sato, Y., Christ, G.J., Horita, H., Adachi, H., Suzuki, N., Tsukamoto, T., 1999. The Shimura, T., Yamamoto, T., Shimokochi, M., 1994. The medial preoptic area is
effects of alterations in nitric oxide levels in the paraventricular nucleus on involved in both sexual arousal and performance in male rats: re-evaluation of
copulatory behavior and reflexive erections in male rats. J. Urol. 162, neuron activity in freely moving animals. Brain Res. 640, 215–222.
2182–2185. Shrenker, P., Barke, A., 1985. Suppression of male copulatory behavior by
Sato, Y., Horita, H., Kurohata, T., Adachi, H., Tsukamoto, T., 1998. Effect of the nitric delta 9-THC is not dependent on changes in plasma testosterone or hypo-
oxide level in the medial preoptic area on male copulatory behavior in rats. Am. J. thalamic dopamine or serotonin content. Pharmacol. Biochem. Behav. 22,
Physiol. 274, R243–R247. 415–420.
Sato, Y., Wada, H., Horita, H., Suzuki, N., Shibuya, A., Adachi, H., Kato, R., Silva, N.L., Boulant, J.A., 1986. Effects of testosterone, estradiol, and temperature on
Tsukamoto, T., Kumamoto, Y., 1995. Dopamine release in the medial preoptic area neurons in preoptic tissue slices. Am. J. Physiol. 250, R625–R632.
during male copulatory behavior in rats. Brain Res. 692, 66–70. Simerly, R.B., 1995. Hormonal regulation of limbic and hypothalalmic pathways.
Sato, Y., Zhao, W., Christ, G.J., 2001. Central modulation of the NO/cGMP pathway In: Micevych, P.E., Hammer Jr., R.P. (Eds.), Neurobiological Effects of Sex
affects the MPOA-induced intracavernous pressure response. Am. J. Physiol. Steroid Hormones. Cambridge University Press, New York, pp. 85–116
Regul. Integr. Comp. Physiol. 281, R269–R278. (Chapter 4).
Sayag, N., Hoffman, N.W., Yahr, P., 1994. Telencephalic connections of the sexually Simerly, R.B., Swanson, L.W., 1986. The organization of neural inputs to the medial
dimorphic area of the gerbil hypothalamus that influence male sexual behavior. preoptic nucleus of the rat. J. Comp. Neurol. 246, 312–342.
Behav. Neurosci. 108, 743–757. Simerly, R.B., Swanson, L.W., 1987. Castration reversibly alters levels of cholecys-
Scaletta, L.L., Hull, E.M., 1990. Systemic or intracranial apomorphine increases tokinin immunoreactivity in cells of three interconnected sexually dimorphic fore-
copulation in long-term castrated male rats. Pharmacol. Biochem. Behav. 37, brain nuclei in the rat. Proc. Natl. Acad. Sci. U.S.A. 84, 2087–2091.
471–475. Simerly, R.B., Swanson, L.W., 1988. Projections of the medial preoptic nucleus:
Scherf, K.S., Smyth, J.M., Delgado, M.R., 2013. The amygdala: an agent of change in a Phaseolus vulgaris leucoagglutinin anterograde tract-tracing study in the rat.
adolescent neural networks. Horm. Behav. 64, 298–313. J. Comp. Neurol. 270, 209–242.
Male Sexual Behavior 55

Simerly, R.B., Chang, C., Muramatsu, M., Swanson, L.W., 1990. Distribution of Steers, W.D., DeGroat, W.C., 1989. Effects of m-chlorophenylpiperazine on penile and
androgen and estrogen mRNA-containing cells in the rat brain: an in situ bladder function in rats. Am. J. Physiol. 257, R1441–R1449.
hybridization study. J. Comp. Neurol. 294, 76–95. Stefanick, M.L., Davidson, J.M., 1987. Genital responses in noncopulators and rats
Simerly, R.B., Gorski, R.A., Swanson, L.W., 1986. Neurotransmitter specificity of cells with lesions in the medial preoptic area or midthoracic spinal cord. Physiol. Behav.
and fibers in the medial preoptic nucleus: an immunohistochemical study in the rat. 41, 439–444.
J. Comp. Neurol. 246, 343–363. Stoleru, S., Gregoire, M.C., Gerard, D., Decety, J., Lafarge, E., Cinotti, L., Lavenne, F.,
Simmons, D., Yahr, P., 2002. Projections of the posterodorsal preoptic nucleus and Le Bars, D., Vernet-Maury, E., Rada, H., Collet, C., Mazoyer, B., Forest, M.G.,
the lateral part of the posterodorsal mudial amygdala in male gerbils, with Magnin, F., Spira, A., Comar, D., 1999. Neuroanatomical correlates of visually
emphasis on cells activated with ejaculation. J. Comp. Neurol. 444, 75–94. evoked sexual arousal in human males. Arch. Sex. Behav. 28, 1–21.
Simmons, D.A., Yahr, P., 2003. GABA and glutamate in mating-activated cells in the Stowers, L., Holy, T.E., Meister, M., Dulac, C., Koentges, G., 2002. Loss of sex
preoptic area and medial amygdala of male gerbils. J. Comp. Neurol. 459, discrimination and male-male aggression in mice deficient for TRP2. Science 295,
290–300. 1493–1500.
Simmons, D.A., Hoffman, N.W., Yahr, P., 2011. A forebrain-retrorubral pathway Struthers, W.M., 2001. Sex-induced fos in the medial preoptic area: projections to the
involved in male sex behavior is GABAergic and activated with mating in gerbils. midbrain. Neuroreport 12, 3065–3068.
Neuroscience 175, 162–168. Succu, S., Mascia, M.S., Melis, T., Sanna, F., Boi, A., Melis, M.R., Argiolas, A.,
Singh, R., Pervin, S., Shryne, J., Gorski, R., Chaudhuri, G., 2000. Castration increases 2006a. Morphine reduces penile erection induced by the cannabinoid receptor
and androgens decrease nitric oxide synthase activity in the brain: physiologic antagonist SR 141617A in male rats: role of paraventricular glutamic acid and
implications. Proc. Natl. Acad. Sci. U.S.A. 97, 3672–3677. nitric oxide. Neurosci. Lett. 404, 1–5.
Sipos, M.L., Nyby, J.G., 1998. Intracranial androgenic activation of male-typical Succu, S., Mascia, M.S., Sanna, F., Melis, T., Argiolas, A., Melis, M.R., 2006b. The
behaviours in house mice: concurrent stimulation of the medial preoptic area cannabinoid CB1 receptor antagonist SR 141716A induces penile erection by
and medial nucleus of the amygdala. J. Neuroendocrinol. 10, 577–586. increasing extra-cellular glutamic acid in the paraventricular nucleus of male rats.
Sisk, C.L., Zehr, J.L., 2005. Pubertal hormones organize the adolescent brain and Behav. Brain Res. 169, 274–281.
behavior. Front. Neuroendocrinol. 26, 163–174. Succu, S., Sanna, F., Argiolas, A., Melis, M.R., 2011. Oxytocin injected into the
Sisk, C.L., Schulz, K.M., Zehr, J.L., 2003. Puberty: a finishing school for male social hippocampal ventral subiculum induces penile erection in male rats by increasing
behavior. Ann. N.Y. Acad. Sci. 1007, 189–198. glutamatergic neurotransmission in the ventral tegmental area. Neuropharmacology
Smith, E.R., Davidson, J.M., 1990. Yohimbine attenuates aging-induce sexual defi- 61, 181–188.
ciencies in male rats. Physiol. Behav. 47, 631–634. Succu, S., Spano, M.S., Melis, M.R., Argiolas, A., 1998. Different effects of omega-
Smith, E.R., Cetrulo, E.J., Donahue, J.P., Sparrow, H., Davidson, J.M., Hoffman, B.B., conotoxin on penile erection, yawning and paraventricular nitric oxide in male rats.
Clark, J.T., 1995. The inhibitory effects of propranolol on genital reflexes in male Eur. J. Pharmacol. 359, 19–26.
rats. Pharmacol. Biochem. Behav. 52, 541–546. Suter, K.J., Pohl, C.R., Plant, T.M., 1998. The pattern and tempo of the pubertal
Smith, E.R., Kacker, S.R., Raskin, A., Yun, P.T., Davidson, J.M., Hoffman, B.B., reaugmentation of open-loop pulsatile gonadotropin-releasing hormone release
Clark, J.T., 1996. Central propanolol and pindolol, but not atenolol nor metoprolol, assessed indirectly in the male rhesus monkey (Macaca mulatta). Endocrinology
inhibit sexual behavior in male rats. Physiol. Behav. 59, 241–246. 139, 2774–2783.
Smith, E.R., Lee, R.L., Schnur, S.L., Davidson, J.M., 1987. Alpha 2-adrenoceptor Swaney, W.T., Curley, J.P., Champagne, F.A., Keverne, E.B., 2007. Genomic
antagonists and male sexual behavior: I. Mating behavior. Physiol. Behav. 41, imprinting mediates sexual experience-dependent olfactory learning in male mice.
7–14. Proc. Natl. Acad. Sci. U.S.A. 104, 6084–6089.
Smith, E.R., Maurice, J., Richardson, R., Walter, T., Davidson, J.M., 1990. Effect of Swaney, W.T., Dubose, B.N., Curley, J.P., Champagne, F.A., 2012. Sexual experience
four beta-adrenergic receptor antagonists on male rat sexual behavior. Pharmacol. affects reproductive behavior and preoptic androgen receptors in male mice. Horm.
Biochem. Behav. 36, 713–717. Behav. 61, 472–478.
Smith, E.R., Stefanick, M.L., Clark, J.T., Davidson, J.M., 1992. Hormones and sexual Swann, J., Rahaman, F., Bijak, T., Fiber, J., 2001. The main olfactory system
behavior in relationship to aging in male rats. Horm. Behav. 26, 110–135. mediates pheromone-induced fos expression in the extended amygdala and pre-
Snoeren, E.M., Ågmo, A., 2013. Female ultrasonic vocalizations have no incentive optic area of the male Syrian hamster. Neuroscience 105, 695–706.
value for male rats. Behav. Neurosci. 127, 439–450. Swann, J.M., 1997. Gonadal steroids regulate behavioral responses to pheromones
Snoeren, E.M., Ågmo, A., 2014. The incentive value of males’ 50-kHz ultrasonic by actions on a subdivision of the medial preoptic nucleus. Brain Res. 750,
vocalizations for female rats (Rattus norvegicus). J. Comp. Psychol. 128, 189–194.
40–55. Swanson, L.W., Petrovich, G.D., 1998. What is the amygdala? Trends Neurosci. 21,
Snoeren, E.M., Veening, J.G., Olivier, B., Oosting, R.S., 2014. Serotonin 1A receptors 323–331.
and sexual behavior in male rats: a review. Pharmacol. Biochem. Behav. 121, Swanson, L.W., Sawchenko, P.E., 1980. Paraventricular nucleus: a site for the inte-
102–114. gration of neuroendocrine and autonomic mechanisms. Neuroendocrinology 31,
Södersten, P., Damassa, D.A., Smith, E.R., 1977. Sexual behavior in developing male 410–417.
rats. Horm. Behav. 8, 320–341. Szczypka, M.S., Zhou, Q.Y., Palmiter, R.D., 1998. Dopamine-stimulated sexual
Somlyo, A.P., Somlyo, A.V., 2000. Signal transduction by G-proteins, rho-kinase and behavior in testosterone dependent mice. Behav. Neurosci. 112, 1229–1235.
protein phosphatase to smooth muscle and non-muscle myosin II. J. Physiol. Szechtman, H., Caggiula, A.R., Wulkan, D., 1978. Preoptic knife cuts and sexual
(Lond.) 522, 177–185. behavior in male rats. Brain Res. 150, 569–591.
Sopko, N.A., Hannan, J.L., Bivalacqua, T.J., 2014. Understanding and targeting the Szechtman, H., Hershkowitz, M., Simantov, R., 1981. Sexual behavior decreases pain
Rho kinase pathway in erectile dysfunction. Nat. Rev. Urol. 11, 622–628. sensitivity and stimulates endogenous opioids in male rats. Eur. J. Pharmacol. 70,
Stafford, S.A., Coote, J.H., 2006. Activation of D2-like receptors induces sympathetic 279–285.
climactic-like responses in male and female anaesthetized rats. Br. J. Pharmacol. Tagliamonte, A., Fratta, W., Gessa, G.L., 1974. Aphrodisiac effect of L-Dopa and
148, 510–516. apomorphine in male sexually sluggish rats. Experientia 30, 381–382.
Stafford, S.A., Bowery, N.G., Tang, K., Coote, J.H., 2006a. Activation by p- Tai, C., Booth, A.M., deGroat, W.C., Roppolo, J.R., 1998. Penile erection produced by
chloroamphetamine of the spinal ejaculatory pattern generator in anaesthetized microstimulation of the sacral spinal cord of the cat. IEEE Trans. Rehabil. Eng. 6,
male rats. Neuroscience 140, 1031–1040. 374–381.
Stafford, S.A., Tang, K., Coote, J.H., 2006b. Activation of lumbosacral 5-HT2C Takanami, K., Sakamoto, H., 2014. The gastrin-releasing peptide receptor (GRPR) in
receptors induces bursts of rhythmic activity in sympathetic nerves to the vas the spinal cord as a novel pharmacological target. Curr. Neuropharmacol. 12,
deferens in male rats. Br. J. Pharmacol. 148, 1083–1090. 434–443.
Staudt, M.D., de Oliveira, C.V., Lehman, M.N., McKenna, K.E., Coolen, L.M., 2011. Tallentire, D., McRae, G., Spedding, M., Clark, R., Vickery, B., 1996. Modulation of
Activation of NMDA receptors in lumbar spinothalamic cells is required for ejac- sexual behavior in the rat by a potent and selective alpha 2-adrenoceptor antag-
ulation. J. Sex. Med. 8, 1015–1026. onist, delequamine (RS-15385-197). Br. J. Pharmacol. 118, 63–72.
Staudt, M.D., Truitt, W.A., McKenna, K.E., de Oliveira, C.V., Lehman, M.N., Tanahashi, M., Karicheti, V., Thor, K.B., Marson, L., 2012. Characterization of bul-
Coolen, L.M., 2012. A pivotal role of lumbar spinothalamic cells in the regulation of bospongiosus muscle reflexes activated by urethral distension in male rats. Am. J.
ejaculation via intraspinal connections. J. Sex. Med. 9, 2256–2265. Physiol. Regul. Integr. Comp. Physiol. 303, R737–R747.
Steers, W.D., 2000. Neural pathways and central sites involved in penile erection: Tanaka, J., Arnold, A.P., 1993. An electrophysiological study of descending projections to
neuroanatomy and clinical implications. Neurosci. Biobehav. Rev. 24, 507–516. the lumbar spinal cord in adult male rats. Exp. Brain Res. 96, 117–124.
56 Male Sexual Behavior

Taziaux, M., Keller, M., Bakker, J., Balthazart, J., 2007. Sexual behavior activity Voigt, C., Ball, G.F., Balthazart, J., 2007. Neuroanatomical specificity of sex differ-
tracts rapid changes in brain estrogen concentrations. J. Neurosci. 27, ences in expression of aromatase mRNA in the quail brain. J. Chem. Neuroanat.
6563–6572. 33, 75–86.
Temple, J.L., Scordalakes, E.M., Bodo, C., Gustafsson, J.A., Rissman, E.F., 2003. Wada, T., Otsu, T., Hasegawa, Y., Mizuchi, A., Ono, H., 1996. Characterization of
Lack of functional estrogen receptor beta gene disrupts pubertal male sexual alpha 1-adrenoceptor subtypes in rat spinal cord. Eur. J. Pharmacol. 312,
behavior. Horm. Behav. 44, 427–434. 263–266.
Thor, K.B., Blitz-Siebert, A., Helke, C.J., 1990. Discrete localization of high-density 5- Wagner, C.K., Clemens, L.G., 1993. Neurophysin-containing pathway from the par-
HT1A binding sites in the midline raphe and parapyramidal region of the ventral aventricular nucleus of the hypothalamus to a sexually dimorphic motor nucleus in
medulla oblongata of the rat. Neurosci. Lett. 108, 249–254. lumbar spinal cord. J. Comp. Neurol. 336, 106–116.
Tobiansky, D.J., Hattori, T., Scott, J.M., Nutsch, V.L., Roma, P.G., Dominguez, J.M., Wagner, C.K., Sisk, C.L., Clemens, L.G., 1993. Neurons in the paraventricular nucleus
2012. Mating-relevant olfactory stimuli activate the rat brain in an age-dependent of the hypothalamus that project to the sexually dimorphic lower lumbar spinal cord
manner. Neuroreport 23, 1077–1083. concentrate 3H-estradiol in the male rat. J. Neuroendocrinol. 5, 545–551.
Toda, N., Ayajiki, K., Okamura, T., 2005. Nitric oxide and penile erection function. Waldherr, M., Neumann, I.D., 2007. Centrally released oxytocin mediates mating-
Pharmacol. Ther. 106, 233–266. induced anxiolysis in male rats. Proc. Natl. Acad. Sci. U.S.A. 104, 16681–16684.
Toner, J.P., Attas, A.I., Adler, N.T., 1987. Transcervical sperm transport in the rat: the Wallace, D.L., Vialou, V., Rios, L., et al., 2008. The influence of deltaFosB in the
roles of pre-ejaculatory behavior and copulatory plug fit. Physiol. Behav. 39, nucleus accumbens on natural reward-related behavior. J. Neurosci. 28, 10272–
371–375. 10277.
Toran-Allerand, C.D., 2004. Minireview: a plethora of estrogen receptors in the brain: Wang, H., Eto, M., Steers, W.D., Somlyo, A.P., Somlyo, A.V., 2002. RhoA-mediated
where will it end? Endocrinology 145, 1069–1074. Ca2þ sensitization in erectile function. J. Biol. Chem. 277, 30614–30621.
Traish, A., Guay, A.T., 2006. Are androgens critical for penile erections in humans? Wang, J., Swann, J.M., 2014. Connections of the magnocellular medial preoptic
Examining the clinical and preclinical evidence. J. Sex. Med. 3, 382–404. nucleus (MPN mag) in male Syrian hamsters. II. The efferents. Neuroscience 274,
Traish, A., Kim, N.N., Moreland, R.B., Goldstein, I., 2000. Role of alpha 102–118.
adrenergic receptors in erectile function. Int. J. Impot. Res. 12 (Suppl. 1), Wang, Z., Hulihan, T.J., Insel, T.R., 1997. Sexual and social experience is associated
S48–S63. with different patterns of behavior and neural activation in male prairie voles. Brain
Travison, T.G., Morley, J.E., Araujo, A.B., O’Donnell, A.B., McKinlay, J.B., 2006. The Res. 767, 321–332.
relationship between libido and testosterone levels in aging men. J. Clin. Endo- Warner, R.K., Thompson, J.T., Markowski, V.P., Loucks, J.A., Bazzett, T.J.,
crinol. Metab. 91, 2509–2513. Eaton, R.C., Hull, E.M., 1991. Microinjection of the dopamine antagonist cis-
Triana-Del Rio, R., Montero-Dominguez, F., Cibrian-Llanderal, T., et al., 2011. Same- flupenthixol into the MPOA impairs copulation, penile reflexes and sexual moti-
sex cohabitation under the effects of quinpirole induces a conditioned socio-sexual vation in male rats. Brain Res. 540, 177–182.
partner preference in males, but not in female rats. Pharmacol. Biochem. Behav. Watson, N.V., Gorzalka, B.B., 1991. DOI-induced inhibition of copulatory behavior in
99, 604–613. male rats: reversal by 5-HT2 antagonists. Pharmacol. Biochem. Behav. 39,
Triemstra, J.L., Nagatani, S., Wood, R.I., 2005. Chemosensory cues are essential for 605–612.
mating-induced dopamine release in MPOA of male Syrian hamsters. Neuro- Watson, N.V., Gorzalka, B.B., 1992. Concurrent wet dog shaking and inhibition of male
psychopharmacology 30, 1436–1442. rat copulation after ventromedial brainstem injection of the 5-HT2 agonist DOI.
Truitt, W.A., Coolen, L.M., 2002. Identification of a potential ejaculation generator in Neurosci. Lett. 141, 25–29.
the spinal cord. Science 297, 1566–1569. Wells, A.B., Coolen, L.M., 2001. Lumbar spinal cord efferents directly contact thalamic
Truitt, W.A., Shipley, M.T., Veening, J.G., Coolen, L.M., 2003. Activation of a subset of neurons that project to MPOA or BNST. Soc. Behav. Neuroendocrinol. Abstr. 5, 354.
lumbar spinothalamic neurons after copulatory behavior in male but not female Wersinger, S.R., Rissman, E.F., 2000. Dopamine activates masculine sexual behavior
rats. J. Neurosci. 23, 325–331. independent of the estrogen receptor alpha. J. Neurosci. 20, 4248–4254.
Tsai, H.W., Shui, H.A., Liu, H.S., Tai, M.Y., Tsai, Y.F., 2006. Monoamine levels in the Wersinger, S.R., Baum, M.J., Erskine, M.S., 1993. Mating-induced FOS-like
nucleus accumbens correlate with male sexual behavior in middle-aged rats. immunoreactivity in the rat forebrain, a sex comparison and a dimorphic effect
Pharmacol. Biochem. Behav. 83, 265–270. of pelvic nerve transection. J. Neuroendocrinol. 5, 557–568.
Tsai, H.W., Tsai, Y.F., Tai, M.Y., Yeh, K.Y., 2014. Regional and subtype-specific loss of Wersinger, S.R., Sannen, K., Villalba, C., Lubahn, D.B., Rissman, E.F., De Vries, G.J.,
GnRH neurons is associated with diminished mating behavior in middle-aged male 1997. Masculine sexual behavior is disrupted in male and female mice lacking
rats. Behav. Brain Res. 258, 112–118. a functional estrogen receptor alpha gene. Horm. Behav. 32, 176–183.
Tzschentke, T.M., 2000. The medial prefrontal cortex as a part of the brain reward Wesson, D.W., McGinnis, M.Y., 2006. Stacking anabolic androgenic steroids (AAS)
system. Amino Acids 19, 211–219. during puberty in rats: a neuroendocrine and behavioral assessment. Pharmacol.
Unda, N.M., Portillo, W., Corona, R., Paredes, R.G., 2016. Sexual stimulation increases Biochem. Behav. 83, 410–419.
the survival of new cells in the accessory olfactory bulb of the male rat. Front. Westberry, J., Meredith, M., 2003. The influence of chemosensory input and
Neurosci. 10, 65. gonadotropin releasing hormone on mating behavior circuits in male hamsters.
Vagell, M.E., McGinnis, M.Y., 1997. The role of aromatization in the restoration of male Brain Res. 974, 1–16.
rat reproductive behavior. J. Neuroendocrinol. 9, 415–421. Wiesenfeld-Hallin, J., Södersten, P., 1984. Spinal opiates affect sexual behavior in
Vagell, M.E., McGinnis, M.Y., 1998. The role of gonadal steroid receptor activation in rats. Nature 309, 257–258.
the restoration of sociosexual behavior in adult male rats. Horm. Behav. 33, Wilbert, D.M., Griffin, J.E., Wilson, J.D., 1983. Characterization of the cytosol androgen
163–179. receptor of the human prostate. J. Clin. Endocrinol. Metab. 56, 113–120.
Van Ree, J.M., Niesink, R.J., Van Wolfswinkel, L., Ramsey, N.F., Kornet, M.M., Van Wilson, R.I., Nicoll, R.A., 2002. Endocannabinoid signaling in the brain. Science 296,
Furth, W.R., Vanderschuren, L.J., Gerrits, M.A., Van den Berg, C.L., 2000. 678–682.
Endogenous opioids and reward. Eur. J. Pharmacol. 405, 89–101. Wingard, C.J., Husain, S., Williams, J., James, S., 2003. RhoA-Rho kinase mediates
Vanderhorst, V.G., Terasawa, E., Ralston 3rd, H.J., Holstege, G., 2000. Monosynaptic synergistic ET-1 and phenylephrine contraction of ratcorpus cavernosum. Am. J.
projections from the lateral periaqueductal gray to the nucleus retroambiguus in the Physiol. Regul. Integr. Comp. Physiol. 285, R1145–R1152.
rhesus monkey: implications for vocalization and reproductive behavior. J. Comp. Wise, R.A., Rompre, P.P., 1989. Brain dopamine and reward. Annu. Rev. Psychol. 40,
Neurol. 424, 251–268. 191–225.
Veening, J.A., Coolen, L.M., 1998. Neural activation following sexual behavior in the Witt, D.M., Insel, T.R., 1994. Increased Fos expression in oxytocin neurons following
male and female rat brain. Behav. Brain Res. 92, 181–193. masculine sexual behavior. J. Neuroendocrinol. 6, 13–18.
Veening, J.G., Coolen, L.M., 2014. Neural mechanisms of sexual behavior in the male Witt, D.M., Young, L.J., Crews, D., 1994. Progesterone and sexual behavior in males.
rat: emphasis on ejaculation-related circuits. Pharmacol. Biochem. Behav. 121, Psychoneuroendocrinology 19, 553–562.
170–183. Witt, D.M., Young, L.J., Crews, D., 1995. Progesterone modulation of androgen-
Verma, S., Chhina, G.S., Mohan Kumar, V., Singh, B., 1989. Inhibition of male sexual dependent sexual behavior in male rats. Physiol. Behav. 57, 307–313.
behavior by serotonin application in the medial preoptic area. Physiol. Behav. 46, Wood, R.I., 1996. Estradiol, but not dihydrotestosterone, in the medial amygdala
327–330. facilitates male hamster sex behavior. Physiol. Behav. 59, 833–841.
Viitamaa, T., Haapalinna, A., Agmo, A., 2006. The adrenergic a2 receptor and Wood, R.I., Coolen, L.M., 1997. Integration of chemosensory and hormonal cues is
sexual incentive motivation in male rats. Pharmacol. Biochem. Behav. 83, essential for sexual behaviour in the male Syrian hamster: role of the medial
360–369. amygdaloid nucleus. Neuroscience 78, 1027–1035.
Male Sexual Behavior 57

Wood, R.I., Newman, S.W., 1993. Mating activates androgen receptor-containing Yang, J., Lin, S., Feng, Y., Wu, G., Hu, J., 2013. Taurine enhances the sexual
neurons in the chemosensory pathways of the male Syrian hamster brain. Brain response and mating ability in aged male rats. Adv. Exp. Med. Biol. 776,
Res. 614, 65–77. 347–355.
Wood, R.I., Newman, S.W., 1995a. Androgen and estrogen receptors coexist within Yeh, K.Y., Pu, H.F., Kaphle, K., Lin, S.F., Wu, L.S., Lin, J.H., Tsai, Y.F., 2008. Ginkgo
individual neurons in the brain of the Syrian hamster. Neuroendocrinology 62, biloba extract enhances male copulatory behavior and reduces serum prolactin
487–497. levels in rats. Horm. Behav. 53, 225–231.
Wood, R.I., Newman, S.W., 1995b. Hormonal influence on neurons of the mating Yells, D.P., Hendricks, S.E., Prendergast, M.A., 1992. Lesions of the nucleus para-
behavior pathway in male hamsters. In: Micevych, P.E., Hammer Jr., R.P. (Eds.), gigantocellularis: effects on mating behavior in male rats. Brain Res. 596, 73–79.
Neurobiological Effects of Sex Steroid Hormones. Cambridge University Press, New Yells, D.P., Prendergast, M.A., Hendricks, S.E., Nakamura, M., 1994. Fluoxetine-
York, pp. 3–39 (Chapter 1). induced inhibition of male rat copulatory behavior: modification by lesions of the
Wood, R.I., Newman, S.W., 1995c. Integration of chemosensory and hormonal nucleus paragigantocellularis. Pharmacol. Biochem. Behav. 49, 121–127.
cues is essential for mating in the male Syrian hamster. J. Neurosci. 15, Yetik-Anacak, G., Sorrentino, R., Linder, A.E., Murat, N., 2015. Gas what: NO is not
7261–7269. the only answer to sexual function. Br. J. Pharmacol. 172, 1434–1454.
Wood, R.I., Williams, S.J., 2001. Steroidal control of male hamster sexual behavior: Yokosuka, M., Hayashi, S., 1996. Co-localization of neuronal nitric oxide synthase and
redundancy without hierarchy. Physiol. Behav. 72, 727–733. androgen receptor immunoreactivity in the premammillary nucleus in rats. Neu-
Wood, R.I., Brabec, R.K., Swain, J.M., Newman, S.W., 1992. Androgen and estrogen rosci. Res. 26, 309–314.
receptor concentrating neurons in chemosensory pathways of the male Syrian Yokosuka, M., Matsuoka, M., Ohtani-Kaneko, R., Iigo, M., Hara, M., Hirata, K.,
hamster brain. Brain Res. 596, 89–98. Ichikawa, M., 1999. Female-soiled bedding induced fos immunoreactivity in the
Woodley, S.K., Baum, M.J., 2004. Differential activation of glomeruli in the ferret’s ventral part of the premammillary nucleus (PMv) of the male mouse. Physiol.
main olfactory bulb by anal scent gland odours from males and females: an early Behav. 68, 257–261.
step in mate identification. Eur. J. Neurosci. 20, 1025–1032. Yokosuka, M., Prins, G.S., Hayashi, S., 1997. Co-localization of androgen receptor and
Woodson, J.C., 2002. Including ‘learned sexuality’ in the organization of sexual nitric oxide synthase in the ventral premammillary nucleus of the newborn rat: an
behavior. Neurosci. Biobehav. Rev. 261, 69–80. immunohistochemical study. Brain Res. Dev. Brain Res. 99, 226–233.
Woolley, S.C., Sakata, J.T., Crews, D., 2004. Evolutionary insights into the regulation Yonezawa, A., Kawamura, S., Ando, R., Tadano, T., Nobunaga, T., Kimura, Y., 1991.
of courtship behavior in male amphibians and reptiles. Physiol. Behav. 83, Biphasic effects of yohimbine on the ejaculatory response in the dog. Life Sci. 48,
347–360. 103–109.
Woolley, S.C., Sakata, J.T., Gupta, A., Crews, D., 2001. Evolutionary changes in Yonezawa, A., Yoshizumi, M., Ebiko, M., et al., 2008. Ejaculatory response induced by
dopaminergic modulation of courtship behaviors in Cnemidophorus whiptail lizards. a 5-HT2 receptor agonist m-CPP in rats: differential roles of 5-HT2 receptor
Horm. Behav. 40, 483–489. subtypes. Pharmacol. Biochem. Behav. 88, 367–373.
Wu, D., Gore, A.C., 2010. Changes in androgen receptor, estrogen receptor alpha, and Zahran, A.R., Vachon, P., Courtois, F., Carrier, S., 2000. Increases in intracavernous
sexual behavior with aging and testosterone in male rats. Horm. Behav. 58, penile pressure following injections of excitatory amino acid receptor agonists in the
306–316. hypothalamic paraventricular nucleus of anesthetized rats. J. Urol. 164,
Xiao, L., Becker, J.B., 1998. Effects of estrogen agonists on amphetamine-stimulated 1793–1797.
striatal dopamine release. Synapse 29, 379–391. Zarrindast, M.R., Mamanpush, S.M., Rashidy-Pour, A., 1994. Morphine inhibits
Yahr, P., 1995. Neural circuitry for the hormonal control of male sexual behavior. dopaminergic and cholinergic induced ejaculation in rats. Gen. Pharmacol. 25,
In: Micevych, P.E., Hammer Jr., R.P. (Eds.), Neurobiological Effects of Sex 803–808.
Steroid Hormones. Cambridge University Press, New York, pp. 40–56 Zarrindast, M.R., Shokravi, S., Samini, M., 1992. Opposite influences of dopaminergic
(Chapter 2). receptor subtypes on penile erection. Gen. Pharmacol. 23, 671–675.
Yahr, P., Gregory, J.E., 1993. The medial and lateral cell groups of the sexually Zehr, J.L., Todd, B.J., Schulz, K.M., McCarthy, M.M., Sisk, C.L., 2006. Dendritic
dimorphic area of the gerbil hypothalamus are essential for male sex behavior and pruning of the medial amygdala during pubertal development of the male Syrian
act via separate pathways. Brain Res. 631, 287–296. hamster. J. Neurobiol. 66, 578–590.
Yahr, P., Ulibarri, C., 1987. Polyadenylated and nonadenylated messenger RNA and Zhang, X.R., Zhang, Z.J., Jenkins, T.A., Cheng, W.R., Reynolds, G.P., 2011. The
androgen control of sexual behavior and scent marking in male gerbils. Horm. dose-dependent effect of chronic administration of haloperidol, risperidone, and
Behav. 21, 53–64. quetiapine on sexual behavior in the male rat. J. Sex. Med. 8, 3345–3353.
Yamada, K., Emson, P., Hökfelt, T., 1996. Immunohistochemical mapping of nitric Zucker, I., Wade, G., 1968. Sexual preferences of male rats. J. Comp. Physiol.
oxide synthase in the rat hypothalamus and colocalization with neuropeptides. Psychol. 66, 816–819.
J. Chem. Neuroanat. 10, 295–316. Zumpe, D., Clancy, A.N., Bonsall, R.W., Michael, R.P., 1996. Behavioral responses to
Yanagimoto, M., Honda, K., Goto, Y., Negoro, H., 1996. Afferents originating from Depo-Provera, Fadrozole, and estradiol in castrated, testosterone-treated cyn-
the dorsal penile nerve excite oxytocin cells in the hypothalamic paraventricular omolgus monkeys (Macaca fascicularis): the involvement of progestin receptors.
nucleus of the rat. Brain Res. 733, 292–296. Physiol. Behav. 60, 531–540.
This page intentionally left blank
1.02 Female Sexual Behavior in Rodents, Lagomorphs, and Goats
Oscar González-Flores and Kurt L Hoffman, Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-
CINVESTAV, Tlaxcala, México
José A Delgadillo, Centro de Investigación en Reproducción Caprina (CIRCA), Universidad Autónoma Agraria Antonio Narro,
Torreón, México
Matthieu Keller, Behavioral & Reproductive Physiology, INRA/CNRS/University of Tours, Nouzilly, France
Raúl G Paredes, Instituto de Neurobiologia, Universidad Nacional Autónoma de México, Querétaro, México
Ó 2017 Elsevier Inc. All rights reserved.

1.02.1 Introduction 59
1.02.1.2 Estrous Behavior in Rodents, Lagomorphs, and Ruminants 60
1.02.1.2.1 Behavioral Aspects of Estrous Behavior 60
1.02.1.2.2 Effect of Steroid Hormones in Rodents 62
1.02.1.2.3 Effect of Steroid Hormones in Lagomorphs (Rabbits) 64
1.02.1.2.4 Seasonality of Reproduction in Goats 65
1.02.1.2.5 Endocrine Control of Sexual Behavior in Goats 66
1.02.1.2.6 Nonsteroidal Agents on Estrous Behavior 66
1.02.1.3 Cellular Processes Related to the Action of Steroid Hormones on Estrous Behavior 67
1.02.1.3.1 ER and PR Expression in the Brain 67
1.02.1.3.2 Cellular Mechanisms Proposed to Explain the Regulation of Estrous Behavior by Different Agents 68
1.02.1.3.3 Protein Kinase A 68
1.02.1.3.4 Leptin and Protein Kinase 69
1.02.1.3.5 Src Tyrosine Kinase, MAPK Pathway, and Lordosis Behavior 69
1.02.1.4 Neuroendocrine Factors Participating in the Behavioral Transition across the Reproductive Cycle of Rats,
Rabbits, and Goats 71
1.02.1.4.1 Anestrous to Estrous in Rodents 71
1.02.1.4.2 Anestrous to Estrous in Rabbits 71
1.02.1.4.3 Postpartum and Lactation in Rats 73
1.02.1.4.4 Postpartum and Lactation in Rabbits 73
1.02.1.4.5 Postpartum Anestrous and Lactation in Goats 73
1.02.1.5 Female Sexual Motivation in Rats and Goats 73
1.02.1.5.1 Paced Mating and Female Sexual Motivation 74
1.02.1.5.2 The Importance of Testing Conditions in the Analysis of Sexual Motivation 74
1.02.1.5.3 The Male Effect 75
1.02.1.5.4 Neurogenesis and Sexual Behavior 76
1.02.1.6 Conclusions 76
Acknowledgments 77
References 77

1.02.1 Introduction However, the estrous cycle is much shorter in duration, lasting
only 4–5 days, as is the case for rats and hamsters. In both of
In general, female mammals show periods of sexual activity these cycles, ovulation occurs in response to a hormonal
(estrous) alternating with periods of sexual refractoriness (dies- change: follicular estrogen secretion, waxing, and waning at
trous). Depending on the effect of coitus on ovulation and on regular intervals. In contrast, in the third type of estrous cycle,
the presence of functional corpora lutea, three types of estrous ovulation occurs following coitus: it is reflexive or induced.
cycles have been distinguished (Beyer et al., 1976; Morali and This type of cycle is displayed by female rabbits, voles, mink,
Beyer, 1979). In the first type, ovulation occurs spontaneously ferrets, cats, camels, to name a few. Mating is followed by
establishing functional corpora lutea with sexual receptivity ovulation with cessation of estrous and formation of active
taking place around ovulation. The estrous cycle is long, with corpora lutea. In contrast to spontaneously ovulating species,
a variable duration among species; for example, 16 days in induced ovulators generally do not show steroid-induced
guinea pigs and 28 days in primates. In the second type of ovulation. For example, in rabbits (Sawyer and Markee,
estrous cycle, ovulation is spontaneous but functional corpora 1959), voles (Milligan, 1978, 1980), and ferrets (Baum,
lutea are not formed unless the animal copulates and becomes 1990), estradiol (E2) treatment, with or without concurrent
pseudopregnant or pregnant. As in the case of the first type of progesterone (P), failed to stimulate ovulation. The cat may
cycle, maximal receptivity is displayed around ovulation. be one exception to this rule, as spontaneous ovulation has

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00002-X 59


60 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

been occasionally observed in group-housed female cats ovulation and the initiation of a functional luteal phase that
(Gudermuth et al., 1997; Lawler et al., 1993). However, as is essential in order to maintain pregnancy (Conaway, 1971).
for any biological phenomenon, the distinction between In this species, copulation is followed by a decrease in female
induced and spontaneous ovulation is sometimes unclear. estrous behaviors, including sexual receptivity and scent-
There is evidence, for example, that spontaneously ovulating marking (González-Mariscal et al., 1997, 1990; Hoffman and
species can also show reflexive ovulation in certain circum- González-Mariscal, 2007).
stances. Under laboratory conditions, during mating, female Although sexual behavior is the result of an ongoing inter-
rats must receive several mounts and intromissions from a sexu- action between the male and the female, each sex displays
ally active male in order for the ovaries to develop fully func- specific, quantifiable behaviors that reflect motivational or
tional corpora lutea. Thus, mating stimulates the release of consummatory aspects of sexual behavior. Among female
prolactin from the anterior hypophysis, which is necessary for mammals, patterns of sexual behavior are generally stereotyped
luteal activity (Terkel, 1988). In this condition, the female within a species, but can be quite variable between species. In
spontaneously exhibits persistent vaginal estrous (Everett, the present chapter we first describe salient behavioral and
1939). hormonal aspects of female sexual behavior in rodents, lago-
In most mammals, female sexual behavior is stereotyped morphs, and ruminants. This section is followed by an over-
and predictable over time, and occurs in organized patterns view of cellular processes underlying the action of sex
that can be very different between species. However, in general, hormones on estrous behavior, as well as neuroendocrine
a complete sequence of sexual behavior generally includes pre- factors participating in behavioral transitions across the repro-
copulatory behavior, copulation, termination, and conse- ductive cycle of rats, rabbits and goats. Finally, the last section
quences of copulation. discusses studies of female sexual motivation in rats and goats,
Precopulatory behaviors: These are species-specific behaviors along with a brief overview of long-lasting changes in neuro-
displayed by males or females in order to attract the opposite genesis induced by sexual behavior.
sex. Thus, the genital contact that characterizes copulation
usually is preceded by sequences of precopulatory or courtship
1.02.1.2 Estrous Behavior in Rodents, Lagomorphs,
behavior, the complexity and duration of which varies from
and Ruminants
species to species. These behaviors are often associated with
morphological and physiological signals such as swelling of 1.02.1.2.1 Behavioral Aspects of Estrous Behavior
the genital area and the production of odoriferous substances 1.02.1.2.1.1 Rodents
in the case of the female and anogenital investigation, scent During estrous, female rodents display a series of stereotyped
marking, and urine investigation by the male. Thus, courtship behaviors in order to attract a male and copulate. Classical
brings together a male and a female of the same species at studies by Beach in 1976 distinguished three typical components
the time when they are likely to mate, enhancing the proba- of female sexual behavior: attractivity, proceptivity, and recep-
bility of successful reproduction. tivity. Attractivity was defined as the female’s “value as a sexual
Copulation: Completion of courtship activity results in a tran- stimulus to evoke a sexual response in the male, which is deter-
sition to copulatory behavior, by which fertilization of the ova mined by a number of factors, including olfactory and visual
is accomplished internally. Behavioral patterns that comprise cues.” Proceptivity was defined as a reaction by the female
copulation are often elaborate and highly stereotyped. Typi- toward the male which constitutes her assumption of initiative
cally, mammals engage in repeated copulations that can be of in establishing or maintaining sexual interaction. Receptivity
long duration, although there are some species in which was defined as “the female responses necessary and sufficient
mating entails a single, brief genital contact. for the male’s success in achieving intravaginal ejaculation”
Termination: When hormonal levels are optimal, copulatory (Beach, 1976). Over the years, these three components of female
activity may persist for a considerable period of time, either sexual behavior have been further characterized and analyzed in
through a single copulation of a long duration, or many brief detail by many different research groups, resulting in a more
series of copulations, the latter characterizing the copulatory detailed account of estrous behavior and female sexual motiva-
behavior of laboratory rats. Thus, in female mammals, the tion (Agmo, 1999). Sexual attractivity of the female rat is neces-
duration of estrous is largely determined by adequate sarily inferred from certain appetitive behaviors that the male rat
hormonal levels, and copulatory activity often wanes when displays in response to signals emitted by the female. These
circulating levels of these hormones decline. In some cases, behaviors include approaching and investigating the female in
even when apparently sexually satiated, a change of mating response to olfactory and visual cues. Experimental procedures
partners often can lead to a reinstatement of copulatory that allow for the investigation of sexual incentive motivation
behavior, the so-called ‘Coolidge effect’ (Tlachi-López et al., (appetitive behavior) independently of copulation (consumma-
2012; Ventura-Aquino et al., 2016). tory behavior) have shown that the female rat is an uncondi-
Consequences of copulation: Pregnancy is the major conse- tioned sexual stimulus for the male. In other words, cues
quence of copulatory activity. In the female, successful fertiliza- emitted by the receptive female rat evoke and maintain approach
tion of the ova and pregnancy depend on a multitude of behavior by the male, and the possibility of copulating with her
endocrinological and physiological processes, including ovula- has no lasting effect on her incentive value (Agmo, 2003).
tion and formation of the corpora lutea, the vaginal deposition Proceptive behavior displayed by female rats has been
of sperm and their transference to the site of fertilization, and described as having different components including approach,
implantation of the fertilized ova in the uterus. For example orientation, and runaway (Beach, 1976; McClintock and Adler,
in rabbits, the appropriate copulatory stimulation induces 1978). During the approach, which is often initiated from
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 61

a distance of at least four body lengths away from the male, the experimentally inducing receptivity in this manner, a second
female brings herself into proximity with the male. The orienta- series of estrogen and P injections was ineffective at stimulating
tion component involves the female positioning herself so that estrous behavior; this phenomenon was termed ‘sequential
her head is near the male, sometimes nudging or grooming him, inhibition’ (Zucker, 1966).
crawling over or dragging herself under his chin, or positioning
herself so that her genital region is near his nose. The female 1.02.1.2.1.2 Lagomorphs (Rabbits)
then suddenly turns and departs from the male while displaying Female sexual behavior in the European rabbit (Oryctolagus
a hopping or darting (zigzagging) behavior – the runaway cuniculus) appears long before the ability to ovulate. However,
component. This often provokes the male to pursue her and such behavior is regarded as prepubertal play, rather than as
attempt a mount or intromission. If the male does not pursue a sign of puberty. In contrast to the rat, the sexually mature
her, the female reapproaches the male and then again with- female rabbit does not display an estrous cycle with regular
draws. The female also can display ‘ear wiggling,’ which is the periods of heat, and ovulation occurs only after mating. In
result of rapid head shaking (Beach, 1976; McClintock and captivity (in the laboratory or on the farm), under controlled
Adler, 1978). In some rodents, proceptive scent-marking behav- long-daylight conditions, unmated does are considered to be
iors are displayed; these behaviors comprise rubbing the surface in a more or less constant state of estrous. However, observa-
of objects in the environment with specialized scent glands or tions of rabbits under natural or seminatural conditions have
by depositing urine (Reynolds, 1971). Another term that has suggested that does may display subtle cycles of waxing and
been introduced is ‘paracopulatory’ behavior, which includes waning receptivity with a cycle length of approximately
proceptive behaviors mentioned above, along with ultrasonic 7 days (Myers and Poole, 1962).
vocalizations. These paracopulatory behaviors are species- Proceptive behaviors in the female rabbit have been much
typical behaviors that are displayed by females, which arouse less studied compared to those of the rat and, as a result,
males and stimulate them to mount (Blaustein, 2009). have not been as thoroughly described and characterized. In
Receptivity in the female is characterized by the display of the wild, on the farm, or under laboratory conditions, procep-
a posture that facilitates penile insertion and ejaculation. In tive female rabbits are observed to display circling of the male
rodents, as in many other mammalian species, this is accom- and ‘flagging’ of the tail (elevation and lateral displacements of
plished by the lordosis reflex, which comprises arching of the the tail). However, specific proceptive behaviors have proven
back, elevation of the pelvis, and deviation of the tail difficult to characterize in the rabbit (Stoufflet and Caillol,
(Komisaruk and Diakow, 1973; Kuehn and Beach, 1963). In 1988). González-Mariscal et al. (1990) proposed that a scent-
this posture, the legs are extended from the initial crouching marking behavior called ‘chinning’ (where the rabbit rubs the
position while the head is elevated above its usual resting posi- undersurface of its chin over objects in its environment, thereby
tion. Usually, lordosis is triggered by the male’s mount, depositing scent gland secretions) might function as a procep-
although tactile stimulation of the perineal region alone can tive behavior, since its heightened expression closely corre-
elicit this reflex in many rodent species such as rats, guinea sponds to periods of sexual receptivity and rapidly declines
pigs, hamsters, etc. (Komisaruk, 1972). The latency to reinitiate following copulation (González-Mariscal et al., 1990, 1997;
proceptive behavior after having received sexual stimulation is Soares and Diamond, 1982).
determined in large part by the intensity and characteristics of Chinning is a frequently observed behavior, being
that stimulation. If the stimulus comprised a mount, intromis- expressed by both females and males within a variety of envi-
sion, and ejaculation, proceptive behavior will be delayed for ronmental, social, and reproductive contexts. The waxy secre-
a longer time than if a mount and intromission were received tion of the submandibular gland is deposited by chinning
without an ejaculation. Likewise, the latency to reinitiate pro- on vertical or irregular surfaces, such as trees, grass, branches,
ceptive behavior is longer when both a mount and intromis- burrow entrances, cages, feeders, waterers, and mates. When
sion are received, compared to a mount alone (Beach, 1976). a rabbit is placed in a new cage, for example, it will explore
Receptivity in the female rat decreases progressively as and chin-mark this unfamiliar environment (Lebas et al.,
a result of repeated stimulation from the male (Hardy and 1986); similarly, when unfamiliar objects are encountered
DeBold, 1972); eventually, the female shows a refractory within a familiar environment, both female and male rabbits
period during which she will no longer mate with the male. preferentially chin-mark these objects, compared to familiar
This inhibition depends on the activity of the corpora ones simultaneously encountered within the same environ-
lutea and was first successfully mimicked in ovariectomized ment (Hoffman and Basurto, 2013, 2014; Hoffman et al.,
(OVX) guinea pigs. Thus, in OVX animals that had received P 2010). In males, chinning apparently serves to delineate
during estrogen priming, the later administration of P was inef- territory, to mark conspecifics within the social group, and
fective at promoting estrous behavior a phenomenon called to promote territorial confidence (Mykytowy, 1965;
‘concurrent inhibition’ (Goy et al., 1968; Zucker, 1966). In Mykytowycz, 1962; Mykytowycz et al., 1976). Chinning has
the guinea pig a ‘biphasic’ effect of P on sexual behavior was been positively associated with social dominance and, in the
also demonstrated. It was observed that, following sponta- male, elevated levels of circulating testosterone (T) (Arteaga
neous receptivity in cycling female guinea pigs, there was et al., 2008).
a refractory period during which exogenous hormone adminis- In the female rabbit, the expression of chinning is closely
tration failed to induce receptivity (Goy et al., 1968; Zucker, tied to reproductive state. Across prepubertal development,
1968). This refractoriness to hormone stimulation was repli- chinning frequency steadily increases until sexual maturity is
cated in OVX guinea pigs that were made receptive by exoge- reached (González-Mariscal et al., 1992). Sexually mature
nous estrogen priming followed 36 h later by P. After does display increased chinning behavior during estrous,
62 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

compared to during pregnancy (González-Mariscal et al., 1990; This behavior attracts males and stimulates sexual activity in
Soares and Diamond, 1982), as this behavior is stimulated by male goats (Shearer and Katz, 2006).
circulating E2 and inhibited by P (Hudson et al., 1990). Stimuli Once in contact, the receptive behavior of the goat when in
associated with mating (e.g., the male’s mounts and intromis- estrous consists of an immobilization. However, this immobi-
sions) engage physiological mechanisms that inhibit chinning lization is not a passive posture but is rather an active response.
across immediate (within minutes after mating), early (across Indeed, the female will actively resist any attempt of movement
the first days postmating), and long-term (from day 4 post- by an external manipulator. This immobilization is rather
mating through the rest of pregnancy) time frames. These subtle and is only observed as a slight arching of the back
mechanisms will be described in more detail in a later section. during immobilization (Fabre-Nys and Gelez, 2007). This
The estrous female rabbit displays a lordosis posture in immobilization is a sign both of proceptivity and receptivity.
response to the male mount, with the back and the hindquar- In the buck, ejaculation is rather rapid and there is no need
ters raised (Lebas et al., 1986). This posture allows intromis- for several intromissions: ejaculation generally occurs after
sion and the immediate ejaculation by the male (unlike rats, the first intromission and lasts a few seconds (Alexander
the male rabbit ejaculates immediately after each intromission et al., 1980). Therefore, the amount of vaginal stimulation
(Beyer et al., 1975; Beyer and Mcdonald, 1973). The lordosis received by females is rather limited in these species, contrary
reflex is stimulated by a characteristic male mounting pattern, to what can be observed in rodents (Rodríguez-Sierra et al.,
in which the male mounts the female from behind, clasping 1977).
her flank region with his forepaws while displaying vigorous
pelvic thrusting directed at the perineal region (Contreras and 1.02.1.2.2 Effect of Steroid Hormones in Rodents
Beyer, 1979; Morali and Beyer, 1992; Morali et al., 2003). 1.02.1.2.2.1 Estrogens
Like chinning, the lordosis reflex is inhibited after ovulation, In females of all subprimate species that have been studied,
whether induced naturally by mating, or experimentally by estrogens are essential for the expression of lordosis behavior
the administration of gonadotropin (Hoffman et al., 2010), (for a review, see Young, 1961). While some species require
and remains repressed throughout gestation (Beyer and only estrogen for the display of sexual behavior, others need
Rivaud, 1969). Similarly, sexual receptivity in female rabbits the sequential action of estrogen and P. Rodent species such
is reduced during lactation. Studies have shown that lactating as rat and guinea pig, which require both estrogen and P,
does consistently refuse the buck after the 12th day of lactation have short periods of estrous or heat and are sexually nonrecep-
(Hammond and Marshall, 1925) or show sexual behavior only tive throughout the major part of the cycle.
occasionally (Yaschine et al., 1967). In one study, only 50% of The estrous cycle of the rat is 4–5 days, comprising the
does sporadically displayed sexual behavior between the third following stages (identified by cellular and histological changes
day of lactation and weaning in both New Zealand white and of the vaginal walls): diestrous I (or metestrus), diestrous II,
California rabbits (Beyer and Rivaud, 1969). Like chinning, proestrous, and estrous. Proestrous is associated with a peak
inhibition of sexual receptivity during lactation was found to in circulating E2, occurring approximately at midday, followed
be related to litter size: suckling a litter of 10 pups resulted in by a sharp peak in P across the afternoon and evening, which
significantly reduced sexual receptivity on lactation day 15, coincides with the release of luteinizing hormone (LH) from
compared to mothers that suckled litters of only one or three the pituitary. Levels of all three hormones decline to low levels
pups (García-Dalman and González-Mariscal, 2012). Thus, by the morning of estrous. Female rat sexual behavior – both
the ‘estrous cycle’ of the doe consists only of alternating periods proceptive and receptive components – is triggered by overlap-
of sexual receptivity and nonreceptivity associated with preg- ping peaks of circulating E2 and P that occur across the after-
nancy, pseudopregnancy, and lactation. noon and evening of proestrous. Likewise, in OVX female
rats, the administration of estradiol benzoate (EB; a synthetic
1.02.1.2.1.3 Goats estrogen with an extended biological half-life), followed 48 h
In natural or seminatural conditions males and females of later by the administration of P, promotes the full expression
ruminant species live most of the year in segregated unisex of female sexual behavior. E2 is conceptualized to prime the
flocks (Alexander et al., 1980). This sexual segregation between neural circuitry that underlies sexual behavior, for its later
males and females disappears around the time of reproduction responsiveness to P. Thus, the full expression of sexual
where males leave their group to look for females. As in other behavior – proceptive as well as receptive components – in
species, estrous behavior is composed of proceptivity and the female rodent has generally been considered to require P
receptivity. Proceptivity consists of seeking out and stimulating in addition to a priming dose of EB (Glaser et al., 1983).
the male partner while receptivity consists in the active immo- In physiological and behavioral studies of estrous behavior
bilization in response to male nudges. At the beginning of that involve exogenous hormone administration, esterified
estrous, proceptivity always precedes receptivity, but later on forms of 17b-E2 are often used, because the circulating levels
these behaviors are expressed simultaneously. The expression of 17b-E2 do not remain elevated for a long time after systemic
of proceptivity and receptivity in ruminant species is quite administration. A variety of esters and other modifications have
different from that of rodents. Although various sensory cues been used in this regard, such as ethynylestradiol (Patisaul
play a role in the attraction of males, the most important cue et al., 2003), E2 valerate (García-Segura et al., 1986), or
for attracting males is the females’ proceptive behavior. An E2 dipropionate (Hlinak, 1993). However, the most
increase in motor activity is observed in most female ruminants common form used is EB, which is 17b-E2 with a benzoic
when estrous starts. Goats express vocalizations and male-type acid esterified in the third carbon position. This form is hydro-
courtship behavior and female–female reciprocal mounting. lyzed in vivo to the physiologically active 17b-E2, and it is more
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 63

potent than free steroids for inducing estrous behavior. Thus, in hormones (such as progestins) because adrenalectomy did
OVX rats, EB is 7 times more potent than E2 for inducing not block the sensitization of sexual behavior. Moreover, EB-
estrous behavior (Powers, 1975). Similarly, exogenously induced sensitization occurred independently of whether or
administered catecholestrogens, which are natural estrogen not they had interacted with a sexually vigorous male during
metabolites, vary in their ability to induce estrous behavior in the course of hormone treatment, indicating that hormonal
OVX female rats (Luttge and Jasper, 1977; Naish and Ball, treatment alone was sufficient to induce behavioral
1981). This variability depends on the route of administration sensitization (Jones and Pfaus, 2014).
or the presence of other estrogens. For example, when
2-hydroxyestradiol and E2 were implanted together in the
hypothalamus, the level of receptivity was higher than when 1.02.1.2.2.2 Progestins
E2 was administered alone (Marrone et al., 1977). Similarly, P, either alone or in combination with other hormones, modu-
Luttge and Jasper (1977) found that intracerebral implants of lates several reproductive processes such as sexual behavior,
2-hydroxyestradiol are ineffective in stimulating lordosis ovulation, gonadotropin secretion, pregnancy, maternal
behavior in female rats but, when combined with intravenous behavior, among others. In intact rodents, estrous behavior is
administration of E2, such implants induced receptivity. normally induced by the combined action of estrogen and P.
Although estrous behavior in the intact female rat is nor- During the 48 h period preceding the onset of sexual receptivity
mally triggered by the combined action of E2 and P, in some in the rat, estrogen levels gradually rise and reach a peak late in
experimental conditions, the administration of E2 or EB can the afternoon of proestrous. At about this time, P levels begin
substitute for P in the induction of female sexual receptivity, to rise rapidly, reaching a peak during the early evening of pro-
in OVX or OVX-adrenalectomized rats (the adrenal gland is estrous. The onset of sexual receptivity is temporally associated
another significant source of P) (Beach, 1942). Thus, a single with this preovulatory peak in circulating P (Freeman, 1994;
high-dose injection of EB can induce sexual behavior, as can Ichikawa et al., 1974). Early experiments carried out in rats
the combination of a single low dose of EB followed by and hamsters demonstrated that lordosis was abolished if the
a second high dose of EB or E2. Moreover, OVX or OVX- ovaries were removed shortly before the preovulatory peak of
adrenalectomized rats displayed sexual behavior after having P, whereas lordosis was consistently displayed by animals
received low doses of EB across several successive days (Beyer that had been ovariectomized after the preovulatory peak in
and Komisaruk, 1971; Blaustein et al., 1987; Kow and Pfaff, P had already occurred (Moreines and Powers, 1977; Powers
1975). Classic early studies demonstrated that various and Valenstein, 1972). Other studies showed that P greatly
estrogenic compounds showed different potencies with facilitated the effect of estrogen on estrous behavior in OVX
respect to their ability to induce estrous (lordosis), after females, but only if the female had been previously primed
subchronic treatment (daily injections, across 10 days) in with estrogen for a minimal period of time, independently of
OVX rats. Thus, E2 was the most potent estrogen in this the amount of estrogen given (Boling and Blandau, 1939;
regard, followed by estrone (E1), estriol (E3), and estrone Dempsey et al., 1936; Quadagno et al., 1972). On the other
sulfate (review in Beyer et al., 1976). The intensity and hand, the duration of the estrogen priming effect does depend
frequency of proceptive behavior in the rat (e.g., ear wiggling on the dose injected: if large dosages of E2 are given, sexual
and hopping) and some characteristics of the lordosis behavior can be facilitated by P administration even 96 h
response are directly correlated with the dosages of estrogen after estrogen administration (Boling and Blandau, 1939;
administered. Thus, by increasing the estrogen dose in OVX Quadagno et al., 1972). Thus, in normal cycling female rats,
rats, the degree of arching of the back during lordosis estrogen acts to prime the subsequent responsiveness to P.
increases, the latency to respond with lordosis to male Rodent species differ widely with respect to the amount of
mounting shortens, and the duration of the lordosis posture systemically administered P that is required to induce lordosis
is extended (Zemlan and Adler, 1977). Likewise, Domínguez- in OVX, estrogen primed females. For example, after being
Ordoñez et al. (2015) showed that lordosis was facilitated by primed with suprathreshold doses of estrogen, the minimum
subcutaneous or intracerebral administration of several doses dose of P per 100 g body weight was 4 mg for guinea pigs
of free E2, in OVX Sprague Dawley female rats primed with (Wade and Feder, 1972), 50 mg for the golden hamster (Frank
EB (Domínguez-Ordonez et al., 2015). Taken together, these and Fraps, 1945), and 100 mg for the rat (Powers and
observations indicate that, in the induction of lordosis in Valenstein, 1972). These differences in potency appear to be
OVX female rats, exogenously administered E2 can act both related to variations in the brain’s capacity to incorporate and
to prime the later responsiveness to P and to facilitate the retain P. Thus, the guinea pig takes up more P than the hamster,
expression of sexual behavior in rats that had previously been which in turn retains more than the rat brain (Wade et al.,
primed with an estrogen. 1973). In estrogen primed rats, P promotes sexual attractivity,
More recently, it has been shown that chronic, repeated as well as the display of proceptive behaviors such as ear
administration of EB (2 or 10 mg) in sexually experienced wiggling and darting (Hlinak and Madlafou, 1969; Hlinak
OVX Long-Evans and Wistar rats induced ‘sensitization’ of and Madlafousek, 1972; Whalen and Gorzalka, 1972). P has
receptivity (Jones et al., 2012; Jones and Pfaus, 2014). Thus, been particularly associated with the display of proceptive
rats administered EB every 4 days, across 32 days, displayed behavior in the OVX female rat: in one study, ear wiggling
significantly higher levels of proceptive behaviors and sexual was highly correlated with the dose of P administered, whereas
receptivity in response to EB administration, compared to no significant relationship was observed between proceptive
rats that had not received chronic EB treatment. EB-induced behavior and the amount of E2 administered (Tennent et al.,
sensitization did not involve the release of adrenal steroid 1980).
64 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

In the brain, P is rapidly reduced in its ring A to 1.02.1.2.3 Effect of Steroid Hormones in Lagomorphs
5a-pregnanedione (a-DHP), which subsequently can be (Rabbits)
reduced in C3 to 3a- or 3b-pregnanolones by the action of 1.02.1.2.3.1 Estrogen
3a- or 3b-hydroxysteroid oxoreductases, respectively (Beyer In the female European rabbit, E2 is the principal gonadal
and González-Mariscal, 1991; Cheng and Karavolas, 1973; hormone that promotes estrous behavior: in contrast to the
Karavolas et al., 1984; Whalen, 1985). A review of the litera- rat, estrous behavior in the rabbit is not associated with
ture reveals large variations between studies in the reported a peak in circulating P levels (Beyer and Mcdonald, 1973). In
capacity of ring A-reduced P metabolites to stimulate estrous unmated female rabbits, under controlled laboratory condi-
behavior. In some cases, these differences among studies can tions, ovarian follicles develop in overlapping waves,
be ascribed to differences in the solvents used and in the producing a relatively constant plasma level of E2. Under these
manner in which the hormone was administered (Gorzalka conditions, does display a state of relatively persistent sexual
and Whalen, 1977; Karavolas et al., 1984; Wade and receptivity. However, in more natural conditions, does were
Feder, 1972). Thus, a-DHP or 5b-pregnanedione, 3a- or 3b- observed to display waxing and waning receptivity, with a cycle
pregnanolones were much less effective than P for inducing length of approximately 7 days, and a heightened attractiveness
lordosis when dissolved in oil and administered subcutane- to males across approximately 1–3 days. The receptivity cycle
ously to OVX rodents primed with estrogen (Czaja et al., length corresponds closely to that of follicular development
1974; Johnson et al., 1976; Pleim and DeBold, 1984; Whalen (reviewed in Beyer and Mcdonald, 1973). Ovulation is induced
and Gorzalka, 1972). However, when a-DHP was dissolved in in receptive females by a combination of diverse stimuli asso-
Tween-80 and administered to OVX rats, it was just as effective ciated with mating, arising from tactile (flank, perineal,
as P at inducing estrous behavior (Gorzalka and Whalen, vaginal), olfactory, visual, and auditory channels (Staples,
1977). On the other hand, additional studies showed that 1967). Immediately following coitus, ovarian output and/or
ring A-reduced progestins were less potent than P when plasma levels of E2, P, 20a-hydroxyprogesterone (20a-OHP),
administered intravenously (Meyerson, 1972). Finally, some and T increase, reaching a peak at approximately 2 h post-
ring A-reduced progestins, such as 5b,3b-pregnanolone mating, and then return to baseline. Ovulation occurs approx-
(5b,3b-Pgl) and 5a,3b-pregnanolone (5a,3b-Pgl), reported imately 10 h postmating (Bakker and Baum, 2000; Hilliard and
to be ineffective when systemically injected (Kubli-Garfias Eaton, 1971; Mills et al., 1981). As a result of ovulation,
and Whalen, 1977), were as effective as P to facilitate lordosis a number of distinct mechanisms become engaged that inhibit
when implanted directly into the ventromedial hypothalamus receptivity during the immediate and early postmating time
(VMH) or the medial preoptic area (MPOA) of OVX, estrogen- frames, across pregnancy (or pseudopregnancy), and during
primed rats (Beyer et al., 1988; González-Mariscal et al., 1989; lactation.
Rodríguez-Manzo et al., 1986). Therefore, the low potency of Contrary to what is observed in rodents, some sexual recep-
these P metabolites to induce estrous when dissolved in oil tivity is retained in a small proportion of adult female rabbits
solutions and administered SC is most likely due to the low for at least 2 months after ovariectomy (Beyer et al., 1969).
bioavailability of these compounds when that route of Moreover, sexual activity was displayed by a small proportion
administration is used (Gorzalka and Whalen, 1977; of adult female rabbits that had been ovariectomized prepuber-
Karavolas et al., 1984). tally; that is, 30 days after birth (for review, see Beyer and
Mcdonald, 1973). Taken together, these results indicate that,
1.02.1.2.2.3 Androgens in some individuals, the ovaries are not indispensable for initi-
In female mammals, androgens are secreted by the ovaries ating and maintaining sexual behavior.
and adrenals and can promote female sexual behavior In OVX rabbits, E2 administration potently promotes sexual
(Dupon and Kim, 1973). In OVX rodents, the most effective receptivity (Beyer et al., 1975). Thus, in OVX New Zealand
androgens in this regard are those that possess the delta 4–3 white rabbits, 3 mg of EB was sufficient to induce receptivity
keto structure, such as T. Such steroids fluctuate across the within 24 h (Palka and Sawyer, 1966). Later, a series of studies
female’s ovarian cycle and show maximal levels during found that the threshold dose for inducing lordosis behavior in
the afternoon of proestrous, temporally corresponding to OVX rabbits was between 250 ng day1 (40% response) and
the preovulatory peak in P (Belanger et al., 1981). Early 1 mg day1 (100% response). In these studies, EB was found
studies showed that testosterone propionate (TP) had to be much more potent than estrone in eliciting lordosis
a weak, but clear stimulatory effect on the lordosis reflex (McDonald et al., 1970).
in OVX rats when administered in large dosages Chin-marking behavior is also increased by E2 in OVX
(6 mg day1, across l0 days) (Beyer and Komisaruk, 1971). rabbits. Hudson et al. (1990) studied the effects of daily injec-
Similar results had previously been described by Ball and tions of 1 or 10 mg on chin-marking and sexual receptivity.
Beach (Ball, 1940; Beach, 1942). However, other studies They found that both doses of EB significantly increased both
in which lower doses of TP were administered (200 mg daily behaviors, over baseline values, relative to the control,
for 9–11 days) failed to observe a stimulatory effect of TP vehicle-treated group (Hudson et al., 1990). Likewise, EB
on lordosis (Pfaff, 1970). Other androgens, including 5a- implants into the VMH or MPOA reliably stimulated chinning
dihydrotestosterone (DHT) and androsterone, also failed in OVX females. In this study, most of the does implanted in
to stimulate the lordosis reflex in the OVX female rat (Beyer the VMH and around half of those that had received EB in
and Komisaruk, 1971). These results were concordant with the MPOA or diagonal band of Broca (DBB) showed lordosis
studies carried out in the OVX female rabbit, discussed in (Melo et al., 2008). These observations suggest that the VMH
the next sections. is an estrogen-sensitive brain area that stimulates both
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 65

chinning and lordosis while the MPOA seems to contain the effects of several types of androgens, both aromatizable
subpopulations of neurons involved in both behaviors. (capable of being converted to estrogen) and nonaromatizable
(unable to be converted to estrogen), on lordosis behavior in
1.02.1.2.3.2 Progestins OVX rabbits. They reported that aromatizable androgens
In contrast to the rat, progestins (e.g., P and 20a-OHP, a P (T, androstenedione, 19-hydroxy-androstenedione, and dehy-
metabolite secreted by ovarian interstitial tissue) have no droepiandrosterone) administered during a 10-day period,
known stimulatory effects on rabbit female sexual behavior. stimulated lordosis and mounting behavior, while nonaroma-
Circulating progestin levels are practically nonexistent during tizable androgens (DHT, androsterone, and 4-chloro-testos-
estrous and do not increase until immediately after ovulation terone) did not. Based on these results, these authors
(in which a brief, sharp peak in 20a-OHP release is observed) proposed that androgens primarily stimulate lordosis behavior
and, later, as a result of the formation of the corpora lutea, in the rabbit through their aromatization, i.e., conversion to E2
beginning around day 4 of pregnancy or pseudopregnancy (reviewed in Morali and Beyer, 1979). These studies were
(Beyer and Mcdonald, 1973; Hilliard, 1973; Hilliard et al., among the first to suggest that some biological effects of T –
1968). In the case of fertile matings, plasma P levels rise in this case, stimulatory effects on female sexual behavior –
steadily from day 4 postmating, reach a peak at approximately might be mediated by its aromatization to E2. This idea was
day 16, and then steadily decline back to baseline (premating) supported by studies showing that an estrogen receptor (ER)
levels by day 31 (parturition) (Hilliard et al., 1968). Sexual antagonist, ethamoxytriphetol (MER-25), blocked the ability
behavior, scent marking as well as sexual receptivity, is abruptly of TP to induce estrous behavior in the rabbit (Beyer and Vidal,
inhibited after mating. Activation of the progesterone receptor 1971).
(PR) appears to participate in the ‘early’ postcoital inhibition of
sexual behavior observed prior to the formation of the corpora 1.02.1.2.4 Seasonality of Reproduction in Goats
lutea, when circulating P levels are negligible, as well as across Reproductive seasonality is a common characteristic of many
pregnancy, when plasma P levels are high (Beyer and Rivaud, mammalian species, allowing females to produce offspring at
1969; Hoffman and González-Mariscal, 2006, 2007). Likewise, the optimal time of year for the survival of the neonate.
in OVX, E2-primed female rabbits, P administration inhibits Thus, female goats originating from temperate or subtropical
sexual receptivity and scent-marking behavior (Beyer et al., latitudes display reproductive seasonality. In several breeds of
1969; Hudson et al., 1990). Beyer and his group studied the goats from the north hemisphere, the breeding season, charac-
effect of 0.5, 1.0, and 2.0 mg of P on sexual receptivity in terized by the display of estrous behavior and ovulation, starts
OVX rabbits that had been treated with twice daily injections in autumn (August–September) and finishes in winter
of 0.5 mg of EB for 4 days. In these experiments, 2 mg of P (February–March). In these females, the anestrous season, char-
inhibited receptivity in all females, while 0.5 mg inhibited acterized by the absence of estrous behavior and ovulation,
this behavior in half of the subjects. This amount of P is compa- occurs during spring and summer (Chemineau et al., 1992;
rable to the daily output of P secreted by the ovaries of mid- Duarte et al., 2008).
pregnant rabbits (Hilliard et al., 1968). Similarly, P inhibited In goats from temperate or subtropical latitudes, the repro-
chinning behavior and sexual receptivity in OVX, EB-primed ductive seasonality is mainly controlled by photoperiod.
does (Hudson et al., 1990), an effect that is primarily Indeed, in experimental conditions where the duration of
mediated by activation of the PR (Hoffman and González- photoperiod is controlled, short days, i.e., 10 or 8 h of light
Mariscal, 2006). per day, stimulate ovulation, whereas long days, i.e., 16
Possible facilitatory effects of ring A-reduced progestins on or 14 h of light per day, inhibit the ovulatory response
female sexual behavior have not been investigated in the (Chemineau et al., 1992; Duarte et al., 2008). In goats exposed
rabbit, but since P itself has no stimulatory effects, it seems to natural light–dark conditions, photoperiod synchronizes an
unlikely that its ring A-reduced metabolites would have such endogenous rhythm of reproduction allowing the alternation
effects. However, it remains possible that 20a-OHP might of the sexual and anestrous seasons (Gebbie et al., 1999;
participate in the early postcoital inhibition of sexual behavior. Gomez-Brunet et al., 2008). During the sexual season,
Thus, 20a-OHP is secreted by the interstitial tissue of the ovary nonpregnant females show several successive estrous cycles of
in very large amounts just prior to ovulation, while at the same a mean duration of 21 days. Nevertheless, short (around
time P levels are very low or nondetectable (Hilliard et al., 8 days), or long (around 39 days) estrous cycles are commonly
1963). These endocrine changes correlate with an abrupt observed. Most estrous cycles are associated with ovulation;
drop in sexual behavior (Beyer et al., 1969), suggesting that however, ovulation without estrous or estrous without ovula-
20a-OHP, which weakly activates the PR, could be involved tion can also occur (Chemineau et al., 1992).
in the early postcoital inhibition of sexual behavior. Consistent Photoperiod modifies the E2 negative feedback on gonado-
with this idea, the administration of a PR antagonist combined tropin secretion between the breeding and anestrous seasons.
with a small dose of EB prevented the early postmating decline During increasing or long days, the feedback of E2 reduces
in sexual receptivity and scent marking in intact female rabbits, dramatically the secretion of gonadotropins (LH and follicular
while neither treatment by itself was able to do so (Hoffman stimulating hormone (FSH)), inhibiting ovulation. By contrast,
et al., 2010). during decreasing or short days, this feedback decreases, stimu-
lating the secretion of gonadotropins, which allow ovulations
1.02.1.2.3.3 Androgens to occur (Chemineau, 1987; Mori et al., 1987). The seasonal
Some androgens are known to stimulate estrous behavior in changes in E2 negative feedback were described in OVX goats
intact and OVX female rabbits. Beyer et al. (1970a,b) evaluated treated with subcutaneous implants constantly releasing
66 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

estradiol-17b (OVX þ E2). In these OVX þ E2 females, high of P itself. These results confirm that the temporal sequence of
and low plasma LH concentrations coincide with ovulatory P and E2 administration affects sexual behavior in OVX goats.
and anovulatory periods of intact goats, respectively (Duarte This sequence of secretion of both E2 and P is quite different
et al., 2008; Henniawati et al., 1995). Therefore, the modifica- from that seen in many rodent species in which the increase
tion of the E2 negative feedback on gonadotropin secretion in P follows the increase in E2. This suggests that the role of
induced by photoperiod constitutes the neuroendocrine mech- these two hormones differs according to the species.
anisms determining the breeding and seasonal anestrous Regarding androgens, it has been shown that aromatizable
seasons. androgens, such as T and androstenedione, stimulate the
expression of sexual behavior in OVX goats (Katz, 2007). In
1.02.1.2.5 Endocrine Control of Sexual Behavior in Goats contrast, a nonaromatizable androgen, such as dihydrotestos-
There is evidence showing the importance of steroids in the terone is not effective (Lindia and Katz, 2003). One study
control of female sexual behavior in goats. First, as in many tested the effect of the androgen receptor antagonist flutamide,
other mammalian species, hormonal changes in female goats a potent androgen receptor antagonist. It was suggested that
include an increase in E2 concentration that is observed around while estrogens facilitate proceptive and maybe attractive
24–48h before the expression of sexual behavior (Thorburn behaviors, an androgen-dependent pathway modulates recep-
and Schneider, 1972). This change suggests that E2 may be tivity in female goats (Imwalle and Katz, 2004).
a good candidate for the control of sexual behavior. In this
regard, it is well known that E2 alone is sufficient to induce 1.02.1.2.6 Nonsteroidal Agents on Estrous Behavior
estrous behavior in French-Alpine goats (Billings and Katz, A number of studies, many of which have been carried out in
1998; Kalivas et al., 2006), even if the amount of E2 used is rodent species, demonstrate that nonsteroidal hormones
in the pharmacological range (around 30 mg), while E2 concen- participate along with ovarian hormones in the regulation of
trations on the day of estrous were reported to be around female sexual behavior. Beginning in the early 1970s, pharma-
15 pg ml1 in intact Surti and Mahwari goats (Pathak et al., cological studies showed that gonadotropin-releasing
1990), or in Shiba goats (Mori and Kano, 1984). In addition, hormone (GnRH) (Moss and McCann, 1973) and prosta-
the proportion of OVX females displaying estrous behavior as glandin E2 (PGE2) (Rodríguez-Sierra and Komisaruk,
well as the intensity of expression of proceptivity and recep- 1977) facilitated lordosis behavior. Since then, a surprising
tivity increase according to the dose of E2 given to the animals variety of nonsteroidal compounds have been shown to stimu-
(Billings and Katz, 1998). Finally, the adequate display of late lordosis behavior. These compounds include certain small
female sexual behavior appears to be dependent on the dura- neuropeptides (e.g., oxytocin (OT), melanocyte-stimulating
tion of E2 treatment rather than on the doses or the maximal hormone), proteins (prolactin, ACTH), and neurotransmitters
concentration achieved (Okada, 1998). (noradrenaline, dopamine, GABA, acetylcholine) (for review,
The increase in E2 concentration appears only after the see Beyer and González-Mariscal, 1986). The diversity of
regression of the corpus luteum, which leads to a decline in P compounds that promote lordosis in the female rat speaks to
concentration around 48 h before the expression of sexual the complex nature of the brain networks that underlie female
behavior. During the breeding season, expression of sexual sexual behavior. In the following paragraphs, we discuss some
behavior is preceded by a 12- to 15-day luteal phase during of these compounds: GnRH, OT, leptin, and nitric oxide (NO).
which plasma concentrations of P are high (between 2 and
5 ng ml1). In addition, it is known that the first estrous of 1.02.1.2.6.1 Gonadotropin-Releasing Hormone
the breeding season may be behaviorally ‘silent’ in some breeds In addition to triggering the release of LH from the anterior
of goats (Jainudeen, 1993). As the concentration of P is very pituitary, ultimately resulting in ovulation, GnRH also facili-
low during the nonbreeding season, this suggests that exposure tates lordosis in the female rat after either systemic or intracere-
to P may be necessary for the full display of sexual behavior at bral administration (Beyer et al., 1997; González-Mariscal
the onset of the breeding season as is the case in sheep. et al., 1993; Mahesh, 1985; McCann, 1977; Moss and McCann,
However, in OVX goats, P priming did not increase the display 1973; Ramirez-Orduna et al., 2007). In OVX, estrogen-primed
of estrous at the end of the anestrous period (Sutherland and rats, infusions of this peptide directly into the MPOA or the
Lindsay, 1991). VMH facilitated lordosis behavior, while infusion of a GnRH
These results should not point to a lack of a role of P in the antibody reduced the frequency of the lordosis response (Pfaff,
display of female sexual behavior. Indeed, it has been also 1993). In these studies, the latency to induce the maximal
shown that the timing between the secretion of P and the rise lordosis response after GnRH infusion was relatively long
in E2 is important for the expression of female sexual behavior (1 h 45 min). However, the effect of GnRH was independent
(Billings and Katz, 1998). In fact, P facilitated E2-induced of pituitary hormones since it persisted in hypophysectomized
attractivity and receptivity in the anestrous period if the P treat- E2-primed rats, and the combined administration of LH, FSH,
ment occurred 48 or 72 h prior to the treatment with E2. During and E2 did not elicit lordosis (Moss and Mccann, 1975).
the breeding season, P facilitated attractivity when treatment Ramirez-Orduña et al. (2007) showed that GnRH (0.005 and
occurred 72 h prior to treatment with E2. By contrast, P has 0.05 mg doses) infused into the right lateral ventricle (intracer-
an inhibitory effect on all sexual behaviors when the treatment ebroventricular (icv)), induced proceptivity and lordosis
occurred 12 or 24 h prior to injection of E2. Finally, P did not behavior in OVX, EB-primed rats (Ramirez-Orduna et al.,
facilitate E2-induced proceptivity in either sexual or anestrous 2007). These results are in agreement with previous studies
seasons. Therefore, the decline in circulating P concentrations that showed that GnRH induced female sexual behavior
prior to the rise in E2 is more important than just the presence when administered either intracerebrally or systemically (Beyer
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 67

et al., 1997; González-Mariscal et al., 1993; Hall and Luttge, concomitant with the LH surge and the period of behavioral
1977; Hall et al., 1975; Moss and Foreman, 1976; Moss and receptivity (Calka, 2006). In OVX female rats primed with E2
McCann, 1973; Rodríguez-Sierra et al., 1977; Rodríguez-Sierra plus P, intracerebroventricular infusion of NOS inhibitors or
and Komisaruk, 1982). NO scavengers attenuated lordosis (Mani et al., 1994).
Conversely, an NO donor facilitated lordosis in E2-primed
1.02.1.2.6.2 Oxytocin OVX rats in the absence of P. Given these observations, Chu
Administration of OT directly into the hypothalamus or MPOA and Etgen (1997) proposed that cGMP, which is synthesized
facilitated lordosis behavior in E2-primed female rats (Caldwell subsequent to the activation of soluble guanylyl cyclase by
et al., 1989, 1994; Schulze and Gorzalka, 1991), whereas treat- NO, could be an important cellular second messenger to stim-
ment with E2 increased OT binding in the VMH (Caldwell et al., ulate lordosis in hormonally primed female rats. In order to
1994). Likewise, in the rat, OT injected into the cerebral ventri- test this idea, they used 1H-[1,2,4]oxadiazolo[4,3-a]quinoxa-
cles stimulated proceptive behavior, while an OT antagonist lin-1-one (ODQ), a selective inhibitor of NO-stimulated
inhibited female sexual behavior (Caldwell et al., 1989, soluble guanylyl cyclase. As predicted, ODQ infused into the
1994; Schulze and Gorzalka, 1991). The stimulatory effects of lateral or third ventricle suppressed lordosis in a dose-
OT on lordosis depended on the time of administration rela- dependent manner in OVX female rats treated with E2 plus P
tive to the light/dark schedule as well as the exact hypothalamic (Chu and Etgen, 1999; Chu et al., 1999).
location of the OT infusion (Caldwell et al., 1989). These vari-
ables appeared to influence measures of lordosis frequency and
duration independently (Schulze and Gorzalka, 1991). For 1.02.1.3 Cellular Processes Related to the Action of Steroid
recent reviews of the effects of OT circulating in the CSF, see Hormones on Estrous Behavior
Veening et al. (2010) and Veening and Olivier (2013).
Intracellular receptors for steroid hormones (e.g., estrogens, P’s,
glucocorticoids, androgens, mineralocorticoids) are members
1.02.1.2.6.3 Leptin
of the nuclear receptor superfamily of ligand-dependent tran-
Leptin is produced primarily in adipose tissue, and it is an
scription factors, which also include receptors for certain other
important hormonal regulator of food intake, weight gain,
nonsteroid lipophilic hormones and ligands (such as thyroid
and energy balance (Campfield et al., 1996). Moreover, leptin
hormone, retinoic acid, 9-cis retinoic acid, vitamin D3 and
has been reported to influence estrous behavior in rodents in
eicosanoids, fatty acids, and lipids), as well as orphan receptors
a manner that appears to depend on the animal’s metabolic
that have no known ligand (Beato, 1987; Brinton et al., 2008).
or nutritional state (Fox and Olster, 2000; García-Juarez
Steroid receptors, cloned in various species (Faber et al., 1976;
et al., 2013, 2012, 2011; Wade et al., 1997). Thus, leptin facil-
Horwitz et al., 1986; Schrader et al., 1981), comprise four main
itated sexual behavior in female hamsters fed ad libitum, but
domains: (1) the DNA-binding domain (DBD), which contains
intensified the fasting-induced inhibition of lordosis in food-
two Zn fingers that can interact with the DNA hormone
deprived animals (Wade et al., 1997). By contrast, in geneti-
response element; (2) the ligand-binding domain (LBD) at
cally obese Zucker rats treated with E2 and P, intracerebrally
the C-terminal of the molecule, which is responsible for
administered leptin did not augment lordosis and inhibited
hormone binding and the interaction with the 90 and 70 kd
proceptivity (Fox and Olster, 2000). Taken together, these
heat shock proteins; (3) the hinge region, between the DBD
studies suggest that this hormone can have either facilitatory
and LBD; and (4) the region located at the N-terminus of the
or inhibitory effects on rodent sexual behavior, apparently
molecule, whose specific functions are unknown, but with
depending at least in part on the animal’s metabolic or nutri-
transactivation properties that can increase gene transcription
tional state. Only a few studies have been carried out on the
regardless of hormone binding (Beato, 1987; Brinton et al.,
modulatory effects of leptin on lordosis in the ad libitum fed
2008; Horwitz et al., 1986; Schrader et al., 1981).
rat. It has been shown that doses of 1 and 3 mg produced signif-
Additionally, steroid receptors contain at least two domains
icant lordosis quotient at 60 min postinjection, with maximal
necessary to mediate transcriptional activation, designated
response being induced at 120 min, without inducing procep-
AF-1 and AF-2. AF-1 is located within the N-terminal
tive behavior at any dose (García-Juarez et al., 2011). The
domain, while AF-2 is found in the C-terminal domain.
observation that leptin can facilitate lordosis while inhibiting
proceptive behavior indicates that this hormone can directly
or indirectly dissociate neural mechanisms underlying female 1.02.1.3.1 ER and PR Expression in the Brain
rodent sexual behavior. 1.02.1.3.1.1 Rodents
The classic intracellular ER and PR are expressed in several brain
1.02.1.2.6.4 Nitric Oxide regions associated with reproductive behavior. Two subtypes of
NO is a diffusible gas neurochemical transmitter that is ERs are synthesized in distinct cell types: ER-a (Greene et al.,
produced from L-arginine, via the action of NO synthase 1986) and ER-b (Kuiper et al., 1996). These isoforms are the
(NOS) (Knowles et al., 1989), and that participates in major product of separate genes, the ER-b protein being somewhat
aspects of brain function related to reproduction (Dawson smaller than ER-a (530 and 595 amino acids, respectively).
and Snyder, 1994; García-Juarez et al., 2012). Studies have Similarly, in rodents, PR has two isoforms, designated PR-A
shown that NOS activity is induced by E2 and is present in and PR-B, and which are coded by a single gene through alter-
neurons of the VMH of female rats (Rachman et al., 1996). nate promoter usage. The PR-A isoform is an N-terminal
In the intact cycling female rat, NO levels in the hypothalamus truncated version of the PR-B isoform (minus 130 amino
doubled between the afternoon and evening of proestrous, acids).
68 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

ER-a and ER-b have distinct patterns of expression in the Dense aggregations of ERa-immunoreactive (IR) neurons
rodent brain (Greco et al., 2001; Osterlund et al., 1998; were also found in the infundibular nucleus (IN), the MPOA,
Shughrue et al., 1997, 1998; Simerly et al., 1990). For example, the bed nucleus of the stria terminalis (BNST), and in the amyg-
in the female rat brain, ER-a mRNA expression was found to be dala. Numerous ERa-IR neurons were observed in the paraven-
most intense in the anterolateral VMH, arcuate, and the ventral tricular hypothalamic nucleus, but not in the supraoptic or
premammillary nucleus. By contrast, the expression of ER- suprachiasmatic nuclei. The hippocampus lacked ERa-IR
b mRNA was greatest in the paraventricular hypothalamic neurons, but the ventral subiculum in the hippocampal forma-
nucleus, supraoptic nucleus, and the ventral premammillary tion had a dense group of such cells (Caba et al., 2003a).
nucleus. Both isoforms were widely expressed in the amygdala, In OVX, E2-treated female rabbits, PR mRNA was also highly
with ER-a expression being highest in the medial posterodorsal expressed in the hypothalamus and preoptic area. The subse-
nucleus, while ER-b expression was greatest in the medial post- quent administration of P resulted in a dramatic reduction of
erodorsal nucleus (Osterlund et al., 1998). PR mRNA in these brain regions (Camacho-Arroyo et al.,
In the intact cycling rat, PR is present in the anteroventral 2007, 1994). Likewise, in OVX, EB-treated does, a high
periventricular nucleus, the MPOA, the arcuate nucleus, and density of PR-IR cells was observed in the nucleus X, preoptic
the VMH during diestrous and across proestrous–estrous region, paraventricular nucleus, and IN. Injections of P
(Numan et al., 1999). During mid-pregnancy, PR expression provoked an intense downregulation of PR immunoreactivity
decreases in the arcuate nucleus and VMH, most likely due to (Caba et al., 2003b). Thus, similar to the rat and guinea pig,
the high levels of circulating P that are present at this time. Like- estrous in the rabbit is associated with increased expression
wise, the expression of the individual PR-A and PR-B isoforms of PR in the MPOA and in the VMH.
is regulated by ovarian steroids, in a brain region-specific
manner, across the ovarian cycle. In OVX female rats, E2 1.02.1.3.2 Cellular Mechanisms Proposed to Explain
administration increased the expression of both PR isoforms the Regulation of Estrous Behavior by Different Agents
in the hypothalamus and preoptic area, whereas the In this section we describe findings regarding the cellular mech-
subsequent administration of P decreased the expression of anisms involved in the regulation of rat female sexual behavior
both isoforms only in the hypothalamus. In intact cycling by a variety of agents. They include specific intracellular path-
rats, PR-A and PR-B were highly expressed in the preoptic ways, such as those that involve protein kinase A (PKA), NO/
area during diestrous, while their expression in the protein kinase G (PKG), and the Src/PR/MAPK system. As
hypothalamus was low (Guerra-Araiza et al., 2003). described below, these pathways participate in estrous behavior
Activation of the PR appears to be necessary for the normal induced by progestins, GnRH, and leptin, and by vaginal-
stimulation of estrous behavior in the rodent species that are cervical stimulation (VCS).
most commonly studied in the laboratory. In OVX, estrogen-
primed female rodents, RU486, a PR antagonist, blocks the 1.02.1.3.3 Protein Kinase A
P-mediated facilitation of sexual behavior. Likewise, P fails to The molecular mechanisms that regulate cell function often
stimulate lordosis in knockout mice lacking the A isoform of involve changes in the specific conformation of proteins (allo-
the PR (Mani and Oyola, 2012). González-Flores and steric processes) or in their chemical composition (e.g., methyl-
colleagues investigated the participation of the PR-A and PR- ation, acetylation, glycosylation, and phosphorylation).
B isoforms in stimulating lordosis in estrogen-primed OVX Protein phosphorylation is a major mechanism through which
female rats. These studies, which used antisense many hormones regulate intracellular processes (Nestler et al.,
oligonucleotides to selectively inhibit the activity of both 1994). The enzymes responsible for protein phosphorylation
isoforms or of PR-B alone, indicated that PR-B activation was are called protein kinases and are specifically activated by
necessary for lordosis induced by P, 5a-DHP, and a number of second messengers, including cyclic AMP
5b,3b-pregnanolone in OVX, estrogen-primed female rats (cAMP), cyclic GMP (cGMP), peptides, calcium, and phospho-
(Guerra-Araiza et al., 2009). Using the same experimental lipids (Beyer et al., 2003; Nestler et al., 1994).
strategy, Carrillo-Martínez et al. (2011) showed that P cAMP is a major intracellular mediator of hormone
induced the expression of tryptophan hydroxylase and responses, through its role as a stimulator of a specific protein
glutamic acid decarboxylase in the hypothalamus of OVX rats kinase, called PKA. Early studies by Beyer et al. (1981) evaluated
via PR-B activation, whereas P reduced the expression of the involvement of the cAMP-PKA signaling pathway for the
tyrosine hydroxylase through the activation of both PR display of estrous behavior of OVX, E2-primed rats, finding
isoforms (Carrillo-Martínez et al., 2011). Taken together, that subcutaneous or intracerebral administration of
these studies indicate that the PR-B isoform is essential for dibutyryl cAMP facilitated lordosis in such animals (Beyer
the display of lordosis behavior in rats, and that, considering et al., 1981). These seminal results were the foundation of
the findings of studies in the mouse, the role of specific PR a unitary model to explain how, in estrogen-primed rats,
isoforms in female sexual behavior may be species specific lordosis could be facilitated by P as well as by a variety of
(Mani and Oyola, 2012; Mani et al., 2009). other hormones and neurotransmitters (Beyer et al., 1981,
2003, 1997; Beyer and González-Mariscal, 1986; Blaustein,
1.02.1.3.1.2 Rabbits 2009; Blaustein and Mani, 2007).
Clear cytoplasmic ER-a immunoreactivity was observed in In the proposed model, estrogen binds to its intracellular
distinct forebrain areas of the female rabbit (Caba et al., receptor, and this ‘hormone-receptor’ complex is translocated
2003a). Thus the nucleus X, located in the ventrolateral part to the nucleus. There, it induces the expression of certain
of the VMH, was very rich in cytoplasmic immunostaining. proteins, most of which are in an inactive condition (inactive
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 69

estrogen-induced proteins, or iEIPs). The subsequent activation agents. They found that icv administration of LNAME (an
of these iEIPs occurs as a result of their phosphorylation by NOS inhibitor) and ODQ (a specific inhibitor of soluble gua-
cAMP-dependent PKA. cAMP can be produced from the nylyl cyclase) suppressed lordosis behavior induced by P and
activation of membrane receptors linked to adenylate cyclase, by 5a-reduced P metabolites, as well as lordosis facilitated by
thus allowing for ‘cross-talk’ between intracellular hormone VCS. Lordosis was suppressed at 2 h after drug administration
receptors and membrane receptors. The first such ‘cross-talk’ but returned to control values by 4 h. Likewise, icv infusion of
model proposed that cAMP – generated from the activation the PKG inhibitor KT5823 significantly inhibited lordosis
of membrane NE receptors linked to adenylate cyclase – induced by all three progestins at 2 h after drug administration.
activates intracellular iEIPs. There is evidence suggesting that Moreover, they found that LNAME and ODQ also blocked
one such iEIP could be the PR. This possibility is supported estrous behavior induced by GnRH, PGE2, and db-cAMP
by a variety of different experimental strategies, including (González-Flores et al., 2009). By contrast, KT5823 reduced
blocking the PR with the antiprogestin RU486, the PGE2- and db-cAMP-induced estrous behavior, but did not
administration of PR antisense oligonucleotides, as well as affect the behavioral response to GnRH. Thus, the NO/
studies in knockout mice lacking the PR (Beyer et al., 2003, cGMP/PKG pathway appears to be one important mechanism
1995, 1997; Blaustein et al., 1987). Studies suggest that by which estrous behavior in estrogen primed rodents can be
kinases (e.g., PKA) that are activated by agents that induce facilitated by a variety of progestins, neurotransmitters, and
lordosis do not directly phosphorylate the PR (Bai et al., neuromodulators, including GnRH and PGE2.
1997), but work indirectly through the activation of MAPK,
which in turn phosphorylates serine residues on the PR 1.02.1.3.4 Leptin and Protein Kinase
(Faivre et al., 2008; Lange et al., 2000; Qiu et al., 2003). As described above, leptin is also a modulator of female sexual
Nevertheless, it is important to note that diverse agents that behavior. The possible interaction of this hormone with the
induce lordosis may act through different protein kinase various signaling pathways implicated in regulating estrous
systems. For example, in OVX, estrogen-primed rats, behavior in the rat has been investigated by González-Flores
pharmacological inhibition of PKC prevented the facilitation et al. (2011). Several doses of leptin, infused icv, significantly
of lordosis by several ring A-reduced progestins, but had no enhanced lordosis behavior in OVX, E2-primed female rats.
effect on lordosis induced by P (González-Flores et al., Additional experiments explored the participation of specific
2006). On the other hand, lordosis induced by P, GnRH, receptors, such as the PR and GnRH-1 receptors, on leptin-
prostaglandin (PG), E2, and db-cAMP were blocked by facilitated lordosis behavior. These experiments revealed that
RpAMPC, an inhibitor of PKA (Ramirez-Orduna et al., 2007). the facilitatory effects of leptin on lordosis are mediated
Therefore, it appears that lordosis is positively modulated by through these receptors, as the administration of RU486 or
the coordinated action of multiple intracellular signaling antide (an antagonist of the GnRH-1 receptor) prior to leptin
systems that converge on the PR, which may be a common administration inhibited the ability of this hormone to facili-
effector for the facilitation of lordosis by both steroidal and tate lordosis (García-Juarez et al., 2011).
nonsteroidal agents. In a follow-up study, the same group investigated the partic-
ipation of the NO-cGMP-PKG pathway in the leptin-induced
1.02.1.3.3.1 NO Pathway facilitation of lordosis (García-Juarez et al., 2012). They
As described above, several lines of evidence indicate that NO is found that LNAME, ODQ, and KT5823 significantly reduced
a modulator of lordosis behavior. Studies show that the stimu- the lordosis behavior induced by the icv administration of
latory effects of NO on lordosis behavior might involve the leptin. Additionally, these authors explored the participation
NO-cGMP pathway. Beyer et al. (1982) found that, in OVX- of several other protein kinases, including JAK2, Src tyrosine
adrenalectomized female rats primed with E2, administration kinase, and MAPK, in leptin-induced lordosis behavior in
of cGMP, dpc-GMP, and 50 GMP stimulated lordosis OVX, E2-primed rats fed ad libitum. Inhibitors of JAK2
behavior. Later, Chu and Etgen (1999) proposed that the (AG490), Src tyrosine kinase (PP2), and MAPK (PD98059)
coupling of hypothalamic a1-adrenoreceptors to the NO- caused a significant decrease in leptin-facilitated lordosis.
cGMP pathway is steroid hormone-dependent and that this These results demonstrate that the stimulation of lordosis
pathway mediates the facilitation of lordosis by NE in OVX behavior in estrogen-primed rats by leptin requires the
rats treated with E2 and P (Chu and Etgen, 1999; Chu et al., simultaneous or sequential activation of several signaling
1999). This idea was supported by experiments showing pathways (see Figure 1), which most likely converge on
that (1) icv infusion of ODQ, a selective inhibitor of NO- hypothalamic neurons that regulate the expression of lordosis
stimulated cGMP, suppressed lordosis in OVX rats treated behavior (García-Juarez et al., 2012). The mechanism
with E2 and P, and (2) lordosis was also inhibited by icv through which the PR participates in the facilitation of
infusion of KT5823, a cell-permeable, highly specific lordosis by leptin is unclear, but it may involve the
inhibitor of PKG. Moreover, the PR apparently also phosphorylation of this receptor.
participates in this pathway, as RU486, a PR antagonist,
inhibited lordosis induced by the administration of 1.02.1.3.5 Src Tyrosine Kinase, MAPK Pathway, and Lordosis
8-br-cGMP (Chu et al., 1999). Behavior
Experiments carried out by González-Flores and colleagues The cellular tyrosine kinase c-Src may be one of the most funda-
(González-Flores et al., 2007, 2009; González-Flores and mental components for the intracellular integration of steroid
Etgen, 2004) explored whether the NO/cGMP/PKG pathway and growth factor signaling. This kinase is expressed by virtu-
participates in the induction of estrous behavior by diverse ally all eukaryotic cells, where its activation is critical for
70 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

Leptin
GnRH neuron (b)

GnRH

(a)
GnRH-1

Antide
JAK2

X
PR PP2
C-Src
RU486

X
X
MAPK

P Gene
PR
Promoter

Lordosis behavior

Figure 1 Direct (a) and indirect (b) effects of leptin potentially involved in lordosis facilitation in OVX, estrogen-primed rats. A direct activation of
the long leptin receptor directly activates JAK2 system that in turn stimulates the Src-MAPK pathway to facilitate lordosis. A potential indirect effect
of leptin is depicted in (b). In this case, leptin releases GnRH through indirect activation of GnRHergic neurons. GnRH, in turn, would activate the Src
kinase (c-Src) pathway, which has been shown to activate lordosis behavior in OVX, estrogen-primed rats. As shown in the figure, specific inhibitors
of PR, RU486, GnRH-1 receptor, antide or PP2, Src inhibitor, interfere with the behavioral action of leptin. The effect of Src could be direct at the
genome or mediated by MAPK. OVX, ovariectomized; GnRH, gonadotropin-releasing hormone; PR, progesterone receptor.

generating the appropriate cellular responses to a variety of the nucleus by import and export mechanisms. In cell culture
extracellular signals, including growth factors, cytokines, adhe- systems, ligand-activated PR has been shown to interact with
sion molecules, and antigen receptors (Brown and Cooper, Src kinase, resulting in the stimulation of the Src/Ras/Raf/
1996; Thomas and Brugge, 1997). All members of the Src MAPK signaling pathway (Boonyaratanakornkit and
kinase family comprise a conserved tyrosine kinase (SH1) Edwards, 2004; Leonhardt et al., 2003; Migliaccio et al.,
domain, SH2, SH3 domains, and an SH4 domain at the 1996, 1998). Thus, Migliaccio et al. (1998), demonstrated
N-terminus that targets the protein to the cell membrane. Src that P induced the rapid activation of MAPK through an
kinase activates the MAPK pathway, which is a prominent Src-dependent mechanism. Subsequent studies indicated
intracellular signaling pathway that is present in virtually all that the ER was required for the interaction between PR and
eukaryotic cells and that can modulate the activity of Src, as well as for the PR-induced activation of the MAPK
a variety of effector molecules, including transcription factors, pathway (Migliaccio et al., 1996, 1998). On the other hand,
cytoskeletal proteins, and protein kinases. studies using the synthetic progestin R5020 as a ligand
Importantly, the PR has the potential to interact with cyto- indicated that, alternatively, PR can activate the Src/MAPK
plasmic signaling molecules as well as with nuclear elements, pathway independently of the ER (Boonyaratanakornkit
as it is actively translocated between the cell cytoplasm and et al., 2008).
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 71

The MAPK pathway appears to be critical for the hormonal Src–MAPK signaling also appears to interact with the classical
facilitation of estrous behavior. Etgen and Acosta-Martinez pathways of PR action by phosphorylating Ser-294 in this
(2003) demonstrated that, in OVX female rats, if MAPK inhib- protein, thereby enhancing its transcriptional activity (Lange
itors (PD98059 and U0126) were infused icv during E2 et al., 2000; Migliaccio et al., 1996, 1998).
priming, subsequent lordosis behavior was completely abol-
ished. Moreover, in E2-primed female rats, icv infusion of
1.02.1.4 Neuroendocrine Factors Participating in the
PD98059 significantly diminished lordosis behavior induced
Behavioral Transition across the Reproductive Cycle of Rats,
by a variety of agents, such as ring A-reduced progestins, the
Rabbits, and Goats
delta opioid agonist DPDPE, GnRH, PGE2, cAMP, leptin, as
well as by VCS (García-Juarez et al., 2013; González-Flores 1.02.1.4.1 Anestrous to Estrous in Rodents
et al., 2009). Interestingly, lordosis induced by the cGMP Anestrous in the female rat is associated with low serum
analog, 8-br-cGMP, was also inhibited by a MAPK inhibitor, FSH levels, compared to those observed in animals that are
as well as by RU486, a PR antagonist. These results suggested in constant estrous, pseudopregnant, or young cycling animals.
that cGMP-mediated facilitation of lordosis involves ligand- Serum LH levels are undetectable in anestrous females (Huang
independent activation of PR in the brain by MAPK et al., 1976, 1978). Likewise, estrogen and progestin levels are
phosphorylation (González-Flores et al., 2004b). also lower during anestrous, relative to those observed in any
A possible role for MAPK phosphorylation in the phenom- other reproductive state. By contrast, serum prolactin levels
enon of sequential inhibition was suggested based on the are sixfold higher among aging anestrous rats, compared to
finding that MAPK phosphorylation of the PR promotes those measured in young (4- to 6-month-old) animals (Huang
receptor downregulation via proteasomal degradation. This and Meites, 1975). Thus, the transition to the anestrous condi-
possibility was tested in OVX, E2-primed female rats by tion coincides with the decrease in plasma gonadotropins and
infusing a MAPK inhibitor (PD98059) icv, 30 min before the an increase in plasma prolactin. It has been suggested that the
first injection of P; 4 h later, these subjects did not display progressive reduction in hypothalamic catecholamines that
lordosis. Then, 24 h later, these same animals received a second occurs during aging might be associated with the anestrous
injection of P. Normally, this second P injection would be inef- state (Huang et al., 1978). Senescent, anestrous animals display
fective at inducing lordosis (sequential inhibition); however, in atrophic ovaries with few primary follicles as well as infantile
the females that had received the MAPK inhibitor along with uteri, suggesting diminished ovarian steroid production
the first P injection showed high levels of lordosis, indicating (Huang and Meites, 1975).
that sequential inhibition had been blocked by inhibiting The transition from anestrous to estrous behavior in the
MAPK (González-Flores et al., 2004b). Downregulation of cycling female rat occurs on the afternoon of proestrous,
hypothalamic PRs was also inhibited by PD98059 (González- approximately 10 h before ovulation. This transition is tempo-
Flores and Etgen, 2004). From these findings, we suggest that rally associated with a rise in P and high circulating levels of E2.
MAPK-dependent phosphorylation of PRs has a dual role in This latter hormone is secreted in increasing amounts across
regulating the actions of P and the PR in female rat sexual day 2 of diestrous, ultimately stimulating a surge of LH, which
behavior: initially, MAPK phosphorylation contributes to the in turn elicits a surge of P, produced by the granulosa cells, in
facilitatory actions of P, but subsequently it targets PR for the early afternoon of proestrous (Freeman, 1994). The P surge
degradation by the 26S proteasome pathway. This wanes across the morning of estrous, concomitant with
mechanism appears to underlie the phenomenon of a decline in estrous behavior. During this phase, hypothalamic
sequential inhibition. Further support for this idea comes PR is also downregulated, a process that participates in the
from the observation that sequential inhibition is likewise termination of estrous behavior (Blaustein and Erskine,
prevented by a proteasome inhibitor (González-Flores and 2002). In the experimental paradigm of sequential inhibition,
Etgen, 2004). a second administration of P, given 24 h after administering
Based on these studies, González-Flores and colleagues E2 and P, does not induce sexual behavior in female rats.
proposed the novel idea that the Src/PR/MAPK pathway is an Sequential inhibition relies on the P-mediated induction of
essential component in the facilitation of estrous behavior the 26S proteasome, a protease that degrades PR (among
(see Figure 2). This model is supported by the finding that other proteins) in the brain (González-Flores et al., 2004a).
the specific Src kinase inhibitor PP2 reduced lordosis behavior Across pregnancy, female rats fail to display estrous behavior
stimulated by progestins (P and its ring A-reduced (Hardy, 1970), an effect that is most likely due to high
metabolites), peptides, and VCS (González-Flores et al., circulating levels of P.
2010; Lima-Hernández et al., 2012). Moreover, these results
suggest that several agents, including GnRH and PGE2, as 1.02.1.4.2 Anestrous to Estrous in Rabbits
well as VCS, act on membrane GPCRs that then activate In the prepubertal or seasonally anestrous rabbit, sexual recep-
serine/threonine kinases (PKs) such as PKA, PKC, and PKG. tivity increases gradually as ovarian follicles mature and begin
Stimulation of these kinases might also activate Src kinase to secrete E2. In the adult female rabbit, under favorable envi-
through several mechanisms. For example, ring A-reduced ronmental conditions, estrous behavior generally persists until
progestins may stimulate Src activity either by acting directly after mating occurs. Thus, the proportion of females that
on a membrane-associated PR or indirectly by stimulating the remain receptive decreases dramatically during the first 24 h
release of GnRH. It appears that PR acts as a scaffold within after mating (Beyer and Rivaud, 1969). Sexual receptivity
a multiprotein Src/MAPK signaling complex, in order remains low across pregnancy, most likely due to P secreted
to recruit a variety of enzymatic effectors. Additionally, by the corpora lutea, which likely counteracts the stimulatory
72 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

ProgesƟns VCS

GnRH PGE2
P

Gα Gα Gα
P

PKs

Cytoplasm
P
c-Src
PR

PP2
P

P MAPK
PR

Nucleus Ser-294 P
CREB STAT

? ?

Estrous behavior
havio
Figure 2 Diagram of the possible mechanisms of action through which several progestins, GnRH, PGE2, and vagino-cervical stimulation (VCS) acti-
vates the PR-Src system to elicit estrous behaviors in OVX, EB-primed rats. As shown in the upper right section of the diagram, VCS stimulates
release of GnRH and PGE2, which act on G protein-coupled membrane receptors (GPCRs). A direct action on the PR–Src neurons is shown on the
left side of the diagram by progestins. GnRH and PGE2 on membrane GPCRs to activate second messenger-regulated several kinases. Progestins
also stimulate Src activity by either acting at the membrane or indirectly through releasing GnRH or directly activating PR. Src signaling through the
MAPK system occurs in the context of a multiprotein complex in which the PR acts as a scaffold to recruit an assortment of enzymatic effectors.
Activation of the Src–MAPK system results in rapid and transient membrane effects on ion channels and membrane receptors and delayed genomic
effects mediated through proteins such as CREB or STATs modulating gene transcription. Src–MAPK signaling also interacts with the classical path-
ways of PR action by phosphorylating Ser-294 in this protein, a reaction enhancing its transcriptional activity. The cross on the arrow indicates the
site of PP2 inhibitory effect on Src action. GnRH, gonadotropin-releasing hormone; PR, progesterone receptor; PGE2, prostaglandin estradiol; PK,
protein kinase.

effects of E2 and T secreted by the ovaries. During the last week brain activity (Hoffman et al., 2010; Kawakami and Sawyer,
of pregnancy, sexual receptivity was reported to increase, as 1959; Sawyer and Kawakami, 1959). The ‘early’ inhibition of
plasma P levels decrease and levels of E2 and T increase (Beyer chinning (observed across the first 4 days following mating)
and Rivaud, 1969; González-Mariscal et al., 1994). After a brief depends on ovulation and endocrine signals from the ovaries,
postpartum estrous, sexual receptivity continues to be inhibited and it is proposed to involve the simultaneous activation of the
during lactation (Beyer and Rivaud, 1969). PR and a decline in E2 receptor activation, the latter possibly
The proceptive scent-marking behavior called ‘chinning’ has being mediated by a temporary postovulatory decline in circu-
been intensively studied with respect to changes in its expres- lating E2 (Hoffman et al., 2010; Hoffman and González-
sion across the female reproductive cycle. Thus, immediately Mariscal, 2007). The long-term ‘luteal’ inhibition of chinning
after mating, chinning is inhibited proportionally to the inten- that occurs across pregnancy is associated with the formation
sity of sexual stimulation that the doe received (González- of the corpora lutea and the high circulating levels of P
Mariscal et al., 1997). This immediate (or ‘acute’) postmating (González-Mariscal et al., 1990). This inhibition is clearly
inhibition of chinning, which is accompanied by an overall mediated by PR activation (Hoffman and González-Mariscal,
decrease in ambulatory activity, occurs independently of ovula- 2006). Notably, in all of the aforementioned studies, low chin-
tion and any other contribution from the ovaries and is most ning frequency was associated with periods of reduced sexual
likely related to changes in electroencephalographic (EEG) receptivity.
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 73

1.02.1.4.3 Postpartum and Lactation in Rats nursing, and a chronic, persistent inhibition of chinning that
Intact cycling rats normally display a postpartum estrous was observed across lactation. Thus, chinning was immediately
between 12 and 14 h after parturition (Connor and Davis, inhibited after each daily nursing bout, in a manner qualita-
1980a,b). Proceptivity and lordosis are displayed during this tively similar to the immediate postmating inhibition of this
time, and mating can result in a new pregnancy (Gilbert, behavior (González-Mariscal et al., 1997). The immediate post-
1984). Thus, at about 3 h postpartum, around the time of suckling inhibition of chinning was observed regardless of the
ovulation, a weak lordosis response is observed; this response total number of pups nursed; that is, independently of the
becomes more intense and is accompanied by proceptive intensity of suckling stimulation. In contrast to the immediate
behaviors by about 10 h postpartum (Connor and Davis, postnursing inhibition of chinning, baseline chinning levels
1980a). A pregnancy resulting from mating during the post- across lactation were inversely related to the total number of
partum estrous normally lasts about 1 week longer than if the pups that were nursed (García-Dalman and González-
fertile mating had occurred during proestrous of a typical cycle Mariscal, 2012).
(Woodside et al., 1981). This is due to a delay in implantation
(Lataste, 1981) that can be induced at any moment by removal 1.02.1.4.5 Postpartum Anestrous and Lactation in Goats
of the suckling litter for 24 h (Zeilmaker, 1964). When the In goats displaying reproductive seasonality, the length of the
female is prevented from nursing on postpartum day 4 and postpartum anestrous depends on several factors, including
then mated, the resulting pregnancy lasts 22 or 23 days, only nursing of the young and season of parturitions (Delgadillo
1 day longer than the duration of a normal pregnancy. In this et al., 1998). The influence of these two factors on the duration
case, there is an overlap of lactation with the last week of the of postpartum anestrous was clearly demonstrated in goats
postpartum pregnancy. P sharply declines to very low levels 2 kidding on different seasons of the year, and nursing their
or 3 days before parturition, while E2 production is maintained young for 2, 30, or 90 days. In seasonal breeds of goats, partu-
or continuously rises. Thus, at parturition, plasma levels of E2 ritions generally occur during spring, and the postpartum anes-
and T are high while P levels are negligible. The initial stimula- trous can coincide with the seasonal anestrous, prolonging its
tory effect of E2 may be enhanced by a subsequent rise in P duration. This prolongation occurred because the onset of the
plasma levels which peak around 12 h postpartum (Connor ovulatory activity after parturitions can only be observed
and Davis, 1980b). when the animals enter the breeding season. As a consequence,
in goats kidding in winter (January) or spring (May), the dura-
1.02.1.4.4 Postpartum and Lactation in Rabbits tion of postpartum anestrous last about 200 and 100 days,
In the case of the rabbit, a brief period of estrous is observed respectively, and it is not modified by the duration of nursing,
during postpartum day 1. In one study, the proportion of i.e., 2, 30, or 90 days. By contrast, in females kidding in autumn
females that were sexually receptive declined to approximately (October), the duration of postpartum anestrous is shorter
40% on lactation day 8, and then to 10% on day 12 than in females kidding in January and May. Moreover, in these
(Hammond and Marshall, 1925). Beyer and Rivaud (1969) cases the duration of nursing modifies the length of the post-
studied female rabbits daily during their first and second lacta- partum anestrous. Therefore, the postpartum anestrous lasts
tions. Hammond and Marshall (1925) also observed an around 46 days in goats kidding in autumn, and this duration
intense postpartum heat in the first days of lactation. Following is not modified by the nursing the young for 2 or 30 days. On
this postpartum estrous, sexual receptivity showed an overall the contrary, this postpartum anestrous lasted 82 days in
decrease, but was still observed in about 50% of the females goats nursing their young for 90 days (Delgadillo et al.,
at some point (Beyer and Rivaud, 1969). In this study, 1998). Interestingly, in nonnursing goats, it seems that the level
a possible correlation between the display of receptivity and of milk production does not modify the length of postpartum
the number of young in the litter was not explored. Yet, the anestrous. Indeed, females of the Saanen breed produce more
fact that some lactating rabbits displayed lordosis behavior milk than Anglo-Nubian ones, but the length of postpartum
suggests that significant plasma levels of E2 and T are main- anestrous does not differ between breeds (Freitas et al.,
tained across days 10 and 25 of lactation. Withdrawal of suck- 2004). These results show that in goats displaying a reproduc-
ling induces a surge of LH and FSH stimulating both follicular tive seasonality, the season of parturition has an important
growth and E2 secretion. This latter observation might explain influence on the time of onset of ovulatory activity following
why the omission of one suckling episode (amounting to the delivery. The effect of nursing is observed only in females
doe’s separation from the pups for 44 h, as female rabbits nor- kidding during their breeding season.
mally nurse only once per day) allows the recovery of sexual
receptivity in 24 h. This phenomenon, called ‘biostimulation’
1.02.1.5 Female Sexual Motivation in Rats and Goats
(preventing the doe from nursing for at least 44 h), was
observed even after nursing was subsequently resumed In this section we focus mostly on rats, a species in which the
(Bonanno, 1999). In fact, biostimulation is a strategy used by study of sexual motivation has been approached by different
rabbit breeders to improve the fertility of lactating does methodologies that reflects the difficulty in defining this
(Theau-Clément, 1998). concept (for reviews, see Agmo, 1999; Pfaus, 1999). We will
Lactation is also marked by a dramatic reduction in chin- also include a description of the male effect in goats which
ning behavior (García-Dalman and González-Mariscal, 2012; clearly demonstrates the plastic capacity of the brain to rapidly
González-Mariscal et al., 1990). This behavior was found to respond to hormonal release with fast behavioral changes.
be inhibited on two levels during lactation: a transient, As described above early studies, Beach, (1976) identified
dramatic, and immediate inhibition that followed each the motivational or appetitive components of female sexual
74 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

behavior as those proceptive or soliciting behaviors (also called hamsters (see Paredes, 2009 and references therein). There
paracopulatory behaviors) displayed by the estrous female, to are also several indirect lines of evidence indicating that
attract and facilitate the sexual interaction with the male opioids are released during sexual behavior in both male
(Blaustein, 2009). These behaviors will be displayed, when and female rats (see Paredes, 2014; Pfaus and Gorzalka,
hormonal levels are adequate, because the male rat is an incen- 1987 for reviews). Therefore, it is not surprising that the
tive stimulus for the female (Agmo, 2003). That is, the female reward state induced by mating in male and female rats is
does not need to associate the male with a previous learning mediated by opioids. Systemic administration of the opioid
experience or a rewarding sexual encounter to approach the antagonist naloxone blocks the CPP induced by mating in
male and display solicitation and proceptive behaviors because males (Agmo and Berenfeld, 1990; Miller and Baum, 1987)
the male is an unconditional incentive. When tested under and females (Paredes and Martínez, 2001). Together with
seminatural or natural conditions, it is the female that usually the data on the effects of administration of naloxone in
initiates contact with the male by approaching and running different brain regions before mating and CPP, it has been
away from him (McClintock and Adler, 1978). This type of suggested that in males and females there is a common opioid
solicitation pattern appears to be the most effective way to acti- system mediating sexual reward in which the MPOA plays
vate pursuit and mating by the male because, under seminat- a crucial role (review in Paredes, 2014). The release of opioids
ural or natural conditions, 90% of the intromissions during sexual behavior, which would induce a CPP, ensures
followed a female solicitation (McClintock and Adler, 1978). that the behavior will be repeated for the simple reason that
This ability to control the rate of the sexual interaction has it is rewarding, favoring the survival of the species. This can
also been observed in laboratory conditions using different be achieved through the social decision-making network
methodologies including pressing a lever to copulate with that regulates adaptive behaviors. This network is formed by
a male rat (Bermant, 1961), mating with tethered males two circuits important in behavioral regulation. One is the
(Edwards and Pfeifle, 1983), copulating in bilevel chambers mesolimbic reward system and the other is the social behav-
(Pfaus et al., 1999), or mating in cages with one hole (Erskine, ioral network composed of several brain regions, including
1989; Paredes and Alonso, 1997) or four holes (Ismail et al., the MPOA, that regulate different forms of social behaviors
2009; Yang and Clemens, 1997) in which the female is able which were already present in early vertebrates (O’Connell
to pace the sexual interaction. and Hofmann, 2011).

1.02.1.5.1 Paced Mating and Female Sexual Motivation 1.02.1.5.2 The Importance of Testing Conditions in the
When females pace the sexual interaction, fewer intromissions Analysis of Sexual Motivation
are needed to induce the progestational state of pregnancy; rats In natural and seminatural conditions, males and females
also show a higher release of prolactin and have a higher have a different temporal pattern of copulation. They mate
number of pups than females not pacing the sexual interaction. in groups when estrous is synchronized in females that live
The behavioral and physiological differences between paced together interacting with several males (McClintock and
and nonpaced mating have been extensively described before Adler, 1978; McClintock and Anisko, 1982; McClintock
(Blaustein and Erskine, 2002; Erskine, 1989). A series of studies et al., 1982). Males and females mate with several partners,
by different groups have shown that the ability to control or changing them in the middle of copulation without an
pace the sexual interaction induces a reward state as evaluated ordered sequence of behavioral events (as is characteristic of
by conditioned place preference (CPP) (Ismail et al., 2009; laboratory testing). A female can receive an ejaculation after
Paredes, 2009; Paredes and Fernández-Guasti, 2008). The few intromissions if she encounters a male that has already
combination of paced mating and CPP reduces the influence experienced intromissions with other females and a male
of confounding variables and enhances the rewarding aspects can achieve intromissions with one or several females. What
of mating. The different factors that contribute to the reward is clear is that both sexes experience a similar amount of copu-
state induced by paced mating have been described before lation (McClintock and Anisko, 1982; McClintock et al.,
(Paredes, 2009; Paredes and Fernández-Guasti, 2008) and 1982), controlling the sexual interaction and assuring that
will not be discussed in the present chapter. One key aspect the rewarding state induced by mating outlasts the execution
is that only when males or females control the sexual interac- of the behavior, making sex rewarding for both sexes
tion they develop a reward state. When males and females (Martínez and Paredes, 2001).
mate without controlling the sexual interaction, no CPP is A recent series of studies have evaluated rat sexual
observed (Martínez and Paredes, 2001) indicative that under behavior in a seminatural environment where groups of three
this conditions the sexual interaction does not induce a reward males and four females are housed together for 8 days and
state. There is one study showing that females that mated evaluating sociosexual interactions during the entire period
without pacing the sexual interaction with only one male in of behavioral estrous. Contrary to what has been shown in
three different conditioning sessions showed CPP (Meerts laboratory conditions female proceptive behaviors were not
and Clark, 2007). In our laboratory, we directly tested whether reduced and rejection behavior did not increase at the end
females that mated with the same male along the conditioning of behavioral estrous. In the same study, it was shown that
sessions developed CPP and no changes were observed, that is, females showed a preference for a male with whom they dis-
no CPP was induced. Possible explanations for these discrep- played the higher number of lordosis. The preferred male also
ancies have been presented before (Camacho et al., 2009). had more intromissions and ejaculated more frequently than
It is also well established that sexual behavior induces the avoided male, the one with which the female displayed
a reward state as evaluated by CPP in male rats, mice, and the lowest number of lordosis. Yet, even from the avoided
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 75

male, the female received mounts, intromissions, and ejacula- 50


tions (Chu and Agmo, 2014). Using the same seminatural

Goats showing estrous (%)


environment, it was shown that male reproductive success is 40
related to the intensity of copulatory behavior, for example,
the higher the number of intromissions and ejaculations the 30
higher the likelihood of producing offspring (Chu et al.,
20
2015). A surprising observation in these series of studies is
that females responded with lordosis to almost every mount
10
by the male from the beginning of estrous until the end of
it (Chu and Agmo, 2014) indicating that the female suddenly
0
changes from a state of complete nonreceptivity to full recep-
tivity and then abruptly changes back to nonreceptivity with 0 5 10 15
males exceptionally mounting the female before the mount Days after buck introduction
was able to induce lordosis (Chu and Agmo, 2015). Accord-
ing to Agmo and colleagues, their results indicate that initia- Figure 3 Daily proportions of goats displaying estrous behavior. One
tion and termination of copulatory behavior are tightly group of females was exposed to male goats subjected to natural
associated with sudden modifications of the amount of changes in day length ( ), and were therefore in sexual rest. The other
female paracopulatory behaviors and male pursuit of the group of females was exposed to males goats subjected to 2.5 months
female. Thus, from the beginning to the end of behavioral of artificially long days (16 h of light per day) from November 1 fol-
lowed by natural day length ( ), and were therefore sexually active.
estrous, the female is fully receptive displaying lordosis in
Females and males were put in contact during the seasonal anestrous.
response to every mount by the male (Chu and Agmo,
Adapted from Delgadillo, J.A., Flores, J.A., Veliz, F.G., Hernandez, H.F.,
2015). These observations are at odds with what has tradi- Duarte, G., Vielma, J., Poindron, P., Chemineau, P., Malpaux, B., 2002.
tionally been observed in laboratory conditions in which Induction of sexual activity in lactating anovulatory female goats using
only one male and one female are tested together and where male goats treated only with artificially long days. J. Anim. Sci. 80 (11),
the transition from nonreceptivity to receptivity (and vice 2780–2786.
versa) have been reported to be gradual. These studies remind
us that the context in which the sexual interaction is observed
can determine how the behavior is expressed. They also exem- the male effect. Indeed, 4-ethyloctanal is the olfactory molecule
plify the complexity of the sexual behavior response, which that activates the GnRH pulse generator (Murata et al., 2014).
can have different expressions depending on the context in Nonetheless, the proportion of goats that ovulate is lower
which it is displayed. when exposed to olfactory cues alone than after exposure to
whole males (Claus et al., 1990; Walkden et al., 1993). This
1.02.1.5.3 The Male Effect difference may be related to the intensity of the sexual behavior
In goats, introduction of males into a group of seasonally of males. In fact, exposure of males to 2.5 months of long days,
anovulatory goats stimulated and synchronizes their sexual starting on November 1, followed by natural photoperiod,
activity during the anestrous season (Chemineau, 1983; Ott stimulates the secretion of T (Figure 4) and increases their
et al., 1980; Shelton, 1960). This phenomenon, where socio-
sexual relationships between males and females impact the
occurrence of ovulation, is known as the ‘male effect’ 20
LD
(Figure 3). The exposure of females to males induces a rapid
increase of LH secretion followed by estrous behavior and
ovulation (Bedos et al., 2014; Hawken and Martin, 2012; 15
Vielma et al., 2009). A variable proportion of goats exposed
to males show estrous behavior at the first ovulation induced 10
by males, between 2 and 5 days after the first contact with
them. Most of these goats display a short ovulatory cycle of
5–7 days of duration, followed by a second ovulation 5
7–10 days after the introduction of males. These second
ovulations are associated with estrous behavior and an 0
estrous cycle of normal duration (Chemineau, 1987; Nov Jan Mar May Jul Sep
Delgadillo et al., 2002; Walkden-Brown et al., 1999).
The endocrine and sexual responses of female goats to the Figure 4 Plasma testosterone concentrations (mean  SEM) in male
male effect depend on various sensory cues emitted by males, goats subjected to natural changes in day length ( ), or to 2.5 months
of artificially long days (LD: 16 h of light per day) from November 1 fol-
including their odor but also the level of male sexual behavior
lowed by natural day length ( ). Blood samples were taken once
(Delgadillo et al., 2009). In fact, the exposure of anovulatory
a week. Stars indicate significant differences between groups. Adapted
goats to hair of males obtained during their breeding season from Delgadillo, J.A., Flores, J.A., Veliz, F.G., Hernandez, H.F., Duarte,
stimulates the activity of GnRH neurons, LH secretion, and G., Vielma, J., Poindron, P., Chemineau, P., Malpaux, B., 2002. Induc-
ovulation (Claus et al., 1990; Hawken and Martin, 2012; tion of sexual activity in lactating anovulatory female goats using male
Okamura et al., 2010). Thus, the olfactory cues are considered goats treated only with artificially long days. J. Anim. Sci. 80 (11),
the primary signal involved in the response of female goats to 2780–2786.
76 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

odor; and sexual behavior during the nonbreeding season, i.e., added groups exposed to banana scent or to a sexually
in March and April. These photo-stimulated, sexually active experienced male but where physical contact was not
males are more efficient than untreated, sexually inactive possible. The proliferation marker BrdU was administered 1 h
ones, to induce ovulation in anovulatory goats. In fact, the before the behavioral test, at the end of the behavioral test
photo-stimulated males induce ovulation in 80–100 % of (which lasts 1 h), and the third injection was administered
goats, whereas only around 10% of females ovulated when 1 h later. Females were then sacrificed at different time
exposed to the untreated males. The higher ovulatory response intervals to evaluate the number of new cells and the number
of goats exposed to the photo-stimulated males seems to be of new neurons. We found that 15 days after mating there is
related with the intensity of the sexual behavior, rather than an increase in the number of new cells in the granular cell
with more intense male olfactory cues. Indeed, goats exposed layer of the AOB in females that paced the sexual interaction
to photo-stimulated males that also displayed an intense sexual in comparison to the other groups, indicating that pacing
behavior, displayed the preovulatory LH surge, and ovulated in behavior promotes an increase in cell proliferation that
>95% of cases. By contrast, goats exposed to the sedated, induces a higher density of new cells in the AOB (Corona
photo-stimulated males to prevent the display of sexual et al., 2011). Indeed when subjects were sacrificed 2 days
behavior showed neither a preovulatory LH surge nor ovula- after the behavioral test, an increase in the number of cells in
tion, even though the males were emitting a strong odor the rostral migratory stream was found in females that
(Martínez-Alfaro et al., 2014). Interestingly, the continuous mated, either pacing or not the sexual interaction, suggesting
presence of the photo-stimulated males prevents the display that sexual behavior induces cell proliferation in females
of seasonal anestrous (Delgadillo et al., 2015). Altogether, independently of the possibility of pacing the sexual
these results strongly suggest that in goats, the intensity of interaction. When females were sacrificed 45 days after the
sexual behavior displayed by males is a critical factor in the behavioral test no differences in the number of cells were
responsiveness of females to the male effect. Finally, it is impor- found in the OB, but a significantly higher number of
tant to mention that the use of photo-stimulated males to neurons were observed in the AOB in the groups that mated
control the sexual activity of females is a sustainable breeding (Corona et al., 2016). In the studies described above, females
technique for goat’s reproduction. mated only once. Yet, if the stimulus is repeated, that is, if
rats are injected with BrdU on the day of the first behavioral
1.02.1.5.4 Neurogenesis and Sexual Behavior test and then mate in three more sessions, a higher number
The incorporation of new neurons that are produced in the of cells are observed in the granular layer of the AOB in
adult brain (neurogenesis) is well documented in several comparison to those females that mated only once.
species including humans. Neurogenesis in the adult mostly Moreover, an increase in the number of cells is observed in
occurs within two brain regions: the subventicular zone other layers of the AOB and MOB in females that paced the
(SVZ) of the lateral ventricles, where the new cells and neurons sexual interaction. When the number of neurons was
migrate to the olfactory bulb system, and the subgranular zone analyzed, a significant increase was observed in both the AOB
(SGZ) of the dentate gyrus of the hippocampus. Several studies and MOB suggesting that repeated paced mating promotes
have clearly demonstrated that social and reproductive behav- the arrival of more newborn neurons to the OB (Arzate et al.,
iors modulate adult neurogenesis (see Peretto and Paredes, 2013). Together, these results indicate that sexual behavior in
2014 and references therein). For example, in female mice, females can induce long-lasting plastic changes in the
where recognition of the sexual partner is essential for the olfactory bulbs. Future studies will need to determine what
display of sexual behavior and maintenance of pregnancy, the function of the new neurons is.
the simple exposure to male pheromones increases the prolif- Adult neurogenesis has also been found in sheep (Brus
eration in the SVZ, the number of new cells in the MOB et al., 2013, 2014). However, the contribution of this phenom-
(Mak et al., 2007) and in the AOB (Oboti et al., 2011). Chronic enon within the context of sexual behavior in ruminants is not
exposure (28 days) to male mice soiled bedding produces known. The only result reported so far in sheep is that the intro-
a significant increase in the number of new neurons in the duction of the ram into a group of anestrous ewes induces an
AOB of adult females that is dependent on vomeronasal organ increase in cell proliferation in the hippocampus (Hawken
activity (Oboti et al., 2009). The newborn granule cells that et al., 2009). However, the functional significance of this effect
reach the AOB are functional, activated by male pheromones, is unclear.
and are important in mate recognition processes such as in
the Bruce effect (reviewed in Oboti et al., 2011; Peretto and
Paredes, 2014). 1.02.1.6 Conclusions
Few studies have evaluated the effect of mating in females
on neurogenesis. For example, in hamsters, new cells generated Most of the research analyzing the interaction between
in the adult are activated by Fos expression after mating hormones, receptors, and cellular mechanisms that trigger
(Huang and Bittman, 2002). Since the olfactory bulbs and female sexual behavior has been done in rodents, rabbits,
the processing of chemosensory relevant cues are crucial for and goats. There are obvious differences, for example, in the
a successful reproduction, we investigated if mating itself can length of the estrous cycle. In the case of the rat, ovulation
increase the number of new cells or neurons along the occurs in response to hormonal changes with E2 and P secre-
SVZ-OB system. To that aim we allowed female rats to pace tion at regular intervals. In the rabbit, ovulation occurs
the sexual interaction or to mate in a condition where they following coitus, being reflexive or induced, while in
were not able to control the sexual stimulation received. We goats the reproductive seasonality is mainly controlled by
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 77

photoperiod. There are also similarities between species References


regarding the physiological control, as well as differences in
behavioral patterns. For example, during estrous, female Agmo, A., 1999. Sexual motivation – an inquiry into events determining the occur-
rodents display a series of stereotyped behaviors including rence of sexual behavior. Behav. Brain Res. 105 (1), 129–150.
Agmo, A., 2003. Unconditioned sexual incentive motivation in the male Norway rat
attractivity, proceptivity, and receptivity in order to attract (Rattus norvegicus). J. Comp. Psychol. 117 (1), 3–14.
a male and copulate. Proceptive female rabbits display circling Agmo, A., Berenfeld, R., 1990. Reinforcing properties of ejaculation in the male rat:
of the male, elevation, and lateral displacements of the tail role of opioids and dopamine. Behav. Neurosci. 104 (1), 177–182.
(flagging), and a scent-marking behavior called ‘chinning.’ In Alexander, G., Signoret, J.P., Hafez, E.S.E., 1980. Sexual, maternal and neonatal
behavior. In: Hafez, E.S.E. (Ed.), Reproduction in Farm Animals. Lea and Febiger,
the female goat (and in most ruminants), motor activity
Philadelphia, pp. 304–334.
increases when estrous starts. Goats express vocalizations and Arteaga, L., Bautista, A., Martínez-Gomez, M., Nicolas, L., Hudson, R., 2008. Scent
male-type courtship behavior and female–female reciprocal marking, dominance and serum testosterone levels in male domestic rabbits.
mounting to attract males and stimulate sexual activity. It is Physiol. Behav. 94 (3), 510–515.
evident then that although the behavioral patterns are different Arzate, D.M., Portillo, W., Corona, R., Paredes, R.G., 2013. Repeated paced mating
promotes the arrival of more newborn neurons in the main and accessory olfactory
among the three species, they are directed to attract the male. bulbs of adult female rats. Neuroscience 232, 151–160.
On the other hand, the receptive response in rats and rabbits Bai, W.L., Rowan, B.G., Allgood, V.E., OMalley, B.W., Weigel, N.L., 1997. Differential
is similar. Both display a clear lordosis posture in response to phosphorylation of chicken progesterone receptor in hormone-dependent and
male mounts, while female goats remain immobile when in ligand-independent activation. J. Biol. Chem. 272 (16), 10457–10463.
Bakker, J., Baum, M.J., 2000. Neuroendocrine regulation of GnRH release in induced
estrous to facilitate mounting by the male.
ovulators. Front. Neuroendocrinol. 21 (3), 220–262.
With respect to effects of hormones on estrous behavior, E2 Ball, J., 1940. The effect of testosterone on the sex behavior of female rats. J. Comp.
is important for the three species but the role of P is different. Psychol. 29 (2), 151–165.
Estrous behavior in female rats is triggered by the combined Baum, M.J., 1990. The ferret as a model for studying the sexual differentiation of
action of E2 and P while in rabbit, E2 is the principal gonadal behavioral and reproductive function. J. Exp. Zool. Suppl. 4, 213–224.
Beach, F.A., 1942. Analysis of factors involved in the arousal, maintenance and
hormone that promotes estrous behavior. In female goats, an manifestation of sexual excitement in male animals. Phychosomatic Med. 4,
increase in E2 concentration is observed before the expression 173–198.
of sexual behavior, and exposure to P may be necessary for the Beach, F.A., 1976. Sexual attractivity, proceptivity, and receptivity in female mammals.
full display of sexual behavior at the onset of the breeding Horm. Behav. 7 (1), 105–138.
Beato, M., 1987. Induction of transcription by steroid hormones. Biochim. Biophys.
season. Intact cycling rats normally display a postpartum
Acta 910 (2), 95–102.
estrous between 12 and 14 h after parturition while in the Bedos, M., Duarte, G., Flores, J.A., Fitz-Rodriguez, G., Hernandez, H., Vielma, J.,
case of the rabbit, a brief period of estrous is observed during Fernandez, I.G., Chemineau, P., Keller, M., Delgadillo, J.A., 2014. Two or 24 h of
postpartum day 1. In goats displaying reproductive seasonal- daily contact with sexually active males results in different profiles of LH secretion
ity, the length of postpartum anestrous depends on several that both lead to ovulation in anestrous goats. Domest. Anim. Endocrinol. 48,
93–99.
factors, including nursing of the young and season of Belanger, A., Cusan, L., Caron, S., Barden, N., Dupont, A., 1981. Ovarian progestins,
parturition. androgens and estrogen throughout the 4-Day estrous-cycle in the rat. Biol.
It is also evident that much research is needed in the field. A Reprod. 24 (3), 591–596.
number of studies, many of which have been carried out in Bermant, G., 1961. Response latencies of female rats during sexual intercourse.
Science 133, 1771–1773.
rodent species, demonstrate that nonsteroidal hormones
Beyer, C., Canchola, E., Larsson, K., 1981. Facilitation of lordosis behavior in the
participate in the regulation of female sexual behavior. These ovariectomized estrogen primed rat by dibutyryl cAMP. Physiol. Behav. 26 (2),
include GnRH, OT, leptin, NO, etc. As well, the cellular mech- 249–251.
anisms involved in the regulation of female sexual behavior by Beyer, C., Cruz, M.L., Rivaud, N., 1969. Persistence of sexual behavior in
a variety of intracellular pathways including PKA, PKC, PKG, ovariectomized-adrenalectomized rabbits treated with cortisol. Endocrinology 85
(4), 790–793.
and the Src/PR/MAPK system have been studied with detailed Beyer, C., de la Torre, L., Larsson, K., Perez-Palacios, G., 1975. Synergistic actions of
in the rat. Future studies need to address the importance of estrogen and androgen on the sexual behavior of the castrated male rabbit. Horm.
these systems in the control of female sexual behavior in other Behav. 6 (3), 301–306.
species. Beyer, C., Fernandez-Guasti, A., Rodríguez-Manzo, G., 1982. Induction of female
sexual-behavior by Gtp in ovariectomized estrogen primed rats. Physiol. Behav. 28
Sexual motivation has mostly been studied in rats where it
(6), 1073–1076.
has been shown that the context in which the animals are tested Beyer, C., González-Flores, O., García-Juarez, M., González-Mariscal, G., 2003. Non-
can modify the expression of the behavior. A clear example of ligand activation of estrous behavior in rodents: cross-talk at the progesterone
the interaction between the context and behavior is also receptor. Scand. J. Psychol. 44 (3), 221–229.
observed in the ‘male effect’ in goats. This is mediated by olfac- Beyer, C., González-Flores, O., González-Mariscal, G., 1995. Ring A reduced
progestins potently stimulate estrous behavior in rats: paradoxical effect through
tion but clearly shows the capacity of the brain to respond to the progesterone receptor. Physiol. Behav. 58 (5), 985–993.
external stimuli modifying the interaction between hormones, Beyer, C., González-Flores, O., González-Mariscal, G., 1997. Progesterone receptor
receptors, and cellular mechanism in different brain regions to participates in the stimulatory effect of LHRH, prostaglandin E2, and cyclic AMP on
modify behavior. The analysis of these interactions requires, lordosis and proceptive behaviours in rats. J. Neuroendocrinol. 9 (8), 609–614.
Beyer, C., González-Mariscal, G., 1986. Elevation in hypothalamic cyclic AMP as
without doubt, a comparative approach.
a common factor in the facilitation of lordosis in rodents: a working hypothesis.
Ann. N.Y. Acad. Sci. 474, 270–281.
Beyer, C., González-Mariscal, G., 1991. Effects of progesterone and natural progestins
Acknowledgments in brain. Reprod. Growth Dev. 71, 199–208.
Beyer, C., González-Mariscal, G., Eguibar, J.R., Gomora, P., 1988. Lordosis facilitation
in estrogen primed rats by intrabrain injection of pregnanes. Pharmacol. Biochem.
Research from Paredes supported by CONACYT: Fronteras 374; Behav. 31 (4), 919–926.
DGAPA: IN210215.
78 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

Beyer, C., Komisaruk, B., 1971. Effects of diverse androgens on estrous behavior, Camacho-Arroyo, I., Perez-Palacios, G., Pasapera, A.M., Cerbon, M.A., 1994. Intra-
lordosis reflex, and genital tract morphology in rat. Horm. Behav. 2 (3), 217–225. cellular progesterone receptors are differentially regulated by sex steroid hormones
Beyer, C., Mcdonald, P., 1973. Hormonal-control of sexual-behavior in female rabbit. in the hypothalamus and the cerebral cortex of the rabbit. J. Steroid Biochem. Mol.
Adv. Reprod. Physiol. 6, 185–219. Biol. 50 (5–6), 299–303.
Beyer, C., McDonald, P., Vidal, N., 1970a. Failure of 5-alpha-dihydrotestosterone to Camacho, F.J., García-Horsman, P., Paredes, R.G., 2009. Hormonal and testing
elicit estrous behavior in the ovariectomized rabbit. Endocrinology 86 (4), conditions for the induction of conditioned place preference by paced mating.
939–941. Horm. Behav. 56 (4), 410–415.
Beyer, C., Morali, G., Larsson, K., Soderstein, P., 1976. Steroid regulation of sexual Campfield, L.A., Smith, F.J., Burn, P., 1996. The OB protein (leptin) pathway–a link
behavior. J. Steroid Biochem. 7 (11–12), 1171–1186. between adipose tissue mass and central neural networks. Horm. Metab. Res. 28
Beyer, C., Rivaud, N., 1969. Sexual behavior in pregnant and lactating domestic (12), 619–632.
rabbits. Physiol. Behav. 4 (5), 753–757. Carrillo-Martínez, G.E., Gomora-Arrati, P., González-Arenas, A., Morimoto, S.,
Beyer, C., Vidal, N., 1971. Inhibitory action of Mer-25 on androgen-induced oestrous Camacho-Arroyo, I., González-Flores, O., 2011. Role of progesterone receptors
behaviour in ovariectomized rabbit. J. Endocrinol. 51 (2), 401–412. during postpartum estrus in rats. Horm. Behav. 59 (1), 37–43.
Beyer, C., Vidal, N., Mijares, A., 1970b. Probable role of aromatization in the induction Chemineau, P., 1983. Effect on oestrus and ovulation of exposing creole goats to the
of estrus behavior by androgens in the ovariectomized rabbit. Endocrinology 87 (6), male at three times of the year. J. Reprod. Fertil. 67 (1), 65–72.
1386–1389. Chemineau, P., 1987. Possibilities for using bucks to stimulate ovarian and estrous
Billings, H.J., Katz, L.S., 1998. Threshold dose of estradiol for inducing sexual cycles in anovulatory goats – a review. Livest. Prod. Sci. 17 (2), 135–147.
receptivity in ovariectomized French-Alpine goats. Appl. Anim. Behav. Sci. 57 Chemineau, P., Daveau, A., Maurice, F., Delgadillo, J.A., 1992. Seasonality of estrus
(1–2), 109–115. and ovulation is not modified by subjecting female alpine goats to a tropical
Blaustein, J.D., 2009. Feminine reproductive behavior and physiology in rodents: photoperiod. Small Ruminant Res. 8 (4), 299–312.
integration of hormonal, behavioral, and environmental influences. Horm. Brain Cheng, Y.J., Karavolas, H.J., 1973. Conversion of progesterone to 5 alpha-pregnane-
Behav. 1–5, 67–107, 2nd Edition. 3,20-dione and 3 alpha-hydroxy-5 alpha-pregnan-20-one by rat medical basal
Blaustein, J.D., Erskine, M.S., 2002. Feminine Sexual Behavior: Cellular Integration of hypothalami and the effects of estradiol and stage of estrous cycle on the
Hormonal and Afferent Information in the Rodent Brain, vol. 1. Academic Press, conversion. Endocrinology 93 (5), 1157–1162.
New York, pp. 139–214. Chu, H.P., Etgen, A.M., 1997. A potential role of cyclic GMP in the regulation of
Blaustein, J.D., Finkbohner, R., Delville, Y., 1987. Estrogen-induced and estrogen- lordosis behavior of female rats. Horm. Behav. 32, 125–132.
facilitated female rat sexual behavior is not mediated by progestin receptors. Chu, H.P., Etgen, A.M., 1999. Ovarian hormone dependence of alpha(1)-adrenoceptor
Neuroendocrinology 45 (2), 152–159. activation of the nitric oxide-cGMP pathway: relevance for hormonal facilitation of
Blaustein, J.M., Mani, S.K., 2007. Feminine sexual behavior from neuroendocrine and lordosis behavior. J. Neurosci. 19 (16), 7191–7197.
molecular neurobiological perspectives. In: Handbook of Neurochemistry and Chu, H.P., Morales, J.C., Etgen, A.M., 1999. Cyclic GMP may potentiate lordosis
Molecular Neurobiology. Springer, New York, pp. 95–149. behaviour by progesterone receptor activation. J. Neuroendocrinol. 11 (2),
Boling, J.L., Blandau, R.J., 1939. The estrogen-progesterone induction of mating 107–113.
responses in the spayed female rat. Endocrinology 25 (3), 359–364. Chu, X., Agmo, A., 2014. Sociosexual behaviours in cycling, intact female rats
Bonanno, A., Alabiso, M., DiGrigoli, A., Alicata, M.L., 1999. Effect of change of cage (Rattus norvegicus) housed in a seminatural environment. Behaviour 151,
and/or 44 h mother-litter separation on productivity of non-receptive lactating 1143–1148.
rabbit does. Preliminary investigation. World Rabbit Sci. 7, 107–111. Chu, X., Agmo, A., 2015. Sociosexual behaviors during the transition from non-
Boonyaratanakornkit, V., Bi, Y., Rudd, M., Edwards, D.P., 2008. The role and receptivity to receptivity in rats housed in a seminatural environment. Behav.
mechanism of progesterone receptor activation of extra-nuclear signaling pathways Processes 113, 24–34.
in regulating gene transcription and cell cycle progression. Steroids 73 (9–10), Chu, X., Guarraci, F.A., Agmo, A., 2015. Sociosexual behaviors and reproductive
922–928. success of rats (Rattus norvegicus) in a seminatural environment. Physiol. Behav.
Boonyaratanakornkit, V., Edwards, D.P., 2004. Receptor mechanisms of rapid extra- 151, 46–54.
nuclear signalling initiated by steroid hormones. Essays Biochem. 40, 105–120. Claus, R., Over, R., Dehnhard, M., 1990. Effect of male odor on Lh-secretion and the
Brinton, R.D., Thompson, R.F., Foy, M.R., Baudry, M., Wang, J., Finch, C.E., induction of ovulation in seasonally anestrous goats. Anim. Reprod. Sci. 22 (1),
Morgan, T.E., Pike, C.J., Mack, W.J., Stanczyk, F.Z., Nilsen, J., 2008. Proges- 27–38.
terone receptors: form and function in brain. Front. Neuroendocrinol. 29 (2), Conaway, C.H., 1971. Ecological adaptation and mammalian reproduction. Biol.
313–339. Reprod. 4 (3), 239–247.
Brown, M.T., Cooper, J.A., 1996. Regulation, substrates and functions of src. Biochim. Connor, J.R., Davis, H.N., 1980a. Postpartum estrus in Norway rats. II. Physiology.
Biophys. Acta 1287 (2–3), 121–149. Biol. Reprod. 23 (5), 1000–1006.
Brus, M., Meurisse, M., Gheusi, G., Keller, M., Lledo, P.M., Levy, F., 2013. Dynamics Connor, J.R., Davis, H.R., 1980b. Postpartum estrus in Norway rats. I. Behavior. Biol.
of olfactory and hippocampal neurogenesis in adult sheep. J. Comp. Neurol. 521 Reprod. 23 (5), 994–999.
(1), 169–188. Contreras, J.L., Beyer, C., 1979. Polygraphic analysis of mounting and ejaculation in
Brus, M., Meurisse, M., Keller, M., Levy, F., 2014. Interactions with the young down- the New Zealand white-rabbit. Physiol. Behav. 23 (5), 939–943.
regulate adult olfactory neurogenesis and enhance the maturation of olfactory Corona, R., Larriva-Sahd, J., Paredes, R.G., 2011. Paced-mating increases the
neuroblasts in sheep mothers. Front. Behav. Neurosci. 8, 53. number of adult new born cells in the internal cellular (granular) layer of the
Caba, M., Beyer, C., Gonzalez-Mariscal, G., Morrell, J.I., 2003a. Immunocytochemical accessory olfactory bulb. PLoS One 6 (5), e19380.
detection of estrogen receptor-alpha in the female rabbit forebrain: topography and Corona, R., Retana-Marquez, S., Portillo, W., Paredes, R.G., February 26, 2016.
regulation by estradiol. Neuroendocrinology 77 (3), 208–222. Sexual behavior increases cell proliferation in the rostral migratory stream and
Caba, M., Rovirosa, M.J., Beyer, C., González-Mariscal, G., 2003b. Immunocyto- promotes the differentiation of the new cells into neurons in the accessory olfactory
chemical detection of progesterone receptor in the female rabbit forebrain: bulb of female rats. Front. Neurosci. 10.
distribution and regulation by oestradiol and progesterone. J. Neuroendocrinol. 15 Czaja, J.A., Goldfoot, D.A., Karavolas, H.J., 1974. Comparative facilitation and
(9), 855–864. inhibition of lordosis in the guinea pig with progesterone, 5-alpha-pregnane-
Caldwell, J.D., Jirikowski, G.F., Greer, E.R., Pedersen, C.A., 1989. Medial preoptic 3,2-dione, or 3-alpha-hydroxy-5-alpha-pregnan-20-one. Horm. Behav. 5 (3),
area oxytocin and female sexual receptivity. Behav. Neurosci. 103 (3), 655–662. 261–274.
Caldwell, J.D., Johns, J.M., Faggin, B.M., Senger, M.A., Pedersen, C.A., 1994. Dawson, T.M., Snyder, S.H., 1994. Gases as biological messengers - nitric-oxide and
Infusion of an oxytocin antagonist into the medial preoptic area prior to proges- carbon-monoxide in the brain. J. Neurosci. 14 (9), 5147–5159.
terone inhibits sexual receptivity and increases rejection in female rats. Horm. Delgadillo, J.A., Flores, J.A., Hernandez, H., Poindron, P., Keller, M., Fitz-
Behav. 28 (3), 288–302. Rodriguez, G., Duarte, G., Vielma, J., Fernandez, I.G., Chemineau, P., 2015.
Calka, J., 2006. The role of nitric oxide in the hypothalamic control of LHRH and Sexually active males prevent the display of seasonal anestrus in female goats.
oxytocin release, sexual behavior and aging of the LHRH and oxytocin neurons. Horm. Behav. 69, 8–15.
Folia Histochem. Cytobiol. 44 (1), 3–12. Delgadillo, J.A., Flores, J.A., Veliz, F.G., Hernandez, H.F., Duarte, G., Vielma, J.,
Camacho-Arroyo, I., Mendoza-Rodriguez, C.A., Mendoza-Garces, L., Vanoye-Carlo, A., Poindron, P., Chemineau, P., Malpaux, B., 2002. Induction of sexual activity in
Cerbon, M., 2007. Progesterone receptor mRNA expression and distribution in the lactating anovulatory female goats using male goats treated only with artificially
female rabbit brain. J. Steroid Biochem. Mol. Biol. 104 (3–5), 100–104. long days. J. Anim. Sci. 80 (11), 2780–2786.
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 79

Delgadillo, J.A., Flores, J.A., Villarreal, O., Flores, M.J., Hoyos, G., Chemineau, P., Glaser, J.H., Rubin, B.S., Barfield, R.J., 1983. Onset of the receptive and proceptive
Malpaux, B., 1998. Length of postpartum anestrus in goats in subtropical Mexico: components of feminine sexual behavior in rats following the intravenous admin-
effect of season of parturition and duration of nursing. Theriogenology 49 (6), istration of progesterone. Horm. Behav. 17 (1), 18–27.
1209–1218. Gomez-Brunet, A., Santiago-Moreno, J., del Campo, A., Malpaux, B., Chemineau, P.,
Delgadillo, J.A., Gelez, H., Ungerfeld, R., Hawken, P.A., Martin, G.B., 2009. The ‘male Tortonese, D.J., Gonzalez-Bulnes, A., Lopez-Sebastian, A., 2008. Endogenous
effect’ in sheep and goats–revisiting the dogmas. Behav. Brain Res. 200 (2), circannual cycles of ovarian activity and changes in prolactin and melatonin
304–314. secretion in wild and domestic female sheep maintained under a long-day
Dempsey, E.W., Hertz, R., Young, W.C., 1936. The experimental induction of oestrus photoperiod. Biol. Reprod. 78 (3), 552–562.
(sexual receptivity) in the normal and ovariectomized guinea pig. Am. J. Physiol. González-Flores, O., Beyer, C., Gomora-Arrati, P., García-Juarez, M., Lima-
116 (1), 201–209. Hernandez, F.J., Soto-Sanchez, A., Etgen, A.M., 2010. A role for Src kinase in
Domínguez-Ordonez, R., García-Juarez, M., Lima-Hernandez, F.J., Gomora- progestin facilitation of estrous behavior in estradiol-primed female rats. Horm.
Arrati, P., Blaustein, J.D., González-Flores, O., 2015. Sexual receptivity Behav. 58 (2), 223–229.
facilitated by unesterified estradiol: dependence on estrogen and progestin González-Flores, O., Beyer, C., Lima-Hernandez, F.J., Gomora-Arrati, P., Gomez-
receptors and priming dose of estradiol benzoate. Behav. Neurosci. 129 (6), Camarillo, M.A., Hoffman, K., Etgen, A.M., 2007. Facilitation of estrous behavior
777–788. by vaginal cervical stimulation in female rats involves alpha1-adrenergic receptor
Duarte, G., Flores, J.A., Malpaux, B., Delgadillo, J.A., 2008. Reproductive seasonality activation of the nitric oxide pathway. Behav. Brain Res. 176 (2), 237–243.
in female goats adapted to a subtropical environment persists independently of González-Flores, O., Etgen, A.M., 2004. The nitric oxide pathway participates in
food availability. Domest. Anim. Endocrinol. 35 (4), 362–370. estrous behavior induced by progesterone and some of its ring A-reduced
Dupon, C., Kim, M.H., 1973. Peripheral plasma levels of testosterone, androste- metabolites. Horm. Behav. 45 (1), 50–57.
nedione, and oestradiol during the rat oestrous cycle. J. Endocrinol. 59 (3), González-Flores, O., Gomora-Arrati, P., García-Juarez, M., Gomez-Camarillo, M.A.,
653–654. Lima-Hernandez, F.J., Beyer, C., Etgen, A.M., 2009. Nitric oxide and ERK/MAPK
Edwards, D.A., Pfeifle, J.K., 1983. Hormonal control of receptivity, proceptivity and mediation of estrous behavior induced by GnRH, PGE2 and db-cAMP in rats.
sexual motivation. Physiol. Behav. 30 (3), 437–443. Physiol. Behav. 96 (4–5), 606–612.
Erskine, M.S., 1989. Solicitation behavior in the estrous female rat: a review. Horm. González-Flores, O., Guerra-Araiza, C., Cerbon, M., Camacho-Arroyo, I.,
Behav. 23 (4), 473–502. Etgen, A.M., 2004a. The 26S proteasome participates in the sequential inhi-
Etgen, A.M., Acosta-Martinez, M., 2003. Participation of growth factor signal trans- bition of estrous behavior induced by progesterone in rats. Endocrinology 145
duction pathways in estradiol facilitation of female reproductive behavior. Endo- (5), 2328–2336.
crinology 144 (9), 3828–3835. González-Flores, O., Ramírez-Orduna, J.M., Lima-Hernandez, F.J., García-Juarez, M.,
Everett, J.W., 1939. Spontaneous persistent estrus in a strain of albino rats. Endo- Beyer, C., 2006. Differential effect of kinase A and C blockers on lordosis facili-
crinology 25, 123–127. tation by progesterone and its metabolites in ovariectomized estrogen-primed rats.
Faber, L.E., Saffran, J., Chen, T.J., Leavitt, W.W., 1976. Mammalian progesterone Horm. Behav. 49 (3), 398–404.
receptors: biosynthesis, structure and nuclear binding. Curr. Top. Mol. Endocrinol. González-Flores, O., Shu, J., Camacho-Arroyo, I., Etgen, A.M., 2004b. Regulation of
4, 68–84. lordosis by cyclic 30 ,50 -guanosine monophosphate, progesterone, and its 5alpha-
Fabre-Nys, C., Gelez, H., 2007. Sexual behavior in ewes and other domestic rumi- reduced metabolites involves mitogen-activated protein kinase. Endocrinology 145
nants. Horm. Behav. 52 (1), 18–25. (12), 5560–5567.
Faivre, E.J., Daniel, A.R., Hillard, C.J., Lange, C.A., 2008. Progesterone receptor rapid González-Mariscal, G., Albonetti, M., Cuamatzi, E., Beyer, C., 1997. Transitory inhi-
signaling mediates serine 345 phosphorylation and tethering to specificity protein 1 bition of scent-marking by copulation in male and female rabbits. Anim. Behav. 53,
transcription factors. Mol. Endocrinol. 22 (4), 823–837. 323–333.
Fox, A.S., Olster, D.H., 2000. Effects of intracerebroventricular leptin administration on González-Mariscal, G., Díaz-Sanchez, V., Melo, A.I., Beyer, C., Rosenblatt, J.S.,
feeding and sexual behaviors in lean and obese female Zucker rats. Horm. Behav. 1994. Maternal behavior in New Zealand white rabbits: quantification of somatic
37 (4), 377–387. events, motor patterns, and steroid plasma levels. Physiol. Behav. 55 (6),
Frank, A.H., Fraps, R.M., 1945. Induction of estrus in the ovariectomized golden 1081–1089.
hamster. Endocrinology 37, 357–361. González-Mariscal, G., González-Flores, O., Beyer, C., 1989. Intrahypothalamic
Freeman, M., 1994. The neuroendocrine control of the ovarian cycle in the rat. In: injection of RU486 antagonizes the lordosis induced by ring A-reduced progestins.
Knobil, E., Neill, J.D. (Eds.), The Physiology of Reproduction. Raven Press, New Physiol. Behav. 46 (3), 435–438.
York, pp. 613–658. González-Mariscal, G., Melo, A.I., Beyer, C., 1993. Progesterone, but not LHRH or
Freitas, W.F., Rondina, D., Nogueira, D.M., Simplicio, A.A., 2004. Post-partum prostaglandin E2, induces sequential inhibition of lordosis to various lordogenic
anoestrus in Anglo-Nubian and Saanen goats raised in semi-arid of North- agents. Neuroendocrinology 57 (5), 940–945.
eastern Brazil. Livest. Prod. Sci. 90 (2–3), 219–226. González-Mariscal, G., Melo, A.I., Zavala, A., Beyer, C., 1990. Variations in chin-
García-Dalman, C., González-Mariscal, G., 2012. Major role of suckling stimulation for marking behavior of New Zealand female rabbits throughout the whole repro-
inhibition of estrous behaviors in lactating rabbits: acute and chronic effects. Horm. ductive cycle. Physiol. Behav. 48 (2), 361–365.
Behav. 61 (1), 108–113. González-Mariscal, G., Melo, A.I., Zavala, A., Beyer, C., 1992. Chin-marking behavior
García-Juarez, M., Beyer, C., Gomora-Arrati, P., Domínguez-Ordonez, R., in male and female New Zealand rabbits – onset, development, and activation by
Lima-Hernandez, F.J., Eguibar, J.R., Galicia-Aguas, Y.L., Etgen, A.M., González- steroids. Physiol. Behav. 52 (5), 889–893.
Flores, O., 2013. Lordosis facilitation by leptin in ovariectomized, estrogen- Gorzalka, B.B., Whalen, R.E., 1977. The effects of progestins, mineralocorticoids,
primed rats requires simultaneous or sequential activation of several protein glucocorticoids, and steroid solubility on the induction of sexual receptivity in rats.
kinase pathways. Pharmacol. Biochem. Behav. 110, 13–18. Horm. Behav. 8 (1), 94–99.
García-Juarez, M., Beyer, C., Gomora-Arrati, P., Lima-Hernandez, F.J., Domínguez- Goy, R.W., Phoenix, C., Young, W.C., 1968. Inhibitory action of the corpus luteum on
Ordonez, R., Eguibar, J.R., Etgen, A.M., González-Flores, O., 2012. The nitric the hormonal induction of estrous behavior in the guinea pig. Gen. Comp. Endo-
oxide pathway participates in lordosis behavior induced by central administration of crinol. 6, 267–275.
leptin. Neuropeptides 46 (1), 49–53. Greco, B., Allegretto, E.A., Tetel, M.J., Blaustein, J.D., 2001. Coexpression of ER beta
García-Juarez, M., Beyer, C., Soto-Sanchez, A., Domínguez-Ordonez, R., Gomora- with ER alpha and progestin receptor proteins in the female rat forebrain: effects of
Arrati, P., Lima-Hernandez, F.J., Eguibar, J.R., Etgen, A.M., González-Flores, O., estradiol treatment. Endocrinology 142 (12), 5172–5181.
2011. Leptin facilitates lordosis behavior through GnRH-1 and progestin receptors Greene, G.L., Gilna, P., Waterfield, M., Baker, A., Hort, Y., Shine, J., 1986. Sequence
in estrogen-primed rats. Neuropeptides 45 (1), 63–67. and expression of human estrogen receptor complementary DNA. Science 231
García-Segura, L.M., Baetens, D., Naftolin, F., 1986. Synaptic remodelling in (4742), 1150–1154.
arcuate nucleus after injection of estradiol valerate in adult female rats. Brain Gudermuth, D.F., Newton, L., Daels, P., Concannon, P., 1997. Incidence of sponta-
Res. 366 (1–2), 131–136. neous ovulation in young, group-housed cats based on serum and faecal
Gebbie, F.E., Forsyth, I.A., Arendt, J., 1999. Effects of maintaining solstice light and concentrations of progesterone. J. Reprod. Fertil. Suppl. 51, 177–184.
temperature on reproductive activity, coat growth, plasma prolactin and melatonin Guerra-Araiza, C., Gomora-Arrati, P., García-Juarez, M., Armengual-Villegas, A.,
in goats. J. Reprod. Fertil. 116 (1), 25–33. Miranda-Martinez, A., Lima-Hernandez, F.J., Camacho-Arroyo, I., González-
Gilbert, A.N., 1984. Postpartum and lactational estrus: a comparative analysis in Flores, O., 2009. Role of progesterone receptor isoforms in female sexual
rodentia. J. Comp. Psychol. 98 (3), 232–245. behavior induced by progestins in rats. Neuroendocrinology 90 (1), 73–81.
80 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

Guerra-Araiza, C., Villamar-Cruz, O., González-Arenas, A., Chavira, R., Camacho- Ichikawa, S., Sawada, T., Nakamura, Y., Morioka, H., 1974. Ovarian secretion of
Arroyo, I., 2003. Changes in progesterone receptor isoforms content in the rat pregnane compounds during the estrous cycle and pregnancy in rats. Endocri-
brain during the oestrous cycle and after oestradiol and progesterone treatments. nology 94 (6), 1615–1620.
J. Neuroendocrinol. 15 (10), 984–990. Imwalle, D.B., Katz, L.S., 2004. Divergent roles for estrogens and androgens in the
Hall, N.R., Luttge, W.G., 1977. Diencephalic sites responsive to prostaglandin E2 expression of female goat sexual behavior. Horm. Behav. 46 (1), 54–58.
facilitation of sexual receptivity in estrogen-primed ovariectomized rats. Brain Res. Ismail, N., Gelez, H., Lachapelle, I., Pfaus, J.G., 2009. Pacing conditions contribute to
Bull. 2 (3), 203–207. the conditioned ejaculatory preference for a familiar female in the male rat. Physiol.
Hall, N.R., Luttge, W.G., Berry, R.B., 1975. Intracerebral prostaglandin E2: effects Behav. 96 (2), 201–208.
upon sexual behavior, open field activity and body temperature in ovariectomized Jainudeen, M.R., Hafez, E.S.E., 1993. Sheep and goats. In: Hafez, E.S.E. (Ed.),
female rats. Prostaglandins 10 (5), 877–888. Reproduction in Farm Animals, sixth ed. Lea & Febiger, Philadelphia, pp. 330–342.
Hammond, J., Marshall, F., 1925. Reproduction in the Rabbit. Oliver and Boyd, Johnson, W.A., Billiar, R.B., Little, B., 1976. Progesterone and 5alpha-reduced
Edinburgh. metabolites: facilitation of lordosis behavior and brain uptake in female
Hardy, D., 1970. Behavior of the female rat during pregnancy, pseudopregnancy, hamsters. Behav. Biol. 18 (4), 489–497.
lactation and following ovariectomy. Horm. Behav. 1, 235–245. Jones, S.L., Ismail, N., King, L., Pfaus, J.G., 2012. The effects of chronic adminis-
Hardy, D.F., DeBold, J.F., 1972. Effects of coital stimulation upon behavior of the tration of testosterone propionate with or without estradiol on the sexual behavior
female rat. J. Comp. Physiol. Psychol. 78 (3), 400–408. and plasma steroid levels of aged female rats. Endocrinology 153 (12),
Hawken, P.A., Jorre, T.J., Rodger, J., Esmaili, T., Blache, D., Martin, G.B., 2009. 5928–5939.
Rapid induction of cell proliferation in the adult female ungulate brain (Ovis aries) Jones, S.L., Pfaus, J.G., 2014. Sensitization of sexual behaviors in ovariectomized
associated with activation of the reproductive axis by exposure to unfamiliar males. Long-Evans rats is induced by a subthreshold dose of estradiol benzoate and
Biol. Reprod. 80 (6), 1146–1151. attenuated by repeated copulation. Horm. Behav. 66 (4), 655–662.
Hawken, P.A., Martin, G.B., 2012. Sociosexual stimuli and gonadotropin-releasing Kalivas, P.W., Peters, J., Knackstedt, L., 2006. Animal models and brain circuits in
hormone/luteinizing hormone secretion in sheep and goats. Domest. Anim. drug addiction. Mol. Interv. 6 (6), 339–344.
Endocrinol. 43 (2), 85–94. Karavolas, H.J., Bertics, P.J., Hodges, D., Rudie, N., 1984. Progesterone processing
Henniawati, Restall, B.J., Scaramuzzi, R.J., 1995. Effect of season on LH secretion in by neuroendocrine structures. In: Celotti, F., Naftolin, F., Martini, L. (Eds.),
ovariectomized Australian cashmere does. J. Reprod. Fertil. 103 (2), 349–356. Metabolism of Hormonal Steroids in the Neuroendocrine Structures. Raven Press,
Hilliard, J., 1973. Corpus-luteum function in guinea pigs, hamsters, rats, mice and New York, pp. 149–170.
rabbits. Biol. Reprod. 8 (2), 203–221. Katz, L.S., 2007. Sexual behavior of domesticated ruminants. Horm. Behav. 52 (1),
Hilliard, J., Archibald, D., Sawyer, C.H., 1963. Gonadotropic activation of prevulatory 56–63.
synthesis and release of progestin in the rabbit. Endocrinology 72, 59–66. Kawakami, M., Sawyer, C.H., 1959. Some hormone-induced alterations in thresh-
Hilliard, J., Eaton Jr., L.W., 1971. Estradiol-17 beta, progesterone and 20-alpha- olds of neuroendocrine function and brain activity in the rabbit. Anat. Rec. 133
hydroxypregn-4-en-3-one in rabbit ovarian venous plasma. II. From mating through (2), 398.
implantation. Endocrinology 89 (2), 522–527. Knowles, R.G., Palacios, M., Palmer, R.M., Moncada, S., 1989. Formation of nitric
Hilliard, J., Scaramuzzi, R.J., Pang, C., Penardi, R., Sawyer, C.H., 1968. Testosterone oxide from L-arginine in the central nervous system: a transduction mechanism for
secretion by the rabbit ovary in vivo. Endocrinology 94 (5), 267–271. stimulation of the soluble guanylate cyclase. Proc. Natl. Acad. Sci. U.S.A. 86 (13),
Hlinak, Z., 1993. Social recognition in ovariectomized and estradiol-treated female 5159–5162.
rats. Horm. Behav. 27 (2), 159–166. Komisaruk, B.R., 1972. Induction of lordosis in ovariectomized rats by stimulation of
Hlinak, Z., Madlafou, J., 1969. A quantitative study of synergistic action of oestradiol the vaginal cervix: hormonal and neural interrelationships. UCLA Forum Med. Sci.
and progesterone in inducing oestrous behaviour of ovariectomized rat. Physiol. 15, 127–135.
Bohemoslov. 18 (5–6), 485–486. Komisaruk, B.R., Diakow, C., 1973. Lordosis reflex intensity in rats in relation to the
Hlinak, Z., Madlafousek, J., 1972. Positive and negative effects of progesterone on the estrous cycle, ovariectomy, estrogen administration and mating behavior. Endo-
precopulatory behavior of ovariectomized rats. Act. Nerv. Super. (Praha) 14 (3), crinology 93 (3), 548–557.
170–171. Kow, L.M., Pfaff, D.W., 1975. Induction of lordosis in female rats: two modes of
Hoffman, K.L., Basurto, E., 2013. One-trial object recognition memory in the domestic estrogen action and the effect of adrenalectomy. Horm. Behav. 6 (3), 259–276.
rabbit (Oryctolagus cuniculus) is disrupted by NMDA receptor antagonists. Behav. Kubli-Garfias, C., Whalen, R.E., 1977. Induction of lordosis behavior in female rats by
Brain Res. 250, 62–73. intravenous administration of progestins. Horm. Behav. 9 (3), 380–386.
Hoffman, K.L., Basurto, E., 2014. Clozapine and glycinamide prevent MK-801-induced Kuehn, R.E., Beach, F.A., 1963. Quantitative measurement of sexual receptivity in
deficits in the novel object recognition (NOR) test in the domestic rabbit (Orycto- female rats. Behaviour 21, 282–299.
lagus cuniculus). Behav. Brain Res. 271, 203–211. Kuiper, G.G., Enmark, E., Pelto-Huikko, M., Nilsson, S., Gustafsson, J.A., 1996.
Hoffman, K.L., Decasa, D.M.H., Ruiz, M.E.B., González-Mariscal, G., 2010. Scent Cloning of a novel receptor expressed in rat prostate and ovary. Proc. Natl. Acad.
marking by the male domestic rabbit (Oryctolagus cuniculus) is stimulated by an Sci. U.S.A. 93 (12), 5925–5930.
object’s novelty and its specific visual or tactile characteristics. Behav. Brain Res. Lange, C.A., Shen, T., Horwitz, K.B., 2000. Phosphorylation of human progesterone
207 (2), 360–367. receptors at serine-294 by mitogen-activated protein kinase signals their degra-
Hoffman, K.L., González-Mariscal, G., 2006. Progesterone receptor activation signals dation by the 26S proteasome. Proc. Natl. Acad. Sci. U.S.A. 97 (3), 1032–1037.
behavioral transitions across the reproductive cycle of the female rabbit. Horm. Lataste, F., 1981. Des variations de duree de la gestation chez les mammiferes et des
Behav. 50 (1), 154–168. circonstances qui determinent ces variations. Mem. Sot. Biol. 43, 21–31.
Hoffman, K.L., González-Mariscal, G., 2007. Relevance of ovarian signaling for the Lawler, D.F., Johnston, S.D., Hegstad, R.L., Keltner, D.G., Owens, S.F., 1993.
early behavioral transition from estrus to pregnancy in the female rabbit. Horm. Ovulation without cervical stimulation in domestic cats. J. Reprod. Fertil. Suppl. 47,
Behav. 52 (4), 531–539. 57–61.
Horwitz, K.B., Wei, L.L., Francis, M.D., 1986. Structural analyses of progesterone Lebas, D.F., Coudert, P., Rouvier, R., Rochambeua, H.D.E., 1986. The Rabbit
receptors. J. Steroid Biochem. 24 (1), 109–117. Husbandry, Health and Production. FAO Animal. Production and Health
Huang, H.H., Marshall, S., Meites, J., 1976. Induction of estrous cycles in old non- Series, Rome.
cyclic rats by progesterone, ACTH, ether stress or L-dopa. Neuroendocrinology Leonhardt, S.A., Boonyaratanakornkit, V., Edwards, D.P., 2003. Progesterone receptor
20 (1), 21–34. transcription and non-transcription signaling mechanisms. Steroids 68 (10–13),
Huang, H.H., Meites, J., 1975. Reproductive capacity of aging female rats. Neuro- 761–770.
endocrinology 17 (4), 289–295. Lima-Hernández, F.J., Beyer, C., Gomora-Arrati, P., García-Juárez, M., Encarnacion-
Huang, H.H., Steger, R.W., Bruni, J.F., Meites, J., 1978. Patterns of sex steroid and Sanchez, J.L., Etgen, A.M., González-Flores, O., 2012. Src kinase signaling
gonadotropin secretion in aging female rats. Endocrinology 103 (5), 1855–1859. mediates estrous behavior induced by 5beta-reduced progestins, GnRH, prosta-
Huang, L., Bittman, E.L., 2002. Olfactory bulb cells generated in adult male golden glandin E2 and vaginocervical stimulation in estrogen-primed rats. Horm. Behav.
hamsters are specifically activated by exposure to estrous females. Horm. Behav. 62 (5), 579–584.
41 (3), 343–350. Lindia, J.A., Katz, L.S., 2003. Estradiol and testosterone stimulation of sexual behavior in
Hudson, R., González-Mariscal, G., Beyer, C., 1990. Chin marking behavior, sexual ovariectomized female goats. Horm. Behav. 44, 62.
receptivity, and pheromone emission in steroid-treated, ovariectomized rabbits. Luttge, W.G., Jasper, T.W., 1977. Studies on the possible role of 2-OH-estradiol in the
Horm. Behav. 24 (1), 1–13. control of sexual behavior in female rats. Life Sci. 20 (3), 419–425.
Female Sexual Behavior in Rodents, Lagomorphs, and Goats 81

Mahesh, V.B., 1985. The dynamic interaction between steroids and gonadotro- Mori, Y., Kano, Y., 1984. Changes in plasma concentrations of LH, progesterone and
pins in the mammalian ovulatory cycle. Neurosci. Biobehav. Rev. 9 (2), oestradiol in relation to the occurrence of luteolysis, oestrus and time of ovulation in
245–260. the Shiba goat (Capra hircus). J. Reprod. Fertil. 72 (1), 223–230.
Mak, G.K., Enwere, E.K., Gregg, C., Pakarainen, T., Poutanen, M., Huhtaniemi, I., Mori, Y., Tanaka, M., Maeda, K., Hoshino, K., Kano, Y., 1987. Photoperiodic modi-
Weiss, S., 2007. Male pheromone-stimulated neurogenesis in the adult female fication of negative and positive feedback effects of oestradiol on LH secretion in
brain: possible role in mating behavior. Nat. Neurosci. 10 (8), 1003–1011. ovariectomized goats. J. Reprod. Fertil. 80 (2), 523–529.
Mani, S.K., Allen, J.M.C., Rettori, V., Mccann, S.M., Omalley, B.W., Clark, J.H., 1994. Moss, R.L., Foreman, M.M., 1976. Potentiation of lordosis behavior by intra-
Nitric-oxide mediates sexual-behavior in female rats. Proc. Natl. Acad. Sci. U.S.A. hypothalamic infusion of synthetic luteinizing hormone-releasing hormone.
91 (14), 6468–6472. Neuroendocrinology 20 (2), 176–181.
Mani, S.K., Oyola, M.G., 2012. Progesterone signaling mechanisms in brain and Moss, R.L., McCann, S.M., 1973. Induction of mating behavior in rats by luteinizing
behavior. Front. Endocrinol. (Lausanne) 3, 7. hormone-releasing factor. Science 181 (4095), 177–179.
Mani, S.K., Portillo, W., Reyna, A., 2009. Steroid hormone action in the brain: cross- Moss, R.L., Mccann, S.M., 1975. Action of luteinizing hormone-releasing factor (Lrf) in
talk between signalling pathways. J. Neuroendocrinol. 21 (4), 243–247. initiation of lordosis behavior in estrone-primed ovariectomized female rat.
Marrone, B.L., Rodriguez-Sierra, J.F., Feder, H.H., 1977. Lordosis: inhibiting effects Neuroendocrinology 17 (4), 309–318.
progesterone in the female rat. Horm. Behav. 8 (3), 391–402. Murata, K., Tamogami, S., Itou, M., Ohkubo, Y., Wakabayashi, Y., Watanabe, H.,
Martínez-Alfaro, J.C., Hernández, H., Flores, J.A., Duarte, G., Fitz-Rodríguez, G., Okamura, H., Takeuchi, Y., Mori, Y., 2014. Identification of an olfactory signal
Fernández, I.G., Bedos, M., Chemineau, P., Keller, M., Delgadillo, J.A., Vielma, J., molecule that activates the central regulator of reproduction in goats. Curr. Biol. 24
2014. Importance of intense male sexual behavior for inducing the preovulatory LH (6), 681–686.
surge and ovulation in seasonally anovulatory female goats. Theriogenology 82 (7), Myers, K., Poole, W.E., 1962. Oestrous cycles in rabbit Oryctolagus cuniculus (L.).
1028–1035. Nature 195 (4839), 358–359.
Martínez, I., Paredes, R.G., 2001. Only self-paced mating is rewarding in rats of both Mykytowy, R., 1965. Further observations on territorial function and histology of
sexes. Horm. Behav. 40 (4), 510–517. submandibular cutaneous (chin) glands in rabbit Oryctolagus cuniculus (L.). Anim.
McCann, S.M., 1977. Luteinizing-hormone-releasing hormone. N. Engl. J. Med. 296 Behav. 13 (4), 400–412.
(14), 797–802. Mykytowycz, R., 1962. Territorial function of chin gland secretion in rabbit, Oryctolagus
McClintock, M.K., Adler, N.T., 1978. The role of the female during copulation in wild cuniculus (L). Nature 193 (4817), 799.
and domestic Norway rats (Rattus norvegicus). Behaviour 67, 67–96. Mykytowycz, R., Hesterman, E.R., Gambale, S., Dudzinski, M.L., 1976. A comparison
McClintock, M.K., Anisko, J.J., 1982. Group mating among Norway rats. I. Sex of the effectiveness of the odors of rabbits, Oryctolagus cuniculus, in enhancing
differences in the pattern and neuroendocrine consequences of copulation. Anim. territorial confidence. J. Chem. Ecol. 2 (1), 13–24.
Behav. 30, 398–409. Naish, S.J., Ball, P., 1981. Catecholestrogens and induction of sexual-behavior in the
McClintock, M.K., Anisko, J.J., Adler, N.T., 1982. Group mating among Norway rats. II. ovariectomized rat. Neuroendocrinology 32 (4), 225–228.
The social dynamics of copulation: competition, cooperation, and mate choice. Nestler, E.J., Guitart, X., Ortiz, J., Trevisan, L., 1994. Second messenger and protein-
Anim. Behav. 30, 410–425. phosphorylation mechanisms underlying possible genetic vulnerability to alco-
McDonald, P.G., Vidal, N., Beyer, C., 1970. Sexual behavior in the ovariectomized holism. Ann. N.Y. Acad. Sci. 708, 108–1118.
rabbit after treatment with different amounts of gonadal hormones. Horm. Behav. Numan, M., Roach, J.K., del Cerro, M.C., Guillamon, A., Segovia, S., Sheehan, T.P.,
67, 161–172. Numan, M.J., 1999. Expression of intracellular progesterone receptors in rat brain
Meerts, S.H., Clark, A.S., 2007. Female rats exhibit a conditioned place preference for during different reproductive states, and involvement in maternal behavior. Brain
nonpaced mating. Horm. Behav. 51 (1), 89–94. Res. 830 (2), 358–371.
Melo, A.I., Chirino, R., Jímenez, A., Cuamatzi, E., Beyer, C., González- O’Connell, L.A., Hofmann, H.A., 2011. The vertebrate mesolimbic reward system and
Mariscal, G., 2008. Effect of forebrain implants of testosterone or estradiol social behavior network: a comparative synthesis. J. Comp. Neurol. 519 (18),
on scent-marking and sexual behavior in male and female rabbits. Horm. 3599–3639.
Behav. 54 (5), 676–683. Oboti, L., Savalli, G., Giachino, C., De Marchis, S., Panzica, G.C., Fasolo, A.,
Meyerson, B., 1972. Latency between intravenous-injection of progestins and Peretto, P., 2009. Integration and sensory experience-dependent survival of newly-
appearance of estrous behavior in estrogen-treated ovariectomized rats. Horm. generated neurons in the accessory olfactory bulb of female mice. Eur. J. Neurosci.
Behav. 3 (1), 1–9. 29 (4), 679–692.
Migliaccio, A., Di Domenico, M., Castoria, G., de Falco, A., Bontempo, P., Nola, E., Oboti, L., Schellino, R., Giachino, C., Chamero, P., Pyrski, M., Leinders-Zufall, T.,
Auricchio, F., 1996. Tyrosine kinase/p21ras/MAP-kinase pathway activation by Zufall, F., Fasolo, A., Peretto, P., 2011. Newborn interneurons in the accessory
estradiol-receptor complex in MCF-7 cells. Eur. Mol. Biol. Organ. J. 15 (6), olfactory bulb promote mate recognition in female mice. Front. Neurosci. 5, 113.
1292–1300. Okada, M.T.,Y., Mori, Y., 1998. Estradiol-dependency of sexual behavior manifestation
Migliaccio, A., Piccolo, D., Castoria, G., Di Domenico, M., Bilancio, A., Lombardi, M., at the post-LH surge period in ovariectomized goat. Jpn. J. Anim. Reprod. 44 (1),
Gong, W., Beato, M., Auricchio, F., 1998. Activation of the Src/p21ras/Erk 53–58.
pathway by progesterone receptor via cross-talk with estrogen receptor. EMBO J. Okamura, H., Murata, K., Sakamoto, K., Wakabayashi, Y., Ohkura, S., Takeuchi, Y.,
17 (7), 2008–2018. Mori, Y., 2010. Male effect pheromone tickles the gonadotrophin-releasing
Miller, R.L., Baum, M.J., 1987. Naloxone inhibits mating and conditioned place hormone pulse generator. J. Neuroendocrinol. 22 (7), 825–832.
preference for an estrous female in male rats soon after castration. Pharmacol. Osterlund, M., Kuiper, G.G., Gustafsson, J.A., Hurd, Y.L., 1998. Differential distribution
Biochem. Behav. 26 (4), 781–789. and regulation of estrogen receptor-alpha and -beta mRNA within the female rat
Milligan, S.R., 1978. The feedback of exogenous steroids on LH release and brain. Mol. Brain Res. 54 (1), 175–180.
ovulation in the intact female vole (Microtus agrestis). J. Reprod. Fertil. 54 (2), Ott, R.S., Nelson, D.R., Hixon, J.E., 1980. Effect of presence of the male on initiation of
309–311. estrous cycle activity of goats. Theriogenology 13 (2), 183–190.
Milligan, S.R., 1980. Effect of bromocriptine treatment on the ovulatory response to Palka, Y.S., Sawyer, C.H., 1966. The effects of hypothalamic implants of ovarian
oestradiol benzoate in the reflex ovulator, Microtus agrestis. J. Endocrinol. 84 (2), steroids on oestrous behaviour in rabbits. J. Physiol. 185 (2), 251–269.
315–316. Paredes, R.G., 2009. Evaluating the neurobiology of sexual reward. ILAR J. 50 (1),
Mills, T., Copland, A., Osteen, K., 1981. Factors affecting the postovulatory surge of 15–27.
FSH in the rabbit. Biol. Reprod. 25 (3), 530–535. Paredes, R.G., 2014. Opioids and sexual reward. Pharmacol. Biochem. Behav. 121,
Morali, G., Beyer, C., 1979. Neuroendocrine control of mammalian estrous behavior. 124–131.
In: Beyer, C. (Ed.), Endocrine Control of Sexual Behavior. Raven Press, New York, Paredes, R.G., Alonso, A., 1997. Sexual behavior regulated (paced) by the female
pp. 33–75. induces conditioned place preference. Behav. Neurosci. 111 (1), 123–128.
Morali, G., Beyer, C., 1992. Motor aspects of masculine sexual-behavior in rats and Paredes, R.G., Fernández-Guasti, A., 2008. Rewarding properties of mating. In:
rabbits. Adv. Study Behav. 21, 201–238. Méndez, M., Mondragón-Ceballos, R. (Eds.), Neural Mechanisms of Drugs of
Morali, G., Soto, M.A.P., Contreras, J.L., Arteaga, M., González-Vidal, M.D., Beyer, C., Abuse and Natural Reinforcers, pp. 159–170.
2003. Detailed analysis of the male copulatory motor pattern in mammals: Paredes, R.G., Martínez, I., 2001. Naloxone blocks place preference conditioning after
hormonal bases. Scand. J. Psychol. 44 (3), 279–288. paced mating in female rats. Behav. Neurosci. 115 (6), 1363–1367.
Moreines, J.K., Powers, J.B., 1977. Effects of acute ovariectomy on lordosis response Pathak, M.M., Patel, A.V., Jaiswal, R.S., Mehta, V.M., 1990. Circulating levels of
of female rats. Physiol. Behav. 19 (2), 277–283. progesterone and estrogen in cyclic goats. Indian J. Anim. Sci. 60 (7), 836–837.
82 Female Sexual Behavior in Rodents, Lagomorphs, and Goats

Patisaul, H.B., Aultman, E.A., Bielsky, I.F., Young, L.J., Wilson, M.E., 2003. Immediate Simerly, R.B., Chang, C., Muramatsu, M., Swanson, L.W., 1990. Distribution of
and residual effects of tamoxifen and ethynylestradiol in the female rat hypothal- androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ
amus. Brain Res. 978 (1–2), 185–193. hybridization study. J. Comp. Neurol. 294 (1), 76–95.
Peretto, P., Paredes, R.G., 2014. Social cues, adult neurogenesis, and reproductive Soares, M.J., Diamond, M., August 1982. Pregnancy and chin marking in the rabbit,
behavior. In: Mucignat-Caretta, C. (Ed.), Neurobiology of Chemical Communication, Oryctolagus cuniculus. Anim. Behav. 30, 941–943.
pp. 363–384. Boca Raton, FL. Staples, R.E., 1967. Behavioural induction of ovulation in the oestrous rabbit.
Pfaff, D., 1970. Nature of sex hormone effects on rat sex behavior: specificity of J. Reprod. Fertil. 13 (3), 429–435.
effects and individual patterns of response. J. Comp. Physiol. Psychol. 73 (3), Stoufflet, I., Caillol, M., 1988. Relation between circulating sex steroid concentrations
349–358. and sexual behaviour during pregnancy and post partum in the domestic rabbit.
Pfaff, D.S.-F.,M., Parhar, I.S., Lauber, A.H., McCarthy, M.M., Kow, L.M., 1993. GnRH J. Reprod. Fertil. 82 (1), 209–218.
neurons and other cellular and molecular mechanisms for simple mammalian Sutherland, S.R.D., Lindsay, D.R., 1991. Ovariectomized does do not require
reproductive behaviors. Recent Prog. Horm. Res. 49, 1–25. progesterone priming for estrous behavior. Reprod. Fertil. Dev. 3 (6), 679–684.
Pfaus, J.G., 1999. Revisiting the concept of sexual motivation. Annu. Rev. Sex Res. Tennent, B.J., Smith, E.R., Davidson, J.M., 1980. The effects of estrogen and
10, 120–156. progesterone on female rat proceptive behavior. Horm. Behav. 14 (1), 65–75.
Pfaus, J.G., Gorzalka, B.B., 1987. Opioids and sexual behavior. Neurosci. Biobehav. Terkel, J., 1988. Neuro-endocrine processes in the establishment of pregnancy and
Rev. 11 (1), 1–34. pseudopregnancy in rats. Psychoneuroendocrinology 13 (1–2), 5–28.
Pfaus, J.G., Smith, W.J., Coopersmith, C.B., 1999. Appetitive and consummatory Theau-Clément, M., Boiti, C., 1998. Biostimulation methods for breeding rabbit does:
sexual behaviors of female rats in bilevel chambers. I. A correlational and factor synthesis of the first results. World Rabbit Sci. 6 (1), 205–208.
analysis and the effects of ovarian hormones. Horm. Behav. 35 (3), 224–240. Thomas, S.M., Brugge, J.S., 1997. Cellular functions regulated by Src family kinases.
Pleim, E.T., DeBold, J.F., 1984. The relative effectiveness of progestins for facilitation Annu. Rev. Cell Dev. Biol. 13, 513–609.
and inhibition of sexual receptivity in hamsters. Physiol. Behav. 32 (5), 743–747. Thorburn, G.D., Schneider, W., 1972. The progesterone concentration in the plasma of
Powers, B., Valenstein, E.S., 1972. Sexual receptivity: facilitation by medial preoptic the goat during the oestrous cycle and pregnancy. J. Endocrinol. 52 (1), 23–36.
lesions in female rats. Science 175 (4025), 1003–1005. Tlachi-López, J.L., Eguibar, J.R., Fernández-Guasti, A., Lucio, R.A., 2012. Copulation
Powers, J.B., 1975. Anti-estrogenic suppression of the lordosis response in female and ejaculation in male rats under sexual satiety and the Coolidge effect. Physiol.
rats. Horm. Behav. 6 (4), 379–392. Behav. 106, 626–630.
Qiu, M., Olsen, A., Faivre, E., Horwitz, K.B., Lange, C.A., 2003. Mitogen-activated Veening, J.G., de Jong, T., Barendregt, H.P., 2010. Oxytocin-messages via the
protein kinase regulates nuclear association of human progesterone receptors. cerebrospinal fluid: behavioral effects; a review. Physiol. Behav. 101 (2), 193–210.
Mol. Endocrinol. 17 (4), 628–642. Veening, J.G., Olivier, B., 2013. Intranasal administration of oxytocin: behavioral and
Quadagno, D.M., McCullough, J., Langan, R., 1972. The effect of varying amounts of clinical effects, a review. Neurosci. Biobehav. Rev. 37 (8), 1445–1465.
exogenous estradiol benzoate on estrous behavior in the rat. Horm. Behav. 3 (3), Ventura-Aquino, E., Baños-Araujo, J., Fernández-Guasti, A., Paredes, R.G., 2016. An
175–179. unknown male increases sexual incentive motivation and partner preference:
Rachman, I.M., Pfaff, D.W., Cohen, R.S., 1996. NADPH diaphorase activity and nitric further evidence for the Coolidge effect in female rats. Physiol. Behav. 158,
oxide synthase immunoreactivity in lordosis-relevant neurons of the ventromedial 54–59.
hypothalamus. Brain Res. 740 (1–2), 291–306. Vielma, J., Chemineau, P., Poindron, P., Malpaux, B., Delgadillo, J.A., 2009. Male
Ramirez-Orduna, J.M., Lima-Hernandez, F.J., García-Juarez, M., González-Flores, O., sexual behavior contributes to the maintenance of high LH pulsatility in anestrous
Beyer, C., 2007. Lordosis facilitation by LHRH, PGE2 or db-cAMP requires acti- female goats. Horm. Behav. 56 (4), 444–449.
vation of the kinase A signaling pathway in estrogen primed rats. Pharmacol. Wade, G.N., Feder, H.H., 1972. Effects of several pregnane and pregnene steroids on
Biochem. Behav. 86 (1), 169–175. estrous behavior in ovariectomized, estrogen-primed guinea pigs. Physiol. Behav. 9
Reynolds, E., 1971. Urination as a social response in mice. Nature 234 (5330), (5), 773–775.
481–483. Wade, G.N., Harding, C.F., Feder, H.H., 1973. Neural uptake of (1,2-3H)progesterone
Rodríguez-Manzo, G., Cruz, M.L., Beyer, C., 1986. Facilitation of lordosis behavior in in ovariectomized rats, guinea pigs and hamsters: correlation with species differ-
ovariectomized estrogen-primed rats by medial preoptic implantation of 5 beta, 3 ences in behavioral responsiveness. Brain Res. 61, 357–367.
beta, pregnanolone: a ring A reduced progesterone metabolite. Physiol. Behav. 36 Wade, G.N., Lempicki, R.L., Panicker, A.K., Frisbee, R.M., Blaustein, J.D., 1997.
(2), 277–281. Leptin facilitates and inhibits sexual behavior in female hamsters. Am. J. Physiol.
Rodríguez-Sierra, J.F., Crowley, W.R., Komisaruk, B.R., 1977. Induction of lordosis 272 (4 Pt 2), R1354–R1358.
responsiveness by vaginal stimulation rats is independent of anterior or posterior Walkden-Brown, S.W., Martin, G.B., Restall, B.J., 1999. Role of male-female inter-
pituitary hormones. Horm. Behav. 8 (3), 348–355. action in regulating reproduction in sheep and goats. J. Reprod. Fertil. 243–257.
Rodríguez-Sierra, J.F., Komisaruk, B.R., 1977. Effects of prostaglandin E2 and Walkden, D., Klugman, K.P., Vally, S., Naidoo, P., 1993. Urinary tract infection with
indomethacin on sexual behavior in the female rat. Horm. Behav. 9 (3), 281–289. Corynebacterium urealyticum in South Africa. Eur. J. Clin. Microbiol. Infect. Dis. 12
Rodríguez-Sierra, J.F., Komisaruk, B.R., 1982. Common hypothalamic sites for acti- (1), 18–24.
vation of sexual receptivity in female rats by LHRH, PGE2 and progesterone. Whalen, R.E., Gorzalka, B.B., 1972. The effects of progesterone and its metabolites on
Neuroendocrinology 35 (5), 363–369. the induction of sexual receptivity in rats. Horm. Behav. 3 (3), 221–226.
Sawyer, C.H., Kawakami, M., 1959. Characteristics of behavioral and electroen- Whalen, R., Yahr, P., Luttge, W.G., 1985. The role of metabolism in hormonal control
cephalographic after-reactions to copulation and vaginal stimulation in the female of sexual behavior. In: Adler, N., Pfaff, D.W., Goy, R.W. (Eds.), Handbook of
rabbit. Endocrinology 65 (4), 622–630. Behavioral NeurobiologyPlenum Press, New York and London, 7, 55, pp.
Sawyer, C.H., Markee, J.E., 1959. Estrogen facilitation of release of pituitary ovulating 609–663.
hormone in the rabbit in response to vaginal stimulation. Endocrinology 65, 614–621. Woodside, B., Wilson, R., Chee, P., Leon, M., 1981. Resource partitioning during
Schrader, W.T., Birnbaumer, M.E., Hughes, M.R., Weigel, N.L., Grody, W.W., reproduction in the Norway rat. Science 211 (4477), 76–77.
O’Malley, B.W., 1981. Studies on the structure and function of the chicken Yang, L.Y., Clemens, L.G., 1997. Function of intromissions on intromission-return
progesterone receptor. Recent Prog. Horm. Res. 37, 583–633. latency of female rats during paced sexual behavior. Physiol. Behav. 61 (6),
Schulze, H.G., Gorzalka, B.B., 1991. Oxytocin effects on lordosis frequency and 889–894.
lordosis duration following infusion into the medial pre-optic area and ventromedial Yaschine, T., Mena, F., Beyer, C., 1967. Gonadal hormones and mounting behavior in
hypothalamus of female rats. Neuropeptides 18 (2), 99–106. the female rabbit. Am. J. Physiol. 213 (4), 867–872.
Shearer, M.K., Katz, L.S., 2006. Female-female mounting among goats stimulates Young, W., 1961. In: Sex and Internal Secretions. Williams & Wilkins Co., Balti-
sexual performance in males. Horm. Behav. 50 (1), 33–37. more, p. 52.
Shelton, M., 1960. Influence of the presence of a male goat on the initiation of estrous Zeilmaker, G.H., 1964. Quantitative studies on the effect of the suckling stimulus on
cycling and ovulation of Angora does. J. Anim. Sci. 19 (2), 368–375. blastocyst implantation in the rat. Acta Endocrinol. (Copenh) 46, 483–492.
Shughrue, P.J., Lane, M.V., Merchenthaler, I., 1997. Comparative distribution of Zemlan, F.P., Adler, N.T., 1977. Hormonal control of female sexual behavior in the rat.
estrogen receptor-alpha and -beta mRNA in the rat central nervous system. Horm. Behav. 9 (3), 345–357.
J. Comp. Neurol. 388 (4), 507–525. Zucker, I., 1966. Facilitatory and inhibitory effects of progesterone on sexual responses
Shughrue, P.J., Scrimo, P.J., Merchenthaler, I., 1998. Evidence for the colocalization of sprayed guinea pigs. J. Comp. Physiol. Psychol. 62 (3), 376–381.
of estrogen receptor-beta mRNA and estrogen receptor-alpha immunoreactivity in Zucker, I., 1968. Biphasic effects of progesterone on sexual receptivity in female
neurons of the rat forebrain. Endocrinology 139 (12), 5267–5270. guinea pig. J. Comp. Physiol. Psychol. 65 (3p1), 472–478.
1.03 Parental Behavior
G González-Mariscal, Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala,
México
M Caba, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Veracruz, México
KL Hoffman and AI Melo, Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala,
Tlaxcala, México
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by G. González-Mariscal, C.H. Kinsley, volume 1, pp. 109–138, Ó 2009, Elsevier Inc.

1.03.1 Introduction 84
1.03.2 Behaviors at Parturition: Combined Actions of Hormones and Somatosensory Stimuli 84
1.03.3 Postpartum Activities 85
1.03.3.1 Nursing: Variability Rules 85
1.03.3.2 Maternal Behavior in Nonlactating Females: What Does It Reveal? 85
1.03.4 Neural Control 86
1.03.4.1 Cortex, Trigeminal Complex, and Olfactory Circuit 86
1.03.4.2 Septum and BNST 86
1.03.4.3 MPOA and Its Connections 86
1.03.4.3.1 Studies Using Lesion, Inactivation, or Deafferentation Techniques 86
1.03.4.3.2 Studies Using Implantation of Hormones and Detection of Their Receptors 87
1.03.4.4 Midbrain Tegmentum, PVN, and Habenular Complex 87
1.03.4.5 Sampling the Active Parental Brain 88
1.03.4.5.1 The Expression of Immediate Early Genes 88
1.03.4.5.2 Other Methods 88
1.03.5 Impact of Maternal Behavior on the Mother: Effects on Neuroanatomy, Cognition, and Emotional Reactivity 89
1.03.5.1 Theoretical Context 89
1.03.5.2 Fear and Anxiety Regulation 89
1.03.5.2.1 Behaviors Affected 89
1.03.5.2.2 Brain Mechanisms 89
1.03.5.3 Regulation of Cognitive Effects 89
1.03.5.3.1 Learning and Memory 89
1.03.5.3.2 Neuroanatomical Changes Observed in the Maternal Brain 89
1.03.6 Timing: A Little-Explored Dimension of Maternal Care 89
1.03.6.1 A ‘Healthy Clock’ Is Necessary for Ovulation and Pregnancy 99
1.03.6.2 Circadian Timing of Parturition 101
1.03.6.3 Circadian Rhythms in Breast and Breast Milk 101
1.03.6.4 Daily Rhythms in the Mother during Lactation 102
1.03.6.4.1 Locomotor Behavior 102
1.03.6.4.2 Core Body Temperature 102
1.03.6.4.3 Rhythms of PER1 Protein in Neuroendocrine Cells 102
1.03.6.4.4 Rhythms of PER1 Protein in Areas Related to Maternal Behavior 103
1.03.7 Beyond Motherhood: Maternal Behavior as an Experimental Model for the Study of Some Neuropsychiatric
Disorders 103
1.03.7.1 Emotional Systems 104
1.03.7.1.1 Motivation: Concepts and Definitions 104
1.03.7.2 Maternal Behavior in the Context of SEEKING 105
1.03.7.3 Maternal Behavior in the Context of the CARE Emotional System 106
1.03.7.3.1 Relationship of the CARE Emotional System to Neuropsychiatric Disorders 106
1.03.7.3.2 Early Parental Preoccupations and Their Relationship to Obsessive–Compulsive Disorder 106
1.03.8 Conclusions and Perspectives 107
Acknowledgments 108
References 108

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00003-1 83


84 Parental Behavior

1.03.1 Introduction contractions of the uterus, changes in the distribution, and/


or number of receptors for specific hormones and neurotrans-
The chapters written for the first and second editions of this mitters in the brain, simultaneously with the multisensorial
book presented a thorough review of the many factors that perception of stimuli from the newborn – favor the consolida-
participate in the onset, maintenance, and decline of parental tion and direct the responsiveness of the mother to the young.
behavior in mammals (González-Mariscal and Poindron, In the case of ungulates an additional process takes place at
2002; González-Mariscal and Kinsley, 2009). In addition, two this time: the recognition of the mother’s own offspring which
books (Numan, 2006; Numan and Insel, 2003), several review determines the exclusive nursing of a particular lamb or kid
chapters in books (González-Mariscal and Melo, 2013; Numan across lactation (see the following section). Parturition has
et al., 2006) and journals (Olazábal et al., 2013a, b), plus long been recognized as a critical period for the consolidation
a special issue of the journal Hormones and Behavior (January of maternal behavior, and recent evidence is beginning to
2016), all devoted to the topic of parental behavior, have unveil the factors underlying this process. Results obtained
appeared within the last 15 years. This vast literature indicates in rats suggest that a pathway similar to learning mediates
that new lines of research have emerged, both from the area of this consolidation (Fleming et al., 1996; Graham et al.,
parental behavior itself and from the finding that the display of 2006; Li and Fleming, 2003). Dopaminergic neurotransmis-
this activity modifies the mother’s brain itself as well as several sion (involving D1 and D2 receptors; Byrnes et al., 2002)
behavioral and physiological functions. Moreover, maternal and OT action in the nucleus accumbens (NAcc) shell (D’Cu-
behavior (MB) is no longer viewed as a ‘reflexive’ activity but, nha et al., 2011) seem to be critical in this regard.
rather, as a complex function involving the coordinated activity Vaginocervical stimulation (VCS), received at parturition,
between neural circuits regulating other functions (e.g., social- plays a major role in the establishment of maternal behavior.
ization, emotionality, fear) but ‘recruited’ to subserve MB, and Peridural anesthesia results in the failure to accept the suckling
those mediating ‘strictly maternal’ activities (e.g., nursing, nest attempts of the newborn in primiparous sheep and goats
building). From this background, for this third edition chapter (Poindron et al., 2007a,b). Experimental evidence suggests
we chose to (1) Highlight topics that have emerged recently, that these effects may be mediated by the release of OT occur-
such as (a) the effects of mother–young contact on the ring at parturition: in rats, icv injections of an OT antagonist at
maternal brain; (b) the time dimension in reproductive physi- this time prevent the onset of maternal behavior (Van Leen-
ology and maternal care; (c) maternal behavior as a model for goed et al., 1987) and in sheep, the disruptive effect of peri-
studying neuropsychiatric disorders. (2) Reflect on what new dural anesthesia is partly overcome by icv injection of OT
questions can be asked regarding parental care and where the (Krehbiel et al., 1987). Similar results have been obtained in
field stands now. nonpregnant animals: VCS (Kendrick et al., 1991) or icv OT
injections (Kendrick et al., 1987) promote maternal behavior
in multiparous ewes primed with ovarian steroids; conversely,
1.03.2 Behaviors at Parturition: Combined Actions an OT antiserum reduces the facilitation of maternal behavior
of Hormones and Somatosensory Stimuli normally seen in virgin rats primed with ovarian steroids
(Pedersen et al., 1985).
Placentophagia and amniotic fluid ingestion are consistently OT has been shown to regulate many forms of social
observed following delivery in most mammals studied behavior, such as maternal nurturing and bonding (Rilling
(Poindron et al., 2007a; Gregg and Wynne-Edwards, 2006; and Young, 2014), social recognition (Ferguson et al., 2001;
Kristal, 1991; Melo and González-Mariscal, 2003). Moreover, Skuse et al., 2014), and pair-bonding (Johnson and Young,
this activity has also been reported in males from some bipa- 2015; Young and Wang, 2004; Numan and Young, 2016).
rental species, e.g., dwarf hamsters (Phodopus campbelli; Gregg There are large variations in the topography of OT receptors
and Wynne-Edwards, 2005; Jones and Wynne-Edwards, 2000), (OTRs) in the mammalian brain, for instance, in rats, OTR
California mice (Peromyscus californicus; Lee and Brown, 2002; binding and mRNA have been detected in the medial preoptic
Perea-Rodríguez and Saltzman, 2014). Despite its prevalence area (MPOA; Numan and Insel, 2003) and the prelimbic cortex
in mothers and in fathers of some species, the biological func- (Krémarik et al., 1993; Young et al., 1997). OTR binding
tion or advantage of placentophagia has not been determined. increases on postpartum day 1 – relative to pregnancy day
The interaction between mother and offspring at parturi- 15 – in the bed nucleus of the stria terminalis (BNST) and
tion is critical to ensure a continuous, ‘unfailing’ maternal ventromedial nucleus of the rat hypothalamus (Insel, 1990),
responsiveness across lactation, especially in primiparous and over the course of pregnancy and lactation, changes in
females. Clear evidence in rabbits (González-Mariscal et al., OTR mRNA have been documented (Young et al., 1997). In
1998b), rats (Bridges, 1975), sheep (Poindron et al., 2007a), the socially monogamous and biparental prairie voles a high
and goats (Poindron et al., 2007b) has shown that interfering abundance of OTR has been observed in the prelimbic cortex,
with mother/young contact at this time disrupts or seriously BNST, NAcc, and lateral amygdala. Moreover, in biparental
alters maternal behavior. A similar interference in early- or prairie voles there is a correlation between individual variation
midlactation provokes milder effects. These results suggest in OTR expression in the NAcc and spontaneous parental
that the particular conditions that exist at parturition – in behavior (Olazábal and Young, 2006a, b). In contrast, the
terms of changes in the concentration of several hormones polygamous, uniparental montane and meadow voles have
in blood (e.g., oxytocin, OT; prostaglandin F2-alpha, PGF2a; few OTR in the NAcc and lateral amygdala but high densities
corticosteroids; prolactine, PRL; progesterone; estradiol; for in the lateral septum (LS; Insel and Shapiro, 1992). Further-
review, see González-Mariscal and Poindron, 2002), more, manipulating the density of OTR in the NAcc provokes
Parental Behavior 85

alterations in pair-bonding (Keebaugh et al., 2015; Keebaugh bout: the normal c.3 min day1 (González-Mariscal et al.,
and Young, 2011; Ross et al., 2009). 1994; Zarrow et al., 1965) requires a minimum litter size of
In rabbits, also polygamous and uniparental (González- five. When a single pup is provided, the time inside the nest
Mariscal et al., 2016), a high density of OTR has been found box (during which nursing occurs) is greatly increased, regard-
in the prefrontal cortex (PFC), preoptic area (POA), LS, hippo- less of the day of lactation (González-Mariscal et al., 2013b). A
campus, and medial amygdala (Jiménez et al., 2015). OT is similar effect is observed when the mother’s nipples are covered
released at parturition (Fuchs and Dawood, 1980) and at each or removed (thelectomy) or when the kits’ mouths are covered
nursing bout (Fuchs et al., 1984). Despite their restricted contact (González-Mariscal, 2007). In sows, suckling stimulation from
with the young throughout lactation (a single nursing bout per the piglets modulates a specific component of the maternal
day, lasting around 3 min), indirect evidence suggests a role of ethogram: the mother’s grunting (Algers and Uvnäs-Moberg,
OT for the regulation of specific aspects of rabbit maternal 2007). As the sow lies down to initiate nursing, she emits
behavior. A minimal number of suckling kits (four to five) is a type of grunting that signals the piglets to her teats. Their
essential for (1) maintaining maternal responsiveness across suckling promotes the initial release of OT which, in turn, trig-
lactation (González-Mariscal et al., 1998, 2013b); (2) express- gers a change in the type of maternal grunting: the piglets
ing nursing behavior with circadian periodicity (González- respond to it by suckling more vigorously, thus initiating
Mariscal et al., 2013a); and (3) allowing a normal duration of milk letdown.
the nursing bout (González-Mariscal et al., 2013b). Moreover, In contrast to the above species, ewes and goats show selec-
suckling increases the number of c-FOS-IR cells in the paraven- tive nursing throughout lactation; that is, they will accept at the
tricular nucleus (PVN; González-Mariscal et al., 2009), and the udder only their own lamb or kid and actively reject alien ones
amount of OT released at each nursing bout is dependent on the (Poindron et al., 2007a,b). The recognition of the mother’s
number of suckling kits (Fuchs et al., 1984). own offspring in these species depends on the perception of
the olfactory signature of the newborn at parturition and its
consolidation as an olfactory memory (Lévy et al., 2004). As
1.03.3 Postpartum Activities lactation progresses, visual and acoustic cues begin to play an
important role in the mother’s recognition of her progeny
1.03.3.1 Nursing: Variability Rules
(Nowak et al., 2011; Poindron et al., 2007b) and also in the
This is undoubtedly the main behavior displayed by all reciprocal process, that is, in the capacity of the young to recog-
mammalian mothers toward their progeny, from parturition nize their own mother (Poindron et al., 2007a,b; Nowak et al.,
until weaning. Yet, the specific characteristics of nursing and 2007). This mutual recognition is essential to adjust the phys-
the display of activities that surround it vary greatly among iology and behavior of both parties to each other across lacta-
species. For instance, in rodents, nursing is preceded by tion. Proximity with the offspring is essential to maintain such
retrieving the pups that have strayed away and grouping maternal memory because ewe–lamb separation for 3 days
them in the nest. Rabbits and sows also nurse within a nest, leads to a loss of maternal responsiveness and rejection of
but they do not retrieve their young (despite their small size); the young (Keller et al., 2005). Remarkably, goat mothers
ewes and goats do not retrieve their large, well-developed continue to recognize the vocalization of their own kids for
offspring, and they nurse in the absence of a nest. The predict- up to 13 months postweaning (Briefer et al., 2012).
able, unfailing display of nursing and its associated behaviors
depends on several hormones that act in late pregnancy (e.g.,
1.03.3.2 Maternal Behavior in Nonlactating Females: What
PRL; Andrews and Grattan, 2002; González-Mariscal et al.,
Does It Reveal?
2000, 2004b) and throughout lactation (e.g., OT and vaso-
pressin; Landgraf and Neumann, 2004; Bosch et al., 2005). Although the hormones of pregnancy prime the maternal brain
The fine-tuning of maternal behavior, that is, the display of to effectively respond to the stimuli she receives from the young
nursing with its associated species-typical ethogram starting at parturition, nonpregnant, nonlactating rodents can
throughout lactation, depends largely on somatosensory behave maternally under specific conditions. Thus, rats,
factors. Thus, in species that give birth to multiple young hamsters, and gerbils gradually behave maternally to foster
(such as rodents, rabbits, and pigs), recognition of the mother’s young after several days of cohabitation with them (for review,
own progeny is irrelevant as they effectively nurse young other see González-Mariscal and Poindron, 2002). Virgin female
than their own (Algers and Uvnäs-Moberg, 2007; González- mice of several strains are spontaneously maternal upon first
Mariscal and Gallegos, 2007; Rosenblatt and Lehrman, exposure to foster pups with a latency of a few minutes
1963). In these animals stimuli coming from the progeny regu- (Gandelman, 1973; Noirot, 1972). This supports the idea
late other aspects of maternal behavior. Specifically, in rats the that, in these rodents, the hormones of pregnancy and lactation
adoption of a high-crouch nursing posture critically depends are irrelevant for the onset of maternal behavior. However,
on the quantity and quality of the ventral stimulation provided important differences exist between the care exhibited by truly
by the suckling pups (Stern and Johnson, 1990; Stern et al., lactating versus sensitized mice (Stolzenberg and Rissman,
1992). Moreover, in rats the number and duration of nursing 2011). Specifically, (1) the latency to retrieve pups and adopt
bouts per day declines from midlactation onward (as does crouching over the litter is significantly smaller in lactating
the amount of milk produced) in relation to the intensity of animals and (2) the number of pups retrieved is larger in
suckling, which becomes more vigorous as the pups grow lactating mice. By contrast, intact doe rabbits do not become
(Mena and Grosvenor, 1972). In rabbits, nipple stimulation maternal even after 2 weeks of daily exposure to kits
plays a major role in regulating the duration of each nursing (González-Mariscal et al., 2004a, 2015). Yet, in both rats
86 Parental Behavior

(Fleming and Rosenblatt, 1974a,b) and rabbits (González- 1.03.4.1 Cortex, Trigeminal Complex, and Olfactory Circuit
Mariscal et al., 2004b, 2015; Chirino et al., 2007), lesions to
Beach and Jaynes (1956) determined that no single sensory
the main or the accessory olfactory systems (MOS, AOS) stim-
modality is essential for the performance of retrieving behavior
ulate the display of maternal behavior. In rats this effect is facil-
in postpartum rats. Rather, they proposed that this behavior is
itated by estrogens (reduced latency to become maternal;
under a multisensorial control because the elimination of
Mayer and Rosenblatt, 1979), whereas in rabbits estrogens
vision, olfaction, or tactile sensitivity of the snout and perioral
are essential (absent in OVX animals; González-Mariscal
region did not interfere with pup retrieval. Later work (Benuck
et al., 2004a; Chirino et al., 2007).
and Rowe, 1975; Herrenkohl and Rosenberg, 1972; Herrenkohl
The above findings indicate that the neural circuits under-
and Sachs, 1972) confirmed the lack of effect of anosmia,
lying the motivation to interact with the progeny and the regu-
blindness, and deafness of lactating rats on maternal behavior.
lation of species-typical behaviors are present in all female rats,
However, desensitization of the perioral region – by either local
mice, and rabbits, regardless of their reproductive status, but
anesthesia or section of the infraorbital branch of the trigem-
are tonically inhibited by the MOS and AOS. The way(s)
inal nerve – does interfere with pup retrieval (Kenyon et al.,
through which that inhibition is overcome at parturition –
1981, 1983; Stern and Kolunie, 1989).
such that the olfactory cues from the young are perceived as
Lesions of the amygdala (a structure that receives major
attractive rather than as repulsive – has been investigated in
input from the MOS and AOS) do not disrupt maternal
sheep (by means of electrophysiology) and in rabbits (through
behavior in lactating mice (Slotnick and Nigrosh, 1975), and
a behavioral assay). Olfactory bulb mitral cells, when recorded
lesions to the stria terminalis (the major efferent pathway of
during pregnancy in ewes, respond mainly to food odors. After
the amygdala) have no effect on the maternal care of lactating
birth, however, a dramatic increase occurs in the number of
rats (Numan, 1974). By contrast, lesions of the corticomedial
cells that respond preferentially to lamb odors (Kendrick
amygdala (DelCerro et al., 1991; Fleming et al., 1980; Numan
et al., 1992). These shifts in the specificity of relevant stimuli
et al., 1993), bed nucleus of the accessory olfactory tract (Del
are associated with changes in the release of gamma aminobu-
Cerro et al., 1991), or stria terminalis in virgin rats stimulate
tyric acid (GABA), glutamate, and OT within the olfactory bulb
maternal behavior (Fleming et al., 1980), while kindling-type
and with the activation of the noradrenergic system that
electrical stimulation of the medial amygdala delays the onset
projects to it (Lévy et al., 1995). In rabbits, virgin animals
of maternal responsiveness to foster pups in experienced
show no sniffing preference for a box containing neutral odors
mother rats (Morgan et al., 1999).
versus one containing pup olfactory cues. However, already on
pregnancy day 7, doe rabbits sniff the ‘pup box’ significantly
more than the neutral one. Interestingly, these differences 1.03.4.2 Septum and BNST
disappear close to parturition (Chirino and González-
Mariscal, 2015). The septal area has been lesioned in mother rats (Fleischer and
Slotnick, 1978; Terlecki and Sainsbury, 1978), mice (Carlson
and Thomas, 1968; Slotnick and Nigrosh, 1975), and rabbits
1.03.4 Neural Control (Cruz and Beyer, 1972). In rodents, these lesions do not
abolish maternal motivation but provoke a spatial disorganiza-
The investigation of the brain structures, fiber pathways, and tion of maternal behavior: mothers pick up pups, carry them
neurotransmitters/neuromodulators involved in regulating around, drop them at random (thus altering crouching and
the expression of the many behavioral patterns characteristic nursing), and may also build several small nests where they
of parental care has required the use of diverse experimental retrieve young (Terlecki and Sainsbury, 1978). Excitotoxic
strategies. As will be described below, brain lesions attempt- amino acid lesions of the BNST disrupt retrieval behavior in
ing to obliterate in the mother the perception of specific postpartum rats (Numan, 1996). Rats lesioned in the septal
cues (olfactory, visual, auditory, and tactile) from the young region also show increased defensiveness, an alteration
assessed the role of the different senses on the display of proposed to explain the disorganization of maternal behavior
maternal behavior. More refined methods (e.g., chemical (Sheehan et al., 2000). In contrast, the behavior of rabbits is
lesions, deafferentation of individual brain regions, intracere- severely altered in all its aspects following septal lesions;
broventricular infusion and intracranial implantation of mothers do not build nests and refuse to enter the nest box
hormones, localization of receptors and neuroactive agents for nursing (Cruz and Beyer, 1972).
in specific brain areas, and electrical stimulation of fiber path-
ways) have provided detailed information on the ways
1.03.4.3 MPOA and Its Connections
through which a complex network of nuclei and tracts may
be activated by hormones and organized to coordinate the 1.03.4.3.1 Studies Using Lesion, Inactivation,
perception of stimuli from the young with the expression of or Deafferentation Techniques
the appropriate behavioral patterns. The ‘active’ parental Much evidence has been accumulated to identify the MPOA as
brain has been sampled by the use of methods (e.g., micro- a critical site for the expression of maternal behavior. Electro-
dialysis, immunostaining of immediate early gene proteins, lytic or radiofrequency lesions of this region disrupt retrieving,
uptake of 2-deoxy-glucose, immunostaining of ‘clock gene’ nest building, and nursing in lactating rats (Gray and Brooks,
proteins) that allow an association between the activity of 1984; Jacobson et al., 1980; Numan, 1974; Numan et al.,
particular brain regions and the display of specific behavioral 1977) and hamsters (Marques et al., 1979; Miceli and
patterns. Malsbury, 1982). A similar effect is provoked by injecting
Parental Behavior 87

N-methyl-D-aspartate (NMDA), which selectively destroys cell by several methodologies (Caba et al., 2003a; Fahrbach and
bodies but spares fibers of passage, into the MPOA of lactating Pfaff, 1986; Giordano et al., 1989, 1990; Wagner and Morrell,
rats (Numan et al., 1988). Moreover, virgin females (ovariecto- 1996; Wagner et al., 1998). Progesterone receptors (PRs) are
mized or intact) lesioned in the MPOA do not show maternal also abundant in the MPOA in rats (Numan et al., 1999), guinea
behavior despite many days of exposure to pups (Gray and pigs (Blaustein and Turcotte, 1989; Blaustein et al., 1988; Ware-
Brooks, 1984; Miceli et al., 1983; Numan et al., 1977). Further- mbourg et al., 1986, 1989), sheep (Scott et al., 2000), and
more, the facilitation of maternal behavior provoked in virgin rabbits (Caba et al., 2003b), but their possible role in the control
rats by lesioning the corticomedial amygdala does not occur if of maternal behavior has been little explored (see below).
such females have also been lesioned in the MPOA (Fleming Regarding PRL, implants of this hormone, placental
et al., 1983). In contrast, by applying kindling-type electrical lactogen-I, or growth hormone into the MPOA of ovariecto-
stimulation to the MPOA, maternal responsiveness to foster mized, bromocriptine-treated rats, given progesterone and
pups is promoted in both experienced mothers and virgins then estradiol, readily facilitate maternal behavior (Bridges
(Morgan et al., 1999), a finding supporting the idea that and Freemark, 1995; Bridges and Mann, 1994; Bridges et al.,
activity of this brain structure is critical for the performance 1990, 1997b). 25I-ovine PRL binding sites have been detected
of maternal behavior. in the hypothalamus (Muccioli and Di Carlo, 1994), and
In sheep, a transitory inactivation of the MPOA at parturi- estrogen-induced PRL receptors were found in the MPOA
tion (provoked by a local injection of lidocaine) severely using immunocytochemistry (Pi and Grattan, 1998) and in
disrupts the display of MB 2 h later in primiparous animals situ hybridization (Pi and Grattan, 1999a), especially at the
(Perrin et al., 2007). end of pregnancy (Bakowska and Morrell, 1997) and during
To investigate which connections of the MPOA were lactation (Pi and Grattan, 1999a,b,c). In late pregnant rabbits
involved in regulating maternal behavior, the rostral, caudal, the intracerebroventricular injection of rabbit PRL counteracts
lateral, or dorsal connections of this region were severed and the inhibitory effects of s.c. bromocriptine on nest building
the effects were assessed in lactating rats. Several studies (Franz and crouching over the litter (González-Mariscal et al.,
et al., 1986; Miceli et al., 1983; Numan, 1974; Numan and 2004b). These findings agree with the detection of PRL
Callahan, 1980; Numan and Corodimas, 1985; Terkel et al., binding sites in the hypothalamus of nonpregnant rabbits
1979) revealed that the lateral projections of the MPOA exert through affinity labeling of 125I-ovine PRL (Di Carlo and
the most important influence. Some of these projections travel Muccioli, 1981) and by autoradiography of 125I-ovine PRL
to the ventral tegmental area (VTA; Barone et al., 1981; Conrad (Walsh et al., 1990).
and Pfaff, 1976; Swanson, 1976) a region that, according to
Numan, plays a major role in the regulation of rat maternal
1.03.4.4 Midbrain Tegmentum, PVN, and Habenular Complex
behavior (Numan and Stolzenberg, 2009; Stack et al., 2002).
Specifically, (1) bilateral electrolytic (though not NMDA) The lateral midbrain tegmentum, which includes the peripe-
lesions of the VTA severely disrupt maternal behavior in post- duncular nucleus, receives input from descending preoptic
partum rats (Gaffori and Le Moal, 1979; Numan and Smith, efferents and also from trigeminal sensory pathways that pass
1984) and (2) bilateral knife cuts through the lateral hypothal- through the VTA and carry input from the perioral region
amus disrupt maternal behavior only when performed at a level (Nadaud et al., 1984; Smith, 1973). Lesions in this area abolish
(dorsal) in which fibers coming from neurons in the lateral maternal aggression toward males, block milk ejection, but do
hypothalamus and going to the VTA are severed (Numan not interfere with nursing behavior (Hansen and Ferreira,
et al., 1985). Efferent dopaminergic fibers from the VTA to 1986; Hansen and Gummesson, 1982; Hansen and Kohler,
the NAcc promote behavioral reactivity to a variety of ‘signifi- 1984). However, knife cuts made caudal to the VTA (thus dis-
cant’ biological stimuli, among them, pup cues. The relevance rupting ascending and descending pathways traveling through
of a circuit involving dopaminergic neurotransmission within the mesencephalon) abolish maternal behavior (Numan and
the MPOA-VTA-NAcc circuit is supported by the finding that Numan, 1991). These results indicate that fibers passing
the infusion of flupenthixol (a D1–D2 antagonist) into the through the VTA and relevant for maternal behavior terminate
NAcc shell antagonizes maternal memory in primiparous at levels lower than the mesencephalon (e.g., caudal periaque-
rats; that is, the latency to reengage in maternal behavior ductal gray (PAG) and central tegmental field). The PAG has
following a 10-day separation is increased relative to vehicle- also been implicated in behavioral switching, that is, as part
injected dams (Parada et al., 2008). of a decision-making apparatus that allows the maternal female
to decide whether to hunt or to act maternal (Felicio and
1.03.4.3.2 Studies Using Implantation of Hormones Canteras, 2008). Apparently, the PAG ‘weighs’ the relative
and Detection of Their Receptors merits of chasing prey versus caring for the offspring, based
Diluted estradiol, implanted into the MPOA of rats that were on concurrent internal states.
ovariectomized–hysterectomized on pregnancy day 16, effec- The PVN seems to participate in the onset of maternal
tively stimulates maternal behavior (Fahrbach and Pfaff, 1986; behavior in several species through the release of OT and its
Numan et al., 1977). Similarly, estradiol benzoate implants parvocellular extrahypothalamic projections. Thus, in rats, (1)
into the MPOA of ovariectomized rabbits promote nest building OT is released in the supraoptic nucleus (SON) and the PVN
when combined with peripheral injections (followed by with- at parturition (Neumann et al., 1993); (2) isolation of the
drawal) of progesterone (González-Mariscal et al., 2005). These mediobasal hypothalamus, a procedure that leaves the PVN
findings coincide with the presence of abundant estradiol and its extrahypothalamic connections intact but severes path-
receptor alpha in the MPOA of rabbits and rats, as determined ways between the PVN and the medial basal hypothalamus,
88 Parental Behavior

abolishes milk output but does not affect maternal behavior for GABA, found in the cytoplasm; Martin and Rimvall,
(Herrenkohl and Rosenberg, 1974); (3) lesions to the PVN 1993). They found that, in lactating rats, interaction with
disrupt rat maternal behavior, although only if performed pups increased the number of colabeled cells in the MPOA,
before its onset (Insel and Harbaugh, 1989; Numan and ventral BNST, and caudal PAG (ventrolateral region), a result
Corodimas, 1985); if performed after parturition PVN lesions suggesting that GABAergic neurons may promote specific
significantly reduce maternal aggression (Consiglio and aspects of maternal behavior by removing tonic inhibitory
Lucion, 1996); and (4) kainic acid-induced lesions of the influences. Similarly, Lonstein et al. (2000) reported that
PVN on postpartum day 2 affect retrieving behavior but not lactating rats allowed to interact with pups showed an
other maternal behavior components (Olazábal and Ferreira, increased number of cells colabeled to c-FOS and ERa than
1997), while lesions performed on day 5 postpartum with ibo- did mothers not given young. These differences were particu-
tenic acid (a procedure that preferentially lesions parvocellular larly evident in the MPOA, lateral habenula, and ventral BNST.
neurons) or infusion of antisense oligonucleotides against OT In virgin Balb/c mice a single brief (30 min) exposure to
increase maternal aggression (Giovenardi et al., 1997, 1998). pups increased the number of FOS-IR cells in the MPOA,
Moreover, OT is released within the PVN during nest defense corticomedial amygdala, entorhinal and piriform cortex, and
(Bosch et al., 2004), particularly in rats bred for high-anxiety- anterior olfactory nucleus, regardless of whether females were
related behavior (Bosch et al., 2005). intact or ovariectomized (Calamandrei and Keverne, 1994).
In sheep there is strong evidence for the participation of OT Interestingly, the presentation of pups inside a dark,
from the PVN in the activation of maternal behavior. Thus, (1) perforated box (that allowed females to perceive only
OT is released in this structure during parturition, and retro- olfactory and auditory signals from the young) stimulated
dialysis of this peptide induces maternal behavior in nonpreg- FOS expression in the olfactory areas but not in the MPOA.
nant steroid-primed females (Da Costa et al., 1996a); (2) birth In contrast, the depletion of noradrenaline in the olfactory
or VCS activates the PVN (as determined by expression of the bulb (provoked by injecting 6-hydroxy-dopamine into the
FOS protein), where oxytocinergic neurons and fibers as well medial olfactory stria) reduced FOS expression only in the
as OTRs have been found (Kendrick et al., 1997); (3) the anterior olfactory nucleus and the piriform cortex. These
number of OT immunoreactive (IR) neurons and mRNA tran- results coincide with those obtained in rats and support the
scripts for the OTR are increased in the PVN at parturition and notion that FOS expression in the MPOA occurs as a result of
following progesterone/estradiol priming (Broad et al., 1993); the active display of maternal behavior.
and (4) previous maternal experience further enhances the In multiparous sheep, the interaction with the lamb for the
expression of OTR mRNA in the PVN (Broad et al., 1999). first 30 min after parturition stimulated c-fos mRNA transcrip-
From this evidence, it has been proposed that, at parturition, tion (detected by in situ hybridization) in the cerebral cortex
OT facilitates its own release (by acting on autoreceptors), (e.g., entorhinal, piriform, and somatosensory), habenula,
not only in the PVN itself but also in the brain regions it hippocampus (CA3 region, dentate gyrus), limbic system
projects to, for example, MPOA and olfactory bulbs (Kendrick (e.g., BNST, LS, and olfactory bulb), MPOA, and several hypo-
et al., 1997). thalamic nuclei (e.g., PVN and SON). Very similar changes
were observed in nonpregnant multiparous ewes treated with
estrogen and progesterone and given 5 min of VCS (Da Costa
1.03.4.5 Sampling the Active Parental Brain
et al., 1997).
1.03.4.5.1 The Expression of Immediate Early Genes In primiparous doe rabbits a single brief (c.3 min) nursing
A large number of studies have used the detection of the c-FOS bout significantly increased the number of c-FOS-IR cells in
protein (product of the c-fos gene) by means of immunocyto- the PVN, SON, and LS on lactation days 1 and 7 (González-
chemistry to explore the brain regions activated during the Mariscal et al., 2009). By contrast, in the POA the number of
display of maternal behavior. Thus, Numan and Numan c-FOS-IR cells did not differ between does given a litter to nurse
(1994) found that lactating rats exposed to pups (following on the day of sacrifice (lactation days 1 or 7) and those not
a 3-day separation) showed significantly higher numbers of given kits on that day. However, both groups of mothers
FOS-IR cells in the MPOA, ventral (though not dorsal) BNST, showed many more c-FOS-IR cells than did virgin animals.
anterior cortical, and posterodorsal medial amygdaloid These results suggest that the POA, though not activated by
nuclei, compared to lactating rats that were given candy suckling itself, is part of a ‘circuit’ underlying the regulation
instead of pups on the day of testing. Similarly, Fleming et al. of maternal behavior which, in rabbits, starts in midpregnancy
(1994) found that postpartum rats exposed to pups showed with the construction of an elaborate maternal nest (see
many more FOS-IR cells in MPOA, piriform cortex, and above).
medial and cortical amygdala than mothers exposed to
a familiar adult female or to food. In addition, sensitized 1.03.4.5.2 Other Methods
adult virgin rats also show an increased number of c-FOS IR The uptake of 14C-2-deoxy-glucose (2DG) has been used as an
cells in the MPOA and ventral BNST following a 2-h index of activity in neuronal terminals but not in cell bodies
interaction with pups (Kalinichev et al., 2000). (Nudo and Masterton, 1986; Sharp et al., 1988). At parturition,
To further understand the meaning of changes in the mother rats showed an increased uptake of 2DG in the MPOA
number of c-FOS-IR cells associated with the display of and VTA, while in sensitized virgins displaying maternal
maternal behavior, Lonstein and De Vries (2000) analyzed behavior a decreased 2DG uptake was observed in several struc-
brain sections costained with FOS and an antiserum against tures of the AOS (i.e., accessory olfactory bulb, medial amygda-
GAD67 (an isoform of the rate-limiting synthesizing enzyme loid nucleus, and bed nucleus of the accessory olfactory tract;
Parental Behavior 89

Del Cerro et al., 1995). These results coincide with the idea that 1.03.5.2 Fear and Anxiety Regulation
the facilitation of maternal behavior in virgin rats occurs as
1.03.5.2.1 Behaviors Affected
a consequence of reducing the impact of aversive olfactory
In the realm of stress regulation and the anxiolytic properties of
stimulation coming from the pups. To distinguish between
reproduction, much research demonstrates altered emotional
excitatory and disinhibitory synaptic relations in specific brain
reactivity in parous females. Many works report a reduction
areas, associated with the display of maternal behavior, the
in stress and fear responses of both primiparous and multipa-
same group of investigators combined the 2DG method with
rous rats, compared with virgins (see specific references in
the immunocytochemical detection of c-FOS (Komisaruk
Tables 1 and 2). Multiparity reduces anxiety, relative to a single
et al., 2000). In the three models of maternal behavior explored
maternal experience, but primiparous rats show much attenu-
(parturient, sensitized virgins, and rats hysterectomized on
ated anxiety responses compared with virgins (see Tables 1A
pregnancy day 16), the MPOA showed increased uptake of
and 2).
2DG and increased expression of c-FOS. This indicates that
the expression of maternal behavior (regardless of how it is
induced) involves excitatory input to the MPOA and excitatory 1.03.5.2.2 Brain Mechanisms
output from it. A different pattern was observed in structures of The changes in hormonal and neurotransmitter systems that
the olfactory system (e.g., bed nucleus of the accessory olfactory may underlie the reductions in fear and anxiety associated
tract, BAOT and medial amygdala): while parturient rats with maternal experience are multiple and involve, mainly,
showed increased 2DG uptake and increased FOS expression (1) modifications in the responses to estradiol and prolactin
(i.e., excitatory/excitatory relations), in sensitized virgins 2DG in the anterior pituitary, hypothalamus, POA, striatum, and
uptake decreased and c-FOS increased. This observation further amygdala; (2) changes in opiate receptors; (3) enhanced gluta-
supports the notion that the expression of maternal behavior matergic and dopaminergic neurotransmission; and (4) other
through sensitization in virgin rats involves a reduction in the (e.g., kisspeptin neurons). These effects (and the corresponding
activity of the olfactory system. references) are detailed in Table 1B.

1.03.5.3 Regulation of Cognitive Effects


1.03.5 Impact of Maternal Behavior on the Mother:
Effects on Neuroanatomy, Cognition, and Emotional 1.03.5.3.1 Learning and Memory
Reactivity Reproductive experience also exerts strong and long-lasting
effects on various forms of learning and memory. As shown
1.03.5.1 Theoretical Context in Table 2, spatial, reference, prospective, working, and
Any new mammalian mother is, on the one hand, the same maternal memory are better in rats that have experienced
female she was before the birth of her offspring. Yet, the tran- motherhood. In addition, functions such as foraging and atten-
sition from nulliparous to parous female involves substantial, tion are more finely tuned in mothers. Not surprisingly,
likely permanent, alterations to both brain and behavior. These maternal behavior per se is more efficient in multiparous
changes are related to the simple recognition that a new set of dams. Moreover, the possibility that such changes, induced
activities emerge in the new mother, that include both behav- by maternal experience, are permanent is supported by reports
iors directly related to pup care and maintenance (the so- showing that aging-related declines in brain function are
called maternal behavior), and others that are ancillary to reduced in multiparous dams, followed by primiparous and
maternal behavior and allow the mother to be efficient in her virgin rats (Table 2). The specific tests used to determine the
search for resources (Kinsley et al., 1999; Lambert et al., 2005). above (as well as the corresponding bibliography) are detailed
In uniparental rodents the mother is confronted with a stark in Table 1C and 1D.
lose–lose situation. She can stay on her nest, safe with her
offspring, and face eventual starvation and the loss of her litter. 1.03.5.3.2 Neuroanatomical Changes Observed in the
Or she can leave the nest and her vulnerable brood in search of Maternal Brain
food and water, in which case she places both herself and the The behavioral responses described above involve the activity
young in danger. Departing the safe confines of the nest for of several neurotransmitter systems in various diencephalic
environments and dangers unknown requires overcoming the and telencephalic regions that regulate enhanced attention
fear inherent to doing so and successfully confronting any risks toward and care of the young (see Section 1.03.3). Table 1E
encountered (predators, rogue males, impassable obstacles, presents a detailed description (and bibliography) of the
etc.). Then, any foraging costs (increased time and effort) could changes in dendritic spine length and density, cell proliferation
be reduced by enhancements to spatial and reference memory. and survival, characteristics, and abundance of glia found in the
These latter changes would ensure a rapid return to the nest and brains of multiparous, primiparous, and virgin rats.
the defenseless offspring. Therefore, reductions in fear and
anxiety and improvements to memory systems should arise
as a consequence of pregnancy and/or lactation. As summa- 1.03.6 Timing: A Little-Explored Dimension
rized in Tables 1 and 2, abundant evidence indicates that of Maternal Care
maternal experience modifies brain neuroanatomy, the respon-
siveness to several hormones, the functioning of some neuro- The suprachiasmatic nucleus (SCN), a small-paired nucleus in
transmitter systems, several forms of memory, and emotional the hypothalamus, is considered the master circadian clock that
reactivity. control circadian rhythms in the body (see specific chapter on
Table 1 Effects of maternal experience on fear and anxiety, neuroendocrinology and neurotransmitters, learning and memory, maternal behavior, and neuroanatomya

90
Primiparousb Multiparousb References

Parental Behavior
A. Fear and anxiety
No [ plasma OT following immobilization (PP 6) Carter and Lightman (1987)
< OT content posterior pituitary Higuchi et al. (1991)
Y OT release by electrical stimulation in isolated posterior pituitary
[ % time and # entries in open-arm EPM (only with pups into the maze) Pereira et al. (2005)
Pups in the maze overrode the Y % time in open arm by haloperidol
\ young (proestrus) Byrnes and Bridges (2006a)
[ time in open-arm EPM
[ travel distance open arms
[ % time center open field
\ middle aged
Opposite effects found in young females
OVX eliminated effect of reproductive experience
>> \ at 18 and 22 months \ 10, 14 months old Love et al. (2005)
[ time in open arms in EPM [ % time in open arms in EPM
[ time contact novel stimuli
After EPM and FS: Neumann et al. (1998)
No [ plasma ACTH and CORT (PP 7)
No [ plasma OT (late pregnancy and lactation)
> CORT plasma (early lactation) < CBG Pawluski et al. (2009)
 CBG
[ time in open arms of EPM Byrnes et al. (2012)
(PPT; ERa agonist treatment) Larsen and Grattan (2010)
[ time open arms of EPM
Y latency to full maternal behavior in a novel cage (PP 2)
\ Pregnancy days 15–18 and lactation days 2–6 Neumann et al. (2000)
OT antagonist induces:
Y entries and time in the open arm (EPM)
No [ CORT and ACTH release
Basal and after EPM and FS tasks
[ OT secretion by stress (only pregnant)
< Corticosterone-binding globulin Shanks et al. (1999)
No [ c-fos mRNA in PVN
No [ AVP mRNA in PVN after LPS endotoxin (lactation days 9–12)
\ PP 7–10 Windle et al. (1997)
No [ plasma CORT or ACTH by white noise stress but, 72 h after weaning,
[ plasma CORT or ACTH by 2 h noise stress
< basal CRF mRNA in PVN but noise stress had no further effect
6 weeks postweaning diazepam vs veh EPM, novel cage tasks: Byrnes and Bridges (2006b)
[ locomotor activity
No anxiolytic effects
< CORT release
> % time
> % distance traveled
\ late pregnancy/early lactation: Da Costa et al. (1996b)
Y plasma CORT by stress
< c-fos mRNA in PVN, lateral septum, cingulate cortex, and MeA
< After weaning: Wartella et al. (2003)
[ < c-fos in CA3 and BLA after 60 min in restraint tube
Y [ # number of center blocks crossed (open field test)
and rearing
Y # freezing (similar results for primigravid and
multigravid \s)
\ OVX Walf and Frye (2008)
[ time spent in open arms in EPM
Y immobility time in FST only in \ pretreated with
17-b E2
\ lactation days 7–8 Miller et al. (2011)
> time light chamber
Y GABA-A receptor activity (by pentylenetetrazol)
B. Neuroendocrinology and neurotransmitters
<  plasma PRL (vs OVX) Anderson et al. (2006)
< estradiol < plasma PRL in mating-induced diurnal/nocturnal peak Bridges et al. (1993)
> pituitary PRL Bridges and Hammer (1992)
(proestrus, diestrus) Bridges and Byrnes (2006)
Byrnes and Bridges (2005, 2007)
Byrnes et al. (2001)
Carvalho-Freitas et al. (2007)
Sider et al. (2003)
\ in proestrus: After EB (125 mg kg1): Bridges and Byrnes (2006)
< estradiol-17b and PRL [ plasma PRL and estradiol
> ERa in anterior pituitary After EB (1 mg kg1):
Y plasma PRL and estradiol
After oPRL (250 mg): Anderson et al. (2006)
< serum PRL and estradiol
[ long-form PRL-R mRNA in MPOA and ARC

Parental Behavior
[ SOCS-1 and SOCS-3 mRNA in MPOA and ARC
(diestrous \)
\ ovx Bridges et al. (1997)
< responsive to HAL (sc) to: Hucke et al. (1998)
Induce PRL release in plasma
Induce DOPAC and HVA
(Continued)

91
Table 1 Effects of maternal experience on fear and anxiety, neuroendocrinology and neurotransmitters, learning and memory, maternal behavior, and neuroanatomyadcont'd

92
Primiparousb Multiparousb References

Parental Behavior
\ proestrus Byrnes and Bridges (2007)
After HAL:
[ D2 long mRNA
[ D2 short mRNA Anterior pituitary
Y D2 long/D2 short mRNA
[ response to apomorphine-induced stereotypy and PPI Byrnes et al. (2001)
Hucke et al. (2001)
(diestrus):
[ DA and DOPAC
Y PRL
[ distance and time moving, total velocity
[ level DA and DOPAC in STR, but not in NAcc (shell)
\ primigravid (day 7) \ multigravid (day 7) Felicio et al. (1996)
[ j DA in STR and hypothalamus and tendency of DOPAC in STR
[ After HAL:
[ PRL in serum j 5-HT in STR
[ PRL in serum
In hypothalamus: Carvalho-Freitas et al. (2007)
Y [ ] DOPAC
Y DOPAC/DA ratio
Y HVA þ DOPAC/DA
(2–3 weeks after weaning)
\ primigravid (day 7–8) \ multigravid (day 7–8) Sider et al. (2003)
<  plasma PRL
[ j DA and NE [ ]
Y k DOPAC/DA
Y k HVA/DA Ratio (light-dark shift)
Y k 5-HIAA/5HT
[[[ Y disrupted effect of b-endorphin (sc and MPOA infusion) on MB Kinsley and Bridges (1988)
[ Y analgesic after morphine injection Mann and Bridges (1992)
[[[ MPOA opiate receptor density (PP 5) Bridges and Hammer (1992)
[[[ Oprm1, Oprk1 gene expression in PAG Teodorov et al. (2011)
[[[ m and k opiate receptors in PAG
[ DA release NAcc (first 8 min pup-exposure) Afonso et al. (2008)
[ response of glutamatergic neurons exposed to pup- Cohen and Mizrahi (2015)
odor (PP 4; mice)
[ kisspeptin neurons colocalized with pSTAT5 in ARC Sjöeholm et al. (2011)
(after icv rat PRL)
[ pSTAT5 IR-cells MPOA, PVN, ARC after PRL (icv)
\ young proestrus Byrnes et al. (2009)
[ # ERa-cells in anterior-dorsal MPOA
Y # ERa-cells in anterior-medial and cortical AMG
\ middle-aged (13 months)
[ ERa-IR cells in dorsal STR
PPT (ERa agonist) treatment: Byrnes et al. (2012)
[ CRH mRNA in PVN
Y in lateral AMG (after EPM task)
Pregnant days 7 and 16: Macbeth et al. (2008a)
> exploration ratio (novel environment)
Y DA, DOPAC, HVA, NE, MHPG, 5-HT, 5-HIAA in PFC
[ NE, HVA, MHPG (PP 9)
Y DOPAC, HVA (PP 18), MHPG (PP 16), 5-HT (PP 16)
in CA1
\ 13 months old Macbeth et al. (2008b)
[ [ ] DA and DOPAC, NE, MHPG, 5HT and 5HIAA in olfactory bulb.
[ [ ] BDNF in CA1 and septum
Pup exposure Akbari et al. (2013)
[ DRD1 and DAT gene expression in MPOA
[ cort-related genes in MeA
[ OPRM1
[ 5HT-R2A in MPOA
C. Learning and memory
< < errors in eight-arm radial Pawluski et al. (2006b)
maze (working memory) Pawluski et al. (2006a)
< errors on spatial learning and memory performance task
Y latency to find baited food (PP 21) > time in closed baited well Franssen et al. (2012)
< time with familiar object Kinsley et al. (1999)
> correct choices to find baited food Lambert et al. (2005)
Pawluski et al. (2006a,b)
Pregnant: WMT task Bodensteiner et al. (2006)
Y latency and distance to find platform, and find new
location of platform
[ time in quadrant of previous platform
Learning sessions: PP 1–4 Darnaudery et al. (2007)
[ latency to reach platform (WMT)
> distance traveled
Recall session: PP 14

Parental Behavior
> % time exploring
Y proliferation cells
< cell decreased over time
(Continued)

93
Table 1 Effects of maternal experience on fear and anxiety, neuroendocrinology and neurotransmitters, learning and memory, maternal behavior, and neuroanatomyadcont'd

94
Primiparousb Multiparousb References

Parental Behavior
2 weeks and 16 months after weaning Lemaire et al. (2006)
Y distance traveled to find hidden platform
[ AMPA/Kainate fEPSP slope
[ NMDAr-mediated (LTP in Hipp)
However
Whether \ was stressed during pregnancy,
No effects on WMT
No effects on LTP
Y \ 6,12, 28, and 24 months old Gatewood et al. (2005)
Y k Latency to find baited food
k Latency to new location of food
(DLM and WMT tasks)
[ \ 5, 9, and 13 months old Love et al. (2005)
Y j # retrieve fruit loops
Y latency to previously baited food
\ 17 and 21 months old vs 9 months
Y latency to reach baited food
Y latency to food reward (DLM; visual and no visual cues, reversal task)
Y predation latency in lactation, mid- and late-pregnancy Kinsley et al. (2014)
[ phosphorylation of CREB and LTP in Hipp slices (mice) by OT Tomizawa et al. (2003)
OT antagonist (icv):
Y # of errors: reference memory task
[ # discrimination ratios of object-in-place memory (foraging) (from lactation Cost et al. (2014)
day 14–42 days postweaning)
\ 13 months old Macbeth et al. (2008b)
> exploration ratio time to new object (object recognition)
> exploration ratio time to object placement
D. Maternal behavior
¼ After cesarean section: Moltz et al. (1966)
¼ ¼ % nest-building
¼ Pup retrieval
Maternal retention test (pup exposure at partum for 2– Bridges (1977)
24 h) Fleming and Korsmit (1996)
Y latency to retrieve pups (test 10, 25–35 days later) Fleming and Sarker (1990)
Stern and Mackinnon (1976)
AVP vs veh (icv) Nephew and Bridges (2008)
Y maternal aggression (PP 5)
Y maternal behavior (PP 15)
Antagonist AVP vs veh
[ maternal aggression (PP 5 and PP 15)
Y maternal behavior (PP 15)
Y grooming (PP 5)
AVP (icv) vs veh Nephew et al. (2010)
k maternal aggression (PP 5)
V1a receptor antagonist vs veh
j aggression (PP 15)
Pregnant days 8, 9, 10, and 20 Serafim and Felicio (2002)
[[[ mammary gland grooming
Y latency to retrieve pups Lopatina et al. (2011)
[ plasma OT
[ hypothalamus
[ pituitary
(CD38/ knockout and wild-type mice)
55 and 80 postweaning days: Scanlan et al. (2006)
Y latency to full maternal behavior
[ exploration and distant travel in an open arm (by pup
exposure)
[ C-fos in cortical AMG, NAcc (shell), BLA
Y prefrontal cortex (4–10 days later)
Y k latency to attacks _s Nephew et al. (2009)
[ j # of attacks
[ j CRH mRNA in lateral septum
Y k OT-Rec mRNA in PVN
Y k OT mRNA in PVN
Y PP 5
k AVP mRNA in PVN
Y k V1a mRNA SO
[ j OT mRNA in lateral septum and BNST
j CRH mRNA in PVN PP 15
k OT-Rec mRNA in central AMG
j time fighting (mice) (6 lactations) Svare and Gandelman (1976)
Nest building (lactating mice > pregnant > nulliparous) Bond et al. (2002)
E. Neuroanatomy
[ Thickness of frontal, somesthetic, and occipital cortex Diamond et al. (1971)
[ Basal and apical spine density in PFC, DG, CA1 Hipp (PP Leuner and Gould (2010)
21)
24–36 h PP þ pups – test: PP 8 Shams et al. (2012)
Y total dendritic length and dendritic intersection in CN
[ body and brain weight
[ dendritic branch in NAcc (shell)
[ # dendrites per branch order in MPOA

Parental Behavior
PP 2 and PP 8 Leuner et al. (2007)
Y BrdU-labeled cells in DG (cell proliferation)
[ CORT
But, pups’s removal for 3 days prevents these effects.
PP 2, 1 week post-BrdU shot
Y BrdU-labeled cells in DG (cell survival)
(Continued)

95
Table 1 Effects of maternal experience on fear and anxiety, neuroendocrinology and neurotransmitters, learning and memory, maternal behavior, and neuroanatomyadcont'd

96
Primiparousb Multiparousb References

Parental Behavior
Pregnancy day 7, lactation days 7 and 14 (mice) Gregg et al. (2007)
[ OPC proliferation and generation of new oligodendrocytes in CC and SC
[ MBP expression
[ myelinated axons (genu of CC)
[ oligodendrocytes, < demyelinated axons, > remyelinated axons, and Y
volume of lesion on pregnancy day 14
[ dendritic spines in CA1 Hipp (late pregnancy, PP 5, and Kinsley et al. (2006)
OVX þ P þ E2 null)
Y cell proliferation and % cell survival in DG of Hipp Y cell proliferation in DG Pawluski and Galea (2007)
(lactation day 2) [ % cell survival than primiparous in DG (PP 2)
OVX \ þ estrogen; 13–14 months old Barha and Galea (2011)
[ cell proliferation in DG
17 a-E2 > estrone > 17 bE2
[ dendritic spine in MeA (anterodorsal) Rasia-Filho et al. (2004)
Y dendritic spine in posterodorsal MeA (after weaning)
[ [[[ # GFAP-IR cell in MPOA Featherstone et al. (2000)
Y YYY # GFAP-IR cells; MeA and habenula (PP 5)
[ [ basic GFAP-IR Cg2 Salmaso and Woodside (2008)
(3 h postpartum and PP 24)
\ Late pregnancy and lactation day 16 Salmaso et al. (2005, 2009)
[ bFGF-IR in PVN, SON, MPOA Salmaso and Woodside (2006, 2008)
[ GFAP-IR in Cg2
[ GFAP-ir and bFGF-ir Cg2 at 3 h and PP days 1, 4,
16, 24
Y vimentin (marker GS) in MPOA (PP 1, PP 4, PP 16) Salmaso et al. (2011)
[ GS-IR (PP 1), glutamate/protein, and glutamine/protein
(3 h, PP 1, PP 16) in Cg2 but not in MPOA
Cell body size: number and length of basal dendritic Keyser-Marcus et al. (2001)
branches in MPOA
Late pregnant and OVX þ P þ E2 > lactating \
[ j apical and basal dendritic length and apical branch points in CA3 and CA1 Pawluski and Galea (2006)
[ of Hipp
j # spine density basal region CA1
Y \ 24 months old Gatewood et al. (2005)
kAPP in CA1
kAPP in DG
Positive correlations: latencies in the mazes (WMT, reversal task) and # APP ir
neurons in CA1 and DG
48 h after KA Franssen et al. (2012)
Y % GFAP-IR cells in HFm, HFI, and CA3 of Hipp
[ cell death Hipp CA3 (16 postshot)
No effects on spatial memory and memory of familiar object
24 h after KA lesion (ip) vs veh, diestrus, OVX:PP 10–14 Monasterio et al. (2008)
< Fos-ir cells in PVN
24 h after KA (ip): PP 18 Vanoye- Carlo et al. (2008)
Dorsal Hipp
< loss of cells (>cellular density) in CA1, CA3, CA4
< # IR-cells positive for caspase-3 in CA1, CA3, CA4
< # positive tunel nuclei in CA1, CA3, CA4
[ Bcl-2 ir (antiapoptotic protein) in CA1, cerebral
cortex, and piriform cortex
24 h after KA (ip): PP 19 vs metestrus \ Vanoye- Carlo et al. (2009)
[ ERb in CA1, CA3 (dorsal Hipp)
Y ERa in CA3
KA lesion (icv): PP 12–14 Cabrera et al. (2009)
24 h postlesion: < Fluoro–Jade C staining (marker of cell
degeneration) in CA3, CA4
72 h postlesion: < Fluoro–Jade C in CA1, CA3, CA4
a
Studies done in rats unless otherwise specified.
b
Relative to virgins unless otherwise specified.
ACTH, adrenocorticotropic hormone; AMG, amygdala; AMPA, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (agonist for glutamate); APP, amyloid precursor protein; ARC, arcuate nuclear; AVP, arginine vasopressin;BDNF, brain-
derived neurotropic factor; bFGF, astrocytic basic fibroblast growth factor-2; BLA, basolateral amygdala; BNST, bed nucleus of stria terminalis; CA1, cornu ammonis (region of hippocampus); CBG, corticosteroid binding globulin; CC, corpus
callosum; Cg2, cingulate cortex (area 2); CN, caudate nucleus; CREB, cAMP response element-binding; CORT, corticosterone; CRH, corticotropin-releasing hormones; DA, dopamine; DAT, dopamine transporter; DG, dentate gyrus; DLM, dry
land maze; DOPAC, 3,4-dihydroxyphenylacetic acid; DRD1, DRD4, dopamine-related genes; E2, estradiol; EB, estradiol benzoate; ERs, estrogen receptors; EPM, elevated plus maze; fEPSPs, field excitatory postsynaptic potential; FS, force
swimming; GABA, gamma-aminobutyric acid; GFAP, glial fibrillary acid protein; GS, glutamine synthetase; 5-HIAA, 5-hydroxyindolacetic acid; 5-HT, 5-hydroxytryptamine (serotonin); 5HT-R2A, 5-HT biosynthesis gene; Hipp, hippocampus;
HVA, homovanillic acid (DA metabolite); KA, kainate acid; L-LTP, long-lasting, long-term potentiation; LPS, lipopolysaccharide (endotoxin); MBP, myelin basic protein; MeA, medial amygdala; MHPG, 3-methoxy-4-hydroxyphenylglycol; MPOA,
medial preoptic area; MWM, Morris water maze; NAcc, nucleus accumbens; NE, norepinephrine; OT, oxytocin; OPC, oligodendrocyte precursor cells; Oprm1, opiod receptor m-1 gene; OVX, ovariectomized; PAG, periaqueductal gray; PFC,
prefrontal cortex; PP, postpartum; PVN, paraventricular nucleus; PRL, prolactine; PPI, prepulse inhibition; PPT, 4,40 ,400 -(4-propyl-[1H]-pyrazole-1,3,5-tryl) trisphenol; PSTAT5, phosphorylated signal transducer and activator of transcription
five; SC, spinal cord; SON, supraoptic nuclear; STR, striatum; SUZ, subventricular zone; V, vasopressin; WMT, water maze task.

Parental Behavior
97
98 Parental Behavior

Table 2 Changes in responses of the whole organism as a consequence of motherhood

Responses affected Comparison References

Stress and fear Multip ¼ primip < null Byrnes et al. (2012)
Carter and Lightman (1987)
Da Costa et al. (1996b)
Neumann et al. (1998, 2000)
Pawluski et al. (2009)
Shanks et al. (1999)
Wartella et al. (2003)
Windle et al. (1997)
Anxiety Multip < primip < null Byrnes and Bridges (2006a)
Byrnes et al. (2009, 2013, 2012)
Love et al. (2005)
Neumann et al. (1998, 2000)
Pereira et al. (2005)
Larsen and Grattan (2010)
Scanlan et al. (2006)
Walf and Frye (2008)
Macbeth and Luine (2010)
Spatial memory Multip > null (>16–24 months: better and faster) Cost et al. (2014)
Gatewoodet al. (2005)
Kinsley et al. (1999)
Lemaire et al. (2006)
Love et al. (2005)
Macbeth et al. (2008a)
Pawluski et al. (2006a,b)
Tomizawa et al. (2003)
Reference memory Primip > null and _s Pawluski et al. (2006a,b)
Working memory Pregnant > virgin Tomizawa et al. (2003)
Bodensteiner et al. (2006)
Maternal memory Primip > null Bridges (1977)
Fleming and Korsmit (1996)
Fleming and Sarker (1990)
Fleming et al. (1999)
Orpen and Fleming (1987)
Lopatina et al. (2011)
Scanlan et al. (2006)
Attention Primip > null Leuner and Gold (2010)
Foraging Multip > primip > null Kinsley et al. (2014)
Love et al. (2005)
Brain plasticity Multip ¼ primip > null Barha and Galea (2011)
Diamond et al. (1971)
Featherstone et al. (2000)
Furuta and Bridges (2005)
Gatewoord et al. (2005)
Gregg et al. (2007)
Kinsley et al. (2006)
Kinsley and Lambert (2006)
Leuner and Gould (2010)
Pawluski and Galea (2006)
Pawluski et al. (2016)
Rasia-Filho et al. (2004)
Shingo et al. (2004)
Salmaso and Woodside (2006, 2008)
Salmaso et al. (2005, 2009, 2011)
Shams et al. (2012)
Sensitivity to E2 Multip < null Bridges et al. (1993)
Bridges and Byrnes (2006)
Byrnes et al. (2009, 2012, 2013)
Parental Behavior 99

Table 2 Changes in responses of the whole organism as a consequence of motherhooddcont'd

Responses affected Comparison References

Brain responsiveness to hormones and peptides (e.g., Primip > null Anderson et al. (2006)
PRL, OT, Glu) Brown et al. (2011)
Carter and Lightman (1987)
Cohen and Mizrahi (2015)
Fleming et al. (1999)
Grattan and Kokay (2008)
Gregg et al. (2007)
Higuchi et al. (1991)
Larsen and Grattan (2010),
Nephew et al. (2009)
Neumann et al. (2000)
Pereira et al. (2005)
Salmaso et al. (2011)
Sjöeholm et al. (2011)
Tomizawa et al. (2003)
Enhanced activity of dopaminergic system Primip > null Afonso et al. (2008)
Bridges et al. (1997a)
Bridges (2016)
Byrnes et al. (2001)
Carvalho-Freitas et al. (2007)
Felicio et al. (1996)
Hucke et al. (2001)
Macbeth et al. (2008a,b)
Sider et al. (2003)
Sensitivity to opioids and benzodiazepines Multip < primip Bridges and Hammer (1992)
Kinsley and Bridges (1988)
Mann and Bridges (1992)
Teodorov et al. (2011)
Byrnes and Bridges (2006b)
Reduced aging-related decline in brain function Multip > primip > null Barha and Galea (2011)
Byrnes et al. (2009)
Gatewood et al. (2005)
Love et al. (2005)
Macbeth et al. (2008b)
Protection from neurotoxics Primip > null Franssen et al. (2012)
Morales (2011)
Shanks et al. (1999)
Vanoye-Carlo et al. (2008, 2009)
Monasterio et al. (2008)
Cabrera et al. (2009)

E2, estradiol; Glu, glutamate; Multip, multiparous mother; Primip, primiparous mother; Null, nulliparous female; OT, oxytocin; PRL, prolactine.

this topic in this book). At the core of these rhythms are a group in brain areas related to reproductive processes. Yet, only
of genes, the clock genes, such as Per, Clock, and Bmal1, and recently has the issue of circadian rhythmicity on reproductive
their proteins that form an autoregulatory negative transcrip- function been explored in laboratory animals and in humans,
tion–translation feedback loop with a periodicity of around e.g., in relation to shift work. Thus, the study of circadian
24 h. This molecular mechanism is found in many cells of rhythms in reproductive processes is a topic of growing
the body and regulates several clock-controlled genes. As importance.
a result there is a timing mechanism perhaps in all tissues
and organs, but the master circadian clock is at the top of the
1.03.6.1 A ‘Healthy Clock’ Is Necessary for Ovulation
hierarchy in the control of circadian oscillations. In this section
and Pregnancy
we will focus on clock genes and circadian rhythms in relation
to reproductive process. Long ago it was established that the Everett and Sawyer (1950) in a seminal publication demon-
SCN plays a key role in the timing of ovulation. Besides that, strated that the preovulatory release of luteinizing hormone
very little is known about the circadian control of other repro- (LH) in the rat could be blocked by barbiturate sedation at
ductive process, namely pregnancy and lactation and the ‘certain critical hours during proestrus,’ usually between 2
outcome of these processes: maternal behavior. As expected, p.m. and 4 p.m. (Everett and Sawyer, 1950). Exogenous admin-
these genes are also present in reproductive organs and also istration of estradiol induces an LH surge, but always in the late
100 Parental Behavior

afternoon, even on multiple successive days (Caligaris et al., as low as 1 lux are enough to drastically decrease MEL concen-
1971). After mating the copulatory stimulus induces the initia- tration in the blood (Zeitzer et al., 2000).
tion of a pattern of biphasic circadian prolactin surges: one It has been calculated that 15–20% of the working popula-
diurnal and one nocturnal of low and high amplitude, respec- tion in Europe and the United States is engaged in shift work
tively (Freeman and Neill, 1972). As expected, the timing of LH (PLoS One Medicine Editors, 2011). Shift work is defined by
and PRL release is controlled by the SCN (Kawakami et al., the International Labor Organization as “a method of organiza-
1980; Poletini et al., 2007). tion of working time in which workers succeed one another at
Rhythmic oscillation of clock genes had been observed in the workplace so that the establishment can operate longer
the ovary, uterus, and oviduct, and it is thought that such than the hours of work of individual workers” (PLoS One
oscillations contribute to the timing of several reproductive Medicine Editors, 2011). Epidemiological studies of working
events (reviewed in Sellix, 2013). By using genetic mouse hours during the night or rotating work schedules have been
models with disruption of the core molecular clock, our associated with a higher risk of developing cardiovascular/
knowledge about the importance of a ‘healthy clock’ on repro- metabolic/gastrointestinal disorders, some types of cancer,
ductive events has significantly advanced. Clock/Clock mutant and mental disorders (Gamble et al., 2013).
mice have a reduced pregnancy rate and even those animals Due to the different styles of altered working hours and
that do become pregnant have an increased rate of midgesta- individual preferences of sleep patterns, it is difficult to have
tion fetal reabsorption and pregnancy failure at full term. This a clear correlation between exposure to abnormal lighting
failure was indicated by severe dystocia, and after 24 h of conditions and reproductive processes. However, studies in
unsuccessful labor females were sacrificed. In all cases the different populations in general agree that altered working
fetuses were fully developed but were dead and showed hours greatly increase the risk of endometriosis, painful and
morphological abnormalities (Miller et al., 2004; Miller and irregular menstrual cycles and menstrual flow, dysmenorrhea
Takahashi, 2014). Similarly, knockout mice of Per1 and Per2 duration of menstrual bleeding, a general delay in ovulation,
show deficiencies in implantation, maintenance of pregnancy, miscarriage, low birth weight, and preterm delivery (reviewed
and/or parturition (Pilorz and Steinlechner, 2008). Moreover, in Gamble et al., 2013; Lohstroh et al., 2003). After birth and
Bmal1 knockout mice show a complete lack of embryonic upon return to their predelivery work schedule, mothers
implantation (Boden et al., 2010). It has been proposed show a low percentage of breastfeeding and early introduction
that these abnormalities are due mainly to an inappropriate to complementary foods, which increase the risk of infections
circadian daily timing to coordinate hypothalamic control and poor nutrition in the newborn (Lakati et al., 2002).
of the specific hormonal milieu of pregnancy (Miller and Recent studies in pregnant rats with controlled chronic
Takahashi, 2014). Indeed, Clock mutant mice have a shortened phase shifts have found specific alterations in hormonal and
pseudopregnancy, indicative of a disruption of PRL release metabolic rhythmic parameters (Varcoe et al., 2013). These
(Miller et al., 2004). Previously it was demonstrated that authors found that timing of corticosterone, leptin, glucose,
blocking of the circadian clock by antisense deoxyoligonu- insulin, free fatty acids, triglycerides, and cholesterol concentra-
cleotides for clock genes in the SCN disrupts the circadian tions were profoundly disrupted. Moreover, gluconeogenic and
rhythm of PRL release (Poletini et al., 2007). In humans circadian clock genes in the maternal and fetal liver became
studies that analyzed polymorphisms in four circadian clock either arrhythmic or were in antiphase relative to control
genes found that a single-nucleotide polymorphism of the animals. This result in the liver is most probably related to
gene ARNTL is associated with an increase in miscarriages altered hours of food consumption as a consequence of phase
(Kovanen et al., 2010). This is the first study to establish a rela- shifts. As already stated, peripheral organs also have functional
tionship between a polymorphism of one clock gene and clock genes, which can be entrained by specific cues others than
fertility in humans, although the reason for this infertility light. Among them, timing of food consumption is a powerful
remains unclear. zeitgeber (entraining cue) for the circadian clock machinery of
The importance of the circadian clock for reproductive the liver and other peripheral organs (Damiola et al., 2000).
processes in humans had been established primarily through Similarly, in women an increased risk of Type 2 diabetes melli-
abundant evidence related to the effects provoked by exposure tus has been reported as a consequence of rotating periods of
to lighting conditions during pregnancy. This procedure imme- night shift work (Pan et al., 2011). These alterations in the
diately affects the normal rhythm of the hormone melatonin hormonal and metabolic parameters of the mother during
(MEL). This hormone is produced in the pineal gland and pregnancy have detrimental effects on their offspring, which
secreted to the blood during the night. Its concentration reaches are glucose intolerant due to whole-body insulin resistance
a peak after midnight, then decreases to reach low, almost (Varcoe et al., 2011). MEL seems to play a central role in this
undetectable levels during the day. The duration of MEL secre- effect because (1) rats born from pinealectomized mothers
tion is affected by the length of the night period and by expo- were glucose intolerant due to hepatic insulin resistance and
sure to a bright light during the night (Pevet, 2003). Lewy et al. (2) offspring from pinealectomized mothers that received
(1980) were the first to demonstrate that the exposure to bright exogenous MEL were not glucose intolerant (Ferreira et al.,
light (2500 lux) during the night inhibits the normal pattern of 2012). These results agree with previous reports that pinealec-
MEL secretion. Now it is known that very low light intensity tomy induces glucose intolerance and increased glucose
(<500 lux), equivalent to the normal illumination of a room production in adult rats (Nogueira et al., 2011). From this
during the night, is sufficient to inhibit the onset of MEL during evidence it is clear that disruption of the normal pattern of
the night (Boivin et al., 1996). Specifically, wavelengths in the MEL during pregnancy has profound consequences on the
range of 446–477 nm, in the blue spectrum of intensities, and fetus. Indeed, exposure to constant light during the night
Parental Behavior 101

suppresses the rhythmic secretion of MEL in pregnant dams MEL, which, in turn, is controlled by the master clock. Thus,
(Mendez et al., 2012). This has important consequences for MEL seems to be a zeitgeber for the timing of parturition
the developing fetus, which is exposed to rhythmic varying (Olcese et al., 2013).
concentrations of MEL, as this hormone crosses the placenta
(Okatani et al., 1998). Because the SCN is not a functional
1.03.6.3 Circadian Rhythms in Breast and Breast Milk
master clock in the fetus, it has been considered that rhythmic
maternal MEL is necessary to drive the fetal circadian system Levels of MEL have a circadian rhythm with higher levels
(Mendez et al., 2012). Consequently, blocking the MEL rhythm during the night, as already mentioned. Concurrently, in breast
in the pregnant mother by exposure to constant light induced milk too, there is a rhythm of MEL. During the day these
intrauterine growth retardation and markedly affected mRNA concentrations are very low; they start to increase during the
expression of clock genes and clock-controlled genes as well early night, reach a peak after midnight, and then decrease
as the rhythm of corticosterone secretion (Mendez et al., (Engler, 2012). MEL itself has important benefits for the
2012). This, in turn, affects normal development of other newborn, for instance, regarding infantile colic. This is
organs because in the fetus corticosterone acts as a peripheral a common pathology worldwide in infants younger than
circadian clock, whose rhythm sustains peripheral clocks in 3 months of age, and the episodes of frequent crying occur in
many tissues of the fetus (Torres-Farfan et al., 2011). In general the evening at about the same hour every day (Savino et al.,
these studies support the proposal that maternal MEL acts as 2014). Colic follows a relationship with the sleep–wake cycle,
a zeitgeber for different fetal peripheral oscillators (Serón- and its alteration seems to be a trigger for colic episodes
Ferré et al., 2016). (Epstein et al., 2013). As mentioned, in utero the fetus is
exposed to a circadian release of MEL coming from the mother
but, after birth, a clear rhythm of the infant’s own MEL is
1.03.6.2 Circadian Timing of Parturition
detected at around 3 months of age (Kennaway et al., 1996),
Parturition in nocturnal rodents usually occurs across their coincident with the consolidation of nighttime sleep
resting period, i.e., during the day or subjective day in animals (Henderson et al., 2010). Consequently, it has been proposed
kept in constant darkness (Olcese et al., 2013; Takayama et al., that MEL, received through breast milk, improves nocturnal
2003). In humans, data of labor onset cluster statistically in the sleep and reduces infantile colic (Engler et al., 2012). This
late nighttime and early morning hours, and parturition occurs datum is important as sales of milk formula substitutes are
12–24 h later, at least in nulliparous women (reviewed in growing yearly by around 10%, mostly in low- and middle-
Olcese et al., 2013). Experiments in the rat revealed that partu- income countries (Piwoz and Huffman, 2015), and some of
rition is controlled by circadian signals as destruction of the them are promoted to increase night sleep, which is not true
maternal SCN disrupts this temporal gating of birth (Reppert (Montgomery-Downs et al., 2010). In cows (as in humans),
et al., 1987). In an elegant experiment, Takayama et al. exposure to artificial light during the night significantly reduces
(2003) demonstrated that the circadian signal is MEL from MEL content in milk, a result that is being considered to
the pineal gland. Pinealectomy eliminated the circadian timing produce ‘chronofunctional milk’ from cows (Asher et al.,
of parturition, which then occurred randomly across the 24-h 2015).
light/dark cycle. Administration of MEL during the night, but Besides MEL, other components of breast milk show
not during the day, restored the temporal daytime phasing of temporal fluctuations. Cytokines, important for the develop-
parturition. Yet, in primiparous rabbits kept under a 14:10 ment of the immune, nervous, and gastrointestinal systems,
photoperiod, parturitions occurred throughout the day show chronobiological variations in breast milk (Morais
(González-Mariscal et al., 2013a). et al., 2015). Additionally, the profile of antibodies and
Evidently, the control of pregnancy maintenance and the complement proteins of the immune system are higher during
necessary physiological events for timing parturition requires the day than at night in human breast milk (Franca et al.,
a complex balance of other hormones. In women, plasma 2010). The chronobiological aspect of breast milk is an impor-
progesterone has a significant diurnal rhythm before week 35 tant and necessary area of research that needs urgent attention,
of pregnancy (Challis et al., 1981; Junkermann et al., 1982), especially considering that suboptimal breastfeeding accounts
but this rhythm is not longer detected by week 38 of gestation for around 800 000 child deaths annually, and one of the
(Challis et al., 1983) coincident with an increase of inhibitory causes is the use of breast-milk substitutes (Piwoz and Huff-
progesterone receptor isoforms (Mendelson, 2009) and pro- man, 2015). Some attempts have been made to reproduce
contractile mechanisms necessary for parturition (Olcese circadian variations of selected carbohydrates, aminoacids,
et al., 2013). and proteins (though not MEL) claiming that they help to
If MEL stimulates nocturnal uterine contractions at parturi- consolidate the sleep/wake rhythm in bottle-fed infants from
tion, then inhibiting its secretion must affect such contractions. 4 to 20 weeks of age (Cubero et al., 2007). At 3 months of
A recent experiment in humans confirmed this proposal age, when the infant’s pineal starts to produce MEL, the addi-
(Olcese et al., 2013). Uterine contractions of pregnant women tion of the amino acid tryptophan (necessary for MEL
>38 weeks of gestation were monitored continuously from 7 synthesis) significantly improves the development of the
p.m. to 7 a.m. under dim light. At 11 p.m. they were exposed sleep/wake rhythms (Aparicio et al., 2007).
to a 10 000 lux full-spectrum lamp. Plasma MEL concentration In agreement with the daily and circadian variations in the
immediately decreased by 45% as did uterine contractions. components of breast milk, the ductal epithelium of the
This evidence strongly supported the proposal of a circadian mammary gland shows robust circadian expression of PER1
control of parturition through the differential secretion of protein in lactating, but not, virgin mice (Metz et al., 2006).
102 Parental Behavior

Detailed genetic analysis in rat dams indicates that from day 20 then onward, reaching lowest levels at around the time of lights
of pregnancy through day 1 of lactation there are significant off (Meza et al., 2008). However, the timing of nursing in
changes in several genes, including clock genes, in the liver lactating does is a powerful stimulus that drastically affects
and mammary gland which the authors suggest are orches- this rhythm. Lactating does spontaneously nurse their kits
trated by the master clock in order to support the increased during the night (Caba and González-Mariscal, 2009; Gonzá-
metabolic demands of milk synthesis at the onset of lactation lez-Mariscal et al., 2013a), but timing of once-a-day nursing
(Casey et al., 2009). In humans a microarray study of RNA ob- can be scheduled either during the day or during the night
tained from breast milk fat globules revealed more than 14 000 (Caba et al., 2008). By using this strategy it was demonstrated
transcripts, 7% of which, including clock genes, changed signif- that the rhythm of locomotor behavior shifts in parallel to the
icantly throughout the day; some were more highly expressed timing of nursing (Meza et al., 2008); that is, locomotor activity
during the day and others during the night (Maningat et al., is maximal at the time of scheduled nursing. Moreover, there is
2009). a clear increase in locomotor behavior even before nursing
The above evidence points to the importance of MEL (and time, which suggests an anticipatory activity. This is very inter-
perhaps other components in mother’s milk that show esting because in other animal models before access to a timed
a rhythmic secretion pattern) for the development of circadian stimulus, such as food or other rewards, animals show an
rhythms in the newborn, when it is no longer exposed to the increase in locomotor behavior, a finding that has been inter-
circadian environment in utero. This deficiency of environ- preted as the behavioral output of an endogenous clock
mental circadian maternal influence after birth is more evident (Caba et al., 2008; Antle and Silver, 2009). Coincident with
in preterm infants. Usually these infants are exposed to this interpretation, a rhythmic expressing of PER1 protein has
constant bright light in the neonatal intensive care units. This been reported in several brain areas of the lactating rabbit brain
condition negatively affects the normal growth and develop- (see below).
ment of infants (Blackburn, 1998). By placing a removable In other species, such as the Djungarian (P. campbelli) and
helmet over the premature infant’s head, the exposure to the Siberian (Phodopus sungorus) hamsters (Scribner and Wynne-
bright light during the night was drastically reduced. The idea Edwards, 1994) and in the rat (Rosenwasser et al., 1987), there
was to create an artificial light–dark cycle instead of the 24-h is a decrease in wheel running or locomotor activity rhythms
constant illumination. As a result, infants gained weight faster during pregnancy and lactation, coincident with an increased
and had a shorter hospital stay than newborns kept in constant time spent in the nest. The locomotor activity gradually
illumination. Importantly, they developed higher concentra- recovers to levels seen before pregnancy at the end of lactation.
tions of MEL during the night than during the day, as opposed
to infants under constant illumination in which this difference 1.03.6.4.2 Core Body Temperature
was not observed (Vásquez-Ruiz et al., 2014). Pregnant rabbits have a circadian rhythm of temperature with
a peak that free-runs with a period length >24 h (Jilge et al.,
2001). In contrast, after parturition the peak of temperature
1.03.6.4 Daily Rhythms in the Mother during Lactation
is shifted to the time of once-a-day nursing. Similar to the loco-
During the transition from pregnancy to lactation, there are motor rhythm, body temperature starts to increase in the hours
tissue-specific changes in molecular clocks. In mice, the ampli- preceding nursing, which also supports the proposal of an
tude and robustness of rhythms change between the SCN, liver, endogenous clock in the control of anticipatory activity.
and mammary gland, as well as the stoichiometric relationship In lactating rodents the dam shows hyperthermia perhaps
between positive (CLOCK) and negative (PER2) component because of the frequent contact bouts with her pups (Woodside
proteins of the core molecular clock. Thus, this relationship and Leon, 1980; Scribner and Wynne-Edwards, 1994). Nursing
remained 1:1 in the liver but was increased to 4:1 in the bouts in the rat occur every 24–54 min without an apparent
mammary gland during this transition (Casey et al., 2014). circadian periodicity (Lincoln et al., 1973). Temperature
Following parturition, all mammals engage in lactation and rhythm is not associated with these nursing events because it
display various forms of maternal behavior. Yet, little is known reaches a peak exclusively during the night (Leon et al.,
about the daily or circadian control of this behavior, except in 1984). Yet, it shows an advance in its rising phase and a reduc-
one species. The rabbit is unique among mammals as the tion in rhythm amplitude during gestation and an increase
mother nurses the kits once a day at around the same hour during lactation (Krittrell and Satinoff, 1988). These changes
throughout the 30-day lactation. This predictive behavior has during lactation are associated with the presence of pups.
important consequences both for the mother and the kits When the litter is removed from the nest, these changes in
(reviewed in González-Mariscal et al., 2016). In this section temperature disappear immediately.
we will review behavioral and physiological parameters, activa-
tion of some neuroendocrine cells, and rhythmic patterns of 1.03.6.4.3 Rhythms of PER1 Protein in Neuroendocrine Cells
clock gene machinery in lactating rabbits. Whenever possible, In rabbits (Caba et al., 1996) and rats (Jirikowski et al., 1989),
literature in other species will also be included. changes in the immunoreactivity of oxytocinergic cells occur in
association with specific reproductive conditions. In doe
1.03.6.4.1 Locomotor Behavior rabbits the number of OT cells increases in the lateral hypotha-
Intact female rabbits kept in nonisolated conditions under lamic area and the PVN after parturition in comparison to
a light/dark cycle have a circadian rhythm of locomotor estrous or pregnant subjects. In the rat there is an increase in
behavior with a significant increase in activity before and after immunostaining for OT cells in several regions of the forebrain,
the first hours of lights on. The activity steadily decreases from associated with the onset of maternal behavior and the
Parental Behavior 103

physiological demands of lactation. By analyzing the expres- rhythm (SCN) and the uncoupling of other cells (OT and dopa-
sion of PER1 protein in OT cells in the rabbit, it was demon- minergic) to a different stimulus. This flexibility in the circa-
strated that the timing of nursing entrains rhythmic activity dian machinery is very useful to sustain specific physiological
of OT cells. In estrous does, PER1 protein has a rhythmic demands, such as the synthesis and/or release of hormones
expression in the supraoptic and paraventricular nuclei, but, necessary to sustain milk production and output.
in lactating rabbits, the timing of nursing entrains the rhythm
of this protein. Specifically, at the time of nursing, PER1 cell 1.03.6.4.4 Rhythms of PER1 Protein in Areas Related
numbers are low in both nuclei, but, 4–8 h after nursing, to Maternal Behavior
they increase significantly and reach a peak. From these results Extensive studies, mainly in rodents, have provided informa-
it was proposed that periodic nursing is a time signal for the tion about several brain areas important for the control of
synthesis and/or release of OT, both at central and peripheral maternal behavior (reviewed in Olazábal et al., 2013a,b). The
levels (Meza et al., 2008). No studies had explored clock genes POA and the LS, among others, have been explored in the
or their proteins in OT cells during lactation in other species. rabbit (reviewed in González-Mariscal et al., 2016). As timing
Besides OT, dopaminergic cells are also entrained by the of nursing entrains neuroendocrine cells (see above), it was
timing of nursing in the rabbit. Dopaminergic cells of TIDA hypothesized that areas related to the control of maternal
(tuberoinfundibular dopaminergic) and PHDA (periventricu- behavior could also be entrained. In fact, timing of nursing
lar hypophyseal dopaminergic) cell groups in the hypothal- induces a robust rhythm of PER1 protein in the POA and in
amus have a rhythmic expression of PER1 protein that peaks the dorsal and ventral regions of the septum in comparison
3 h after lights-off in estrous subjects. The timing of lactation to estrous does. The effect in the POAs was most dramatic as
entrains both dopaminergic populations, and now higher no rhythm at all was detected in controls (Meza et al., 2015).
levels of PER1 are attained 4–8 h after the time of scheduled Similar to neuroendocrine cells this effect seems to be main-
nursing (Meza et al., 2011). This shift in the rhythmic activity tained by suckling particularly in the LS as the number of
of dopaminergic cells is probably associated with the daily PER1 cells significantly decreases after 24–48 h of nursing
release of prolactin, which is released massively to the plasma deprivation (Meza et al., 2015). The authors suggested that
in response to the suckling stimulus of the kits (Fuchs et al., the POA and LS are part of an ‘entrainable maternal circuit’
1984). In contrast, no change in the number of PER1 cells that supports maternal behavior in the rabbit. Future studies
was found in the incertohypothalamic dopaminergic cell group are warranted to determine the phenotypical identity of these
in the diencephalon, which does not project to the anterior PER1 rhythmic cells, which could help to understand the
pituitary and has not been associated with the control of almost unexplored control of the daily nursing of the rabbit.
prolactin secretion, as TIDA and PHDA cell groups (Cheung Besides these studies in the rabbit, there is little evidence in
et al., 1998). other species about changes in clock genes in relation to the
It had been suggested that suckling stimulus could be the neural control of behavioral and physiological changes during
entraining signal for the observed rhythms in OT and dopami- pregnancy or lactation. Changes in the rhythms of PER2 and c-
nergic cells during lactation. When lactating females were pre- FOS proteins were found in subregions of the SCN as well as in
vented from nursing for 24 or 48 h, there was a significant the ventral subparaventricular zone in early pregnancy
decrease of PER1 at peak time in those cells (Meza et al., (Schrader et al., 2010). Additionally c-FOS expression was
2008, 2011). Indeed, the intensity of suckling, determined by reduced and its daily rhythm was lost or attenuated in other
the number of suckling pups, is essential to maintain the circa- regions such as the medial septum, diagonal band of Broca,
dian periodicity of nursing: does with litters smaller than four lateral hypothalamus, and MPOA (Schrader et al., 2013). The
kits show a disruption in the normal pattern of daily periodic authors suggest that these changes may support the above re-
nursing. From this finding it was proposed that suckling could ported adaptations of locomotor behavior and temperature
be an entraining signal for periodicity of nursing (González- during pregnancy and parturition and also perhaps the rhythm
Mariscal et al., 2013a) and for neuroendocrine cells (Meza of sleep/wake cycle.
et al., 2008, 2011). Moreover, virgin rabbits – that do not
produce milk – induced to behave maternally by lesioning their
olfactory epithelium show nursing behavior (toward foster kits) 1.03.7 Beyond Motherhood: Maternal Behavior
with a periodicity of c.24 h in 55% of cases (González-Mariscal as an Experimental Model for the Study of Some
et al., 2015). This finding suggests that exposure to the Neuropsychiatric Disorders
hormones of pregnancy and lactation is not essential for
the operation of a system that determines the occurrence of Scientific investigations are often grounded on the use of exper-
the doe–litter interaction with a periodicity of c.24 h. imental models. Models are experimentally accessible represen-
In contrast to the effects of timing of nursing on OT and tations of a natural phenomenon that is being investigated, and
dopaminergic neurons, no change in the rhythm of PER1 in information obtained by observing and manipulating such
the SCN was observed. However, the number of PER1 cells models can be used to generate hypotheses about its nature.
significantly decreases at peak time in this nucleus (Meza A model’s validity refers to the extent to which information ob-
et al., 2008, 2011), which suggest that perhaps suckling is tained from the model is useful for generating accurate hypoth-
a nonphotic stimulus for the SCN (Maywood et al., 1999), eses. One important application of experimental models is to
but this has not been explored properly. The studies in lactating understand the biological underpinnings of pathological
rabbits clearly illustrate the differential control of clock gene conditions in which core mental or behavioral processes are
rhythms in the brain, with some cells locked to the light/dark disrupted. In humans, such disruptions can result in a wide
104 Parental Behavior

range of neuropsychiatric symptoms including (1) psycho- fear is proposed to have arisen together with the adaptive
motor dysfunction, e.g., hyperactivity, tics, and compulsive behavioral responses of freezing or flight, and autonomic
behavior; (2) emotional disturbances such as depression and responses such as increases in blood pressure, heart rate, and
anxiety; (3) cognitive alterations including psychosis, obses- reactivity to startling stimuli. Importantly, neurobiological
sional thinking and alterations in attentional processes; and studies overwhelmingly indicate that the fundamental brain
(4) altered motivational states ranging from compulsive drug processes that underlie these emotional systems are primarily
seeking in the case of addiction, to avolition and anhedonia subcortical and are well conserved across mammalian species,
characteristic of depressive disorders and schizophrenia. and perhaps in some cases (FEAR and PANIC, for example)
Although there is some reason to believe that humans are across vertebrate species in general (Davis et al., 2010; Stewart
uniquely capable of consciously reflecting on and experiencing et al., 2012; Sufka et al., 2009). In the context of animal
these behavioral and mental states as psychopathological modeling, this theoretical framework provides a foundation
symptoms (e.g., Craig, 2002), the core brain processes that for the use of specific behaviors in nonhuman animals as
underlie these states also operate in nonhuman animals and proxies, or ‘endo-psycho-phenotypes’ for human emotional
are similarly subject to pathological disruption. In this context, states, both normal and pathological (Panksepp, 2006).
valid animal models would include those in which informa- Maternal care involving direct interactions with the young,
tion can be obtained, and useful hypotheses can be generated, such as feeding, grooming, carrying, and defending, is ubiqui-
about these core processes and their disruption (Hoffman, tous among mammals, as described in this chapter. According
2015, 2016). to the theoretical framework proposed by Panksepp (2006),
Most investigators that use animal models in order to learn mammalian maternal behavior is the core of the CARE
about human psychopathologies recognize the difficulties – or emotional system, while the SEEKING emotional system –
impossibility – of reproducing a human mental disorder in encompassing general motivational processes – is suggested
a nonhuman animal. Given these limitations, some investiga- to be a fundamental component of all emotional systems.
tors have emphasized modeling strategies in which core symp- There are many converging lines of evidence indicating that
toms or endophenotypes of a given neuropsychiatric disorder the brain mechanisms associated with these emotional systems
are convincingly replicated in the animal: for example, anhe- are conserved across mammalian species and comprise path-
donia and alterations in sleep patterns in the case of models ways that interconnect the anterior cingulate, orbitofrontal,
of depression (Gottesman and Gould, 2003; Uys et al., 2003; and medial prefrontal cortices with the basal ganglia, medial
Hoffman, 2011, 2013). Many emphasize pharmacological thalamus, hypothalamus, and midbrain (PAG and VTA)
responsiveness of the animal model to drugs that successfully (Panksepp, 2006, 2011). In the following sections, we will
treat the human disorder as an important validating criteria discuss mammalian maternal behavior in the context of the
(Willner, 1991; Wolmarans et al., 2013). Others have proposed SEEKING and CARE emotional systems.
that important information can be obtained from considering
human neuropsychiatric symptoms as the consequence of 1.03.7.1.1 Motivation: Concepts and Definitions
alterations in fundamental, evolutionarily adaptive psycho- The term motivation encompasses behavioral and psychological
physiological processes that are conserved between human processes by which an animal regulates the proximity and avail-
and nonhuman animals. According to this view, studying the ability of a certain motivating stimulus. Such stimuli include
biological underpinnings of these core processes in controlled external ones that are important for survival and which are
experimental conditions, and using a comparative approach in approached, as in the case of food or a mate, as well as those
which a variety of animal species are studied, can give impor- that signal danger and which are avoided, such as a predator.
tant insights into the genetic, environmental, and neurobiolog- Interacting with motivating stimuli provokes species-typical
ical processes that underlie human psychopathology consummatory behaviors, or unconditioned responses, that are impor-
(Hoffman, 2015, 2016; Matthysse, 1986; Kas et al., 2007, tant for survival and adaptation to the environment. The expres-
2011; Uys et al., 2003; Insel, 2007; Panksepp, 2006, 2011). sion of an unconditioned response is associated with a particular
affective state. These affective states are suggested to be generated
from the basolateral amygdala (BLA) and the central nucleus of
1.03.7.1 Emotional Systems
the amygdala (CeA), which modulate hypothalamic and brain
The concept of evolutionarily conserved psychophysiological stem mechanisms that control unconditioned autonomic and
processes has been applied to understanding the roots of behavioral responses (Cardinal et al., 2002).
human emotions. Panksepp (2006) proposes that the brains Motivated behavior (also referred to as goal-directed or instru-
of all mammals possess conserved neural substrates for seven mental behavior) is described as having appetitive and consum-
fundamental, adaptive, and evolutionarily ancient ‘emotional matory phases. During the appetitive phase, the animal
systems.’ The names of these systems, by convention written actively regulates the proximity of the motivating stimulus.
in upper case in order to distinguish them from the common, The consummatory phase is when the animal interacts with
everyday meaning of these words, are LUST, CARE, PANIC, the motivating stimulus, displaying behaviors that are often
PLAY, FEAR, RAGE, and SEEKING. These brain emotional stereotypical and species-specific. The psychobiological
systems are proposed to have arisen concomittantly with brain construct of motivation is considered to have components of
systems that underlie basic adaptive behavioral and physiolog- ‘wanting’ and ‘liking,’ each arising from separate brain
ical responses: in other words, an adaptive behavior and its processes (Berridge, 2004). Wanting is manifested by the
associated affective experience are ‘two sides of the same animal’s behavior directed toward the motivating stimulus,
coin.’ For example, the subjective emotional experience of while liking comprises the subjective experience of pleasure
Parental Behavior 105

in response to interacting with the motivating stimulus. Inter- of the pups to the nest. However, unlike most commonly
fering with dopamine neurotransmission in the NAcc markedly studied operant conditioning paradigms, pup retrieval itself is
reduces the animal’s willingness to work to gain access to an innate, unconditioned response. The experimental variables
a motivating stimulus (wanting), while liking responses are that are measured are the latency to begin collecting, propor-
mediated by endogenous opioid neurotransmission in the tion of pups returned to the nest site, and duration of pup-
NAcc and ventral pallidum and are independent of dopamine carrying behavior. These measures clearly reflect the ‘wanting’
(Smith et al., 2011; Castro et al., 2015). component of motivated behavior, as they involve behavioral
Importantly, goal-directed behavior must be flexible and activation and ‘working’ to achieve a specific outcome. More-
adaptable to the animal’s internal physiological state and to over, similar to conditioned instrumental behavior, dopamine
external contingencies. This flexibility is determined in large in the NuAcc augments the vigor and persistence of this uncon-
part by brain processes that represent the incentive value, or moti- ditioned behavior (Hansen, 1994; Keer and Stern, 1999).
vational value of a given stimulus. The incentive value of a stim- In other experiments, rat or mouse dams were conditioned
ulus refers to the relative capacity of a given stimulus to elicit to display bar-pressing in order to gain access to a live pup
goal-directed behavior. The incentive value of a given stimulus (Hauser and Gandelman, 1985; Lee et al., 2000; Van Hemel,
varies according to the animal’s internal state, prior experiences 1973). These studies demonstrated that, for rodents, contact
with that stimulus, and current external contingencies. Most with a live pup can serve as a rewarding stimulus that can
laboratory studies of goal-directed behavior utilize operant support behavioral conditioning. Similarly, maternal rats
conditioning paradigms, in which the animal (most often, preferred pup contact over cocaine during the early postnatal
a rodent) is trained to perform a specific arbitrary behavior period – but no longer during the late postnatal period – in
(e.g., pressing a bar) in order to receive a motivating stimulus conditioned place preference paradigms where the female
(e.g., a food or sweet ‘reward’) (Berridge, 2004). In these test chose between spending time in a chamber previously associ-
paradigms, the amount of work that the animal is required to ated with pups and an alternative chamber previously associ-
do in order to receive the reward can be experimentally varied, ated with cocaine (reviewed in Pereira and Morrell, 2011).
as can the incentive value of the reward. The former is experi- In other words, similar to food or cocaine reward, contact
mentally varied by increasing or decreasing the number of with a pup apparently elicits an unconditioned pleasure
bar presses that the animal is required to do in order to receive response, or ‘liking.’ Likewise, the incentive value of the pup
the reward (thereby increasing or decreasing the amount of changes across the reproductive cycle, and the behavioral
‘work’ it is required to do), and the latter is varied by prior response of the female changes accordingly. Changes in the
deprivation of the reward (increasing its incentive value), or incentive value of maternally relevant stimuli are most likely
by satiating the animal to the reward (decreasing its incentive related to changing circulating levels of certain hormones of
value by allowing the animal to eat it ad libitum just prior to pregnancy. One particularly clear demonstration of this
the test). These measures assess the ‘wanting’ component of phenomenon is the maternal behavior of the female domestic
motivation. In the specific case of sweet rewards, ‘liking’ is rabbit.
outwardly manifested by the animal’s unconditioned response In nature, the pregnant female European rabbit (Oryctolagus
of tongue protrusion and smacking of the lips (Berridge, 2000). cuniculus) digs a burrow, ending in a nest chamber, approxi-
To condense the findings of a substantial body of literature mately 1–2 weeks before parturition (gestation in the rabbit
into a pair of sentences: the incentive value of a particular is 31 days). When finished, she exits the burrow and conceals
reward is associated with activity of the orbitofrontal cortex its entrance by covering it with soil. Then, a few days before
(OFC) and is determined by interactions between this region parturition, she reopens the burrow and begins to collect dry
and the BLA; while the vigor and persistence to work in order grass in her mouth and carries it into the chamber, where she
to obtain the reward (‘wanting’) is augmented by dopamine constructs a nest. About 20–40 trips are necessary in order to
release in the NuAcc core (Baxter et al., 2000; Berridge, 2000, collect enough grass to finish the nest (Hoffman and Rueda-
2004; Cardinal et al., 2002; Salamone and Correa, 2002; Morales, 2009). When finished, she leaves the burrow, again
Kringelbach et al., 2003; Kringelbach and Rolls, 2004; concealing its entrance. Later, on the night of parturition, she
Schoenbaum et al., 2011; Balleine, 2011). The unconditioned reopens the burrow, pulls out hair from her ventrum and lines
affective response to the goal (‘liking’) appears to be modulated the inside of the nest with it, and then gives birth to the kits
by endogenous opioid neurotransmission in the NuAcc and inside the maternal nest. She eats the placentae (Melo and Gon-
ventral pallidum (Castro et al., 2015). zález-Mariscal, 2003), cleans the newborn and nurses them,
and then leaves the nest burrow and conceals its entrance.
One time per day thereafter, she returns to the nest to nurse
1.03.7.2 Maternal Behavior in the Context of SEEKING
her kits for a very brief time (approximately 5 min), each
Studies of maternal behavior can offer a complementary time reopening and concealing the burrow’s entrance (Myers
perspective on motivational processes and the neural under- and Poole, 1961).
pinnings of goal-directed behavior. Let us consider one This behavioral pattern is faithfully replicated in the labora-
frequently used measure to assess maternal behavior in the tory. A series of studies have shown that the rise in circulating
rat: pup retrieval. In the laboratory, when a dam’s pups are progesterone (against a background of constant estradiol
removed from the nest site and scattered around on the floor levels) during pregnancy stimulates the motivation to dig while
of the cage, she quickly begins to pick them up and carry the subsequent fall in the levels of this hormone, which occurs
them, one by one, back to the nest site. This behavior is goal- a few days before parturition, triggers the drive to collect straw,
oriented in that it is aimed toward a specific end: to return all carry it into the nest box, and construct a straw nest. A surge in
106 Parental Behavior

prolactin levels just before parturition is associated with the the OFC in the positive affective response to stimuli
onset of hair pulling and the incorporation of this hair into associated with their own infant (discussed later).
the straw nest (González-Mariscal et al., 1994, 1996, 2004b).
Controlled experiments strongly suggest that these behavioral
1.03.7.3 Maternal Behavior in the Context of the CARE
changes are associated with changes in the incentive value of
Emotional System
specific external stimuli. Thus, several days before parturition
the pregnant rabbit displays a clear preference to collect and 1.03.7.3.1 Relationship of the CARE Emotional System
carry straw over collecting real rabbit hair or synthetic material to Neuropsychiatric Disorders
that resembles rabbit hair, when both materials are freely avail- Some evolutionary theories suggest that mother–infant attach-
able. By contrast, just before parturition, she preferentially ment is the most basic and evolutionarily ancient example of
collects and carries real or synthetic hair (González-Mariscal affiliative behavior. A reasonable hypothesis is that the
et al., 1998). Moreover, the motivation to collect and carry mammalian CARE emotional system arose along with lacta-
straw is also modulated by external cues associated with the tion, around 200 million years ago. Comparative investiga-
nest: whether constructed by the rabbit herself, or by the exper- tions of mammalian maternal behavior in placental
imenter, interacting with a finished nest inside the nest box mammals, as well as in marsupials and monotremes, can illu-
quenches her motivation to collect and carry straw. Conversely, minate the inner workings of the CARE emotional system.
straw carrying motivation is prolonged during experimental Components of this fundamental system are most likely dis-
manipulations in which the rabbit is able to collect and carry rupted in neuropsychiatric conditions such as autism, which
straw but unable to interact with a completed nest. In other is characterized by difficulties in forming social attachments,
words, the incentive value of straw is adjusted according to and schizophrenia, which often involves symptoms of disin-
external cues associated with the status of nest completion terest in social interactions and difficulties in processing social
(Hoffman and Rueda Morales, 2009). Perhaps most strikingly, stimuli. In both disorders, some studies suggest that pharmaco-
the female rabbit will eat meat only during a very brief period logical manipulations of oxytocinergic neurotransmission
just after parturition, when she would normally consume the might alleviate these symptoms (Hofman et al., 2015). Brain
placentae. During the rest of her life, the rabbit is a very strict processes associated with the sudden dissolution of a strong
herbivore (Melo and González-Mariscal, 2003). The theme social bond (e.g., death of a loved one or the termination of
running through this body of work is that the incentive value a romantic relationship) are proposed to be similar to those
of specific stimuli (straw, hair, meat) is determined by a combi- associated with depressive disorders as well as drug withdrawal
nation of internal physiological cues (e.g., hormone levels) and symptoms in opioid addiction (Panksepp, 2006).
external cues (e.g., the presence or absence of a completed nest
inside the nest box). What is the neural substrate underlying 1.03.7.3.2 Early Parental Preoccupations and Their
such dramatic changes in the incentive value of these stimuli? Relationship to Obsessive–Compulsive Disorder
Studies using operant conditioning paradigms, in which the In nonhuman animals, the CARE emotional system is activated
incentive value of the reinforcer (most often, a food reward) is by a combination of internal endocrine and neurochemical
experimentally controlled, point to OFC-BLA connections as signals and external stimuli. For example, in the case of the
the substrate for determining the incentive value of food female rabbit, the interactions between internal hormonal
reward (Baxter et al., 2000; Kringelbach et al., 2003; milieu and specific external stimuli, and their affects on moti-
Kringelbach and Rolls, 2004). Are these areas similarly vation and behavior, have been elegantly demonstrated. In
involved in determining the incentive value of unconditioned the case of humans the situation is much more complex, but
maternally related stimuli? A current model for the it is apparent that becoming a new parent is associated with
neurocircuitry of maternal behavior implicates the BLA and significant emotional, cognitive, and behavioral changes.
PFC as regions that process and relay information about Leckman et al. (1999) and Abramowitz et al. (2007) described
maternally relevant stimuli to the NuAcc and ventral thought and behavior patterns in new parents that strikingly
pallidum, which gate the motor response to these stimuli resemble the symptoms of obsessive–compulsive disorder
(Olazábal et al., 2013a,b). In support of this circuit model, (OCD). Persons with OCD suffer from uncontrollable intrusive
lesions to the BLA severely disrupted bar-pressing for pups in thoughts (obsessions) that provoke the performance of specific
maternal rats (Lee et al., 2000) and temporary inactivation of repetitive behaviors or behavioral rituals (compulsions).
the basomedial nucleus of the amygdala (BMA) disrupted Approximately 80–90% of the new mothers and fathers re-
pup retrieval (Numan et al., 2010), suggesting that these ported experiencing recurrent and intrusive thoughts related
manipulations reduced the incentive value of pup contact. to some harm occurring to the baby. Moreover, similar propor-
Likewise, temporary inactivation of the medial prefrontal tions of new parents in both studies (approximately 30–40%)
cortex (mPFC, infralimbic or paralimbic regions) reduced the reported having intrusive thoughts about intentionally harm-
preference for pup contact over cocaine administration in ing the baby. Other intrusive thoughts included ‘misplacing’
a conditioned place preference paradigm (Perreira and the baby (for example, leaving it at the supermarket checkout;
Morell, 2011) and greatly increased the latency to retrieve experienced by 50% of new parents), illness (30%),
pups (Febo et al., 2010). However, the specific involvement contamination (50%), and sexual thoughts about the baby
of OFC-BLA connections in determining the incentive value (10%) (Leckman et al., 1999; Abramowitz et al., 2007).
of maternally relevant stimuli apparently has not been Leckman et al. (1999) propose that such ‘early parental preoc-
investigated in laboratory animals. On the other hand, there cupations and behavior,’ or EPPB, are triggered by enhanced
is a growing body of work in human mothers that implicates activity of highly conserved neural pathways that link
Parental Behavior 107

heightened error detection and risk assessment to harm avoi- appear to be times of increased vulnerability to the onset
dant behaviors. They also propose that OCD involves similar of clinical OCD in women. Several studies have reported
alterations in these same systems. that a considerable proportion of women that suffer from
Functional magnetic resonance imaging (fMRI) studies in OCD retrospectively associate the onset or worsening of their
human mothers have implicated certain cortical and subcor- symptoms with pregnancy or childbirth (Forray et al., 2010;
tical regions in maternal responsiveness to stimuli associated Neziroglu et al., 1996; Maina et al., 1999; Labad et al., 2005;
with their child (Hipwell et al., 2015; Kim et al., 2011; Guglielmi et al., 2014; reviewed in Abramowitz et al., 2004;
Lorderbaum et al., 1999, 2002; Swain et al., 2008; Leibenluft McGuinness et al., 2011). Likewise, the results of two
et al., 2004; Nitchke et al., 2003; Noriuchi et al., 2008; Ranote prospective studies were consistent with the proposal that
et al., 2004). Baby-associated cues almost invariably activate pregnancy and the postpartum period are times of increased
the OFC and/or anterior cingulate cortices. Activation in the risk for the onset or exacerbation of obsessive–compulsive
anterior cingulate cortex is frequently associated with hearing symptoms (Chaudron and Nirodi, 2011; Abramowitz et al.,
a baby’s cries, compared to hearing white noise (the control 2007). Moreover, women who reported OCD symptom
sound), along with activations in motor-associated regions worsening during pregnancy or the postpartum period were
(premotor, supplementary motor, primary motor). Interest- more likely to also have suffered from premenstrual wors-
ingly, activations in the anterior cingulate do not seem to ening of OCD symptoms, suggesting that reproductive
distinguish between cries of the mother’s own baby and those hormones might play a role in OCD symptom exacerbation
of a different child. On the other hand, activation of the OFC (Forray et al., 2010). Consistent with this idea, some studies
was also associated with hearing a baby’s cry, but in this case have reported an increase in subclinical obsessive–
activations were greater for the mother’s own baby compared compulsive-like thoughts and behaviors – typically having
to cries from an unfamiliar baby. Similarly, when mothers to do with contamination and cleaning – during the premen-
were shown pictures or video clips of their own child or an struum. These obsessive–compulsive-like experiences were
unfamiliar child, seeing her own child elicited greater activa- positively correlated with salivary progesterone concentra-
tions in the OFC and posterior cingulate cortex. In one study, tions (Fleischman and Fessler, 2011; Dillon and Brooks,
OFC activation correlated positively with the mother’s self- 1992). Fleischman and Fessler (2011) propose that such
reported ratings of pleasant mood evoked by the photograph cognitive and behavioral responses to elevated progesterone
of her child (Nitchke et al., 2003). Additionally, specific subcor- might be evolutionarily adaptive responses to progesterone’s
tical regions were also activated in response to baby-associated inhibitory effects on the immune system: behavioral
cues. Subcortical areas that were activated in this study compensations to minimize contact with pathogens. There-
included many components of the cortico-striatal-thalamo- fore, a detailed understanding of the fundamental brain
cortical looped circuits: the caudate and NuAcc, globus pal- mechanisms that underlie how the hormones of pregnancy
lidus, thalamus, VTA, and substantia nigra, as well as the amyg- alter the female’s perception of specific stimuli should shed
dala and hypothalamus. light on possible pathological mechanisms of OCD (Hoff-
It is striking many of these very same regions have been man and Rueda-Morales, 2009, 2012).
associated with the symptoms of OCD. The results of many
fMRI studies concur: obsessive–compulsive symptoms are
associated with increased activity in the OFC, anterior cingu- 1.03.8 Conclusions and Perspectives
late and insular cortices, caudate and putamen, globus pal-
lidus, and medial thalamus. Some studies have also pointed The study of mammalian maternal behavior has expanded
to increased activity in the amygdala and ventral striatum much since the last edition of this book. New areas of research
(NuAcc) (Rotge et al., 2008; Menzies et al., 2008). Together, have flourished, e.g., the modifications to the female’s brain
these regions form looped circuits that are proposed to serve that occur as a consequence of her engaging in maternal
as an interface between mechanisms that perceive and inter- behavior. An abundance of data have revealed that the
pret emotionally salient stimuli and those that initiate and offspring (via their combination of sensory load) can perma-
maintain the appropriate motor response to these stimuli nently change the mother’s emotional reactivity, her capacity
(Burquière et al., 2015; Haber, 2016). In this context it is for learning and memory, and her subsequent maternal
compelling that, although the specific characteristics of obses- behavior. These findings agree with the notion that plasticity
sions and compulsions in an individual can be quite ideosyn- is an inherent property of the mammalian brain that is not
cratic, they virtually always have at their core adaptive restricted to the infantile–juvenile stage.
processes of harm avoidance (to self, loved ones, or one’s A new dimension has been added to the study of maternal
home and belongings), contamination and uncleanliness, behavior: time. In both animals and humans many works are
and symmetry and order in one’s personal space (Hoffman, exploring the relationship between the circadian system and
2011, 2015). The hypothesized CARE emotional system, particular aspects of reproductive physiology (e.g., parturition,
which is proposed to have arisen as a mechanism to allow milk production) and reproductive behavior. The underlying
the formation and maintenance of the mother–infant bond subcellular mechanisms are beginning to be revealed by
as well as to promote behaviors in the mother that increase showing the activation of several ‘clock genes’ in particular
the chances that her offspring will survive, might participate brain regions that participate in the regulation of maternal
at some level in each of these processes. behavior and nursing. The rabbit is emerging as a particularly
In further support of a relationship between the CARE good species for exploring this topic given its natural once-a-
emotional system and OCD, pregnancy and childbirth day nursing pattern.
108 Parental Behavior

The richness of the behavioral repertoire displayed by Balleine, B.W., 2011. Sensation, incentive learning, and the motivational control of
mothers (human and animal), plus the accompanying changes goal-directed action. In: Gottfried, J.A. (Ed.), Neurobiology of Sensation and
Reward. CRC Press/Taylor and Francis, Boca Raton, FL (Chapter 13).
in emotionality, has emerged as a useful model for exploring
Barha, C.K., Galea, L.A.M., 2011. Motherhood alters the cellular response to estrogens
some aspects of particular neuropsychiatric diseases. As the in the hippocampus later in life. Neurobiol. Aging 32, 2091–2095.
key processes that underlie symptoms such as hyperactivity, Barone, F.C., Weiner, M.J., Scharoun, S.L., et al., 1981. Afferent connection of the
deficits in attention, and anxiety also operate in animals, lateral hypothalamus: a horseradish peroxidase study in the rat. Brain Res. Bull. 7,
specific models have been generated in rodents and rabbits to 75–88.
Baxter, M.G., Parker, A., Lindner, C.C.C., et al., 2000. Control of response selection by
obtain objective information for generating hypothesis about reinforcer value requires interaction of amygdala and orbital prefrontal cortex.
the disruption of particular neuronal networks in specific J. Neurosci. 20, 4211–4319.
neuropsychiatric diseases. Beach, F.A., Jaynes, J., 1956. Studies of maternal retrieving in rats. III. Sensory
In summary, the investigation of maternal behavior has cues involved in the lactating female’s response to her young. Behaviour 10,
104–125.
spilled over its traditional borders. The varied repertoire of activ-
Benuck, I., Rowe, F.A., 1975. Centrally and peripherally induced anosmia: influences
ities (and associated physiological changes) shown by mamma- on maternal behavior in lactating female rats. Physiol. Behav. 14, 439–447.
lian mothers as they care for their offspring is now viewed from Berridge, K.C., 2004. Motivation concepts in behavioral neuroscience. Physiol. Behav.
the perspectives of developmental psychobiology, chronobi- 81, 179–209.
ology, biological psychiatry, and, of course, behavioral neuroen- Berridge, K.C., 2000. Measuring hedonic impact in animals and infants: microstructure
of affective taste reactivity patterns. Neurosci. Biobehav. Rev. 24, 173–198.
docrinology. The authors of this chapter hope that, from reading Blackburn, S., 1998. Environmental impact of the NICU on developmental outcomes.
our text, young investigators will be recruited to continue J. Pediatr. Nurs. 13, 279–289.
studying maternal behavior and thus broaden our under- Blaustein, J.D., Turcotte, J.C., 1989. Estradiol-induced progestin receptor immuno-
standing of this most complex, essential mammalian activity. reactivity is found only in estrogen receptor-immunoreactive cells in guinea pig
brain. Neuroendocrinology 49, 454–461.
Blaustein, J.D., King, J.C., Toft, D.O., et al., 1988. Immunocytochemical localization
of estrogen-induced progestin receptors in guinea pig brain. Brain Res. 474,
Acknowledgments 1–15.
Boden, M.J., Varcoe, T.J., Voultsios, A., Kennaway, D.J., 2010. Reproductive biology
The authors recognize the work of the late Craig Kinsley, who coau- of female Bmal1 null mice. Reproduction 139, 1077–1090.
Bodensteiner, K.J., Cain, P., Ray, A.S., et al., 2006. Effects of pregnancy on spatial
thored this chapter for the second edition of this book. His valuable
cognition in female hooded Long-Evans rats. Horm. Behav. 49, 303–314.
contributions to the field will forever remain a testimony to the crea-
Boivin, B.D., Duffy, J.F., Kronauer, R.E., et al., 1996. Dose-response relationships for
tivity and originality of his academic thinking. We thank Francisco resetting of human circadian clock by light. Nature 379, 540–542.
Torres Quintanar for his generous help in the preparation of the Bond, T.L.Y., Neumann, P.E., Mathieson, W.B., et al., 2002. Nest building in nulli-
bibliography. gravid, primigravid and primiparous C57BL/6J and DBA/2J mice (Mus musculus).
Physiol. Behav. 75, 551–555.
Bosch, O., Meddle, S.I., Beiderbeck, D.I., et al., 2005. Brain oxytocin correlates with
maternal aggression: link to anxiety. J. Neurosci. 25, 6807–6815.
References Bosch, O.J., Krömer, S.A., Brunton, P.J., et al., 2004. Release of oxytocin in the
hypothalamic paraventricular nucleus, but not central amygdala or lateral septum in
Abramowitz, J.S., Nelson, C.A., Rygwall, R., et al., 2007. The cognitive mediation of lactating residents and virgin intruders during maternal defence. Neuroscience
obsessive-compulsive symptoms: a longitudinal study. J. Anxiety Disord. 21, 124, 439–448.
91–104. Bridges, R.S., 1975. Long-term effects of pregnancy and parturition upon maternal
Abramowitz, J.S., Schwartz, S.A., Moore, K.M., et al., 2004. Obsessive-compulsive responsiveness in the rat. Physiol. Behav. 14, 245–249.
symptoms in pregnancy and the puerperium: a review of the literature. Bridges, R.S., 1977. Parturition: its role in the long-term retention of maternal behavior
J. Anxiety Disord. 17, 461–478. in the rat. Physiol. Behav. 18, 487–490.
Afonso, V.M., Grella, S.L., Chatterjee, D., et al., 2008. Previous maternal experience Bridges, R.S., 2016. Long-term alterations in neural and endocrine processes induced
affects accumbal dopaminergic responses to pup-stimuli. Brain Res. 1198, by motherhood in mammals. Horm. Behav. 77, 193–203.
115–123. Bridges, R.S., Byrnes, E.M., 2006. Reproductive experience reduces circulating
Akbari, E.M., Shams, S., Belay, H.T., et al., 2013. The effects of parity and maternal estradiol-17b and prolactin levels during proestrus and alters oestrogen sensitivity
behavior on gene expression in the medial preoptic area and the medial amygdala in female rats. Endocrinology 147, 2575–2582.
in postpartum and virgin female rats: a microarray study. Behav. Neurosci. 127, Bridges, R.S., Freemark, M.S., 1995. Human placental lactogen infusions into the
913–922. medial preoptic area stimulate maternal behavior in steroid-primed, nulliparous
Algers, B., Uvnäs-Moberg, K., 2007. Maternal behavior in pigs. Horm. Behav. 52, female rats. Horm. Behav. 29, 216–226.
78–85. Bridges, R.S., Hammer Jr., R.P., 1992. Parity-associated alterations of medial preoptic
Anderson, G.M., Grattan, D.R., van den Ancker, W., et al., 2006. Reproductive opiate receptors in female rats. Brain Res. 578, 269–274.
experience increases prolactin responsiveness in the medial preoptic area and Bridges, R.S., Mann, P.E., 1994. Prolactin–brain interactions in the induction of
arcuate nucleus of female rats. Endocrinology 147, 4688–4694. material behavior in rats. Psychoneuroendocrinology 19, 611–622.
Andrews, Z.B., Grattan, D.R., 2002. Opioid control of prolactin secretion in late Bridges, R.S., Felicio, L.F., Pellerin, L.J., et al., 1993. Prior parity reduces post-coital
pregnant rats is mediated by tuberoinfundibular dopamine neurons. Neurosci. Lett. diurnal and nocturnal prolactin surges in rats. Life Sci. 53, 439–445.
328, 60–64. Bridges, R.S., Henriquez, B.M., Sturgis, J.D., et al., 1997a. Reproductive experience
Antle, M.C., Silver, R., 2009. Neural basis of timing and anticipatory behaviors. Eur. J. reduces haloperidol-induced prolactin secretion in female rats. Neuroendocrinology
Neurosci. 30, 1643–1649. 66, 321–326.
Aparicio, S., Garau, C., Esteban, S., et al., 2007. Chrononutrition: use of dissociated Bridges, R.S., Numan, M., Ronsheim, P.M., et al., 1990. Central prolactin infusions
day/night infant milk formulas to improve the development of the wake-sleep stimulate maternal behavior in steroid-treated, nulliparous female rats. Proc. Natl.
rhythms. Effects of tryptophan. Nutr. Neurosci. 10, 137–143. Acad. Sci. U.S.A. 87, 8003–8007.
Asher, A., Shabtay, A., Brosh, A., et al., 2015. “Chrono-functional milk”: the difference Bridges, R.S., Robertson, M.C., Shiu, R.P., Sturgis, J.D., Henriquez, B.M., Mann, P.E.,
between melatonin in night-milk versus day-milk under different night illumination 1997b. Central lactogenic regulation of maternal behavior in rats: steroid depen-
conditions. Chronobiol. Int. 32, 1409–1416. dence, hormone specificity, and behavioral potencies of rat prolactin and rat
Bakowska, J.C., Morrell, J.I., 1997. Atlas of neurons that express mRNA for the long placental lactogen I. Endocrinology 138, 756–763.
form of the prolactin receptor in the forebrain of the female rat. J. Comp. Neurol. Briefer, E.F., Padilla de la, T., McElligot, A.G., 2012. Mother goats do not forget their
386, 161–177. kids’ calls. Proc. R. Soc. B Biol. Sci. 279, 3749–3755.
Parental Behavior 109

Broad, K.D., Kendrick, K.M., Sirinathsinghji, D.J., et al., 1993. Changes in oxytocin Castro, D.C., Cole, S.L., Berridge, K.C., 2015. Lateral hypothalamus, nucleus
immunoreactivity and mRNA expression in the sheep brain during pregnancy, accumbens, and ventral pallidum roles in eating and hunger: interactions between
parturition and lactation and in response to oestrogen and progesterone. homeostatic and reward circuitry. Front. Syst. Neurosci. 9. Article 90.
J. Neuroendocrinol. 5, 435–444. Challis, J.R., Workewych, J.V., Patrick, J.E., 1981. Diurnal changes in the concen-
Broad, K.D., Lévy, F., Evans, G., et al., 1999. Previous maternal experience potentiates tration of progesterone in the plasma of women at 34–35 weeks of gestation.
the effect of parturition on oxytocin receptor mRNA expression in the para- J. Endocrinol. 89, 337–341.
ventricular nucleus. Eur. J. Neurosci. 11, 3725–3737. Challis, J.R.G., Sprague, C., Patrik, J.E., 1983. Relation between diurnal changes in
Brown, R.S., Herbison, A.E., Grattan, D.R., 2011. Differential changes in responses of peripheral plasma progesterone, cortisol, and estriol in normal women at 30–31,
hypothalamic and brainstem neuronal populations to prolactin during lactation in 34–35, and 38–39 weeks of gestation. Gynecol. Obstet. Investig. 16, 33–44.
the mouse. Biol. Reprod. 84, 826–836. Chaudron, L.H., Nirodi, N., 2011. The obsessive-compulsive spectrum in the perinatal
Burquière, E., Monteiro, P., Mallet, L., et al., 2015. Striatal circuits, habits, and period: a prospective pilot study. Arch. Women’s Ment. Health 13, 403–410.
implications for obsessive-compulsive disorder. Curr. Opin. Neurobiol. 30, 59–65. Cheung, S., Ballew, J.R., Moore, K.E., et al., 1998. Contribution of dopamine neurons
Byrnes, E.M., Bridges, R.S., 2005. Lactation reduces prolactin levels in reproductively in the medial zona incerta to the innervations of the central nucleus of the
experienced female rats. Horm. Behav. 48, 278–282. amygdala, horizontal diagonal band of Broca and hypothalamic paraventricular
Byrnes, E.M., Bridges, R.S., 2006a. Reproductive experience alters anxiety-like nucleus. Brain Res. 808, 174–181.
behavior in the female rat. Horm. Behav. 50, 70–76. Chirino, R., Beyer, C., González-Mariscal, G., 2007. Lesion to the main olfactory
Byrnes, E.M., Bridges, R.S., 2006b. Reproductive experience reduces the sedative, but epithelium facilitates maternal behavior in virgin rabbits. Behav. Brain Res. 180,
not anxiolytic effects of diazepam. Psychoneuroendocrinology 31, 988–996. 127–132.
Byrnes, E.M., Bridges, R.S., 2007. Reproductive experience and expression of Chirino, R., González-Mariscal, G., 2015. Changes in responsiveness to kit odors
Dopamine D2 receptor mRNA: a possible mechanism for reduced prolactin across pregnancy: relevance for the onset of maternal behavior. World Rabbit Sci.
secretion in primiparous rats. J. Neuroendocrinol. 19, 773–778. 23, 103–109.
Byrnes, E.M., Babb, J.A., Bridges, R.S., 2009. Differential expression of oestrogen Cohen, L., Mizrahi, A., 2015. Plasticity during motherhood: changes in excitatory and
receptor a following reproductive experience in young and middle-aged female inhibitory layer 2/3 neurons in auditory cortex. J. Neurosci. 35, 1806–1815.
rats. J. Neuroendocrinol. 21, 550–557. Conrad, L.C.A., Pfaff, D.W., 1976. Efferences from medial basal forebrain and
Byrnes, E.M., Byrnes, J.J., Bridges, R.S., 2001. Increased sensitivity of dopamine hypothalamus in the rat. I. Medial preoptic area. J. Comp. Neurol. 169, 185–220.
systems following reproductive experience in rats. Pharmacol. Biochem. Behav. 68, Consiglio, A.R., Lucion, A.B., 1996. Lesion of hypothalamic paraventricular nucleus
481–489. and maternal aggressive behavior in female rats. Physiol. Behav. 59, 591–596.
Byrnes, E.M., Casey, C., Carini, L.M., et al., 2013. Reproductive experience alters Cost, K.T., Lobell, T.D., Williams-Yee, Z.N., et al., 2014. The effects of pregnancy,
neural and behavioural responses to acute oestrogen receptor a activation. lactation, and primiparity on object-in-place memory of female rats. Horm. Behav.
J. Neuroendocrinol. 25, 1280–1289. 65, 32–39.
Byrnes, E.M., Casey, K., Bridges, R.S., 2012. Reproductive experience modifies the Craig, A.D., 2002. How do you feel? Interoception: the sense of the physiological
effects of estrogen receptor alpha activity on anxiety-like behavior and corticotropin condition of the body. Nat. Rev. Neurosci. 3, 655–666.
releasing hormone mRNA expression. Horm. Behav. 61, 44–49. Cruz, M.L., Beyer, C., 1972. Effects of septal lesions on maternal behavior and
Byrnes, E.M., Rigero, B.A., Bridges, R.S., 2002. Dopamine receptor antagonists during lactation in the rabbit. Physiol. Behav. 9, 361–365.
parturition disrupt maternal care and the retention of maternal behavior in rats. Cubero, J., Narciso, D., Terrón, P., et al., 2007. Chrononutrition applied to formula
Pharmacol. Biochem. Behav. 73, 869–875. milks to consolidate infant’s sleep/wake cycle. Neuro Endocrinol. Lett. 28,
Caba, M., González-Mariscal, G., 2009. The rabbit pup, a natural model of nursing- 360–366.
anticipatory activity. Eur. J. Neurosci. 30, 1697–1706. D’Cunha, T.M., King, S.J., Fleming, A.S., et al., 2011. Oxytocin receptors in the
Caba, M., Beyer, C., González-Mariscal, G., et al., 2003a. Immunocytochemical nucleus acumens shell are involved in the consolidation of maternal memory in
detection of estrogen receptor alpha in the female rabbit forebrain: topography and postpartum rats. Horm. Behav. 59, 14–21.
regulation by estradiol. Neuroendocrinology 77, 208–222. Da Costa, A.P., Broad, K.D., Kendrick, K.M., 1997. Olfactory memory and maternal
Caba, M., Rovirosa, M.J., Beyer, C., et al., 2003b. Immunocytochemical detection of behaviour-induced changes in c-fos and zif/268 mRNA expression in the sheep
progesterone receptor in the female rabbit forebrain: distribution and regulation by brain. Mol. Brain Res. 46, 63–76.
oestradiol and progesterone. J. Neuroendocrinol. 15, 855–864. Da Costa, A.P., Guevara-Guzman, R.G., Ohkura, S., et al., 1996a. The role of oxytocin
Caba, M., Silver, R., González-Mariscal, G., 1996. Oxytocin and vasopressin immu- release in the paraventricular nucleus in the control of maternal behaviour in the
noreactivity in rabbit hypothalamus during estrus, late pregnancy, and postpartum. sheep. J. Neuroendocrinol. 8, 163–177.
Brain Res. 720, 7–16. Da Costa, A.P., Wood, S., Ingram, C.D., et al., 1996b. Region-specific reduction in
Caba, M., Tovar, A., Silver, R., et al., 2008. Nature’s food anticipatory experiment: stress-induced c-fos mRNA expression during pregnancy and lactation. Brain Res.
entrainment of locomotor behavior, suprachiasmatic and dorsomedial hypothalamic 742, 177–184.
nuclei by suckling in rabbit pups. Eur. J. Neurosci. 27, 432–443. Damiola, F., Le Minh, N., Preitner, N., et al., 2000. Restricted feeding uncouples
Cabrera, V., Cantúa, D., Ramos, E., et al., 2009. Lactation is a natural model of circadian oscillators in peripheral tissues from the central pacemaker in the
hippocampus neuroprotection against excitotoxicity. Neurosci. Lett. 461, suprachiasmatic nucleus. Genes Dev. 14, 2950–2961.
136–139. Darnaudery, M., Perez-Martin, M., Del Favero, F., et al., 2007. Early motherhood in
Calamandrei, G., Keverne, E.B., 1994. Differential expression of Fos protein in the rats is associated with a modification of hippocampal function. Psychoneur-
brain of female mice dependent on pup sensory cues and maternal experience. oendocrinology 32, 803–812.
Behav. Neurosci. 108, 113–120. Davis, M., Walker, D.L., Miles, L., et al., 2010. Phasic vs sustained fear in rats and
Caligaris, L., Astrada, J.J., Taleisnik, S., 1971. Release of luteinizing hormone induced humans: role of the extended amygdala in fear vs anxiety. Neuropsychopharmacol.
by estrogen injection into ovariectomized rats. Endocrinology 88, 810–815. Rev. 35, 105–135.
Cardinal, R.N., Parkinson, J.A., Hall, J., et al., 2002. Emotion and motivation: the role Del Cerro, M.C., Izquierdo, M.A., Collado, P., et al., 1991. Bilateral lesions of the bed
of the amygdala, ventral striatum, and prefrontal cortex. Neurosci. Biobehav. Rev. nucleus of the accessory olfactory tract facilitate maternal behavior in virgin female
26, 321–352. rats. Physiol. Behav. 50, 67–71.
Carlson, N.R., Thomas, G.J., 1968. Maternal behavior of mice with limbic lesions. Del Cerro, M.C., Izquierdo, M.A., Rosenblatt, J.S., et al., 1995. Brain 2-deoxyglucose
J. Comp. Physiol. Psychol. 66, 731–737. levels related to maternal behavior-inducing stimuli in the rat. Brain Res. 696,
Carter, D.A., Lightman, S.L., 1987. Oxytocin responses to stress in lactating and 213–220.
hyperprolactinemic rats. Neuroendocrinology 46, 532–537. Di Carlo, R., Muccioli, G., 1981. Presence of specific prolactin binding sites in the
Carvalho-Freitas, M.I.R., Anselmo-Franci, J.A., Teodorov, E., et al., 2007. Repro- rabbit hypothalamus. Life Sci. 28, 2299–2307.
ductive experience modifies dopaminergic function, serum levels of prolactin, and Diamond, M.C., Johnson, R.E., Ingham, C., 1971. Brain plasticity induced by envi-
macrophage activity in female rats. Life Sci. 81, 128–136. ronment and pregnancy. Environ. Pregnancy, Int. J. Neurosci. 2, 4–5.
Casey, T., Crodian, J., Erickson, E., et al., 2014. Tissue-specific changes in molecular Dillon, K.M., Brooks, D., 1992. Unusual cleaning in the luteal phase. Psychol. Rep. 70,
clocks during the transition from pregnancy to lactation in mice. Biol. Reprod. 90, 35–39.
1–15. Engler, A.C., Hadash, A., Shehadeh, N., 2012. Breastfeeding may improve nocturnal
Casey, T., Patel, O., Dykema, K., et al., 2009. Molecular signatures reveal circadian sleep and reduce infantile colic: potential role of breast milk melatonin. Eur. J.
clocks may orchestrate the homeorhetic response to lactation. PLoS One 4, e7395. Pediatr. 171, 729–732.
110 Parental Behavior

Epstein, L.G., Phyllis, C., Zee, M.D., 2013. Infantile colic and migraine. JAMA 309, Gaffori, O., Le Moal, M., 1979. Disruption of maternal behavior and appearance of
1–2. cannibalism after ventral mesencephalic tegmentum lesions. Physiol. Behav. 23,
Everett, J.W., Sawyer, C.H., 1950. A 24-hour periodicity in the “LH-release apparatus” 317–323.
of female rats, disclosed by barbiturate sedation. Endocrinology 47, 198–218. Gamble, K.L., Resuehr, D., Johnson, C.H., 2013. Shift work and circadian dysregu-
Fahrbach, S.E., Pfaff, D.W., 1986. Effect of preoptic region implants of dilute estradiol lation of reproduction. Front. Endocrinol. 4 http://dx.doi.org/10.3389/
on the maternal behavior of ovariectomized, nulliparous rats. Horm. Behav. 20, fendo.2013.00092.
354–363. Gandelman, R., 1973. The ontogeny of maternal responsiveness in female Rockland-
Featherstone, R.E., Fleming, A.S., Ivy, G.O., 2000. Plasticity in the maternal circuit: Swiss mice. Horm. Behav. 4, 257–268.
effects of experience and partum condition on brain astrocyte number in female Gatewood, J.D., Morgan, M.D., Eaton, M., et al., 2005. Motherhood mitigates aging-
rats. Behav. Neurosci. 114, 158–172. related decrements in learning and memory. Brain Res. Bull. 66, 91–98.
Febo, M., Feliz-Ortiz, A.C., Johnson, T.R., 2010. Inactivation or inhibition of neuronal Giordano, A.L., Ahdieh, H.B., Mayer, A.D., et al., 1990. Cytosol and nuclear estrogen
activity in the medial prefrontal cortex largely reduces pup retrieval and grouping in receptor binding in the preoptic area and hypothalamus of female rats during
maternal rats. Brain Res. 1325C, 77–88. pregnancy and ovariectomized, nulliparous rats after steroid priming: correlation
Felicio, L.F., Canteras, N., 2008. Maternal choices: neural mediation – caring for with maternal behavior. Horm. Behav. 24, 232–255.
young or hunting? In: Bridges, R.S. (Ed.), Neurobiology of the Parental Brain. Giordano, A.L., Siegel, H.I., Rosenblatt, J.S., 1989. Nuclear estrogen receptor binding
Academic Press, San Diego, CA, pp. 75–82. in the preoptic area and hypothalamus of pregnancy-terminated rats: correlation
Felicio, L.F., Florio, J.C., Sider, L.H., et al., 1996. Reproductive experience increases with the onset of maternal behavior. Neuroendocrinology 50, 248–258.
striatal and hypothalamic dopamine levels in pregnant rats. Brain Res. Bull. 40, Giovenardi, M., Padoin, M.J., Cadore, L.P., et al., 1997. Hypothalamic paraventricular
253–256. nucleus, oxytocin, and maternal aggression in rats. Ann. N.Y. Acad. Sci. 807,
Ferguson, J.N., Aldag, J.M., Insel, T.R., 2001. Oxytocin in the medial amygdala is 606–609.
essential for social recognition in the mouse. J. Neurosci. 21, 8278–8285. Giovenardi, M., Padoin, M.J., Cadore, L.P., et al., 1998. Hypothalamic paraventricular
Ferreira, D.S., Amaral, F.G., Mesquita, C.C., et al., 2012. Maternal melatonin nucleus modulates maternal aggression in rats: effects of ibotenic acid lesion and
programs the daily pattern of energy metabolism in adult offspring. PLoS One 7, oxytocin antisense. Physiol. Behav. 63, 351–359.
e38795. González-Mariscal, G., 2007. Mother rabbits and their offspring: timing is everything.
Fleischer, S., Slotnick, B.M., 1978. Disruption of maternal behavior in rats with lesions Dev. Psychobiol. 49, 71–76.
of the septal area. Physiol. Behav. 21, 189–200. González-Mariscal, G., Caba, M., Martínez-Gómez, M., et al., 2016. Mothers and
Fleischman, D.S., Fessler, D.M.T., 2011. Progesterone’s effects on the psychology of offspring: the rabbit as a model system in the study of mammalian maternal
disease avoidance: support for the compensatory behavioral prophylaxis hypoth- behavior and sibling interactions. Horm. Behav. 77, 30–41.
esis. Horm. Behav. 59, 271–275. González-Mariscal, G., Gallegos, J.A., 2007. New Zealand white rabbits show non-
Fleming, A., Morgan, H.D., Walsh, C., 1996. Experiential factors in postpartum selective nursing in various types of nests. World Rabbit Sci. 15, 167–172.
regulation of maternal care. In: Rosenblatt, J.S., Snowdon, C.T. (Eds.), Advances in González-Mariscal, G., Poindron, P., 2002. Parental care in mammals: immediate and
the Study of Behavior, vol. 25. Academic Press, San Diego, CA, pp. 295–332. sensory factors of control. In: Pfaff, D., Arnold, A.P., Etgen, A.M., Fahrbach, S.E.,
Fleming, A.S., Korsmit, M., 1996. Plasticity in the maternal circuit: effects of maternal Rubin, R.T. (Eds.), Hormones, Brain and Behavior, first ed., vol. 1. Academic Press,
experience on Fos-Lir in hypothalamus, limbic and cortical structures in the San Diego, CA, pp. 215–298 (Chapter 2).
postpartum rat. Behav. Neurosci. 110, 567–582. González-Mariscal, G., Chirino, R., Beyer, C., et al., 2004a. Removal of the accessory
Fleming, A.S., Rosenblatt, J.S., 1974a. Olfactory regulation of maternal behavior in olfactory bulbs facilitates maternal behavior in virgin rabbits. Behav. Brain Res.
rats. I. Effects of olfactory bulb removal in experienced and inexperienced lactating 152, 89–95.
and cycling females. J. Comp. Physiol. Psychol. 86, 221–232. González-Mariscal, G., Chirino, R., Flores-Alonso, J.C., et al., 2004b. Intra-
Fleming, A.S., Rosenblatt, J.S., 1974b. Olfactory regulation of maternal behavior in cerebroventricular injections of bromocryptine counteract the antagonistic effect
rats. I. Effects of peripherally induced anosmia and lesions of the lateral olfactory of bromocriptine on rabbit maternal behaviour. J. Neuroendocrinol. 16,
tract in pup-induced virgins. J. Comp. Physiol. Psychol. 86, 233–246. 949–955.
Fleming, A.S., Sarker, J., 1990. Experience-hormone interactions and maternal González-Mariscal, G., Chirino, R., Rosenblatt, J.S., et al., 2005. Forebrain implants of
behavior in rats. Physiol. Behav. 47, 1165–1173. estradiol stimulate maternal nest-building in ovariectomized rabbits. Horm. Behav.
Fleming, A.S., Miceli, M., Moretto, D., 1983. Lesions of the medial preoptic area 47, 272–279.
prevent the facilitation of maternal behavior produced by amygdala lesions. Physiol. González-Mariscal, G., Cuamatzi, E., Rosenblatt, J.S., 1998a. Hormones and external
Behav. 31, 503–510. factors: are they “on/off” signals for maternal nest-building in rabbits? Horm.
Fleming, A.S., O’Day, D.H., Kraemer, G.W., 1999. Neurobiology of mother–infant Behav. 33, 1–8.
interactions: experience and central nervous system plasticity across development González-Mariscal, G., Díaz-Sánchez, V., Melo, A.I., et al., 1994. Maternal behavior in
and generations. Neurosci. Biobehav. Rev. 23, 673–685. New Zealand White rabbits: quantification of somatic events, motor patterns, and
Fleming, A.S., Suh, E.J., Korsmit, M., et al., 1994. Activation of Fos-like immunore- steroid plasma levels. Physiol. Behav. 55, 1081–1089.
activity in the medial preoptic area and limbic structures by maternal and social González-Mariscal, G., Jiménez, A., Chirino, R., et al., 2009. Motherhood and nursing
interactions in rats. Behav. Neurosci. 108, 724–734. stimulate c-FOS expression in the rabbit forebrain. Behav. Neurosci. 123,
Fleming, A.S., Vaccarino, F., Luebke, C., 1980. Amygdaloid inhibition of maternal 731–739.
behavior in the nulliparous female rat. Physiol. Behav. 25, 731–743. González-Mariscal, G., Kinsley, C.H., 2009. From indifference to ardor: the onset,
Forray, A., Focseneanu, M., Pittman, B., et al., 2010. Onset and exacerbation of maintenance and meaning of the maternal brain. In: Pfaff, D., Arnold, A.P.,
obsessive-compulsive disorder in pregnancy and the postpartum period. J. Clin. Etgen, A.M., Fahrbach, S.E., Rubin, R.T. (Eds.), Hormones, Brain and Behavior,
Psychiatry 71, 1061–1068. second ed., vol. 1. Academic Press, San Diego, CA, pp. 109–136 (Chapter 3).
Franca, E.L., Nicomedes, T.R., Calderón, I.M.P., et al., 2010. Time-dependent alter- González-Mariscal, G., Lemus, A.C., Aguilar-Roblero, R., 2015. Contribution of
ations of soluble and cellular components in human milk. Biol. Rhythms Res. 41, suckling stimulation to the daily periodic display of nursing behavior in non-lactating
333–347. virgin rabbits. J. Neurol. Neurophysiol. 6, 327. http://dx.doi.org/10.4172/2155-
Franssen, R.A., Rzucidlo, A.M., Franssen, C.L., et al., 2012. Reproductive experience 9562.1000327.
facilitates recovery from kainic acid-induced neural insult in female Long–Evans González-Mariscal, G., Lemus, A.C., Vega-González, A., et al., 2013a. Litter size
rats. Brain Res. 1454, 80–89. determines circadian periodicity of nursing in rabbits. Chronobiol. Int. 30,
Franz, J.R., Leo, R.J., Steuer, M.A., et al., 1986. Effects of hypothalamic knife cuts 711–718.
and experience on maternal behavior in the rat. Physiol. Behav. 38, 629–640. González-Mariscal, G., Melo, A.I., 2013. Parental behavior. In: Pfaff, D. (Ed.),
Freeman, M.E., Neill, J., 1972. The pattern of prolactin secretion during pseudo- Neuroscience in the 21st Century, first ed., vol. 4. Springer-Verlag, New York,
pregnancy in the rat: a daily nocturnal surge. Endocrinology 90, 1292–1294. pp. 2069–2100.
Fuchs, A.R., Cubile, L., Dawood, M.Y., et al., 1984. Release of oxytocin and prolactin González-Mariscal, G., Melo, A.I., Chirino, R., et al., 1998b. Importance of mother/
by suckling rabbits throughout lactation. Endocrinology 114, 462–469. young contact at parturition and across lactation for the expression of maternal
Fuchs, A.R., Dawood, M.Y., 1980. Oxytocin release and uterine activation during behavior in rabbits. Dev. Psychobiol. 32, 101–111.
parturition in rabbits. Endocrinology 107, 1117–1126. González-Mariscal, G., Melo, A.I., Jiménez, P., et al., 1996. Estradiol, progesterone,
Furuta, M., Bridges, R.S., 2005. Gestation-induced cell proliferation in the rat brain. and prolactin regulate maternal nest-building in rabbits. J. Neuroendocrinol. 8,
Dev. Brain Res. 156, 61–66. 901–907.
Parental Behavior 111

González-Mariscal, G., Melo, A.I., Parlow, A.F., et al., 2000. Pharmacological evidence Hofman, S.G., Fang, A., Brager, D.N., 2015. Effect of intranasal oxytocin adminis-
that prolactin acts from late gestation to promote maternal behavior in rabbits. tration on psychiatric symptoms: a meta-analysis of placebo-controlled studies.
J. Neuroendocrinol. 12, 983–992. Psychiatry Res. 228, 708–714.
González-Mariscal, G., Toribio, A., Gallegos-Huicochea, J.A., et al., 2013b. The Hucke, E.E.T.S., Cruz-Casallas, P.E., Florio, J.C., et al., 1998. Reproductive experience
characteristics of suckling stimulation determine the daily duration of mother-young reduces striatal dopaminergic responses in freely moving female rats. Neuroreport
contact and milk output in rabbits. Dev. Psychobiol. 55, 809–817. 16, 3589–3593.
Gottesman II, Gould, T.D., 2003. The endophenotype concept in psychiatry: etymology Hucke, E.E.T.S., Cruz-Casallas, P.E., Sider, L.H., et al., 2001. Reproductive experience
and strategic intentions. Am. J. Psychiatry 160, 636–645. modulates dopamine-related behavioral responses. Pharmacol. Biochem. Behav.
Graham, M.D., Rees, S.L., Steiner, M., et al., 2006. The effects of adrenalectomy and 68, 575–582.
corticosterone replacement on maternal memory in postpartum rats. Horm. Behav. Insel, T.R., 1990. Regional changes in brain oxytocin receptors post-partum: time-
49, 353–361. course and relationship to maternal behaviour. J. Neuroendocrinol. 2, 539–545.
Grattan, D.R., Kokay, I.C., 2008. Prolactin: a pleiotropic neuroendocrine hormone. J. Insel, T.R., 2007. From animal models to model animals. Biol. Psychiatry 62,
Neuroendocrinol. 20, 752–763. 1337–1339.
Gray, P., Brooks, P.J., 1984. Effect of lesion location within the medial preoptic- Insel, T.R., Harbaugh, C.R., 1989. Lesions of the hypothalamic paraventricular nucleus
anterior hypothalamic continuum on maternal and male sexual behaviors in disrupt the initiation of maternal behavior. Physiol. Behav. 45, 1033–1041.
female rats. Behav. Neurosci. 98, 703–711. Insel, T.R., Shapiro, L.E., 1992. Oxytocin receptor distribution reflects social organi-
Gregg, C., Shikar, V., Larsen, P., et al., 2007. White matter plasticity and enhanced zation in monogamous and polygamous voles. Proc. Natl. Acad. Sci. U.S.A. 89,
remyelination in the maternal CNS. J. Neurosci. 27, 1812–1823. 5981–5985.
Gregg, J.K., Wynne-Edwards, K.E., 2005. Placentophagia in naïve adults, new fathers, Jacobson, C.D., Terkel, J., Gorski, R.A., et al., 1980. Effects of small medial preoptic
and new mothers in the biparental dwarf hamster, Phodopus campbelli. Dev. area lesions on maternal behavior: retrieving and nest building in the rat. Brain Res.
Psychobiol. 47, 179–188. 194, 471–478.
Gregg, J.K., Wynne-Edwards, K.E., 2006. In uniparental Phodopus sungorus new Jilge, B., Kuhnt, B., Landerer, W., et al., 2001. Circadian temperature rhythms in
mothers and fathers, present during the birth of their offspring, are the only rabbit pups and in their does. Lab. Anim. 35, 364–373.
hamsters that readily consume fresh placenta. Dev. Psychobiol. 48, 528–536. Jiménez, A., Young, L.J., Triana-Del Río, R., et al., 2015. Neuroanatomical distribution
Guglielmi, V., Vulink, N.C.C., Denys, D., et al., 2014. Obsessive-compulsive disorder of oxytocin receptor binding in the female rabbit forebrain: variations across the
and female reproductive cycle events: results from the OCD and reproduction reproductive cycle. Brain Res. 1629, 329–339.
collaborative study. Depress. Anxiety 31, 979–987. Jirikowski, G.F., Caldwell, J.D., Pilgrim, C., et al., 1989. Changes in immunostaining
Haber, S., 2016. Corticostriatal circuitry. Dialogues Clin. Neurosci. 18, 7–21. for oxytocin in the forebrain of the female rat during late pregnancy, parturition and
Hansen, S., 1994. Maternal behavior of female rats with 6-OHDA lesions in the ventral early lactation. Cell Tissue Res. 256, 411–417.
striatum: characterization of the pup retrieval deficit. Physiol. Behav. 55, 615–620. Johnson, Z.V., Young, L.J., 2015. Neurobiological mechanisms of social attachment
Hansen, S., Ferreira, A., 1986. Food intake, aggression, and fear behavior in the and pair bonding. Curr. Opin. Sci. 3, 38–44.
mother rat: control by neural systems concerned with milk ejection and maternal Jones, J.S., Wynne-Edwards, K.E., 2000. Paternal hamsters mechanically assist the
behavior. Behav. Neurosci. 100, 64–70. delivery, consume amniotic fluid and placenta, remove fetal membranes, and
Hansen, S., Gummesson, B.M., 1982. Participation of the lateral midbrain tegmentum provide parental care during the birth process. Horm. Behav. 37, 116–125.
in the neuroendocrine control of sexual behavior and lactation in the rat. Brain Res. Junkermann, H., Mangold, H., Vecsel, P., et al., 1982. Circadian rhythm of serum
251, 319–325. progesterone levels in human pregnancy and its relation to the rhythm of cortisol.
Hansen, S., Kohler, C., 1984. The importance of the peripeduncular nucleus in the Acta Endocrinol. (Copenh.) 10, 98–104.
neuroendocrine control of sexual behavior and milk ejection in the rat. Neuroen- Kalinichev, M., Rosenblatt, J.S., Nakabeppu, Y., et al., 2000. Induction of c-Fos-like
docrinology 39, 563–572. and FosB-like immunoreactivity reveals forebrain neuronal populations involved
Hauser, H., Gandelman, R., 1985. Lever pressing for pups: evidence for hormonal differentially in pup-mediated maternal behavior in juvenile and adult rats.
influence upon maternal behavior of mice. Horm. Behav. 19, 454–468. J. Comp. Neurol. 416, 45–78.
Henderson, J.M., France, K.G., Owens, J.L., Blampied, N.M., 2010. Sleeping through Kas, M.J.H., Fernandes, C., Schalkwyk, L.C., et al., 2007. Genetics of behavioural
the night: the consolidation of self-regulated sleep across the first year of life. domains across the neuropsychiatric spectrum; of mice and men. Mol. Psychiatry
Pediatrics 126, e1081–e1087. 12, 324–330.
Herrenkohl, L.R., Rosenberg, P.A., 1972. Exteroceptive stimulation of maternal Kas, M.J.H., Krishnan, V., Gould, T.D., et al., 2011. Advances in multidisciplinary and
behavior in the naive rat. Physiol. Behav. 8, 595–598. cross-species approaches to examine the neurobiology of psychiatric disorders.
Herrenkohl, L.R., Rosenberg, P.A., 1974. Effects of hypothalamic deafferentation late Eur. Neuropsychopharmacol. 21, 532–544.
in gestation on lactation and nursing behavior in the rat. Horm. Behav. 5, 33–41. Kawakami, M., Arita, J., Yoshioka, E., 1980. Loss of estrogen-induced surges of
Herrenkohl, L.R., Sachs, B.D., 1972. Meeting report: sensory regulation of maternal prolactin and gonadotropins by suprachiasmatic nucleus lesions in ovariectomized
behavior in mammals. Physiol. Behav. 9, 689–692. rats. Endocrinology 106, 1087–1092.
Higuchi, T., Bicknell, R.J., Leng, G., 1991. Reduced oxytocin release from the neural Keebaugh, A.C., Young, L.J., 2011. Increasing oxytocin receptor expression in the
lobe of lactating rats is associated with reduced pituitary content and does not nucleus accumbens of pre-pubertal female prairie voles enhances alloparental
reflect reduced excitability of oxytocin neurons. J. Neuroendocrinol. 3, 297–302. responsiveness and partner preference formation as adults. Horm. Behav. 60,
Hipwell, A.E., Guo, C., Phillips, M.L., et al., 2015. Right frontoinsular cortex and 498–504.
subcortical activity to infant cry is associated with maternal mental state talk. Keebaugh, A.C., Barrett, C.E., Laprairie, J.L., et al., 2015. RNAi knockdown of oxytocin
J. Neurosci. 35, 12725–12732. receptor in the nucleus accumbens inhibits social attachment and parental care in
Hoffman, K.L., 2011. Animal models of obsessive compulsive disorder: recent findings monogamous female prairie voles. Soc. Neurosci. http://dx.doi.org/10.1080/
and future directions. Expert Opin. Drug Discov. 6, 725–737. 17470919.2015.1040893.
Hoffman, K.L., 2013. Role of murine models in psychiatric illness drug discovery: Keer, S.E., Stern, J.M., 1999. Dopamine receptor blockade in the nucleus accumbens
a dimensional view. Expert Opin. Drug Discov. 8, 865–877. inhibits maternal retrieval and licking, but enhances nursing behavior in lactating
Hoffman, K.L., 2015. Modeling Neuropsychiatric Disorders in Laboratory Animals, first rats. Physiol. Behav. 67, 659–669.
ed. Woodhead Publishing, Cambridge. Keller, M., Meurisse, M., Lévy, F., 2005. Mapping of brain networks involved in
Hoffman, K.L., 2016. New dimensions in the use of rodent behavioral tests for novel consolidation of lamb recognition memory. Neuroscience 133, 359–369.
drug discovery. Expert Opin. Drug Discov. 11, 343–353. Kendrick, K.M., Da Costa, A.P., Broad, K.D., et al., 1997. Neural control of maternal
Hoffman, K.L., Rueda-Morales, R.I., 2009. Toward an understanding of the neurobi- behaviour and olfactory recognition of offspring. Brain Res. Bull. 44, 383–395.
ology of “just right” perceptions: nest building in the female rabbit as a possible Kendrick, K.M., Keverne, E.B., Baldwin, B.A., 1987. Intracerebroventricular OT stim-
model for compulsive behavior and task completion. Behav. Brain Res. 204, ulates maternal behaviour in the sheep. Neuroendocrinology 46, 56–61.
182–191. Kendrick, K.M., Lévy, F., Keverne, E.B., 1991. Importance of vaginocervical stimulation
Hoffman, K.L., Rueda-Morales, 2012. RI D1 and D2 dopamine receptor antagonists for the formation of maternal bonding in primiparous and multiparous parturient
decrease behavioral bout duration, without altering the bout’s repeated behavioral ewes. Physiol. Behav. 50, 595–600.
components, in a naturalistic model of repetitive and compulsive behavior. Behav. Kendrick, K.M., Lévy, F., Keverne, E.B., 1992. Changes in the sensory processing of
Brain Res. 230, 1–10. olfactory signals induced by birth in sheep. Science 256, 833–836.
112 Parental Behavior

Kennaway, D.J., Goble, F.C., Stamp, G.E., 1996. Factors influencing the development Lemaire, V., Billard, J.M., Dutar, P., 2006. Motherhood-induced memory improvement
of melatonin rhythmicity in humans. J. Clin. Endocrinol. Metab. 81, 1525–1532. persists across lifespan in rats but is abolished by a gestational stress. Eur. J.
Kenyon, P., Cronin, P., Keeble, S., 1983. Role of the infraorbital nerve in retrieving Neurosci. 23, 3368–3374.
behavior in lactating rats. Behav. Neurosci. 97, 255–269. Leon, M., Adels, L., Coopersmith, R., et al., 1984. Diurnal cycle of mother-young
Kenyon, P., Cronin, P., Keeble, S., 1981. Disruption of maternal retrieving by perioral contact in Norway rats. Physiol. Behav. 32, 999–1003.
anesthesia. Physiol. Behav. 27, 313–321. Leuner, B., Gould, E., 2010. Dendritic growth in medial prefrontal cortex and cognitive
Keyser-Marcus, L., Stafisso-Sandoz, G., Gerecke, K., et al., 2001. Alterations of flexibility are enhanced during the postpartum period. J. Neurosci. 30, 13499–
medial preoptic area neurons following pregnancy and pregnancy-like steroidal 13503.
treatment in the rat. Brain Res. Bull. 55, 737–745. Leuner, B., Mirescu, C., Noiman, L., et al., 2007. Maternal experience inhibits the
Kim, P., Feldman, R., Mayes, L.C., et al., 2011. Breastfeeding, brain activation to own production of immature neurons in the hippocampus during the postpartum period
infant cry, and maternal sensitivity. J. Child Psychol. Psychiatry 52, 907–915. through elevations in adrenal steroids. Hippocampus 17, 434–442.
Kinsley, C.H., Bridges, R.S., 1988. Parity associated reductions in behavioral sensitivity Lévy, F., Keller, M., Poindron, P., 2004. Olfactory regulation of maternal behavior in
to opiates. Biol. Reprod. 39, 270–278. mammals. Horm. Behav. 46, 284–302.
Kinsley, C.H., Lambert, K.G., 2006. The maternal brain: pregnancy and motherhood Lévy, F., Kendrick, K.M., Goode, J., et al., 1995. OT and vasopressin release in the
change the structure of the female mammal’s brain making mothers attentive to olfactory bulb of parturient ewes: changes with maternal experience and effects on
their young and better at caring for them. Sci. Am. 294, 72–79. acetylcholine, gamma-aminobutyric acid, glutamate and noradrenaline release.
Kinsley, C.H., Blair, J.C., Karp, N.E., et al., 2014. The mother as hunter: significant Brain Res. 669, 197–206.
reduction in foraging costs through enhancements of predation in maternal rats. Lewy, A.J., Wehr, T., Goodwin, F.K., Newsome, D.A., Markey, S.P., 1980. Light
Horm. Behav. 66, 649–654. suppresses melatonin secretion in humans. Science 210, 1267–1269.
Kinsley, C.H., Madonia, L., Gifford, G.W., et al., 1999. Motherhood improves learning Li, M., Fleming, A.S., 2003. Differential involvement of nucleus accumbens shell and
and memory: neural activity in rats is enhanced by pregnancy and the demands of core subregions in maternal memory in postpartum female rats. Behav. Neurosci.
rearing offspring. Nature 402, 137–138. 117, 426–445.
Kinsley, C.H., Trainer, R., Stafisso-Sandoz, G., et al., 2006. Motherhood and the Lincoln, D.W., Hill, A., Wakerley, J.D., 1973. The milk-ejection reflex on the rat: an
hormones of pregnancy modify concentrations of hippocampal neural dendritic intermittent function not abolished by surgical levels of anesthesia. J. Endocrinol.
spines. Horm. Behav. 49, 131–142. 57, 459–476.
Komisaruk, B.R., Rosenblatt, J.S., Barona, M.L., et al., 2000. Combined c-fos and Lohstroh, P.N., Chen, J., Ba, J., et al., 2003. Bone resorption is affected by follicular
14
C-2-deoxyglucose method to differentiate site-specific excitation from disinhibi- phase length in female rotating shift workers. Environ. Health Perspect. 111,
tion: analysis of maternal behavior in the rat. Brain Res. 859, 262–272. 618–622.
Kovanen, L., Saarikoski, S.T., Aromaa, A., et al., 2010. ARNTL (BMAL1) and NPAS2 Lonstein, J.S., De Vries, G.J., 2000. Maternal behaviour in lactating rats stimulates
gene variants contribute to fertility and seasonality. PLoS One 5, e10007. http:// c-fos in glutamate decarboxylase synthesizing neurons of the medial preoptic area,
dx.doi.org/10.1371/journal.pone.0010007. ventral bed nucleus of the stria terminalis, and ventrocaudal periaqueductal gray.
Krehbiel, D., Poindron, P., Levy, F., et al., 1987. Peridural anesthesia disturbs maternal Neuroscience 100, 557–568.
behavior in primiparous and multiparous parturient ewes. Physiol. Behav. 40, Lonstein, J.S., Greco, B., De Vries, G., et al., 2000. Maternal behavior stimulates c-fos
463–472. activity within estrogen receptor alpha-containing neurons in lactating rats.
Krémarik, P., Freund-Mercier, M.J., Stoeckel, M.E., 1993. Histoautoradiographic Neuroendocrinology 72, 91–101.
detection of oxytocin- and vasopressin-binding sites in the telencephalon of the rat. Lopantina, O., Inzhutova, A., Pichugina, Y.A., et al., 2011. Reproductive experience
J. Comp. Neurol. 333, 343–359. affects parental retrieval behaviour associated with increased plasma oxytocin
Kringelbach, M.L., Rolls, E.T., 2004. The functional anatomy of the human orbitofrontal levels in wild-type and Cd38-knockout mice. J. Neuroendocrinol. 23,
cortex: evidence from neuroimaging and neuropsychology. Prog. Neurobiol. 72, 1125–1133.
341–372. Lorderbaum, J.P., Newman, J.D., Dubno, J.R., et al., 1999. Feasibility of using fMRI to
Kringelbach, M.L., O’Doherty, J., Rolls, E.T., et al., 2003. Activation of the human study mothers responding to infant cries. Depress. Anxiety 10, 99–104.
orbitofrontal cortex to a liquid food stimulus is correlated with its subjective Lorderbaum, J.P., Newman, J.D., Horwitz, A.R., et al., 2002. A potential role for
pleasantness. Cereb. Cortex 13, 1064–1071. thalamocingulate circuitry in human maternal behavior. Biol. Psychiatry 51,
Kristal, M.B., 1991. Enhancement of opioid-mediated analgesia: a solution to the 431–445.
enigma of placentophagia. Neurosci. Biobehav. Rev. 15, 425–435. Love, G., Torrey, N., McNamara, I., et al., 2005. Maternal experience produces long-
Krittrell, E.M., Satinoff, E., 1988. Diurnal rhythms of body temperature, drinking and lasting behavioral modifications in the rat. Behav. Neurosci. 119, 1084–1096.
activity over reproductive cycle. Physiol. Behav. 42, 477–484. Macbeth, A.H., Luine, V.N., 2010. Changes in anxiety and cognition due to repro-
Labad, J., Menchón, J.M., Alonso, P., et al., 2005. Female reproductive cycle and ductive experience: a review of data from rodent and human mothers. Neurosci.
obsessive-compulsive disorder. J. Clin. Psychiatry 66, 428–435. Biobehav. Rev. 34, 452–467.
Lakati, A., Binns, C., Stevenson, M., 2002. The effect of work status on exclusive Macbeth, A.H., Gautreaux, C., Luine, V.N., 2008a. Pregnant rats show enhanced
breastfeeding in Nairobi. Asia Pac. J. Public Health 14, 85–90. spatial memory, decreased anxiety, and altered levels of monoaminergic neuro-
Lambert, K.G., Berry, A.E., Griffin, G., et al., 2005. Pup exposure differentially transmitters. Brain Res. 1241, 136–147.
enhances foraging ability in primiparous and nulliparous rats. Physiol. Behav. 84, Macbeth, A.H., Scharfman, H.E., MacLusky, N.J., et al., 2008b. Effects of multiparity
799–806. on recognition memory, monoaminergic neurotransmitters, and brain-derived
Landgraf, F., Neumann, I.D., 2004. Vasopressin and oxytocin release within the brain: neurotrophic factor (BDNF). Horm. Behav. 54, 7–17.
a dynamic concept of multiple and variable modes of neuropeptide communication. Maina, G., Albert, U., Bogetto, F., et al., 1999. Recent life events and obsessive-
Front. Neuroendocrinol. 25, 150–176. compulsive disorder (OCD): the role of pregnancy/delivery. Psychiatry Res. 89,
Larsen, C.M., Grattan, D.R., 2010. Prolactin-induced mitogenesis in the sub- 49–58.
ventricular zone of the maternal brain during early pregnancy is essential for Maningat, P.D., Sen, P., Rijnkels, M., et al., 2009. Gene expression in the human
normal postpartum behavioral responses in the mother. Endocrinology 151, mammary epithelium during lactation: the milk fat globule transcriptome. Physiol.
3805–3814. Genom. 37, 12–22.
Leckman, J.F., Mayes, L.C., Feldman, R., et al., 1999. Early parental preoccupations Mann, P.E., Bridges, R.S., 1992. Neural and endocrine sensitivities to opiods decline
and behaviors and their possible relationship to the symptoms of obsessive- as a function of multiparity in rats. Brain Res. 580, 241–248.
compulsive disorder. Acta Psychiatr. Scand. Suppl. 396, 1–26. Marques, D.M., Malsbury, C.W., Daood, J., 1979. Hypothalamic knife cuts dissociate
Lee, A., Clancy, S., Fleming, A.S., 2000. Mother rats bar-press for pups: effects of maternal behaviors, sexual receptivity and estrous cyclicity in female hamsters.
lesions of the MPOA and limbic sites on maternal behavior and operant responding Physiol. Behav. 23, 347–355.
for pup-reinforcement. Behav. Brain Res. 108, 215–231. Martin, D.L., Rimvall, K., 1993. Regulation of gamma-aminobutyric acid synthesis in
Lee, A.W., Brown, R.E., 2002. The presence of the male facilitates parturition in the brain. J. Neurochem. 60, 395–407.
California mice (Peromyscus californicus). Can. J. Zool. 80, 926–933. Matthysse, S., 1986. Animal models in psychiatric research. Prog. Brain Res. 65,
Leibenluft, E., Gobbini, M.I., Harrison, T., et al., 2004. Mother’s neural activation in 259–270.
response to pictures of their children and other children. Biol. Psychiatry 56, Mayer, A.D., Rosenblatt, J.S., 1979. Hormonal influences during the ontogeny of
225–232. maternal behavior in female rats. J. Comp. Physiol. Psychol. 93, 879–898.
Parental Behavior 113

Maywood, E.S., Mrosovsky, N., Field, M.D., et al., 1999. Rapid down-regulation of Nephew, B.C., Bridges, R.S., Lovelock, D.F., et al., 2009. Enhanced maternal
mammalian period genes during behavioral resetting of the circadian clock. Proc. aggression and associated changes in neuropeptide gene expression in multipa-
Natl. Acad. Sci. U.S.A. 96, 15211–15216. rous rats. Behav. Neurosci. 123, 949–957.
McGuinness, M., Blisset, J., Jones, C., 2011. OCD in the perinatal period: is post- Nephew, B.C., Byrnes, E.M., Bridges, R.S., 2010. Vasopressin mediates
partum OCD (ppOCD) a distinct subtype? A review of the literature. Behav. Cogn. enhanced offspring protection in multiparous rats. Neuropharmacology 58,
Psychother. 39, 285–310. 102–106.
Melo, A.I., González-Mariscal, G., 2003. Placentophagia in rabbits: incidence across Neumann, I.D., Johnstone, H.A., Hatzinger, M., et al., 1998. Attenuated neuroendo-
the reproductive cycle. Dev. Psychobiol. 43, 37–43. crine responses to emotional and physical stressor in pregnant rats involve
Mena, F., Grosvenor, C.E., 1972. Effects of suckling and exteroceptive stimulation adenohypophysial changes. J. Physiol. 58, 289–300.
upon prolactin release in the rat during late lactation. J. Endocrinol. 52, 11–22. Neumann, I.D., Russell, J.A., Landgraf, R., 1993. Oxytocin and vasopressin release
Mendelson, C.R., 2009. Minireview: fetal-maternal hormonal signaling in pregnancy within the supraoptic and paraventricular nuclei of pregnant, parturient and
and labor. Mol. Endocrinol. 23, 947–954. lactating rats: a microdialysis study. Neuroscience 53, 65–75.
Mendez, N., Abarzua-Catalan, L., Vilches, N., et al., 2012. Timed maternal melatonin Neumann, I.D., Torner, L., Wigger, A., 2000. Brain oxytocin: differential inhibition of
treatment reverses circadian disruption of the fetal adrenal clock imposed by neuroendocrine stress responses and anxiety-related behaviour in virgin, pregnant
exposure to constant light. PLoS One 7, e42713. http://dx.doi.org/10.1371/ and lactating rats. Neuroscience 95, 567–575.
journal.pone.0042713. Neziroglu, F., Anemone, R., Yaryura-Tobias, J.A., 1996. Onset of obsessive-
Menzies, L., Chamberlain, S.R., Laird, A.R., et al., 2008. Integrating evidence compulsive disorder in pregnancy. Am. J. Psychiatry 149, 947–950.
from neuroimaging and neuropsychological studies of obsessive-compulsive Nitchke, J.B., Nelson, E.E., Rusch, B.D., et al., 2003. Orbitofrontal cortex tracks
disorder: the orbitofronto-striatal model revisited. Neurosci. Biobehav. Rev. positive mood in mothers viewing pictures of their newborn infants. NeuroImage
32, 525–549. 21, 583–592.
Metz, R.P., Qu, X., Laffin, B., et al., 2006. Circadian clock and cell cycle gene Nogueira, T.C., Lellis-Santos, C., Jesus, D.S., 2011. Absence of melatonin induces
expression in mouse mammary epithelial cells and in the developing mouse night-time hepatic insulin resistance and increased gluconeogenesis due to
mammary gland. Dev. Dyn. 235, 263–271. stimulation of nocturnal unfolded protein response. Endocrinology 152,
Meza, E., Aguirre, J., Waliszewski, S., et al., 2015. Suckling induces a daily rhythm in 1253–1263.
the preoptic area and lateral septum but not in the bed nucleus of the stria ter- Noirot, E., 1972. The onset of maternal behavior in rats, hamsters and mice. In:
minalis in lactating rabbit does. Eur. J. Neurosci. 41, 196–204. Lehrman, D.S., Hinde, R.A., Shaw, E. (Eds.), Advances in the Study of Behavior.
Meza, E., Juárez, C., Morgado, E., et al., 2008. Brief daily suckling shifts locomotor Academic Press, New York, pp. 107–145.
behavior and induces PER1 protein in paraventricular and supraoptic nuclei, but Noriuchi, M., Kiduchi, Y., Senoo, A., 2008. The functional neuroanatomy of maternal
not in the suprachiasmatic nucleus of rabbit does. Eur. J. Neurosci. 28, love: mother’s response to infant’s attachment behaviors. Biol. Psychiatry 63,
1394–1403. 415–423.
Meza, E., Waliszewski, S.M., Caba, M., 2011. Circadian nursing induces PER1 protein Nowak, R., Keller, M., Lévy, F., 2011. Mother-young relationships in sheep: a model
in neuroendocrine tyrosine hydroxylase neurons in the rabbit doe. for a multidisciplinary approach of the study of attachment in mammals. J. Neu-
J. Neuroendocrinol. 23, 472–480. roendocrinol. 23, 1042–1053.
Miceli, M.O., Malsbury, C.W., 1982. Sagittal knife cuts in the near and far lateral Nowak, R., Keller, M., Val-Laillet, D., Lévy, F., 2007. Perinatal visceral events and
preoptic area–hypothalamus disrupt maternal behavior in female hamsters. Physiol. brain mechanisms involved in the development of mother–young bonding in sheep.
Behav. 28, 857–867. Horm. Behav. 52, 92–98.
Miceli, M.O., Fleming, A.S., Malsbury, C.W., 1983. Disruption of maternal behaviour in Nudo, R.J., Masterton, R.B., 1986. Stimulation-induced 14C-2-deoxyglucose
virgin and postparturient rats following sagittal plane knife cuts in the preoptic labeling of synaptic activity in the central auditory system. J. Comp. Neurol.
area–hypothalamus. Behav. Brain Res. 9, 337–360. 245, 553–565.
Miller, B.H., Oleon, S.L., Turek, F.W., et al., 2004. Circadian clock mutation disrupts Numan, M., 1974. Medial preoptic area and maternal behavior in the female rat.
estrous cyclicity and maintenance of pregnancy. Curr. Biol. 4, 1367–1373. J. Comp. Physiol. Psychol. 87, 746–759.
Miller, S.M., Piasecki, C.C., Lonstein, J.S., 2011. Use of the light–dark box to compare Numan, M., 1996. A lesion and neuroanatomical tract-tracing analysis of the role of
the anxiety-related behavior of virgin and postpartum female rats. Pharmacol. the bed nucleus of the stria terminalis in retrieval behavior and other aspects of
Biochem. Behav. 100, 130–137. maternal responsiveness in rats. Dev. Psychobiol. 29, 23–51.
Miller, B.H., Takahashi, J.S., 2014. Central circadian control of female reproductive Numan, M., 2006. Neurobiology of Social Behavior. Academic Press, San Diego.
function. Front. Endocrinol. 4. http://dx.doi.org/10.3389/fendo.2013.00195. Numan, M., Callahan, E.C., 1980. The connections of the medial preoptic region and
Moltz, H., Robbins, D., Parks, M., 1966. Caesarean delivery and maternal behavior of maternal behavior in the rat. Physiol. Behav. 25, 653–665.
primiparous and multiparous rats. J. Comp. Physiol. Psychol. 61, 455–460. Numan, M., Corodimas, K.P., 1985. The effects of paraventricular hypothalamic
Monasterio, N., Ramos, E., Morales, T., 2008. Changes in c-Fos and NOS expression lesions on maternal behavior in rats. Physiol. Behav. 35, 417–425.
in the PVH of lactating rats in response to excitotoxicity and stress. Stress, Numan, M., Insel, T.R., 2003. The Neurobiology of Parental Behavior. Springer,
Neurotransmitters, and Hormones Ann. N.Y. Acad. Sci. 1148, 161–164. New York.
Montgomery-Downs, H., Clawges, H.M., Santy, E.E., 2010. Infant feeding methods Numan, M., Numan, M.J., 1991. Preoptic–brainstem connections and maternal
and maternal sleep and daytime functioning. Pediatrics 126, e1562–1568. behavior in rats. Behav. Neurosci. 105, 1013–1029.
Morais, T.C., Honorio-Franca, A.C., Rodríguez, R., 2015. Temporal fluctuations of Numan, M., Numan, M.J., 1994. Expression of Fos-like immunoreactivity in the
cytokine concentrations in human milk. Biol. Rhythms Res. 46, 811–821. preoptic area of maternally behaving virgin and postpartum rats. Behav. Neurosci.
Morales, T., 2011. Recent findings on neuroprotection against excitotoxicity in the 108, 379–394.
hippocampus of female rats. J. Neuroendocrinol. 23, 994–1001. Numan, M., Numan, M.J., 1995. Importance of pup-related sensory inputs and
Morgan, H.D., Watchus, J.A., Milgram, N.W., et al., 1999. The long lasting effects of maternal performance for the expression of fos-like immunoreactivity in the pre-
electrical simulation of the medial preoptic area and medial amygdala on maternal optic area and ventral bed nucleus of the stria terminalis of postpartum rats. Behav.
behavior in female rats. Behav. Brain Res. 99, 61–73. Neurosci. 109, 135–149.
Muccioli, G., Di Carlo, R., 1994. Modulation of prolactin receptors in the rat hypo- Numan, M., Smith, H.G., 1984. Maternal behavior in rats: evidence for the involvement
thalamus in response to changes in serum concentration of endogenous prolactin of preoptic projections to the ventral tegmental area. Behav. Neurosci. 98,
or to ovine prolactin administration. Brain Res. 663, 244–250. 712–727.
Myers, K., Poole, W.E., 1961. A study of the biology of the wild rabbit, Oryctolagus Numan, M., Stolzenberg, D.S., 2009. Medial preoptic area interactions with dopamine
cuniculus (L), in confined populations. II. The effect of season and population neural systems in the control of the onset and maintenance of maternal behavior in
increase on behaviour. CSIRO Wildl. Res. 6, 1–41. rats. Front. Neuroendocrinol. 30, 46–64.
Nadaud, D., Simon, H., Herman, J.P., et al., 1984. Contributions of the mesencephalic Numan, M., Young, L.J., 2016. Neural mechanisms of mother-infant bonding and pair
dopaminergic system and the trigeminal sensory pathway to the ventral tegmental bonding: similarities, differences, and broader implications. Horm. Behav. 77,
aphagia syndrome in rats. Physiol. Behav. 33, 879–887. 98–112.
Nephew, B.C., Bridges, R.S., 2008. Central actions of arginine vasopressin and a V1a Numan, M., Bress, J.A., Ranker, L.R., et al., 2010. The importance of the basolateral/
receptor antagonist on maternal aggression, maternal behavior, and grooming in basomedial amygdala for goal-directed maternal responses in postpartum rats.
lactating rats. Pharmacol. Biochem. Behav. 91, 77–83. Behav. Brain Res. 214, 368–376.
114 Parental Behavior

Numan, M., Corodimas, K.P., Numan, M.J., Factor, E.M., Piers, W.D., 1988. Axon- Pereira, M., Uriarte, N., Agrati, D., et al., 2005. Motivational aspects of maternal
sparing lesions of the preoptic region and substantia innominata disrupt anxiolysis in lactating rats. Psychopharmacology (Berlin) 180, 241–248.
maternal behavior in rats. Behav. Neurosci. 102, 381–396. Perrin, G., Meurisse, M., Lévy, F., 2007. Inactivation of the medial preoptic area of the
Numan, M., Fleming, A.S., Lévy, F., 2006. Maternal behavior. In: Neill, J.D. (Ed.), bed nucleus of the stria terminalis differentially disrupts maternal behavior in
Knobil and Neill’s Physiology of Reproduction, third ed., vol. 2. Elsevier, San Diego, sheep. Horm. Behav. 52, 461–473.
CA, pp. 1921–1923 (Chapter 35). Pevet, P., 2003. Melatonin: from seasonal to circadian signal. J. Neuroendocrinol. 15,
Numan, M., Morrell, J.I., Pfaff, D.W., 1985. Anatomical identification of neurons 422–426.
in selected brain regions associated with maternal behavior deficits induced Pi, X., Grattan, D.R., 1999a. Expression of prolactin receptor mRNA is increased in the
by knife cuts of the lateral hypothalamus in rats. J. Comp. Neurol. 237, preoptic area of lactating rats. Endocrine 11, 91–98.
552–564. Pi, X.J., Grattan, D.R., 1998. Distribution of prolactin receptor immunoreactivity in the
Numan, M., Numan, M.J., English, J.B., 1993. Excitotoxic amino acid injections into brain of estrogen-treated, ovariectomized rats. J. Comp. Neurol. 394, 462–474.
the medial amygdala facilitate maternal behavior in virgin female rats. Horm. Pi, X.J., Grattan, D.R., 1999b. Increased prolactin receptor immunoreactivity in the
Behav. 27, 56–81. hypothalamus of lactating rats. J. Neuroendocrinol. 11, 693–705.
Numan, M., Roach, J.K., Del Cerro, M.C., et al., 1999. Expression of intracellular Pi, X.J., Grattan, D.R., 1999c. Increased expression of both short and long forms of
progesterone receptors in rat brain during different reproductive states, and prolactin receptor mRNA in hypothalamic nuclei of lactating rats. J. Mol. Endo-
involvement in maternal behavior. Brain Res. 830, 358–371. crinol. 23, 13–22.
Numan, M., Rosenblatt, J.S., Komisaruk, B.R., 1977. Medial preoptic area and onset Pilorz, V., Steinlechner, S., 2008. Low reproductive success in Per1 and Per2 mutant
of maternal behavior in the rat. J. Comp. Physiol. Psychol. 91, 146–164. mouse females due to accelerated ageing? Reproduction 135, 559–568.
Okatani, Y., Okamoto, K., Hayashi, K., et al., 1998. Maternal-fetal transfer of melatonin Piwoz, E.G., Huffman, S.L., 2015. The impact of marketing of breast-milk substitutes
in pregnant women near term. J. Pineal Res. 25, 129–134. on WHO-recommended breastfeeding practices. Food Nutr. Bull. 36, 373–386.
Olazábal, D.E., Ferreira, A., 1997. Maternal behavior in rats with kainic acid-induced PLoS One Medicine Editors, 2011. Poor diet in shift workers: a new occupational
lesions of the hypothalamic paraventricular nucleus. Physiol. Behav. 61, 779–784. health hazard? PLoS Med. 8, e1001152. http://dx.doi.org/10.1371/
Olazábal, D.E., Pereira, M., Agrati, D., 2013a. Flexibility and adaptation of the neural journal.pmed.1001152.
substrate that supports maternal behavior in mammals. Neurosci. Biobehav. Rev. Poindron, P., Lévy, F., Keller, M., 2007a. Maternal responsiveness and maternal
37, 1875–1892. selectivity in domestic sheep and goats: the two facets of maternal attachment.
Olazábal, D.E., Pereira, M., Agrati, D., et al., 2013b. New theoretical and experimental Dev. Psychobiol. 49, 54–70.
approaches on maternal motivation in mammals. Neurosci. Biobehav. Rev. 37, Poindron, P., Terrazas, A., Navarro Montes de Oca, M.L., et al., 2007b. Sensory and
1860–1874. physiological determinants of maternal behavior in the goat (Capra hircus). Horm.
Olazábal, D.E., Young, L.J., 2006a. Species and individual differences in juvenile Behav. 52, 99–105.
female alloparental care are associated with oxytocin receptor density in the Poletini, M.O., McKee, D.T., Kennett, J.E., et al., 2007. Knockdown of clock genes in
striatum and the lateral septum. Horm. Behav. 49, 681–687. the suprachiasmatic nucleus blocks prolactin surges and alters FRA expression in
Olazábal, D.E., Young, L.J., 2006b. Oxytocin receptors in the nucleus accumbens the locus coeruleus of female rats. Am. J. Physiol. Endocrinol. Metab. 293,
facilitate “spontaneous” maternal behavior in adult female prairie voles. Neuro- E1325–E1334.
science 141, 559–568. Ranote, S., Elliot, R., Abel, K.M., et al., 2004. The neural basis of maternal respon-
Olcese, J., Lozier, S., Paradise, C., 2013. Melatonin and the circadian timing of human siveness to infants: an fMRI study. Neuroreport 15, 1825–1829.
parturition. Reprod. Sci. 20, 168–174. Rasia-Filho, A.A., Fabian, C., Rigoti, K.M., et al., 2004. Influence of sex estrous cycle
Orpen, B.G., Fleming, A.S., 1987. Experience with pups sustains maternal responding and motherhood on dendritic spine density in the rat medial amygdala revealed by
in postpartum rats. Physiol. Behav. 40, 47–54. the Golgi method. Neuroscience 126, 839–847.
Pan, A., Schernhammer, E.S., Sun, Q., et al., 2011. Rotating night shift work and risk Reppert, S.M., Henshaw, D., Schwartz, W.J., 1987. The circadian-gated timing of birth
of type 2 diabetes: two prospective cohort studies in women. PLoS Med. 8, in rats: disruption by maternal SCN lesions or by removal of the fetal brain. Brain
e1001141. http://dx.doi.org/10.1371/jounrnal.pmed.1001141. Res. 403, 398–402.
Panksepp, J., 2006. Emotional endophenotypes in evolutionary psychiatry. Prog. Rilling, J.K., Young, L.J., 2014. The biology of mammalian parenting and its effect on
Neuropsychopharmacol. Biol. Psychiatry 30, 774–784. offspring social development. Science 345, 771–776.
Panksepp, J., 2011. Toward a cross-species neuroscientific understanding of the Rosenblatt, J.S., Lehrman, D.S., 1963. Maternal behavior in the laboratory rat. In:
affective mind: do animals have emotional feelings? Am. J. Primatol. 73, Rheingold, H.L. (Ed.), Maternal Behavior in Mammals. Wiley, New York, pp. 8–57.
545–561. Rosenwasser, A.M., Hollander, S.J., Adler, N.T., 1987. Effects of pregnancy and
Parada, M., King, S., Li, M., et al., 2008. The roles of accumbal dopamine D1 and D2 parturition on free-running circadian activity rhythms in the rat. Chronobiol. Int. 4,
receptors in maternal memory in rats. Behav. Neurosci. 122, 368–376. 183–187.
Pawluski, J.L., Galea, L.A.M., 2006. Hippocampal morphology is differentially affected Ross, H.E., Freeman, S.M., Spiegel, L.L., et al., 2009. Variation in oxytocin receptor
by reproductive experience in the mother. J. Neurobiol. 66, 71–81. density in the nucleus accumbens has differential effects on affiliative behaviors in
Pawluski, J.L., Galea, L.A.M., 2007. Reproductive experience alters hippocampal monogamous and polygamous voles. J. Neurosci. 29, 1312–1318.
neurogenesis during the postpartum period in the dam. Neuroscience 149, Rotge, J.-Y., Guehl, D., Dilharreguy, B., et al., 2008. Provocation of obsessive-
53–67. compulsive symptoms: a quantitative voxel-based meta-analysis of functional
Pawluski, J.L., Charlier, T.D., Lieblich, S.E., et al., 2009. Reproductive experience neuroimaging studies. J. Psychiatry Neurosci. 33 (5), 405–412.
alters corticosterone and CBG levels in the rat dam. Physiol. Behav. 96, 108–114. Salamone, J.D., Correa, M., 2002. Motivational views of reinforcement: implications
Pawluski, J.L., Vanderbyl, B.L., Ragan, K., et al., 2006a. First reproductive experience for understanding the behavioral functions of nucleus accumbens dopamine.
persistently affects spatial reference and working memory in the mother and these Behav. Brain Res. 137, 3–25.
effects are not due to pregnancy or ‘mothering’ alone. Behav. Brain Res. 175, Salmaso, N., Woodside, B., 2006. Upregulation of astrocytic basic fibroblast growth
157–165. factor in the cingulate cortex of lactating rats: time course and role of suckling
Pawluski, J.L., Walker, S.K., Galea, L.A., 2006b. Reproductive experience differentially stimulation. Horm. Behav. 50, 448–453.
affects spatial reference and working memory performance in the mother. Horm. Salmaso, N., Woodside, B., 2008. Fluctuations in astrocytic basic fibroblast growth
Behav. 49, 143–149. factor in the cingulate cortex of cycling, ovariectomized and postpartum animals.
Pawlusky, J.L., Lambert, K.G., Kinsley, C.H., 2016. Neuroplasticity in the maternal Neuroscience 154, 932–939.
hippocampus: relation to cognition and effects of repeated stress. Horm. Behav. Salmaso, N., Cossette, M.P., Woodside, B., 2011. Pregnancy and maternal behavior
77, 86–97. induce changes in glia, glutamate and its metabolism within the cingulate cortex.
Pedersen, C.A., Caldwell, J.D., Johnson, M.F., et al., 1985. OT antiserum delays onset PLoS One 6, e23529. http://dx.doi.org/10.1371/journal.pone.0023529.
of ovarian steroid-induced maternal behavior. Neuropeptides 6, 175–182. Salmaso, N., Nadeau, J., Woodside, B., 2009. Steroid hormones and maternal
Perea-Rodríguez, J.P., Saltzman, W., 2014. Differences in placentophagia in relation experience interact to induce glial plasticity in the cingulate cortex. Eur. J. Neu-
to reproductive status in the california mouse (Peromyscus californicus). Dev. rosci. 29, 786–794.
Psychobiol. 56, 812–820. Salmaso, N., Popeski, N., Peronace, L.A., et al., 2005. Differential effects of repro-
Pereira, M., Morrell, J., 2011. Functional mapping of the neural circuitry of rat ductive and hormonal state on basic fibroblast growth factor and glial fibrillary acid
maternal motivation: effects of site-specific transient neural inactivation. protein immunoreactivity in the hypothalamus and cingulate cortex of female rats.
J. Neuroendocrinol. 23, 1020–1035. Neuroscience 134, 1431–1440.
Parental Behavior 115

Savino, F., Ceratto, S., De Marco, A., et al., 2014. Looking for new treatments of Stern, J.M., Dix, L., Bellomo, C., et al., 1992. Ventral trunk somatosensory deter-
infantile colic. Ital. J. Pediatr. 40, 1–6. minants of nursing behavior in Norway rats. II. Role of nipple and surrounding
Scanlan, V.F., Byrnes, E.M., Bridges, R.S., 2006. Reproductive experience and acti- sensations. Psychobiology 20, 71–80.
vation of maternal memory. Behav. Neurosci. 120, 676–686. Stewart, A., Gaikwad, S., Kyzar, E., 2012. Modeling anxiety using adult zebrafish:
Schoenbaum, G., Takahashi, Y., Liu, T.-L., et al., 2011. Does the orbitofrontal cortex a conceptual review. Neuropharmacology 62, 135–143.
signal value? Ann. N.Y. Acad. Sci. 1239, 87–99. Stolzenberg, D.S., Rissman, E.F., 2011. Oestrogen independent, experience-induced
Schrader, J.A., Nunez, A.A., Smale, L., 2010. Changes in and dorsal to the rat maternal behaviour in female mice. J. Neuroendocrinol. 23, 345–354.
suprachiasmatic nucleus during early pregnancy. Neuroscience 171, Sufka, K.J., Warnick, J.E., Pulaski, C.N., et al., 2009. Antidepressant efficacy
513–523. screening of novel targets in the chick anxiety-depression model. Behav. Phar-
Schrader, J.A., Smale, L., Nunez, A.A., 2013. Pregnancy affects FOS rhythms in brain macol. 20, 146–154.
regions regulating sleep/wake state and body temperature in rats. Brain Res. Svare, B., Gandelman, R., 1976. A longitudinal analysis of maternal aggression in
1480, 53–60. rockland-swiss albino mice. Dev. Psychobiol. 9, 437–446.
Scott, C.J., Pereira, A.M., Rawson, J.A., et al., 2000. The distribution of progesterone Swain, J.E., Tasgin, E., Mayes, L.C., et al., 2008. Maternal brain response to own
receptor immunoreactivity in the preoptic area and hypothalamus of the ewe: baby-cry is affected by cesarean section delivery. J. Child Psychol. Psychiatry 49,
upregulation of progesterone receptor mRNA in the mediobasal hypothalamus by 1042–1052.
oestrogen. J. Neuroendocrinol. 12, 565–575. Swanson, L.W., 1976. An autoradiographic study of the efferent connections of the
Scribner, S.J., Wynne-Edwards, K.E., 1994. Disruption of body temperature and preoptic region in the rat. J. Comp. Neurol. 167, 227–256.
behavior rhythms during reproduction in dwarf hamsters (Phodopus). Physiol. Takayama, H., Nakamura, Y., Tamura, H., et al., 2003. Pineal gland (melatonin) affects
Behav. 55, 361–369. the parturition time, but not luteal function and fetal growth, in pregnant rats.
Sellix, M.T., 2013. Clocks underneath: the role of peripheral clocks in the timing of Endocr. J. 50, 37–43.
female reproductive physiology. Front. Endocrinol. 4. http://dx.doi.org/10.3389/ Teodorov, E., Bernardi, M.M., Ferrari, M.F.R., et al., 2011. Plasticity of opioid receptors
fendo.2013.00091. in the female periaqueductal gray: multiparity-induced increase in the activity of
Serafim, A.P., Felicio, L.F., 2002. Reproductive experience influences grooming genes encoding for mu and kappa receptors and a post-translational decrease in
behavior during pregnancy in rats. Braz. J. Med. Biol. Res. 35, 391–394. delta receptor expression. J. Mol. Neurosci. 43, 175–181.
Serón-Ferré, M., Richter, H.G., Valenzuela, G.J., et al., 2016. Circadian rhythms in the Terkel, J., Bridges, R.S., Sawyer, C.H., 1979. Effects of transecting lateral neural
fetus and newborn: significance of interactions with maternal physiology and the connections of the medial preoptic area on maternal behavior in the rat: nest
environment. In: Walker, D.W. (Ed.), Prenatal and Postnatal Determinants of building, pup retrieval and prolactin secretion. Brain Res. 169, 369–380.
Development. Neuromethods, vol. 109. Springer Science þ Business Media New Terlecki, L.J., Sainsbury, R.S., 1978. Effects of fimbria lesions on maternal behavior in
York, New York, NY, pp. 147–164. the rat. Physiol. Behav. 21, 89–97.
Shams, S., Pawluski, J.L., Chatterjee-Chakrabortyc, M., et al., 2012. Dendritic Tomizawa, K., Iga, N., Lu, Y., et al., 2003. Oxytocin improves long-lasting spatial
morphology in the striatum and hypothalamus differentially exhibits experience- memory during motherhood through MAP kinase cascade. Nat. Neurosci. 6,
dependent changes in response to maternal care and early social isolation. 384–390.
Behav. Brain Res. 233, 79–89. Torres-Farfan, C., Mendez, N., Abarzua-Catalan, L., et al., 2011. A circadian clock entrained
Shanks, N., Windle, R.J., Perks, P., et al., 1999. The hypothalamic-pituitary-adrenal by melatonin is ticking in the rat fetal adrenal. Endocrinology 152, 1891–1900.
axis response to endotoxin is attenuated during lactation. J. Neuroendocrinol. 11, Uys, J.D.K., Stein, D.J., Daniels, W.M.U., et al., 2003. Animal models of anxiety
857–865. disorders. Psychiatry Rep. 5, 274–281.
Sharp, J.S., González, M.F., Morton, M.T., et al., 1988. Decreases of cortical and Van Hemel, S.B., 1973. Pup retrieving as a reinforcer in nulliparous mice. J. Exp. Anal.
thalamic glucose metabolism produced by parietal cortex stimulation in the rat. Behav. 19, 233–238.
Brain Res. 438, 357–362. Van Leengoed, E., Kerker, E., Swanson, H.J., 1987. Inhibition of postpartum maternal
Sheehan, T.P., Cirrito, J., Numan, M.J., et al., 2000. Using c-Fos immunocyto- behavior in the rat by injecting an OT antagonist into the cerebral ventricles.
chemistry to identify forebrain regions that may inhibit maternal behavior in rats. J. Endocrinol. 112, 275–282.
Behav. Neurosci. 114, 337–352. Vanoye-Carlo, A., Mendoza-Rodríguez, C.A., Morales, T., et al., 2009. Estrogen
Shingo, T., Gregg, C., Enwere, E., et al., 2004. Pregnancy-stimulated neurogenesis in receptor increased expression during hippocampal neuroprotection in lactating rats.
the adult female forebrain. Science 299, 117–120. J. Steroid Biochem. Mol. Biol. 116, 1–7.
Sider, L.H., Hucke, E.E.T.S., Florio, J.C., et al., 2003. Influence of time of day on Vanoye-Carlo, A., Morales, T., Ramos, E., et al., 2008. Neuroprotective effects of
hypothalamic monoaminergic activity in early pregnancy: effect of a previous lactation against kainic acid treatment in the dorsal hippocampus of the rat. Horm.
reproductive experience. Psychoneuroendocrinology 28, 195–206. Behav. 53, 112–123.
Sjoeholm, A., Bridges, R., Grattan, D.R., et al., 2011. Region-, neuron-, and signaling Varcoe, T.J., Boden, M.J., Voultsios, A., et al., 2013. Characterisation of the maternal
pathway-specific increases in prolactin responsiveness in reproductively experi- response to chronic phase shifts during gestation in the rat: implications for fetal
enced female rats. Endocrinology 152, 1979–1988. metabolic programming. PLoS One 8, e53800. http://dx.doi.org/10.1371/
Skuse, D.H., Lori, A., Cubells, J.F., et al., 2014. Common polymorphism in the journal.pone.0053800.
oxytocin receptor gene (OXTR) is associated with human social recognition skills. Varcoe, T.J., Wright, N., Voultsios, A., et al., 2011. Chronic phase shifts of the
Proc. Natl. Acad. Sci. U.S.A. 111, 1987–1992. photoperiod throughout pregnancy programs glucose intolerance and insulin
Slotnick, B.M., Nigrosh, B.J., 1975. Maternal behavior of mice with cingulate cortical, resistance in the rat. PLoS One 6, e18504. http://dx.doi.org/10.1371/
amygdala, or septal lesions. J. Comp. Physiol. Psychol. 88, 118–127. journal.pone.0018504.
Smith, K.S., Berridge, K.C., Aldridge, J.W., 2011. Disentangling pleasure from Vásquez-Ruiz, S., Maya-Barrios, J.A., Torres-Narváez, P., et al., 2014. A light/dark
incentive salience and learning signals in brain reward circuitry. Proc. Natl. Acad. cycle in the NICU accelerates body weight gain and shortens time to discharge in
Sci. U.S.A. 108, E255–E264. preterm infants. Early Hum. Dev. 90, 535–540.
Smith, R.L., 1973. The ascending fibers from the principal trigeminal sensory nucleus Wagner, C.K., Morrell, J.I., 1996. Levels of estrogen receptor immunoreactivity are
in the rat. J. Comp. Neurol. 148, 423–446. altered in behaviorally-relevant brain regions in female rats during pregnancy. Mol.
Solum, D.T., Handa, R.J., 2002. Estrogen regulates the development of brain-derived Brain Res. 42, 328–336.
neurotrophic factor mRNA and protein in the rat hippocampus. J. Neurosci. 22, Wagner, C.K., Silverman, A.J., Morrell, J.I., 1998. Evidence for estrogen receptor in
2650–2659. cell nuclei and axon terminals within the lateral habenula of the rat: regulation
Stack, E.C., Balakrishnan, R., Numan, M.J., et al., 2002. A functional neuroanatomical during pregnancy. J. Comp. Neurol. 392, 330–342.
investigation of the role of the medial preoptic area in neural circuits regulating Walf, A.A., Frye, C.A., 2008. Parity and estrogen-administration alter affective behavior
maternal behavior. Behav. Brain Res. 131, 17–36. of ovariectomized rats. Physiol. Behav. 93, 351–356.
Stern, J.M., Johnson, S.K., 1990. Ventral somatosensory determinants of nursing Walsh, R.J., Mangurian, L.P., Posner, B.I., 1990. The distribution of lactogen receptors
behavior in Norway rats (Rattus norvegicus). Physiol. Behav. 47, 993–1011. in the mammalian hypothalamus: an in vitro autoradiographic analysis of the rabbit
Stern, J.M., Kolunie, J.M., 1989. Perioral anesthesia disrupts maternal behavior during and rat. Brain Res. 530, 1–11.
early lactation in Long-Evans rats. Behav. Neural Biol. 52, 20–38. Warembourg, M., Jolivet, A., Milgrom, E., 1989. Immunohistochemical evidence
Stern, J.M., Mackinnon, D.A., 1976. Postpartum, hormonal, and nonhormonal of the presence of estrogen and progesterone receptors in the same
induction of maternal behavior in rats: effects on t-maze retrieval of pups. Horm. neurons of the guinea pig hypothalamus and preoptic area. Brain Res. 480,
Behav. 7, 305–316. 1–15.
116 Parental Behavior

Warembourg, M., Logeat, F., Milgrom, E., 1986. Immunocytochemical localization of Woodside, B., Leon, M., 1980. Thermoendocrine influences on maternal nesting
progesterone receptor in the guinea pig central nervous system. Brain Res. 384, behavior in rats. J. Comp. Physiol. Psychol. 94, 41–60.
121–131. Young, J.L., Wang, Z., 2004. The neurobiology of pair bonding. Nat. Neurosci. 7,
Wartella, J., Amory, E., Madonia-Lomas, L., et al., 2003. Single or multiple repro- 1048–1054.
ductive experiences attenuate neurobehavioral stress and fear responses in the Young, L.J., Muns, S., Wang, Z., et al., 1997. Changes in oxytocin receptor mRNA in
female rat. Physiol. Behav. 79, 373–381. rat brain during pregnancy and the effects of estrogen and interleukin-6.
Willner, P., 1991. Methods for assessing the validity of animal models of human J. Neuroendocrinol. 9, 859–865.
psychopathology. In: Boulton, A., Baker, G., Martin-Iverson, M. (Eds.), Neuromethods, Zarrow, M.X., Denenberg, V.H., Anderson, C.O., 1965. Rabbit: frequency of suckling in
Animal Models in Psychiatry I, vol. 18. Humana Press, New York, pp. 1–23. the pup. Science 150, 1835–1836.
Windle, R.J., Wood, S., Shanks, N., et al., 1997. Endocrine and behavioural responses Zeitzer, J.M., Dijk, D.J., Kronauer, R.E., 2000. Sensitivity of the human circadian
to noise stress: comparison of virgin and lactating female rats during non-disrupted pacemaker to nocturnal light: melatonin phase resetting and suppression.
maternal activity. J. Neuroendocrinol. 9, 407–414. J. Physiol. 526, 695–702.
Wolmarans, W., Brand, L., Stein, D.J., et al., 2013. Reappraisal of spontaneous
stereotypy in the deer mouse as an animal model of obsessive-compulsive disorder
(OCD): response to escitalopram treatment and basal serotonin transporter (SERT)
density. Behav. Brain Res. 256, 545–553.
1.04 The Neurobiology of Social Affiliation and Pair Bonding
CS Carter, The Kinsey Institute, Indiana University, Bloomington, IN, USA
EB Keverne, University of Cambridge, Cambridge, UK
Ó 2017 Elsevier Inc. All rights reserved.

1.04.1 Introduction 117


1.04.2 The Natural History of Pair Bond Formation 118
1.04.2.1 Experiences Associated with Pair Bonding 118
1.04.2.2 Measurement of Social Bonding 119
1.04.3 Oxytocin and Vasopressin – Classical Neuroendocrine Peptides 119
1.04.3.1 Oxytocin and Vasopressin Receptors 120
1.04.3.2 Opioid, Oxytocin Interactions in the Brain 120
1.04.4 Mother–Infant Bonding 121
1.04.4.1 Mechanisms Underlying Maternal State 121
1.04.4.2 Oxytocin, Infant Recognition, and Bonding 122
1.04.4.3 Frontal Cortex: Role in Social and Parental Reward 122
1.04.4.4 Social and Maternal Bonding in Primates 124
1.04.5 Adult Pair Bonds 125
1.04.5.1 Mating Systems and Pair Bonds 125
1.04.5.2 Social Systems 125
1.04.5.3 Sexual versus Social Monogamy 125
1.04.5.4 Social Monogamy in Prairie Voles 126
1.04.5.5 Experimental Analysis of Partner Preferences 126
1.04.5.6 Selective Aggression as a Component of Pair Bonding 126
1.04.5.7 Affiliative Behaviors and Positive Sociality 127
1.04.5.8 Selective Social Behavior, a Role for Oxytocin and Vasopressin 128
1.04.6 Stress and Pair Bond Formation 128
1.04.6.1 Dopamine and Pair Bonding 130
1.04.7 Responses to Separation 130
1.04.7.1 Consequences of Isolation May Be Mediated through Oxytocin 131
1.04.7.2 Social Behavior, the Polyvagal Theory and Oxytocin 132
1.04.8 Phylogenetic and Ontogenetic Factors Influencing Pair Bonding 132
1.04.9 Peptides and Human Behavior 133
1.04.10 Clinical Implications of a Peptidergic Theory of Social Attachment 134
1.04.11 Attachment and Bonding in Primates: An Evolutionary Perspective for Humans 135
1.04.11.1 The Evolutionary Basis of Bonding 136
1.04.11.2 Genomic Imprinting, Social Behavior, and Cortical Growth 137
1.04.11.3 Oxytocin and Love 137
1.04.11.4 Oxytocin and Development 138
1.04.12 Summary 138
Acknowledgments 138
References 138

1.04.1 Introduction either positive, including affiliations and social bonds, or


agonistic, including aggressive or defensive behaviors. Selective
Positive social interactions, including affiliations and social social behaviors and social bonds are critical components of
bonds, dominate the behavioral repertoire of humans and long-lasting social interactions.
many higher vertebrates. Social behaviors are the product of The purpose of this review is to examine neuroendocrine
evolution and result from phytogenetic, ontogenetic, and mechanisms underlying social affiliation and social bonds in
epigenetic processes. Social behaviors provide a context for mammals. The biological substrates of social behaviors are
reproduction and can facilitate survival. Social behaviors also shared with those necessary for birth and lactation. As with
are inherently species-typical and variable across individuals. reproduction, sociality and social bond formation rely on
Social behaviors by definition involve two or more individuals neural systems that incorporate neuropeptides, including
and require the willingness of individuals to aggregate and oxytocin, vasopressin, opioids, corticotropin-releasing factor
remain together. Social behaviors are typically identified as (CRF), and related hormones. These peptides also serve to

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00004-3 117


118 The Neurobiology of Social Affiliation and Pair Bonding

coordinate the physiological and behavioral consequences of Old World primate societies are frequently referred to as being
positive social experiences, thus implicating reward systems ‘female-bonded.’ Such primate matrilines not only provide for
and especially dopamine. These molecules will be the focus group cohesion and social continuity, but kin also benefit from
of this chapter. the learning experience gained when helping mother. The
neurobiology of same-sex social bonds remains largely unex-
plored, but may rely on the underlying neural systems that
1.04.2 The Natural History of Pair Bond Formation regulate other forms of social bonding.
Alloparenting and male parental behavior are traits of
Social bonds may form between a parent and infant, between species, ranging from birds to rodents, canids, and New World
two adults, and among other members of a social group primates, which are defined as socially monogamous (reviewed
(Table 1). These apparently very different bonding relation- in Solomon and French, 1997). Although both related and
ships have a common function – namely to enhance reproduc- unrelated animals may aid in the care of young, alloparenting
tion and ensure reproductive success. This is achieved in pair is most common among genetically related individuals. Selec-
bonding and social group bonding by providing a readily avail- tive social behaviors and social bonds are defining characteris-
able mate and predator defense, while mother–infant bonding tics of species that engage in alloparenting or paternal behavior,
and social group bonding secure the nurture of infants and suggesting that common underlying mechanisms may regulate
their protection. Pair bonding is especially favored in those social bonding and other social traits in species that tend to
mammalian species where offspring survival cannot be assured form communal families.
without the help of mates. Social bonds also may provide direct benefits to the
Clear examples of pair bonding and the physiological basis members of a pair or group, by serving to buffer the indi-
for social bond formation are provided from studies on voles, vidual from stress. The physiological mechanisms for stress
marmosets, and other species that are identified as “monoga- buffering are related to those responsible for the formation
mous” (Kleiman, 1977). In domesticated species, social bonds of pair bonds.
may cross species barriers (e.g., dogs and humans) where
a combination of tameness, docility, and dependence, selected
1.04.2.1 Experiences Associated with Pair Bonding
for by human intervention, has secured their survival and
enhanced reproductive success. The essential first steps in social bond formation involve prox-
In promiscuous mammalian species, where parental males imity and social engagement (Porges, 2011). Potential part-
are frequently absent, mother–infant bonding may help to ners must interact in a species- and sex-appropriate manner.
guarantee infant survival. This often involves individual infant The choice of a social partner, the tendency to engage in social
recognition, and examples of filial bonding are found among behaviors, and patterns of social behavior differ among
ungulate species. Research on maternal bonding has taken species and individuals, and between males and females
advantage of the fact that precocial ungulates, including sheep, (Carter et al., 1995; DeVries et al., 1996). In addition, because
are excellent subjects for neurobiological investigation and do the outcome of social interactions can be positive or negative,
develop selective mother–infant bonding. the emotional experience of a particular social encounter may
In social living mammals and especially in primate socie- determine whether that encounter leads to pair bond
ties, an alternative strategy for promoting reproductive success formation.
through infant survival is seen in the assistance given by other Both positive social interactions and social bonds may func-
females (Hrdy, 2009). Serving as surrogate parents may involve tion to provide a sense of safety and reduce anxiety or the nega-
a self-sacrifice for reproductive potential and often is under- tive feelings associated with stressful experience. In species that
taken by females that are genetically related to the offspring. do tend to form pair bonds, stressful experiences (such as preg-
nancy and parturition), anxiety, neophobia, and isolation
often precede the formation of long-lasting social attachments
Table 1 Animal models for studying the biological bases of pair (Carter, 1998). Threatening situations may encourage return to
bonding
a ‘secure base’ or otherwise strengthen social bonds (Bowlby,
Maternal–infant bonding: 1969). These circumstances may increase social ‘drive’ or ‘moti-
Most commonly studied in animals in which the mother is selective. vation,’ and subsequent social interactions, and thus increase
Mother usually cares for only her own young. the tendency to form a social attachment.
Example: Sheep. The association of pair bonding with overcoming stressful
Maternal behavior: experiences also offers clues to the physiology of pair bond
Not selective, but may share physiological substrates with pair formation. Hormones can reduce fear or behavioral inhibition
bonding. and permit the subsequent expression of selective social behav-
Examples: Rats and other rodents, rabbits, pigs.
iors, such as those necessary for pair bonding. The neurochem-
Adult pair bonding:
Most commonly studied in adult monogamous mammals. Most ical processes that are capable of overcoming neophobia also
commonly studied in adult monogamous mammals. may be needed to permit the formation of new social attach-
Examples: Prairie voles and titi monkeys. ments. Although sharing some common substrates, the neural
substrates for social engagement and selective social bonding
Reproduced from Carter, C.S., Keverne, E.B., 2002. The neurobiology of social are probably not identical (Cho et al., 1999), and it is likely
affiliation and pair bonding. In: Pfaff, D., Arnold, A., Etgen, A., Fahrbach, S., Rubin,
R. (Eds.), Hormones, Brain, and Behavior. Academic Press, New York, pp. 299–337, that the processes leading to social interactions differ from
with permission from Elsevier. those necessary for a bond to form.
The Neurobiology of Social Affiliation and Pair Bonding 119

Table 2 Measurements of pair bonding pair bonding, the neural systems that incorporate these neuro-
peptides are potential candidates for a role in the mediation
Selective contact or physical proximity in the presence of an attachment
of social behavior and especially social bonding (Table 3).
object
Oxytocin and vasopressin are neuropeptides composed of
Aggression in defense of the attachment object
Autonomic and/or endocrine responses nine amino acids, including a six amino acid ring with a three
Distress after separation amino acid tail. Oxytocin and vasopressin differ from each
Crying or vocalization other in two amino acids, one in the ring and one in the tail,
Increased release of stress hormones (HPA activation) and may have evolved from a common ancestral peptide
(Van Kesteren et al., 1992; Acher et al., 1997). The gene for
Reproduced from Carter, C.S., Keverne, E.B., 2002. The neurobiology of social each of these two peptides is located on the same chromosome.
affiliation and pair bonding. In: Pfaff, D., Arnold, A., Etgen, A., Fahrbach, S.,
Rubin, R. (Eds.), Hormones, Brain, and Behavior. Academic Press, New York, The stereochemical similarities between these peptides allow
pp. 299–337, with permission from Elsevier. functional interactions with each other’s receptors. Oxytocin
and vasopressin are the predominant neurohypophyseal
hormones in mammals, although structurally similar peptides,
1.04.2.2 Measurement of Social Bonding
such as vasotocin, mesotocin, and isotocin, have been identi-
Social bonds are hypothetical constructs, and thus measure- fied in other vertebrates (Acher et al., 1997). In addition, struc-
ments require analysis of a complex assortment of behaviors turally related peptides with consequences for reproductive
over long periods of time. However, selective behavioral, behavior are found in invertebrates, suggesting that variations
hormonal, or autonomic responses to social stimuli, or their on these molecules appeared prior to the evolution of verte-
absence, may be used to infer social bonds (Table 2). Labora- brates (Van Kesteren et al., 1992; Garrison et al., 2012).
tory analyses of social bonding often involve preference tests Oxytocin is released in a coordinated fashion within the
in which experimental animals are allowed to choose between brain and at the posterior pituitary, into general circulation
or express differential responses to social stimuli. (Neumann and Landgraf, 2012; Gainer, 2012). It is likely
Differential responses to the entire animal, visual cues, that the ability of oxytocin to have exceptionally broad behav-
vocalizations, or odors can be used to assess preferences. ioral and physiological consequences is related to this capacity
The most compelling evidence for positive social preference for movement throughout the brain and body (Stoop, 2012;
is usually obtained in tests in which an experimental animal Grinevich et al., 2016).
is allowed to simultaneously or sequentially make a choice Oxytocin is produced tonically. In typical humans, basal
between a familiar animal or an otherwise similar unfamiliar levels of oxytocin in plasma are notably consistent across
conspecific (or stimuli associated with those individuals). time (Gouin et al., 2010; Dai et al., 2012; Weisman et al.,
Physical proximity and positive social interactions are 2013). However, oxytocin also can be released as pulses, thus
commonly used to index preference. Testing conditions in promoting muscle contractions in the uterus and mammary
which the experimental animal is allowed elective physical gland. The pulsatile release of oxytocin neurons may be related
contact with the stimulus animals are especially sensitive to the notable plasticity of these cells. In adult rats, oxytocin
(Williams et al., 1992). In addition, conditions that allow neurons undergo physical transformations in response to
the experimental animal to remain alone are desirable, hormonal and social stimulation. During pregnancy and lacta-
because voluntary social preferences may differ from forced tion, and perhaps under other conditions such as dehydration
choices. or sexual stimulation (Carter, 1992), glial processes that nor-
Responses to separation or reunion can also be evidence mally separate the oxytocin-containing neurons are retracted
for social bonding. In mammals and birds, ‘distress’ vocaliza- allowing electrical coupling and then the pulsatile release of
tions may increase following separation and decline oxytocin. Vasopressin-containing neurons typically do not
following reunion. Secretion of hormones from the hypotha-
lamic–pituitary–adrenal (HPA) axis, usually cortisol or corti-
costerone or adrenocorticotrophic hormone (ACTH), also
Table 3 Endocrine mediators of pair bonding
follows separation from the ‘attachment figure’ and HPA
activity tends to return to normal upon reunion (DeVries Steroids
et al., 1996). However, vocalizations, changes in the HPA Gonadal steroids – not essential
axis, and autonomic responses may be triggered by either But may modulate effects of peptides
social or nonsocial stressors. The most compelling evidence Adrenal steroids – not essential
for social bonding is obtained when various measures May modulate effects of peptides
converge and when those measures can be interpreted in light Effects may be sexually dimorphic
Tends to be elevated then decline
of the biology of the entire organism and especially the neuro-
Peptides
endocrine system.
Oxytocin – may be especially important in females
Vasopressin – may be especially important in males
Corticotropin-releasing hormone – may modulate effects of stress
1.04.3 Oxytocin and Vasopressin – Classical Opioids – modulation of separation responses
Neuroendocrine Peptides
Reproduced from Carter, C.S., Keverne, E.B., 2002. The neurobiology of social
affiliation and pair bonding. In: Pfaff, D., Arnold, A., Etgen, A., Fahrbach, S., Rubin,
Because oxytocin and vasopressin are released during social R. (Eds.), Hormones, Brain, and Behavior. Academic Press, New York, pp. 299–337,
and sexual interactions, and under conditions that result in with permission from Elsevier.
120 The Neurobiology of Social Affiliation and Pair Bonding

show this form of plasticity and pulsatile release. Furthermore, and areas of the caudal brain stem. Androgens facilitate the
oxytocin-producing cells are sensitive to oxytocin itself; thus synthesis and release of vasopressin, particularly in the
a form of autocrine feedback regulates the functions of mAMY and lateral septum (LS) (DeVries and Panzica, 2006),
oxytocin-producing cells. Stimulation of the oxytocin system accounting for clear sex differences in the abundance of vaso-
may ‘feed forward’ to release more oxytocin, and in some cases, pressin in the CNS.
administration of oxytocin appears to enhance the synthesis of Although vasopressin plays an important role in social
endogenous oxytocin (Grippo et al., 2012). behavior, many of the functions of stimulation of the vaso-
Oxytocin exposure is of particular significance during pressin receptor differ from those of oxytocin (Neumann
development. Specifically oxytocin can influence cellular and Landgraf, 2012; Stoop, 2012; Albers, 2015). For example,
growth, death or motility, inflammation, or differentiation in maternal behavior, oxytocin is critical to nursing and
(Gutkowska and Jankowski, 2012). Oxytocin treatments in important to nurture, while vasopressin has been implicated
early life can have dose-dependent effects on physiology and in maternal (Bosch and Neumann, 2012) and paternal
behavior (Carter et al., 2009). Low doses of exogenous defense of the young (Kenkel et al., 2012, 2013). Some
hormone facilitate pair bonding, as well as the expression of aspects of vasopressin’s functions within the nervous system
the endogenous oxytocin, and inhibit the expression of the are sexually dimorphic, with possible implications for sex
vasopressin (V1a) receptor. Furthermore, the oxytocin receptor differences in the tendency to show defensive behaviors and
is susceptible to epigenetic regulation, for example, by for disorders, such as autism, that are male-biased (reviewed
silencing genes via methylation. Changes in the expression Carter, 2007).
of the oxytocin receptor as a function of early experiences
could help to account for species and individual differences
1.04.3.1 Oxytocin and Vasopressin Receptors
in sociality and emotionality. As just one example, hyperme-
thylation of the oxytocin receptor gene has been detected in The oxytocin receptor is a member of the seven transmembrane
autism (Gregory et al., 2009). Through mechanisms such as G-protein coupled receptors and is distinguished by differences
these, the developmental effects of oxytocin may fine-tune in its anatomical distribution across different mammalian
the nervous system, including the capacity for pair bonding species. There is at present only evidence for one oxytocin
and attachment in later life. receptor, which is similar across species. However, species
The major sources of systemic oxytocin and vasopressin in differences do exist in the regulation of the oxytocin receptor
mammals are the magnocellular neurons of the supraoptic (Freeman and Young, 2016), allowing a variety of other
nucleus (SON) and paraventricular nucleus of the hypothal- neuroendocrine factors, including steroids, to determine
amus (PVN) (reviewed in Russell and Leng, 1998). Oxytocin species-typical patterns of receptor distribution (Witt et al.,
and vasopressin, in conjunction with neurophysin carrier 1991; Insel and Shapiro, 1992; Jiménez et al., 2015).
proteins, are transported from magnocellular neurons in the Oxytocin receptors have been identified by receptor autora-
SON and PVN to the posterior pituitary, where they are secreted diography and receptor mRNA expression in several brain
into the blood stream. The release of oxytocin into the systemic areas. Among the areas that contain high concentrations of
circulation is typically pulsatile. In addition, oxytocin and vaso- the oxytocin receptor in rodents are the olfactory bulbs, ante-
pressin are released within the central nervous system (CNS) rior olfactory nucleus (AON), olfactory tubercle, nucleus
from smaller, parvocellular neurons, located in the PVN and accumbens (NAcc), prelimbic cortex, ventral subiculum,
other brain areas. The release of peptides within the CNS and bNST, central nucleus of the amygdala, ventromedial nucleus
posterior pituitary can occur independently, although central of the hypothalamus (VMH), LS, cingulate cortex (CC), dorsal
and peripheral release patterns also may be coordinated motor nucleus of the vagus (DMX), and the nucleus tractus sol-
(Kendrick et al., 1986; Landgraf and Neumann, 2004; itarius (NTS) (Barberis and Tribollet, 1996). These areas have
Grinevich et al., 2016). been implicated in both social behavior and the regulation of
In addition to the oxytocin that is produced in the SON and the autonomic nervous system.
PVN, cell bodies and fibers staining for oxytocin also have been Although three isoforms of the vasopressin receptor have
identified in the bed nucleus of the stria terminalis (bNST), the been identified, most of the behavioral effects have been attrib-
anterior commissural nucleus, and the spinal cord (Sofroniew, uted to the V1a receptor, which is expressed in the brain. Vaso-
1983). The latter fibers terminate within the CNS or release pressin V1a binding has been identified in the granule cell layer
oxytocin into the cerebrospinal fluid (CSF). Oxytocin gene of olfactory bulbs, accessory olfactory bulbs, the diagonal band
expression increases in the PVN and SON during lactation of Broca, CC, central and lateral aspects of the amygdala, the
and around birth or under hormonal conditions that mimic dorsolateral thalamus, and superior colliculus (Insel, 1997).
birth. Oxytocin also is produced in other tissues including Like oxytocin, the distribution of V1a receptors is highly
the ovary, uterus, placenta, and thymus. The multiple actions species-specific, and follows a pattern that implicates this
of this hormone may integrate a variety of peripheral and receptor in social behavior and pair bonding (Hammock and
central processes, including those involved in reproduction Young, 2005).
and social behavior.
As with oxytocin, the most abundant sources of vasopressin
1.04.3.2 Opioid, Oxytocin Interactions in the Brain
are found in the PVN and SON. Vasopressin is synthesized and
released within the CNS by parvocellular neurons in the PVN. The interactions between opioids and oxytocin in the brain are
Vasopressinergic cell bodies also have been identified in the complex. During pregnancy, it is important that oxytocin is not
suprachiasmatic nucleus, medial amygdala (mAMY), bNST, released from the magnocellular neurons into peripheral
The Neurobiology of Social Affiliation and Pair Bonding 121

circulation as this produces a risk for preterm labor. Release of time period that maternal care becomes exclusive. Thus in
oxytocin from neurohypophysial terminals is sensitive to inhi- sheep, the ewe, having ‘bonded,’ accepts only her own lamb
bition by opioid receptor agonists (Ortiz-Miranda et al., 2005). at suckling and rejects, often violently, any strange lamb
Thus, in pregnancy, a central inhibitory opioid mechanism that attempts to suckle. Because sheep are seasonal breeders
restrains magnocellular oxytocin neurons from responding to with synchronized multiple births occurring throughout the
stress and cytokine release, minimizing the risk of preterm flock, all within a few weeks, it is important that the mother
labor (Brunton et al., 2006). recognizes and only permits her own lamb to suckle.
A number of findings indicate that endogenous opioids as Nurturing is energetically costly to the mother, and ‘bonding’
well as oxytocin are active during parturition. Plasma concen- ensures that this energetic sacrifice is only made to her own
trations of b-endorphin increase during labor and birth in genetically related offspring.
humans (Goland et al., 1981), and in the rat brain, b-endor- The mechanisms which underlie the formation of the
phin concentrations are elevated in the hypothalamus, mother–infant bond involve the female being in a maternal
midbrain, and amygdala during pregnancy and parturition state, being able to recognize her offspring dependent on
(Wardlaw and Frantz, 1983). The terminal areas of distribu- unique sensory cues, and activation of the sensitive window
tion for endorphin neurons have much in common with for recognition contingent upon the appearance of the
oxytocin neurons, and in certain areas the release of oxytocin offspring. The recognition window is activated by parturition
is strongly regulated by b-endorphin. However, considerable and delivery of the neonate.
species differences have been demonstrated with respect to
the receptor concentrations in different areas of the brain
1.04.4.1 Mechanisms Underlying Maternal State
(Mansour et al., 1988). Synthesis of proopiomelanocortin,
the precursor peptide for b-endorphin, increases in ewes The ovarian hormones of importance in maternal behavior are
following estradiol or progesterone replacement therapy estrogens and progestins, and because there is no restriction to
which mimics the effects of pregnancy hormones. In sheep, the passage of steroid hormones from the vascular to the cere-
but not in rats, expression of proopiomelanocortin mRNA bral compartment, high-affinity binding neurons for these
during lactation increases significantly. Also in ewes, levels hormones will be activated in all parts of the brain simulta-
of preproenkephalin messenger RNA expression are signifi- neously (Kendrick and Keverne, 1991). Although studies have
cantly higher during lactation compared to late pregnancy tended to focus attention on the hypothalamus as a site of
and parturition (Kendrick and Keverne, 1992). Taken together, action for steroid hormones, it should be remembered that
these findings provide an anatomical basis for opioid– steroid binding neurons have a widespread distribution
oxytocin interactions during pregnancy and lactation. A func- throughout the limbic and olfactory brain. The pattern of secre-
tional relationship between these peptides has also been tion of steroid hormones during pregnancy is remarkably
demonstrated in the context of maternal behavior (Keverne similar among all nonprimate mammals and is characterized
and Kendrick, 1991) and also in pair bond formation in voles by high levels of progesterone in the postimplantation period,
(Burkett et al., 2011). which decreases prior to parturition with a concomitant
increase in estradiol. This prolonged priming of the brain by
exposure to high progesterone and low estrogens is important
1.04.4 Mother–Infant Bonding for the suppression of sexual behavior in mammals and for
genomic activation promoting the synthesis of hypothalamic
The most notable form of an enduring social bond is that peptides (oxytocin, CRF, prolactin, and b-endorphin). Proges-
between a mother and her infants. Bonding, although integral terone decline followed by the sequential increase in estradiol
to maternal behavior, can be distinguished from this. This is important for synthesis of receptors for these peptides in
distinction is most obvious in a postpartum mother that is many parts of the limbic brain. In other words, steroid
highly maternal but rejects her offspring through a failure to hormone receptors have one of their actions in the brain by
bond. This dissociation of maternal behavior and infant rejec- binding to DNA motifs on the promoter regions of certain
tion occurs following failure in the recognition process. genes, many of which code for neuropeptides and their recep-
Recognition is prerequisite for bonding, and in ungulate tors (Table 4).
species a sensitive period following birth provides the The steroid hormones of pregnancy are integral to the
window of opportunity for this recognition bond to occur. induction of maternal responding but do not exclusively
A failure to make this recognition within the sensitive period, code for these events; they also influence sexual behavior,
as sometimes occurs in primiparous mothers, or multiparous feeding behavior, and exploratory behavior, and thus have
mothers if their infant is experimentally removed, results in the recruiting capacity for a wide range of neural systems. The
a rejection of the offspring (Poindron et al., 2007). It might ovarian steroids are therefore perhaps best viewed as part of
seem counterintuitive that a maternally motivated female, a widely distributed addressing system, provided with a degree
having experienced the hormones of pregnancy and parturi- of specificity in the context of maternal behavior by the events
tion, would reject her genetically related infant. Indeed, this which occur at parturition (Keverne and Kendrick, 1992). The
only makes sense when we view maternal rejection behavior prolonged action of progesterone throughout pregnancy
in its appropriate biological context. Within minutes after suppresses estrous cycles and behavior, while parturition itself
birth in ungulates, the mother starts to lick the newborn, recruits other neural systems for the initiation of maternal
which can usually stand and suckle within 2 h of birth behavior, thereby synchronizing maternal care with the birth
(Poindron and Le Neindre, 1980). It is around this same of infants.
122 The Neurobiology of Social Affiliation and Pair Bonding

Table 4 Mechanisms through which steroids and/or peptides may the whole brain may be flooded with the peptide, activating
influence social bonding those receptors where axon terminals fail to project; however,
there is increasing evidence that oxytocin is neuronally secreted
Developmental regulation of species-typical traits (Often including sexual
more widely than was previously recognized (Grinevich et al.,
dimorphism in nervous system and endocrine function)
Alterations in peptide sensitivity or reactivity by altering: 2016).
Hormones or neurotransmitters and/or their receptors Whether secreted locally or through wide perfusion of the
In adulthood: nervous system, it may be the case that the exceptionally high
Peptide synthesis and/or release levels of oxytocin released into CSF at parturition act both as
Peptide receptors and/or binding transmitter and neurohumoral factors. This may permit
Altering behaviors that release peptides oxytocin to orchestrate maternal responses that require activity
Peptide–peptide interactions throughout widespread areas of the brain, including the olfac-
Steroid–steroid interactions tory bulbs, for integration of complex sensorimotor programs.
Steroid–peptide–neurotransmitter interactions
Administration of oxytocin to CSF in nonparturient ewes
Hormonal effects on the autonomic nervous system
evokes the complete complement of sensorimotor patterns
Reproduced from Carter, C.S., Keverne, E.B., 2002. The neurobiology of social that make up full maternal responsiveness (Kendrick et al.,
affiliation and pair bonding. In: Pfaff, D., Arnold, A., Etgen, A., Fahrbach, S., Rubin, 1987).
R. (Eds.), Hormones, Brain, and Behavior. Academic Press, New York, pp. 299–337,
with permission from Elsevier.
The main source of intracerebral oxytocin is the parvocellu-
lar neurons in the PVN, which project widely throughout the
brain. Not only is the PVN the main source of intracerebral
oxytocin, but it also seems to be one of the targets for oxytocin.
1.04.4.2 Oxytocin, Infant Recognition, and Bonding
The relative importance of this oxytocin feedback for the induc-
Pedersen and Prange (1979) first tested the hypothesis that tion of maternal behavior has been shown following low doses
central oxytocin has a role in maternal behavior. It is an attrac- of oxytocin infused directly to the PVN (Da Costa et al., 1996).
tive idea that both the central and peripheral components of The consequent induction of behavior is comparable to the
a neuroendocrine system may be activated simultaneously as induction of maternal behavior by vaginal–cervical stimulation
part of a coordinated behavioral and neuroendocrine response. and by ICV infusions of high doses of oxytocin. Oxytocin
Infusions of oxytocin and its analogue, tocinoic acid, had release in the PVN at parturition is probably facilitating a posi-
a significant effect in promoting maternal behavior within tive feedback on both parvocellular and magnocellular neurons
a 2 h period (Pedersen et al., 1982). Subsequent studies have to coordinate high levels of oxytocin release that are important
shown that the onset of maternal behavior can be delayed by for the induction of maternal behavior, infant recognition, and
intracerebroventricular (ICV) treatment with oxytocin antisera bonding.
or a synthetic oxytocin antagonist (Fahrbach et al., 1985), The half-life of peptides such as oxytocin in CSF is consider-
while olfactory stimuli stimulate maternal care through ably longer than that found in peripheral plasma. Nevertheless,
increased expression of oxytocin receptors in specific brain the duration of its behavioral effectiveness is approximately
areas (Francis et al., 2002). In contrast, long periods of 1 h, a finding in parallel with a rapid waning of maternal
mother–infant separation inhibit maternal care, again through behavior in parturient ewes that fail to bond because their
modulation of oxytocin receptors (Pedersen and Boccia, 2002). lambs are removed (Poindron and Le Neindre, 1980). In this
In the sheep, vaginocervical stimulation and parturition context, it is interesting to note that not only parturition but
increase the levels of oxytocin within the brain, even though also suckling increases CSF oxytocin in sheep (Keverne and
the blood–brain barrier is relatively impermeable to oxytocin Kendrick, 1992), while chemosensory cues from anogenital
(Keverne and Kendrick, 1992). Vaginocervical stimulation licking of pups maintain the efficiency of oxytocin release in
can itself promote a rapid onset of maternal behavior in non- rats in response to suckling. Likewise, a baby’s crying can
gestant ewes (Keverne et al., 1983), while ewes that deliver induce oxytocin release and milk ‘let-down’ in lactating
under epidural anesthesia show no interest in lambs until women. Hence the initiation and maintenance of maternal
such ewes are subsequently given intracerebral oxytocin care appear to differ in important ways. For initiation, oxytocin
(Levy et al., 1992). Also in sheep, administration of oxytocin and its receptors depend largely on steroid hormone priming,
directly to the brain’s ventricular system stimulates a rapid while sustained release of oxytocin is evoked by somatosensory
onset of maternal behavior and induces the ewe to maintain stimulation at parturition. During the maintenance of maternal
proximity with lambs in an open field (Kendrick et al., behavior, steroid levels are low and oxytocin sustains its own
1987). receptors, while its release can be evoked by suckling and other
Microdialysis studies provide direct evidence for the release sensory stimuli from the offspring. The duration of the mainte-
of oxytocin at sites in the brain known to be involved in nance phase is therefore dependent on the frequency of sensory
maternal behavior. In ewes, oxytocin is released during labor, stimulation from the offspring, which diminishes in both type
birth, and suckling in the bNST, medial preoptic area and frequency as the infant becomes self-sufficient.
(mPOA) and olfactory bulbs (Kendrick et al., 1992). For
some time, anatomists have been confused by the mismatch
1.04.4.3 Frontal Cortex: Role in Social and Parental Reward
between neurotransmitter terminals and their receptors in the
brain. Indeed, under basal conditions, oxytocin may only be The prefrontal cortex has shown progressive enlargement
effective at terminal areas. During critical life events such as throughout mammalian evolution (Carter, 2014). In small-
mating and parturition, however, it has been proposed that brained rodents, the medial frontal cortex (mPFC) matures
The Neurobiology of Social Affiliation and Pair Bonding 123

around day 13–15 postpartum (Diergaade et al., 2005), while behavior (Broad et al., 2002). It seems likely that the primary
in the human, this development continues late into adoles- action of the local anesthetic under these circumstances is to
cence or later (Sowell et al., 2001). In rats, this area of the cortex cause a shutdown of neuronal activity in the mPFC, which
mainly receives a strong olfactory input via the mediodorsal promotes local large increases in inhibitory transmitter concen-
thalamic nucleus, and lesions to this system decrease the trations (notably GABA and serotonin). Tetracaine has similar
frequency with which the mother protects her pups by attack- effects to localized cooling on inhibiting membrane conduc-
ing intruders who threaten to cannibalize her infants (Ferreira tance (Luzzati et al., 1999) and so it is likely that behavioral
et al., 1987). Unlike males, females rarely initiate aggression and neurochemical actions observed with tetracaine infusions
to others unless they are nursing offspring (Bosch and result from inhibition of these mPFC neurones, disabling their
Neumann, 2012). The mPFC synchronizes this response to communication with the ventral striatum. Infusions of tetra-
odor cues via the release of CRF from the bNST and PVN where caine in the mPFC, which prevent overt motor rejection of
the majority of CRF neurons are found (Spencer et al., 2005). A strange lambs, do not interfere with recognition memory nor
second major output from mPFC is to the ventral striatum prevent memory retrieval for generating emotional vocal
(NAcc), an area of the brain concerned with reward. Hence, responses, but this treatment does prevent downloading the
in the context of emotionally rewarding behaviors associated information necessary for initiating an aggressive motor
with maternal care, olfactory signals relayed in the mPFC are response via the striatum. Hence the process of maternal
endowed with incentive salience. The reward value provided bonding in the sheep brain is strongly driven by olfactory
by the NAcc requires a biological context signaled by the cues processed in the mPFC. Ungulates are intrinsically averse
hormonal state of the female and activated by release of to strangers, and mothers are especially aggressive to the
opioids and dopamine in the NAcc. It is well established that approach of strange lambs. Therefore proactive maternal care
estrogens increase synthesis of preproenkephalin in the NAcc and subsequent bonding require an active suppression of the
(LeSaux and DiPaolo, 2005) and that the dopaminergic projec- aggressive rejection driven by unfamiliar olfactory signals.
tions from the mesolimbic neurons of the midbrain are also Interestingly, the frontal cortex projection to the ventral stria-
immunoreactive for estrogen receptors (Creutz and Kritzer, tum is known not only to activate reward, but also to be acti-
2004). vated in the context of punishment which also involves
In rats, low levels of maternal care are associated with dopamine release in the ventral stratum (Schultz, 2007). Hence
reduced dopamine release in the NAcc in response to pup an integral component of reward-based bonding, initially
cues (Champagne et al., 2004). In contrast, drugs which act dependent upon olfaction and subsequently expanded to other
in the frontal cortex and NAcc that are associated with reduced social cues (visual and auditory), employs the frontal–striatal
dopamine show reduced PFC response to pup suckling as reward pathways, but first requires the aversive intrinsic
revealed by fMRI (Ferris et al., 2005). Oxytocin has also been circuitry to be gated and silenced.
shown to be active in brain regions associated with mother– The evolutionary enlargement of the frontal cortex in
pup bonding during suckling (Febo et al., 2005; Ferris et al., primates plays a notable role in social interactions and activa-
2015). tion of social reward associated with bonding. Nursery-reared
In sheep that have already given birth, exposure to lamb rhesus monkeys deprived of a maternal upbringing demon-
odors provokes intense c-fos activity in the mPFC as well as strate reduced oxytocin secretion and reduced social interac-
in the pyriform and cortical amygdala, central relays for main tions, but show increased aggressive and stereotypical
olfactory inputs known to be important in lamb recognition behaviors. Increased stereotypical behaviors are often associ-
and selective bonding. However, in sheep, reversible inactiva- ated with disruption of frontal cortical function and often
tion of the mPFC (medial limbic area) using tetracaine infu- result from an inability to suppress inappropriate behavioral
sions demonstrates this region of the brain is not required for responses (Winslow et al., 2003). Squirrel monkeys, 4 years
the performance of proactive maternal behavior or for own after experiencing separations from their mothers early in
lamb recognition. It is, however, required to enable the ewe development, exhibit differences in emotional behavior, stress
to execute an aggressive motor action in response to the odors physiology, and development of the brain, notably in the
of strange lambs. This reversible inactivation does not interfere mPFC. Behavior tests have subsequently shown these monkeys
with other forms of motor activity related to maternal care to be impaired in reward-related memory tasks (Lyons and
including emotionally generated rejection vocalizations, Schatzberg, 2003). Electrophysiological recordings from
walking, approaching, licking and suckling lambs, and/or normal adult monkeys have shown the mPFC mediates the
feeding (Broad et al., 2002). Rejection behavior directed at achievement of goals (Matsumoto and Tanaka, 2004). The
strange lambs is associated with increases in c-fos mRNA precise involvement of the PFC in reward-related behavior
expression in the large output cells of layer V of the mPFC has also been examined in humans using imaging techniques.
(which project to the ventral striatum). This suggests that in Interestingly, the detection of unfavorable outcomes, response
order to execute the aggressive motor response underlying conflict, and decision uncertainty elicit overlapping clusters of
rejection, novel, anxiety provoking olfactory information needs activation foci in the mPFC (Ridderinkhof et al., 2004).
to be routed by the projections from the frontal cortex to the Choosing between actions associated with uncertain rewards
ventral striatum (Lidow et al., 1998). and punishments in humans is mediated by neural circuitry
Aggressive responses to the odors of strange lambs are also involving frontal cortex, anterior cingulate, and striatum
associated with increased levels of dopamine, serotonin, gluta- (Rogers et al., 2004). Showing mothers alternating videos of
mate, and gamma-aminobutyric acid (GABA) in this cortical their own child, versus that of a stranger, provokes the greatest
region, increases that do not occur in the absence of rejection signal contrasts in the mPFC and orbitofrontal cortex. These
124 The Neurobiology of Social Affiliation and Pair Bonding

distinctions require face recognition and emotional processing, declines with opioid receptor blockade. Mother is not the
which correlate with activation in the visual cortex, temporal normal attentive caregiver, and mother–infant interactions
pole, and amygdala (Ranote et al., 2004). are invariably infant-initiated (Keverne et al., 1997; Martel
Recent human neuroimaging research documents the extent et al., 1993, 1995).
to which social cognition, the ability to make inferences about Primates and other mammals have in common opioid
psychological aspects of other people, relies on distinct frontal involvement in maternal care, but the consequences of opioid
cortical areas. For example, regions of the mPFC appear to blockade in small-brained mammals are much greater for the
contribute to the recognition of different emotions (Hornak biological aspects of maternal behavior. In sheep, interference
et al., 1996; Shamay-Tsoory et al., 2005) and to the task of with the endogenous opioid system severely impairs maternal
forming a ‘person impression.’ This area is activated specifically behavior, including suckling, whereas monkeys neglect to show
when a subject is asked to form an impression of a person, but a focused preoccupation with infant care but still permit suck-
not when they are asked to form an impression of an inanimate ling (Kendrick and Keverne, 1989; Martel et al., 1993). These
object, suggesting that this region specifically indexes social differences may reflect the degree of emancipation from endo-
cognition (Mitchell et al., 2005). Studies of the perception of crine determinants that maternal behavior has undergone in
animacy also implicate activation in the medial mPFC and primates, and the increased importance of ‘emotional reward’
related brain regions. In one recent study, individual differ- for the bonding process.
ences in plasma levels of oxytocin were strongly related to acti- If the endogenous opioid system in monkeys is positively
vation in these areas critical for social cognitive processes linked to mother–infant bonding, then heroin addiction,
including the mPFC, suggesting that oxytocin may help to which acts at the same opioid receptor, would be predicted
explain individual differences in social perception (Lancaster to have severe consequences for human maternal bonding.
et al., 2015). Women who are heroin addicts have many aspects of their
social and economic life disrupted, making the data difficult
to disentangle. Nevertheless, by 1 year of age, nearly 50% of
1.04.4.4 Social and Maternal Bonding in Primates
children are living away from their biological mothers, and
The evolutionarily conserved biology that underpins mother– by school age, only 12% remain with their biological mothers
infant bonding in mammals raises the question as to what (Mayes, 1995). These infants have been abandoned for adop-
neural changes have occurred in the maternal behavior of tion or are taken into the care of their grandparents and other
humans and certain nonhuman primates that enables female kin. Moreover, methadone-maintained mothers, when
female–infant bonding to occur outside the context of preg- compared with controls matched for ethnicity, socioeconomic
nancy and parturition and in the absence of lactation. In Old status, infant birth weight, and gestational age, are less likely to
World monkeys, apes and humans, the hormones of preg- have remained the child’s primary parent. Their children are
nancy, parturition, and lactation are not necessary for maternal significantly more likely to have been referred to child protec-
or alloparental care, as females of these species can be motherly tive care or special service agencies for neglect, abandonment,
toward infants without ever being pregnant. Nevertheless, of or abuse.
major significance in primate and human maternal care is the Integral to the bonding process in large brained primates is
endogenous opioid system. Indeed, it has been suggested that the action of the endogenous opioid system on receptors local-
the activation of this system at parturition and during suckling ized to the ventral striatum (Koob and Le Moal, 1997). This
promotes the positive affect arising from maternal behavior area of the brain is involved in ‘reward’ and also requires the
(Broad et al., 1993; Martel et al., 1993, 1995; Franceschini mesolimbic dopamine projection, which detects rewarding
et al., 1989). Studies on naloxone (an opioid receptor antago- stimuli and also when they occur at unpredicted times to
nist) treatment of postpartum rhesus monkey mothers living in enable ‘updating’ of the stimulus (Schultz and Dickinson,
social groups have addressed the importance of opioids in 2000). In primates, the oxytocin system is also important in
maternal bonding. bonding, and in humans, oxytocin release is increased at birth,
In the early postpartum period, a mother’s social interac- following female orgasm and following exposure to neonatal
tions are almost exclusively with her new infant, and opioid images or sounds (Carmichael et al., 1987; McNeilly et al.,
receptor blockade in the mother has marked effects on this 1983). Indeed, as reviewed earlier in this chapter, oxytocin,
mother–infant relationship. This treatment reduces the the endogenous opioids, and their receptors are intimately
mother’s caregiving and protective behavior shown toward coordinated by the hormones of pregnancy and interlinked
their infants. In the early weeks following birth when infant in the context of mother–infant bonding. The mother–infant
retrieval is normally very high, mothers treated with an opioid bonding process in monkeys entails obsessive grooming espe-
receptor blocker neglect their infants and show little retrieval cially to hands, face, and genitalia by mothers, and these are
even when the infant moves a distance away. As the infants the morphological traits of infant monkeys that show the
approach 8 weeks of age, when a bonded grooming relation- greatest changes during development. Because primates
ship normally develops between mother and infant, mothers show extended postpartum care, offspring recognition
again treated with an opioid receptor blocker fail to groom requires the continual updating of any changes in these
their infant. Moreover, they permit other females to groom morphological features and in behavioral development. This
their infants, while saline-treated control females are very updating of infant recognition involves visual cues and
possessive and protect their infants from contact with others prefrontal–ventral striatal pathways, which are also intimately
at this stage. Mothers do not reject from suckling, but the linked to the emotional brain via the amygdala. The positive
mother’s usual possessive preoccupation with the infant emotional responses that infants generate in females enable
The Neurobiology of Social Affiliation and Pair Bonding 125

parental care to occur without the continual priming by preg- In mammals, female reproduction is constrained by gesta-
nancy and parturition. tion, birth, and lactation. In contrast, males have the option
Human mothers also experience preoccupations and rituals to invest less than females in their offspring, with the minimal
in the context of maternal care, and even before the birth of investment being a contribution of sperm and genetic material.
their baby, they are obsessive with cleaning and creating It has been conventional to describe mating systems in terms of
a safe environment. After birth, safety is the major concern, male fitness or reproductive success. However, defining mating
and mothers frequently check on their baby even at times systems in terms of male behavior tends to ignore female
when they know the baby is fine. The evolution of these obses- fitness, female–female interactions, and the importance of the
sive psychological and behavioral states can be seen as a devel- larger family unit. In addition, DNA fingerprinting has revealed
opmental extension of the preoccupations all primates show that at least some of the offspring raised by seemingly monog-
for their infants. Thus it is notable that areas of the human amous partners of most species are in fact due to ‘extra-pair
brain that MRI findings shown to be responsive to babies’ copulations’ or EPCs (Okhovat et al., 2015). The high inci-
crying include the brain’s reward structures (mesolimbic dopa- dence of EPCs and extra-pair paternity creates a problem for
mine inputs from the ventral tegmental area to the ventral stria- theories that focus on monogamy purely as a mating system-
tum and amygdala) (Lorberbaum et al., 2002). These same and have encouraged scientists to shift their attention to social
regions of the brain are also active in the context of romantic systems.
love (Fisher et al., 2015).
The evolutionary progression away from hormonal centric
1.04.5.2 Social Systems
determinants of maternal behavior to emotional, reward-ful-
filling activation, probably involves dopaminergic and opioi- A vast array of social systems have been described, many of
dergic activity in the ventral striatum. The enhanced role of which are based directly on mating systems (such as polygamy
this circuitry for regulating behavior in humans may also or monogamy), while others may be defined by the living
provide vulnerability to various forms of psychopathology arrangements or genetic relationship among the members of
such as obsessive compulsive disorder (OCD) and substance a group. In comparison to mating systems, definitions of social
abuse. Mild forms of addictive behavior (gambling, video systems are more likely to take into account the role of females
games, internet use, and consumption of caffeine and choco- and offspring. For example, as described above, many mamma-
late) may be such indicators of this neurological predisposition lian social systems can be described as matrilinear, and a variety
for obsessive behavior seen in humans (Greenberg et al., 1999). of mammalian species live in groups in which young animals
In vivo neuroimagining studies identify the orbital frontal may remain in the natal family. The tendency to form extended
cortex, head of the caudate, and closely associated ventral stria- families, often, but not always, based around a monogamous
tum and anterior cingulate as being involved in OCD (Rauch, pair, has been termed communal or cooperative breeding
2000), while acquired OCD occurs later in life in patients (reviewed Solomon and French, 1997).
with striatal lesions (Chacko et al., 2000). There is also
evidence that CSF levels of the neuropeptide, oxytocin, are
1.04.5.3 Sexual versus Social Monogamy
elevated in OCD, and this peptide also plays a fundamental
role in many obsessive aspects of maternalism (Leckman Most of what has been written regarding monogamy focuses on
et al., 1994). Not surprisingly, therefore, OCD is more sexual monogamy. The awareness that – at the species level –
common in women. The influence of gonadal hormones on sexual exclusivity is rarely created a challenge for the concept
periodicity of OCD (Weiss et al., 1995) and the postpartum of sexual monogamy (Carter et al., 1995). Nonetheless, selec-
exacerbation of OCD symptoms in women suggest that the tive social behaviors and pair bonds between adults do provide
course of this disorder may be influenced by the hormones a social matrix for reproductive behaviors, including sexual
of pregnancy (Williams and Koran, 1997; Labad et al., 2005). behavior and care of offspring, supporting the welfare of indi-
Hence some components of human behavior that occur in viduals, groups, and species.
the postpartum maternal period are influenced by hormones, The traits of mammalian social monogamy typically occur
but interestingly they relate to areas of the brain concerned together, forming what has been called a ‘syndrome’ (Kleiman,
with reward and not with the direct execution of maternal 1977). The cardinal characteristic of social monogamy is
behavior per se. a male–female pair bond, characterized by selective partner
preferences and social interactions. Selective aggression is
another feature of social monogamy. Monogamous mammals
1.04.5 Adult Pair Bonds are highly social within their families, although they can show
lethal aggression toward strangers (Carter et al., 1995; Bowler
1.04.5.1 Mating Systems and Pair Bonds
et al., 2002). Aggression is most common toward members
One common method for categorizing species is based on of the same sex and usually occurs during or after pair bond
mating systems, characterized by the presumed choice or formation. The capacity of socially monogamous partners to
number of sexual partners, and often focused on male defend their mates, family, or territory may influence reproduc-
behavior. The most common mating system in mammals is tive fitness and their own survival. Incest avoidance, a tendency
called polygamy (many mates) or polygyny (many wives). for reproductive suppression of the young, who remain as allo-
The less common alternative, monogamy (one mate) is esti- parents in communal families, and a relative absence of phys-
mated to occur in about 3% of mammalian species (Kleiman, ical sexual dimorphism also characterize monogamous
1977), while polyandry (many husbands) is even rarer. mammals.
126 The Neurobiology of Social Affiliation and Pair Bonding

1.04.5.4 Social Monogamy in Prairie Voles for the familiar male. However, even in postpartum estrus, if
given an opportunity, a significant proportion of females will
Prairie voles (Microtus ochrogaster) exhibit all of the features of
mate with an unfamiliar male. Once females are in estrus,
social monogamy including the formation of pair bonds
sexual postures allowing mating are reflexive and may be less
between adult conspecifics. Prairie voles, which are small
selective than social contact (Carter et al., 1995).
rodents found in grasslands throughout midwestern North
Pair-bonded males become highly aggressive following
America, have attracted particular interest because they can be
mating and probably patrol the runways that lead to their
studied under both field and laboratory conditions. Field
nest. If successful in defending his partner, then the male
studies begun in the 1960s provided the first evidence that
may father the offspring that his partner delivers. In nature,
prairie voles form long-lasting pair bonds (Carter et al.,
when one member of the pair dies or abandons the nest, fewer
1995). Male and female pairs of this species maintain
than 20% of the remaining partners form a new pair bond
a common nest and territory and tend to enter live traps
(Getz et al., 1993); thus for most prairie voles, pair bonds
together as long as both members of the pair are alive. By
last until the pair is separated by death.
contrast, less social, polygamous meadow voles (Microtus pen-
nysylvanicus) and montane voles (Microtus montanus) show
few indications of pair bonding; males and females of these 1.04.5.5 Experimental Analysis of Partner Preferences
species have separate nests and territories and are usually trap-
ped alone. In the laboratory, nonmongamous species are less Prairie voles adapt easily to the laboratory, and therefore it has
likely to engage in social contact and less likely to exhibit selec- been possible to investigate various aspects of the behavioral
tive partner preferences. and neuroendocrine control of pair bonding. Voles may be
Prairie voles live in nature in communal family groups placed in a large three-chambered apparatus that allows the
comprised primarily of a male and female breeding pair and experimental animal to spend time with either a stimulus
their offspring. About 70–75% of young voles do not leave animal designated by familiarity as the ‘partner,’ or with
their natal family (Getz et al., 1993). The original breeding a comparable ‘stranger.’ or elect to be alone in a neutral cage
pair within a family has a reproductive advantage, while (Williams et al., 1992). Female stimulus animals are tethered
most other members of the communal groups are reproduc- loosely, allowing them to move only within their own
tively inactive offspring. These offspring serve as helpers at chamber, whereas the experimental male is free to explore the
the nest or alloparents, presumably gaining reproductive entire test apparatus. This testing paradigm yields reliable social
advantages through inclusive fitness. Familiarity inhibits repro- preferences that can be studied as a function of physiological
duction in young prairie voles, and incest is avoided through and experimental manipulations. In female prairie voles, social
several mechanisms, including reluctance to mate with a family preferences for the familiar partner are established more quickly
member. Young males remain sexually suppressed within the if mating occurs prior to testing. Nonsexual cohabitation also is
family nest and must leave the family group to reproduce eventually followed by a significant partner preference (DeVries
(Carter et al., 1995; Okhovat et al., 2015). New pairs are and Carter, 1999). As mentioned above, however, social prefer-
most likely to form when previously naïve males and females ence may bias reproduction to favor the familiar or resident
leave their group, meet an unfamiliar member of the opposite male; laboratory studies do not indicate that newly paired
sex, develop new pair bonds, and mate and generate their own females form a strong sexual preference for familiar or unfa-
families. Pair bond formation in prairie voles commences miliar males (Carter et al., 1995). In fact during the early stages
within hours, or sooner, after meeting. In addition, when of pair bonding, females are equally likely to mate with familiar
young males and females meet strangers of the opposite sex, and unfamiliar males. For this reason, social contact is more
a cascade of events is initiated which leads to reproductive acti- selective and probably more useful as an index of pair bonding
vation in both sexes and eventual estrus induction in the than is sexual preference (Carter and Getz, 1993).
female (Carter et al., 1995). Female prairie voles do not exhibit
ovarian cycles, and spontaneous estrus is rare. Mating usually
1.04.5.6 Selective Aggression as a Component of Pair
follows within a day or two of male–female pairing. In newly
Bonding
formed pairs, intermittent mating bouts continue for about
24–48 h followed by a 21-day gestation. After an initial mating Sexually naïve prairie voles of either sex are rarely aggressive
and pregnancy, prairie vole females experience a postpartum toward conspecifics. By contrast, after approximately
estrus, allowing them to mate again and nurse one litter while 12–24 h of sexual experience, male prairie voles become
they are pregnant with a second litter (Witt et al., 1989). extremely aggressive toward unfamiliar males (Winslow
Attempts to use sexual preferences as an index of pair et al., 1993). Sexually experienced ‘breeder’ males are
bonding have not been successful. However, significant social socially dominant and may drive away or even kill male
preferences for physical contact with a new partner can be strangers. However, they are usually not aggressive to
measured within hours following an initial pairing (Carter familiar animals, and breeder males will allow their own
et al., 1995). The onset of social preferences is facilitated by offspring to remain in the family, at least as long as these
mating, but can occur in the absence of mating and even in offspring are sexually naïve. Cohabitation with a nonrepro-
gonadectomized animals, albeit less readily (Williams et al., ductive female, even for several days, usually does not
1992). produce a reliable increase in male aggression; however,
Females in postpartum estrus that have remained with nonsexual cohabitation does cause hormonal changes that
a male for one or more litters may exhibit a sexual preference are associated with reproductive activation. Females also
The Neurobiology of Social Affiliation and Pair Bonding 127

become aggressive after mating, but postcopulatory aggres- heterologous receptor (oxytocin plus AVPV1aA or vaso-
sion develops more slowly in females than in males. In pressin plus OTA). However, when animals are treated with
females the onset and intensity of aggression is variable either an OTA or an AVPV1aA, they no longer show a selec-
and does not require sexual interactions (Bowler et al., tive partner preference. These results indicate that either
2002). If two unrelated females are placed with a male, oxytocin or vasopressin, at least when applied at high doses,
both may mate with the male (Witt et al., 1989). Under can stimulate nonselective social contact in prairie voles of
these conditions, one female becomes socially dominant, either sex; however, it is possible that both types of receptors
and the nondominant female is blocked from social contact must be accessible and stimulated to allow these peptides to
with the male (Firestone et al., 1991). facilitate the formation of a selective social preference. Cross-
In both sexes, postpartum aggression is common and may reactivity between oxytocin and vasopressin and their recep-
be directed toward both unfamiliar females and unfamiliar tors probably occurs in most mammals and under many
males (Getz et al., 1981). However, female prairie voles do conditions that regulate dynamic social interactions (Albers,
not express aggression toward their own offspring. Female– 2015).
female aggression, sometimes leading to death of one female, Vasopressin facilitates partner preference formation, espe-
is also a feature of the behavior of paired tamarins and other cially in male prairie voles. ICV infusion of an AVPV1aA
monogamous mammals (Kleiman, 1977). prevents mating-induced partner preference formation as well
Partner preferences and selective aggression characterize as the selective aggression associated with mating in male
established pair bonding in prairie voles while sexual and prairie voles (Winslow et al., 1993). Prairie voles have higher
social experiences regulate the onset of the pair bond (Carter levels of V1a receptors in several brain regions including the
et al., 1995). However, partner preferences can develop even ventral pallidum, medial dorsal thalamus, and mAMY, espe-
in the absence of gonadal hormones. The initial development cially in comparison to nonmonogamous montane or meadow
of a partner preference usually occurs prior to mating during voles.
a time when gonadal hormones are comparatively low. Subse- In male prairie voles, infusing the AVPV1aA into the ventral
quent sexual experience may consolidate the pair bonding pallidum, but not other regions, prevents partner preference
process (Williams et al., 1992). Hormonal events associated formation (Lim and Young, 2004). In addition, upregulating
with social interactions therefore are likely contributors to the V1a receptor in this region facilitates the formation of
pair bond formation. Prior to these studies, it was known partner preferences. Species differences in pair bonding may
that neuropeptides, including oxytocin and vasopressin, are arise from species differences in the expression pattern of the
released by both sexual (reviewed Carter, 1992) and nonsexual V1a receptor in males. In male meadow voles, which do not
contact (Uvnas-Moberg, 1998). In addition, as described normally form partner preferences, overexpression by viral
above, oxytocin is implicated in both nonselective maternal vector of the prairie vole V1a receptor in the ventral pallidum
behavior and maternal bonding. resulted in the development of partner preferences (Lim
Research on pair bonding has focused on factors capable of et al., 2004). A separate study found that infusing AVPV1aA
affecting the formation of a pair bond and the induction of into the LS inhibits partner preference (Liu et al., 2001). Septal
selective aggression. Relatively, little is known regarding the V1a receptors also regulate selective social recognition in male
mechanisms involved in the maintenance of these behaviors, mice (Bielsky et al., 2005). The LS projects heavily to the NAcc
although there is reason to believe that the factors responsible and may allow social signals to activate reward pathways and
for the induction of pair bonding can differ from those facilitate pair bonding.
involved in its maintenance. For example, compounds that Based on the behavioral effects of vasopressin and species
block vasopressin prevent the development, but not the expres- differences in the vasopressin receptor, Young and his associ-
sion, of selective aggression (Winslow et al., 1993). In addition, ates (1999) have created transgenic mice overexpressing the
as detailed below, the induction and maintenance of pair gene for the vasopressin receptor. These transgenic animals
bonds may rely on different dopamine receptors subtypes express the vasopressin receptor (V1a) in a pattern that was
(D2 versus D1) (Aragona et al., 2006). different from wild-type mice and similar in some brain
regions to prairie voles. For example, V1a receptor binding
is elevated in transgenic mice in the CC, claustrum, laterodor-
1.04.5.7 Affiliative Behaviors and Positive Sociality
sal and ventroposterior thalamic nuclei, and especially the
Most socially monogamous mammals show high levels of olfactory bulbs. Oxytocin receptor distribution is not altered
social contact within the family. Sociality can be either selec- in the V1a transgenic mice. When the V1a transgenic male
tive, and thus in some cases indicative of social bonds, or animals are treated with exogenous vasopressin, they also
nonselective. Oxytocin and vasopressin can increase the show high levels of social contact with female stimulus
tendency to show both selective and nonselective social animals.
behaviors. For example, treatment (ICV) with either oxytocin A highly repetitive microsatellite DNA element is found in
or vasopressin approximately doubles nonselective social the promoter of the prairie vole; this gene promoter is virtually
contact in male and female prairie voles (Cho et al., 1999). absent in the montane and meadow voles. Variations in this
Pretreatment with a selective antagonist for either peptide microsatellite can alter gene expression in a cell-specific
(oxytocin plus an OT antagonist (OTA), or vasopressin manner (Hammock and Young, 2004). In prairie voles, V1a
plus an AVP V1a antagonist (AVPV1aA)) reduces social receptor abundance varies among individuals (Okhovat et al.,
contact. High levels of social behavior are still seen if either 2015). The microsatellite in the gene promoter varies signifi-
of these peptides is combined with an antagonist for the cantly among individuals (Hammock and Young, 2002,
128 The Neurobiology of Social Affiliation and Pair Bonding

2005). Prairie voles have been bred based on the length of their 1.04.5.8 Selective Social Behavior, a Role for Oxytocin
microsatellites. Those with a long microsatellite have higher and Vasopressin
levels of V1a receptor expression in several brain regions,
Taken together, these data suggest that neural systems respon-
including the olfactory bulb and LS, and are more likely to
sible for the expression of sociality can be stimulated by either
display partner preferences than males with short microsatel-
vasopressin or oxytocin. However, the formation of social
lites (Hammock and Young, 2005). The repeat sequences
bonds may require both peptides and/or the activation of addi-
found in microsatellites often recruit methylation and gene
tional neural systems, beyond those required to facilitate
silencing, which is opposite to the findings demonstrated in
nonselective social behaviors (Cho et al., 1999).
these studies. Moreover, the generality of this finding remains
In prairie voles, sexual experience can hasten the onset of
uncertain. A survey of other nonmonogamous vole species sug-
pair bonding (Williams et al., 1992) and, in males, induces
gested that the microsatellite is also present in several other
aggression toward strangers (Winslow et al., 1993). It is also
nonmonogamous rodent species, and therefore is not solely
known that oxytocin and vasopressin can be released during
responsible for differences in social organization across other
mating (reviewed in Carter, 1992). These observations sug-
vole species (Fink et al., 2006).
gested the hypothesis that oxytocin and/or vasopressin could
One study reports that an OTA, but not an AVPV1aA,
promote social bonding. Oxytocin infusions, when centrally
prevents mating-induced pair bonding in females (Insel and
administered, do indeed facilitate the onset of partner prefer-
Hulihan, 1995). A separate study found that an AVPV1aA,
ences in sexually naïve female and male prairie voles (Williams
but not an OTA, significantly inhibits partner preference in
et al., 1994; Cho et al., 1999). Moreover, oxytocin antagonists
males (Winslow et al., 1993). These data suggest a sex differ-
reduce the behavioral effects of exogenous oxytocin and also
ence in the relative role of endogenous oxytocin and vaso-
block partner preference formation during prolonged cohabita-
pressin in pair bond formation. However, in male prairie
tion. In addition, in male prairie voles, vasopressin is necessary
voles, infusions of either oxytocin or vasopressin can facilitate
to the onset of stranger-directed aggression, which in nature
partner preferences after a brief cohabitation (Cho et al.,
would promote mate guarding, another essential aspect of
1999). In addition, infusion of an OTA into the LS blocks
pair bonding.
partner preference formation in male prairie voles (Liu
Experimental evidence for a role of vasopressin in males
et al., 2001). Taken together, these studies suggest that activa-
comes from the fact that pretreatment with a V1a vaso-
tion of both the oxytocin receptor and V1a receptor may be
pressin antagonist inhibits pair bonding that normally
necessary in selective partner preference formation in prairie
results from sexual experience (Winslow et al., 1993). In
voles.
addition, exogenous vasopressin administration facilitated
In general, sex differences have not been reported in
aggression induction in this species. When tested under
oxytocin or vasopressin V1a receptor binding in the prairie
comparable conditions, infusions of vasopressin were not
vole brain (Insel and Shapiro, 1992; Wang et al., 1996). Quan-
effective in facilitating female aggression. Thus, the physi-
tification of oxytocin synthesis in the PVN and SON does not
ology of female aggression in pair bonding species remains
reveal sex differences (Yamamoto et al., 2004), although the
to be understood.
release of oxytocin may, under some conditions, be sexually
dimorphic (Kramer et al., 2004). However, in various species
including voles, vasopressin synthesis and release is higher in
males in the mAMY, BNST, and LS, brain regions that may be 1.04.6 Stress and Pair Bond Formation
important in a variety of social and defensive behaviors (De
Vries and Panzica, 2006). It is also possible that, under some Oxytocin and social behaviors are critical components of the
conditions, oxytocin is preferentially released in the female capacity of the mammalian body to manage and downregulate
(Kramer et al., 2004), which might be especially relevant reactivity to stressful experiences. Oxytocin has effects on the
during the vaginocervical stimulation associated with copula- regulation of emotion, the mammalian autonomic nervous
tion. In addition, oxytocin is essential for social recognition, system, homeostasis, coping, and healing, helping to explain
which is implicit in pair bonding in both sexes. the consequences of the presence or absence of social engage-
Additional information regarding the neural basis of social ment and attachment. For example, oxytocin has been impli-
behavior comes from studies of ‘knockout’ mice, created with cated in human wound healing (Gouin et al., 2010) and is
selective genetic lesions. Deletions of the exon that encodes protective against cardiovascular dysfunction, acting to repair
the oxytocin peptide in mice produce an animal (OTKO) tissue (Karelina and DeVries, 2011). Oxytocin also has antiox-
capable of all aspects of reproduction except milk ejection idant and anti-inflammatory properties across the life span
(Nishimori et al., 1996). However, the social behavior of (Jankowski et al., 2016). Acute stressors, especially of high
OTKO mice also is not normal. OTKO mice tend to be more intensity, can release oxytocin in both sexes (Neumann and
defensively aggressive and reactive to stressors than normal Landgraf, 2012), possibly helping the organism to adapt in
mice (Winslow et al., 2000; Amico et al., 2004). OTKO mice the face of a challenge. These adaptive properties of oxytocin
also have deficiencies in ‘social memory,’ which are restored help to explain the capacity of social bonds to protect and
by injections of oxytocin (Ferguson et al., 2000). Social recog- heal in the face of stress and adversity. Concurrently, stressors
nition is necessary for pair bond formation, and other studies seem to play a major role in the formation of social bonds.
suggest that both oxytocin and vasopressin play a role in social Evidence for a role for hormones of the HPA axis in pair
recognition in male rodents (Bielsky and Young, 2004; Bielsky bond formation comes from studies which reveal the repeated
et al., 2005). association between stressful experiences and social bonding
The Neurobiology of Social Affiliation and Pair Bonding 129

(Carter, 1998; Sachser et al., 1998). More direct evidence can be change was not detected in sexually experienced males for
found in laboratory studies of the effects of stressful stimuli or which this situation may have been less novel (Hillegaart
administration of hormones of the HPA axis on the formation et al., 1998). Oxytocin treatments may reduce anxiety as
of partner preferences. measured by exploration of a novel environment in rats
Evidence for the role of biochemistry in the formation of an (McCarthy et al., 1992; Uvnas-Moberg, 1997). In humans
attachment comes from behavioral changes associated with (Chiodera et al., 1991) and prairie voles (DeVries et al.,
mammalian birth, lactation, and sexual interactions. In addi- 1997), oxytocin inhibits the secretion of glucocorticoids. Social
tion, novel or stressful experiences may encourage increased contact also can inhibit HPA axis activity in prairie voles
social behaviors and attachment. Comparatively high levels (DeVries et al., 1995, 1996). In reproductively naïve prairie
of HPA axis activity or indications of sympathetic arousal, voles, either oxytocin (ICV) or social contact produce a 50%
and the subsequent release of oxytocin, have been measured decline in corticosterone, which occurs within 30–60 min. In
under conditions that commonly precede or are associated male prairie voles, either mating (Insel et al., 1995) or vaso-
with the formation of social bonds. pressin treatment (Dharmadhikari et al., 1997) is associated
Mammalian birth is a uniquely stressful experience. In the with increased exploration in the open arm of a plus maze,
mother, the physiological events preceding and during parturi- a measure often considered indicative of reduced anxiety.
tion involve exceptionally high levels of adrenal activity (both The unanticipated finding that socially naïve prairie voles
glucocorticoids and catecholamines) and the subsequent respond to exposure to a novel stranger of the opposite sex
release of peptides, including endogenous opioids, oxytocin, with a decline in corticosterone suggests that pair bonding in
and vasopressin (Keverne and Kendrick, 1992; Kendrick, prairie voles might be inhibited by hormones of the HPA axis
2000; Landgraf et al., 1991). Infants also experience parturi- (DeVries et al., 1995). This has been shown to be the case
tional stress or ‘birth trauma’ and probably experience when, in naïve female prairie voles, removal of the adrenal
increased exposure to maternal oxytocin during labor. glands facilitates the development of a partner preference.
Oxytocin has a large profile of neural consequences which Adrenalectomized females form a significant preference for
may help to reduce the vulnerability of infants during the birth the familiar male within 1 h or less of cohabitation compared
process, although whether this applies to exogenous oxytocin, with the usual requirement of in excess of 3 h. The facilitatory
administered, for example, to induce birth, requires further effect of adrenalectomy is reversed by corticosterone replace-
study. Hormonal experiences associated with birth may affect ment prior to female exposure. In addition, increasing cortico-
the tendency of young animals to form social bonds, although sterone levels in females, by exogenous injection or pellets of
such effects remain to be demonstrated. corticosterone, produce a dose-dependent inhibition of the
In species that form heterosexual pair bonds, including preference formation.
prairie voles, sexual interactions are associated with the forma- In contrast, in socially naïve male prairie voles, the behav-
tion of social attachments (Carter et al., 1995). Sexual behavior ioral effects of corticosterone, when examined in experiments
also can be physiologically stressful for both sexes. Adrenal that paralleled those in females, produce strikingly different
steroids, vasopressin, oxytocin, and endogenous opioids are results (DeVries et al., 1996). Injections of corticosterone, or
released during sexual behavior (reviewed in Carter, 1992). the stress of swimming, facilitated the development of partner
Stress or corticosterone facilitates pair bond formation in preferences in males. By contrast, removal of the adrenal
male prairie voles (DeVries et al., 1996). Although female gland was followed by failure to form a partner preference,
prairie voles do not form pair bonds with familiar males which was subsequently restored by corticosterone
following stressful treatments, stress does encourage the devel- replacement.
opment of preferences for other females, consistent with the Sex differences in the behavioral effects of oxytocin and
communal breeding pattern of this species (DeVries, George vasopressin are consistent with a larger literature implicating
and Carter, unpublished data). Steroid hormones, including oxytocin in various aspects of positive sociality and female
glucocorticoids, can influence the synthesis of, release of, reproduction and vasopressin in territoriality and defensive
and/or receptors for neuropeptides, including oxytocin. In behaviors. As described above, vasopressin is more abundant
addition, steroid and peptide hormones may regulate or in males, and vasopressin production, especially within the
interact with each other to influence behavior. mAMY and bNST, is increased by androgens. Sex differences
Hormones can reduce fear or behavioral inhibition and also exist in pair bonding (DeVries et al., 1996), and it has
permit the expression of social behaviors, such as those been suggested that pair bonding in female prairie voles
necessary for pair bonding, maternal behavior (Chirino and depends primarily on oxytocin, while males rely on vaso-
González-Mariscal, 2015; Fleming et al., 1989; McCarthy pressin (Winslow et al., 1993; Insel and Hulihan, 1995).
et al., 1992), or sexual behavior (Carter, 1992). The neuro- However, there are no dramatic sex differences in oxytocin
chemical processes that overcome neophobia also may be or vasopressin receptors in prairie voles (Insel, 1997). As
needed to permit the formation of new social attachments. Pro- mentioned above, steroid-dependent increases in hypotha-
social behavior or social contact is facilitated, and aggression is lamic oxytocin receptors have been reported in rats
diminished following central oxytocin treatments in estrogen- (Schumacher et al., 1990). In female prairie voles, estrogen
treated female prairie voles (Witt et al., 1990). Increases in treatments do not change hypothalamic oxytocin receptors;
social contact also follow oxytocin treatment in both male only the AON shows an obvious estrogen induction of
and female rats (Witt et al., 1992). In addition, in sexually naïve oxytocin receptor expression (Witt et al., 1991). Furthermore,
male rats, a brief (15 min) heterosexual interaction is followed when given as an exogenous treatment, either oxytocin or vaso-
by an approximate doubling of serum oxytocin levels; this pressin can facilitate pair bonding in both male and female
130 The Neurobiology of Social Affiliation and Pair Bonding

prairie voles, and receptors for both peptides may need to be activation of D1-type receptors in NAcc block partner prefer-
available to allow pair bond formation (Cho et al., 1999). ence formation induced either by mating or by D2-type
The finding, described elsewhere, that the effects of stressful receptor activation (Aragona et al., 2003, 2006), whereas
experiences on prairie vole pair bonding are sexually dimorphic blockade of D1-type receptors in the NAcc of pair-bonded
(DeVries et al., 1996) is most easily interpreted in the context of males (paired with a female for 2 weeks; a manipulation that
natural history. Male prairie voles must leave the family to upregulates D1-type receptors within the NAcc) diminished
breed and must be capable of forming new pair bonds and aggressive behavior toward a novel conspecific female
mating under stressful conditions. In contrast, females of this (Aragona et al., 2006). This selective aggression is considered
species may respond to stressful conditions by remaining as an additional behavioral index of monogamy and pair
within the natal family (Getz et al., 1993). Thus, sexually bonding (Winslow et al., 1993). Together, these data demon-
different behavioral strategies and the hormonal response to strate that in NAcc, D2-type activation is important for pair
stress from the HPA axis may both reflect and support a sex bond formation. In contrast, D1-type activation prevents
difference in reproductive strategies. partner preference formation and is related to high levels of
CRF regulates the release of ACTH and also is known to selective aggression toward a conspecific stranger and thus
have a number of behavioral effects including increased may play a role in the maintenance of an established pair
arousal. In addition, in rats, glucocorticoids are capable of bond (Aragona et al., 2006).
enhancing the behavioral effects of CRF (Schulkin, 1999). It is likely that dopamine also plays an important role in
CRF, administered ICV and in moderate doses, is capable of human social bonding. For example, dopamine receptor-rich
facilitating pair bond formation in male prairie voles (DeVries areas of brain have been implicated in romantic love in
et al., 2002). There is recent evidence from male voles that loss humans (Fisher et al., 2015).
of a partner disrupts the capacity to cope with stress through
pathways that involve the NAcc; this disruption can be restored
by application of oxytocin into the shell of the NAcc in which 1.04.7 Responses to Separation
oxytocin receptors are colocalized with receptors for CRF
(Bosch et al., 2016). One indication of pair bond formation is the degree to which
These findings are consistent with the observation in sheep animals show behavioral or autonomic responses to the
that CRF can facilitate the maternal acceptance of a lamb absence of a familiar or preferred partner. Because responses
(Keverne and Kendrick, 1991). In addition, there is evidence to separation are species-typical, they are best understood in
in rats that CRF is colocalized with oxytocin (Levin and Saw- the context of social organization and ecology.
chenko, 1993), suggesting that functional interactions between Separation from an attachment figure may be associated
these peptides are likely. with various behavioral and physiological changes (Reite and
Bocca, 1994; Hennessy, 1997; Sachser et al., 1998; Mendoza
and Mason, 1997; Mason and Mendoza, 1998). In young
1.04.6.1 Dopamine and Pair Bonding
animals, vocalizations, in either the audible or ultrasonic range,
Dopamine modulates pair bonding behavior in prairie voles often increase following separation. Measurements of these
(Gingrich et al., 2000; Aragona et al., 2003, 2006; Liu and vocalizations have been used as indices of ‘distress’ and may
Wang, 2003). The effects of dopamine may be due to effects be indicative of attachment (Nelson and Panksepp, 1998).
on a wide variety of events that are important for partner pref- Physiological changes, including increased secretion of gluco-
erences, such as sensory processing, reward, memory forma- corticoids and/or ACTH, cardiovascular measures, and
tion/consolidation, and behavioral expression. Additionally, immune system parameters, also have been described
dopamine is released during mating (Gingrich et al., 2000), following social separation in primates (Reite and Boccia,
which in turn facilitates pair bonding (Williams et al., 1992). 1994). Cortisol responses are sometimes used to assess the
Reward associated with the formation of social bonds is regu- intensity of separation ‘distress’ and/or to examine the hypoth-
lated by the dopamine transmission in the NAcc. Intra-NAcc esis that the presence of a partner may provide a form of social
administration of a general dopamine receptor antagonist buffering (Hennessy, 1997; Mendoza and Mason, 1997; Mason
blocks mating-induced partner preferences, indicating that and Mendoza, 1998).
dopamine transmission within the NAcc is necessary for pair Social separation in both male and female prairie voles is
bonding. In addition, intra-NAcc administration of a general followed by an increase in glucocorticoid (corticosterone)
dopamine receptor agonist dose-dependently induces this levels (DeVries et al., 1996). When reunited with a partner,
behavior without mating, showing that dopamine receptor corticosterone levels drop to below baseline in previously
activation is sufficient to produce pair bond formation paired males and females. However, if previously paired
(Aragona et al., 2003, 2006). animals and separated animals are placed with an unfamiliar
Dopamine acts on two distinct groups of receptors, D1 and partner of the opposite sex, corticosterone levels remain
D2. Dopamine D1-type and D2-type receptors have different elevated.
effects on pair bonding in both male and female prairie voles. Social separation for a period of a few days in both male
Blockade of D2-type receptors in NAcc blocks mating-induced and female prairie voles is followed by an increase in corticoste-
partner preferences. Conversely, activation of D2-type receptors rone levels, as well as CRF (Ruscio et al., 2007). When reunited
in NAcc induces partner preferences without mating (Gingrich with a partner, corticosterone levels drop to below baseline in
et al., 2000). Such effects are not found following a similar previously paired males and females. However, if previously
manipulation of D1-type receptors in NAcc. In addition, paired and then separated animals are placed with an
The Neurobiology of Social Affiliation and Pair Bonding 131

unfamiliar partner of the opposite sex, corticosterone levels voles, offer useful models for examining the biology of isola-
remain elevated (DeVries et al., 1996). tion because they share with humans the capacity to form
Prolonged separation or chronic isolation over a period of long-lasting social relationships. For example, prairie voles
weeks has many consequences including increases in heart exhibit social monogamy (Carter et al., 1995). They also have
rate, reductions in parasympathetic activity, and increases in a humanlike autonomic nervous system, with high levels of
behavioral responses, such as immobility in a forced swim parasympathetic–vagal activity and a dependence on social
test, anhedonia, and reduced exploration in an elevated plus behavior for emotion regulation (Kenkel et al., 2013). Because
maze. These behavior measures are considered to index depres- the parasympathetic nervous system mediates many of the
sion and anxiety (Grippo et al., 2007a, 2008, 2009; Bosch et al., consequences of social interactions, this aspect of prairie vole
2016). In experiments subjecting voles to chronic isolation, biology helps us understand the high levels of sensitivity to
corticosterone is not elevated. However, chronic isolation, the presence or absence of social experiences in this species.
especially in female prairie voles, is associated with elevations In prairie voles, isolation from a partner for a few weeks
in endogenous oxytocin and oxytocin synthesis, which may produced significant increases in several behavioral measures
help to downregulate the HPA axis (Grippo et al., 2007b). of depression and anxiety (Sun et al., 2014). Isolated animals
Support for the latter hypothesis comes from experiments in were less exploratory, showed increases in anhedonia (indexed
voles showing that chronic treatment with exogenous oxytocin by a loss of preference for sweet liquids), and were more likely
(administered peripherally) prevents or reverses several of the to show immobility in response to a stressor. In prairie voles,
detrimental cardiac and behavioral effects of isolation (Grippo separation from a partner, followed by prolonged isolation, is
et al., 2009; Bosch et al., 2016). associated with increases in heart rate, decreases in parasympa-
Social experiences also may release oxytocin. In adult, thetic function, and increased behavioral reactivity to stressors,
nonreproductive prairie voles, pup exposure can facilitate pair such as the presence of a social intruder. As one example,
bond formation, possibly through the release of oxytocin following a 5-min social stressor (intruder), isolated prairie
(Kenkel et al., 2012; 2013). Social stimuli from a pup exposure voles required an average of more than 15 h for heart rate to
also can increase cell proliferation including neurogenesis as return to baseline. In contrast, animals living in sibling pairs
measured by BrdU; these effects are most pronounced in the required about 2.5 h for their heart rate to recover. In the
dentate gyrus of the hippocampus (Ruscio et al., 2008). Inter- absence of a social partner, the neuropeptide hormone oxytocin
estingly, in male voles, those that attack pups have the highest increased in blood in female prairie voles (Grippo et al., 2007c)
level of neurogenesis and also have more OT-ir neurons and and centrally in male prairie voles (Sun et al., 2014). Elevated
fibers in and around the PVN. oxytocin may be protective against the negative consequences
Experiments on separation ‘distress’ have tested the capacity of isolation, which include reductions in the expression of the
of peptides to prevent behavioral changes during separation. oxytocin receptor (Pournajafi-Nazarloo et al., 2013) and
The intense calling behavior of isolated domestic chicks increases in both vasopressin and CRF (Sun et al., 2014). In
declines following various treatments including, injections of the context of personal safety, the release of oxytocin could
oxytocin/vasotocin, opioids that stimulate m receptors, and encourage social interactions including those associated with
prolactin (Panksepp et al., 1997). Opiate injections also detecting and responding to the emotions or experiences of
diminish distress vocalizations in guinea pigs (Herman and others. We also tested this hypothesis by treating isolated or
Panksepp, 1978), but separation cries of infant rats do not paired prairie voles with exogenous oxytocin. Oxytocin injec-
show the predicted decline following opiate treatments tions over a period of weeks were capable of reversing the
(Winslow and Insel, 1991). However, in rat pups, separation cardiac and behavioral effects of isolation, including protecting
cries are inhibited by central treatments with oxytocin or vaso- against the increases in heart rate and reductions in vagal tone
pressin (Insel and Winslow, 1991; Winslow and Insel, 1993). that typically accompany isolation (Grippo et al., 2009).
In squirrel monkeys, there is evidence that both vasopressin In postmenopausal women, increases in oxytocin also have
and oxytocin can reduce isolation calling, although the effects been associated with ‘gaps in social relationships’ (Taylor et al.,
are dependent on social status and high doses of the peptides 2006). Releasing oxytocin may be a component of a self-
were necessary to obtain behavioral effects (Winslow and Insel, regulatory process that helps mammals deal with isolation or
1991). other stressful experiences. Such responses also might facilitate
seeking out social engagement or relationships, functions that
could be especially adaptive in females who may be less able
1.04.7.1 Consequences of Isolation May Be Mediated
than males to live and maintain a family alone (Taylor et al.,
through Oxytocin
2000).
In the context of the shared physiology among social and Monogamous monkeys, such as titi monkeys (Mendoza
emotional behaviors, it is not surprising that social interactions and Mason, 1997; Mason and Mendoza, 1998), form social
and isolation have powerful physiological consequences. Indi- bonds that can be observed in the laboratory. In fact, in this
viduals with a perceived sense of social support are more likely species, pairs typically sit in physical contact with their long
to avoid or survive illness and have longer lives than otherwise tails braided together. In titi monkeys, increases in cortisol in
similar people who live alone, especially those who experience response to disturbances are attenuated by the presence of
a sense of loneliness (Cacioppo et al., 2006). a familiar companion. Titi monkeys also respond to the
Experiments in animals provide an opportunity to examine absence of adult partners. In contrast, there is little evidence
in more depth the physiological consequences of the absence that titi monkeys differentiate between familiar and unfamiliar
of a social partner. Highly social mammals, including prairie infants of comparable ages, and it is reported that this species
132 The Neurobiology of Social Affiliation and Pair Bonding

typically will select their adult mate over their infant. The pres- In addition, the afferent or sensory vagus provides a major
ence of an adult partner could have survival consequences in source of input to the brain from visceral organs, via the NTS.
this monogamous species, since maintenance of adult social At least some of this feedback via the autonomic nervous
bonds permits subsequent reproduction. system, and especially the visceral, unmyelinated vagus, is
Squirrel monkeys are small New World primates that experienced by the cortex as vague but powerful emotions.
usually live in unisex groups. Behavioral changes in squirrel Although difficult to cognitively control or interpret, visceral
monkeys do occur following the removal of companions, experiences modulate arousal and play a major role in state
although these responses may reflect general arousal or physi- regulation and social cognition (Porges, 2011), which in turn
ological adjustments to being alone, rather than the loss of are important to social behaviors such as attachment and pair
a particular companion. Although squirrel monkeys appear bonding.
highly social, physiological measures do not suggest selective Oxytocin appears to regulate both sympathetic and vagal
pair bonding or stress buffering by companions (reviewed branches of the autonomic nervous system. The central cells
Mendoza and Mason, 1997; Hennessy, 1997). of origin of the autonomic nervous system and autonomic inte-
grative centers, such as the PVN, synthesize and respond to
oxytocin. Exposure to exogenous oxytocin, especially in a social
1.04.7.2 Social Behavior, the Polyvagal Theory and Oxytocin
context, neurally coupled activity in the PVN to brain stem
The evolutionary origins of human behavior are especially nuclei implicated in the regulation of the autonomic nervous
informed by awareness of the ontogeny and phylogeny of system (Yee et al., 2016). In addition most, if not all, of the
the physical structures that are necessary for social communi- peripheral target organs of the autonomic nervous system
cation (Porges, 2011). Ancient gill arches and associated contain receptors for oxytocin. Behavioral, physiological and
muscles and nerves gave rise to human face, larynx and anatomic data from rodents and humans (Grewen and Light,
pharynx, and middle ear. The detachment from the skull of 2011) suggest that the ‘antistress’ effects of chronic oxytocin
the middle-ear bones occurred in the transition from reptiles may include downregulating the sympathetic nervous system,
to mammals, possibly serving to permit a higher level of while supporting the protective and behavioral functions of
acoustic social communication in mammals. Of particular the vagal systems. As one behavioral example, individuals
importance to social behavior was the evolved expansion of with higher levels of parasympathetic activity showed more
the human face and skull, formation of the detached rapid increases in self-described positive emotions and a sense
middle-ear bones, larynx, pharynx, and eventually the expan- of connectedness (Koh and Fredrickson, 2010).
sion of the neocortex. Together, these structures and their
neural control systems allowed the emergence of human
cognition, speech, and language. 1.04.8 Phylogenetic and Ontogenetic Factors
Components of this complex system are under voluntary, Influencing Pair Bonding
corticomotor control. However, these same structures may be
influenced by the autonomic nervous system. The social The expression of attachment behaviors varies widely among
engagement system, critical to social communication and species, and the mechanisms responsible for these behaviors
cognition, is innervated in part by the parasympathetic compo- also must have species-specific components (Sachser et al.,
nent of the autonomic nervous system. 1998). Peptide hormones, including oxytocin and vasopressin,
The cranial part of the parasympathetic nervous with species-typical patterns of peptide production, receptor
system consists of the vagus nerve, which exits the skull as distributions, and functions (Insel and Shapiro, 1992; Insel
the 10th cranial nerve. The vagus is functionally a coaxial cable et al., 1997; Wang, 1995; Wang et al., 1996, 1997; Witt et al.,
with two source nuclei, containing both motor and sensory 1991; Young et al., 1996, 1999), are particularly well posi-
fibers. The myelinated motor component of the vagus arises tioned to influence behaviors, such as pair bonding, that vary
in the ventral vagal complex (VVC) and supports the metabolic among different species (Carter et al., 1995, 1997). Both
demands of the upper part of the body, including the face and general patterns of oxytocin receptor expression and binding
brain. Actions of the VVC allow the reduction in fear and the and receptor responses to steroids differ among species (Insel
concurrent emergence of calm states necessary for social and Shapiro, 1992; Insel et al., 1994, 1997; Jiménez et al.,
engagement and bonding. Also originating in the VVC, the 2015; Tribollet et al., 1992; Witt, 1997; Young et al., 1996).
special visceral efferent (SVE) pathways regulate the striated For example, estrogen increases oxytocin binding within the
muscles of the face and head. The myelinated SVE system VMH in rats, but not in prairie voles (Witt et al., 1991). In
allows dynamic and rapid autonomic input in social interac- rabbits (polygamous, monoparental), ovariectomy increases
tions, including eye movements and auditory communication, oxytocin binding in the prefrontal cortex but has the opposite
which are necessary for mammalian social engagement. effect in the POA (Jiménez et al., 2015).
Primary features of emotion and affect (i.e., neural regulation Species differences in peptide receptor activity are presum-
of facial muscles and the heart) are regulated by this system. ably an important source of interspecific variation in the behav-
The more ancient unmyelinated vagus arises from the dorsal ioral effects of oxytocin and vasopressin including the capacity
vagal complex (including the DMX). The unmyelinated vagus to form social bonds. Species-typical variations in peptide
primarily serves visceral organs below the diaphragm. receptors are apparent in early development. For example,
However, some organs including the heart and immune system vasopressin receptor binding increased rapidly in the second
have dual or ‘polyvagal’ – myelinated and unmyelinated – week of life in the LS of nonmonogamous montane voles,
innervation. but not prairie voles (Wang et al., 1997). Insel, Young, and
The Neurobiology of Social Affiliation and Pair Bonding 133

their colleagues compared the genes for oxytocin receptors in these changes in turn may affect the subsequent tendency of
prairie voles and montane voles and found that these receptors a species or individual to develop social bonds.
were ‘virtually identical’ in genetic structure (Insel, 1997; Ontogenetic experiences, including levels of perinatal stress
Young et al., 1996, 1998). However, promoter elements can and varying amounts of parent–young interaction, can
regulate the expression of these receptor genes in particular contribute to the development of species-typical patterns of
tissues; subtle, but potentially important, species differences social behavior (Carter, 2003, 2007). As one example, reduc-
in base sequences may be responsible for the interspecific vari- tions in early handling during the first week of life, probably
ations in peptide receptor distributions. Based on rodent work, mediated by differential maternal stimulation, were associated
especially in voles, Wang et al. (1996) suggest that “neuroendo- with a reduced capacity later in life to form social bonds in
crine systems may evolve by changes in receptor distribution female prairie voles (Bales et al., 2007b). This form of differen-
rather than by restructuring the presynaptic pathway.” Compar- tial handling is also associated with lower levels of central
isons among related vole species with very different patterns of oxytocin (Bales et al., 2007a). Behavioral flexibility, such as
social behavior offer insights into the role of peptides and their that seen in prairie voles, and possibly mediated by peptide–
receptors in species-typical social behaviors (Insel, 1997; steroid interactions during development, allows animals to
Hammock and Young, 2005). individually adapt their social systems to accommodate early
Among the traits of monogamous mammals is a relative experiences and environmental demands.
absence of physical sexual dimorphism in appearance In the transition from reptiles to mammals, and especially
(Kleiman, 1977). It is well known that steroid hormones, to primates, high levels of encephalization and cognition
including testosterone and dihydrotestosterone, play an impor- appeared (Somel et al., 2013). Sophisticated autonomic
tant role in the development of masculine genitalia. Thus, the systems also emerged in mammals capable of supporting the
absence of physical sexual dimorphism in monogamous physiological demands of the expanding cortex for oxygen
species may reflect a comparative absence of, or insensitivity (Porges, 2011). The neuroendocrine systems that regulate
to sex steroids, especially in early life (Carter and Roberts, social interactions evolved in conjunction with the autonomic
1997). It is possible that social behaviors, including the nervous system include processes necessary for homeostasis.
tendency to form pair bonds, which characterize monogamous These in turn facilitate adaptation to ever-changing social and
mammals, also emerge in the relative absence of develop- physical environments (Dunbar, 2009).
mental exposure to high levels of androgens. For example, In humans the capacity to adapt to environmental demands
male parental care is a trait of some, but not all, pair bonding often relies on the neocortex and cognition, in ways that exceed
species. There is evidence from a variety of species that exposure that of other mammals. However, the neocortex rests physically
to androgens in early life and in some cases in adulthood can and functionally upon an ancient brain stem, relying for suste-
inhibit the tendency to be parental. nance on autonomic mechanisms (Porges, 2011). The neural
In the relative absence of high levels of androgens, some substrates for social behavior and emotion involve not only
species may rely more directly on neuropeptides, including the cortex, but also subcortical brain structures, below
vasopressin, to determine ‘masculine’ patterns of physiology conscious awareness. Voluntary and conscious aspects of social
and behavior – for example, selective aggression. Neonatal behavior and social engagement behaviors may be directly
exposure to vasopressin also can facilitate aggression especially influenced by the same neural systems, including the central
in male prairie voles (Stribley and Carter, 1999). Peptide treat- and autonomic nervous systems, which regulate survival and
ments either in development (Boer, 1993; Swaab and Boer, homeostasis. For example, neonatally decorticated rats appear
1994) or in adulthood (Poulain and Pittman, 1993) may alter remarkably normal and continue to engage in social contact
the sensitivity of the nervous system to subsequent hormonal (Carter et al., 1982). In contrast, lower brain stem lesions
experiences (reviewed in Carter, 2003, 2007). For example, in generally are fatal since they interrupt the autonomic supply
rats, treatment with vasopressin during the first week of life lines for oxygen and nutrients to the cortex.
reduced gene expression for the oxytocin receptor in the PVN
during adulthood (Ostrowski, 1998). Because vasopressin is
part of the HPA axis and is sensitive to androgens, this finding 1.04.9 Peptides and Human Behavior
suggests that developmental changes associated with perinatal
stress or sex-dependent androgenization could alter the subse- Two aspects of mammalian life, birth and lactation, are clearly
quent sensitivity of the oxytocinergic system. associated with the release of oxytocin. Among the neuroendo-
It is possible that the presence or absence of androgens at crine adaptations that accompany both birth and subsequent
various times in the life span plays a role in the development lactation are hormonal changes that may promote selective
of the capacity to form pair bonds. It is known that exposure social interactions, including high levels of physical contact
to steroids, including testosterone or corticosterone during between parent and offspring and maternal–infant bonding.
perinatal life, can alter the tendency of young prairie voles to Although, it is likely that the experiences associated with birth
prefer a sibling versus a novel stranger (Roberts et al., 1997). facilitate maternal bonding in humans as they do in other
In addition, even in prairie voles, there is a sex difference in animals, studies of the physiology of mother–infant interac-
the initial preference for male or female stimulus animals tions are at present primarily correlational.
(DeVries and Carter, 1999), which may reflect the develop- Birth is a hormonally complex event that is difficult to
mental effects of steroids in this species. During development, study. In addition, it is well known that strong social attach-
varying exposure to peptides and steroids may ‘retune’ the ments between adults and children can occur in the absence
nervous system, altering thresholds for sociality and aggression; of parturition. Thus, the hormonal events of birth are not
134 The Neurobiology of Social Affiliation and Pair Bonding

essential for social bonding. However, as described above, (SNPs) in the gene for the oxytocin receptor have been linked
research on bonding in sheep provides strong evidence for a to individual differences in human behavior (Jacob et al.,
role for vaginocervical stimulation and birth-related hormonal 2007) and in vagal functions relevant to social behavior
events (Kendrick, 2000). Recent studies in women reveal (Kanthak et al., 2016). The strength of these relationships varies
a stability of individual differences in plasma oxytocin; in among SNPs, among ethnic groups, and among individuals.
this study, oxytocin is positively correlated with several indices However, genetic patterns are emerging suggesting that some
of maternal interactions including gaze, positive affect, affec- individuals are highly susceptible to both peptides and social
tionate touch, and attachment-related thoughts (Feldman factors, while others are relatively insensitive to the impact of
et al., 2007). the social environment and perhaps also relatively insensitive
Lactation is a defining property of mammals and, until to the effects of peptides.
modern times, was necessary for mammalian reproduction.
Oxytocin plays an essential role in milk let-down, but not
maternal behavior (Nishimori et al., 1996; Russell and 1.04.10 Clinical Implications of a Peptidergic
Leng, 1998). However, lactating women interact more posi- Theory of Social Attachment
tively with their babies, directing more touching and smiling
toward their infants than do bottle-feeding mothers. It is also The presence or absence of attachments has broad conse-
reported that nursing mothers are more likely to describe quences across the life span. Like other mammals, humans
positive mood states and be less reactive to stressful experi- rely on positive social interactions for both safety and reproduc-
ences than bottle-feeding mothers (reviewed in Carter and tion. It has been argued that the tendency to form pair bonds or
Altemus, 1997). social attachments is a universal human characteristic (Money,
Oxytocin injections, given peripherally to the mother, can 1980; Fisher, 2016; Hazan and Shaver, 1987). Social support
facilitate nipple attachment by young rat pups, suggesting has documented health benefits, and the absence of positive
that oxytocin may change the response of a young animal to social interactions or social bonds typically is associated with
its mother (Singh and Hofer, 1978). Rat pups also show pref- both physical and mental illness (Ryff and Singer, 1998).
erences for specific odors that are associated with exposure to Forced social separations or the absence of social attach-
their mothers. Preferences for the mother do not develop in ments can trigger stress, anxiety, fear, and even depression
animals that are pretreated with oxytocin antagonists (Nelson (Sachser et al., 1998). The behaviors and physiological changes
and Panksepp, 1996). Thus, oxytocin may act on both the associated with bereavement or grief are similar to those used
mother and infant to influence the response of young animals, to define depression (Reite and Boccia, 1994). Understanding
including humans, to their mother. the nature of physiological processes that regulate social attach-
Interacting with an infant may release oxytocin in adult ment also could be of value in the treatment or prevention of
humans, including human males (Feldman, 2012), as well as disorders, such as autism, depression, or schizophrenia, which
in male prairie voles (Kenkel et al., 2012). Oxytocin has been can involve dysfunctional social attachment (Kirkpatrick, 1997;
shown to enhance the processing of social information and Henry and Wang, 1998).
in some cases to facilitate a sense of trust (Kosfeld et al., In primate infants the absence of adequate maternal care
2005) or empathy. Oxytocin may mediate the buffering effects and opportunities to form attachments has been associated
of social support and modulate reactivity to stressful experi- with growth retardation, social withdrawal, inadequate inter-
ences, playing a role in what humans experience as ‘love’ personal relationships, and inhibited verbal communication
(Carter, 1998; Carter and Porges, 2013). In general, oxytocin (Harlow, 1971). This complex of symptoms has even been
tends to support a sense of safety and social behaviors charac- recognized in human development as a medical syndrome,
terized by immobility without fear (Porges, 2011). termed ‘Reactive Attachment Disorder of Infancy.’ Although
Understanding the actions of peptides in humans will the concept of attachment disorder has begun to generate treat-
require greater knowledge of the capacity of the peptide to ment strategies, the relationship between this ‘disorder’ and
affect the peptide receptors. Individuals vary in the sensi- normal human attachment remains to be described.
tivity of their receptors to oxytocin and vasopressin. It has been proposed that autism, which can be character-
Research on pair bonding species, such as prairie voles, ized by atypical social behavior and a failure to form social
suggest that these differences are likely the result of selec- attachments, may involve abnormal activity in endogenous
tion pressures acting on receptors, including those for peptidergic systems. For example, a variety of clinical studies
oxytocin and vasopressin. Receptor variation, in turn, may implicate opioids in autism (Bouvard et al., 1995). Treatment
permit adaptive individual or species differences in the with naltrexone produces some clinical benefits and alters
sensitivity to a given peptide, and hence in the capacity to biochemical profiles in a subset of autistic children. Studies
exhibit pair bonding or other characteristics of social also have begun to explore the role of oxytocin in autism (Insel,
monogamy (Okhovat et al., 2015). 1997). In autistic children, deficits in oxytocin (Modahl et al.,
The effects of oxytocin on behaviors such as pair bonding 1998; Guastella and Hickie, 2016), genetic variation in (Jacob
are due to actions on both the oxytocin and the related vaso- et al., 2007) or epigenetic silencing of the oxytocin receptor
pressin receptor (Cho et al., 1999; Albers, 2015). The relative (Gregory et al., 2009), have been correlated with some symp-
importance of these across species, between males and females, toms of autism. However, the possible role for oxytocin, vaso-
and among individuals is increasing being examined in a search pressin, endogenous opioids (or receptors for these) in
for the source of differences in the capacity for sociality and pair selective human social attachments, more analogous to pair
bonding. For example, single nucleotide polymorphisms bonding, remains less well understood.
The Neurobiology of Social Affiliation and Pair Bonding 135

Other neurochemicals, such as catecholamines and sero- examples of experiences that have been shown to influence
tonin, have effects on behavior and influence the release and the endogenous production, release, or actions of these
actions of oxytocin and vasopressin. For example, selective peptides.
serotonin reuptake inhibitors can influence peptidergic systems Many components of human behavior, such as motor
(Li et al., 1993; Uvnas-Moberg et al., 1999). The clinical effects patterns, are regulated at least in part, by cognitive processes.
of these chemicals in the context of attachment are largely However, the decision to engage in a given behavior and
unknown, although drugs that affect these systems could influ- various mood states are strongly determined by visceral
ence social bonding. processes or autonomic states, which may in turn motivate
Forced isolation, anxiety, fear, and other forms of stressful the occurrence of specific behaviors (Porges, 2011). Peptides,
conditions are associated with increased levels of HPA activity. including oxytocin and vasopressin, regulate these visceral
Such conditions or experiences normally tend to encourage states or emotional feelings. Steroids, opioids, oxytocin, and/
social interactions. Both the human and animal literature or vasopressin may induce a physiological process or ‘social
(Carter, 1998) suggest an association between HPA activation motivation’ that increases the probability of social interactions
and stressful experiences and the development of social attach- and the formation of social bonds. In addition, oxytocinergic
ments. The role of hormones of the HPA axis in attachment is processes, possibly due to their unique central position in the
probably not linear, because both animals and humans under nervous system, may help to modulate autonomic and behav-
extreme conditions may become either self-protective or ioral reactivity. Thus, by understanding the neurobiology of
immobilized – conditions which are at odds with the forma- peptides, such as oxytocin and vasopressin, we may gain
tion of social bonds (Porges, 1998, 2011). Excessively stressful a better understanding of the processes through which social
conditions, such as those that compromise survival or intense bonds promote physical and emotional health.
grief, may lead to a breakdown of social relationships (Reite Hormones, including oxytocin and vasopressin, are directly
and Boccia, 1994). Thus, chronic or extreme stress could inhibit and indirectly manipulated by various medical practices. For
subsequent attachment. However, research with rodents example, large doses of ‘Pitocin,’ a synthetic version of
suggests that within a homeostatic range, stress-related physio- oxytocin, are routinely used to hasten childbirth, with unex-
logical processes, including hormones of the HPA axis, can plored effects on the social behavior or propensity to attach-
promote the subsequent development of social bonds, espe- ment of both the mother and child (Boer, 1993). Long
cially in males (DeVries et al., 1996). In addition, positive labors, caesarian sections, and the decision to breast or bottle
social interactions, including social bonds, may help to create feed are indirectly peptidergic manipulations (Kenkel et al.,
physiological states that are anxiolytic or stress reducing. 2014). Only recently has attention begun to be directed to
Oxytocin, perhaps released by positive social interactions, the behavioral or hormonal consequences of these peptide-
has the capacity to produce both acute and chronic reductions related events that can have profound effects on behavioral,
in the activity of the HPA axis (Carter, 1998; Petersson et al., homeostatic, and emotional systems for both the parent and
1999). Studies of lactating women support this hypothesis in the child.
humans (Altemus et al., 1995). Thus, oxytocin, with both
central and peripheral processes, is part of an endogenous
homeostatic system. This system has the concurrent capacity 1.04.11 Attachment and Bonding in Primates: An
to increase social attachment and other positive social behav- Evolutionary Perspective for Humans
iors, providing the additional indirect benefits of sociality.
The role of vasopressin in human social behavior is more Human infants form attachment bonds with their caregiver
difficult to characterize. Vasopressin is believed to be similar who is usually the mother, for it is she who provides the secure
to the ancestral peptide from which oxytocin and vasopressin base for fostering exploration, play, and other social behaviors
were derived. Vasopressin binds to several types of receptors, (Klaus et al., 1995). The nature of this attachment figure
including the oxytocin receptor. Vasopressin has been impli- becomes more visible under conditions that are fearful or novel
cated in territoriality, agonistic behaviors, and HPA and threatening to the infant. By seeking protection through
arousal and may be part of a more primitive adaptive system attachment, the immature offspring are believed to increase
for mobilization and self-defense (Carter, 1998; Porges, 1998, their likelihood of survival. Infant attachment is not indepen-
2007; 2011). In some cases the functions of vasopressin are dent of caregiving and therefore it is important that mothers
apparently similar to those of oxytocin. However, under other should find ‘satisfaction with the parental role,’ and across
conditions, oxytocin and vasopressin may have antagonistic many cultures this appears to be the case. To the extent that
actions (Engelmann et al., 1996). Dynamic and complex inter- such ‘satisfaction’ may help to maintain investment in rearing
actions between oxytocin and vasopressin (Albers, 2015), offspring to reproductive age, maternal caregiving behavior
working in the presence of a more slowly changing steroid must have evolved with infant attachment behavior. Also
background, could help to regulate human visceral states and common to all cultures are the changes in hormonal status
emotions. during pregnancy and parturition that predispose women to
Awareness of the importance of peptides, including oxytocin respond to an infant’s signals.
and vasopressin, in human behavior is comparatively recent, The quality of mothering, particularly in times of fear and
but of considerable importance. Many aspects of daily life can stress, contributes substantially to the infant’s security of
affect the release of the peptides. Social or sexual contact, food attachment. Bowlby (1969) argued that infant attachment
intake (Uvnas-Moberg, 1994), the use of steroid hormones, or and maternal bonding provided the foundation for human
drugs of abuse or alcohol (Kovacs et al., 1998) are only a few attachment behavior from the cradle to the grave. Although
136 The Neurobiology of Social Affiliation and Pair Bonding

the ‘internalized’ bond is secured by the various forms of behavior from endocrine determinants has only been evolu-
attachment behavior that contribute to it, touching and tionarily possible with the development of a bigger brain,
huddling are features of attachment behavior that persist into because decision-making processes, especially in the context
adult life, particularly in times of distress, fear, and illness. of maternal behavior, are complex and need to be strategically
A question of some importance is how this relationship, correct. Progression away from the synchronization of maternal
engendered by maternal bonding and reciprocated by infant behavior with the hormones of pregnancy and replacement
attachment, and which extends into adult relationships and with a system of cognitive control require exceptional cognitive
bonds, is subserved at the neural level. If, as Bowlby suggests, abilities. These abilities are not inherited, but a larger brain
all meaningful relationships are structured on the foundations enables social factors to take over from hormonal factors in pre-
of early attachment, are there common underlying mecha- dicting reproductive success. This would explain the overriding
nisms, and if so how have they evolved? The biology of importance of social and maternal experience in order to
mammalian bonding, as exemplified by monogamous prairie achieve successful maternal care, experience that is acquired
voles and sheep with their lambs, focuses heavily on the impor- during early social development under the watchful eye of
tance of peripheral steroid hormones (estradiol, progesterone, mothers, but outside the immediate context of pregnancy
testosterone, and corticosterone) and their effects on central and parturition (Kim et al., 2016).
release of peptides including vasopressin, oxytocin, CRF, and From the available fossil records, it appears that many
opioids, and the synthesis of receptors for these peptides. A mammalian lineages have evolved increased cranial capacity,
primary site of action for these peptides, although not exclu- but because it is claimed that the push for an exceptionally
sive, may be on olfactory recognition that is important in the larger neocortex in primates has developed from complex
bonding process. It is likely that attachments or bonding in social living, then differences in maternal and paternal lifestyles
humans are more complex and probably more dependent on may have subjected brain evolution to differential selection
cognitive processes and mental representations rather than pressures. In Old World monkeys, females provide social
the olfactory processes that occur in rodents or sheep (Kim stability and group cohesion, are more affiliative than males,
et al., 2016). However, some insight regarding the processes and maintain the continuity of the group over successive gener-
involved in social bonding in humans may come from studies ations. Females are the primary caregivers with social rank of
in nonhuman primates. daughter, but not sons, being related to the matriline. This
Many of the basic features in human infant behavior that kind of matrilineal inheritance is compatible with genomic
helped Bowlby formulate his attachment theory can be imprinting, which not only results in the development of
observed in the mother directed behavior of infant monkeys. a larger forebrain from maternally expressed alleles (Keverne
This sharing of behavioral propensities and emotional labilities et al., 1996a), but also the advantages of genomically
of monkey and human infants (sucking, clinging, crying, and imprinted inheritance are transmitted to both sons and daugh-
following) is consistent with their close evolutionary histories. ters. Hence, the differential selection pressures operating
The first detailed studies of attachment relationships were through the matriline could have resulted in a larger neocortex
undertaken in the 1960s on rhesus monkeys and provided giving greater cognitive control over behavior (Keverne et al.,
descriptions of infant behavioral development that generalize 1996b). For such neocortical control over behavior to be
to many primate species (Harlow, 1971; Kraemer, 1992). successful, the process of attachment and early learning is
Primate mothers provide their infants with nourishment, essential for the subsequent development of normal affiliative
protection, and warmth; protection from predators; and relationships.
a secure base to which they can retreat when disruptions occur The development of a larger brain has been particularly
in the social group. The attachment that a rhesus monkey infant important to enable the lifetime attachments such as those
develops with its mother is especially enduring in females, that occur in humans. For the operation of ‘attachments’ to
lasting a lifetime, while in infant males, it rarely lasts beyond sustain relationships, and endure a human lifetime, a brain
puberty. that can develop growing insights into self as well as the
maternal attachment figure is clearly an advantage. As young
children develop, their knowledge base expands especially in
1.04.11.1 The Evolutionary Basis of Bonding
regard to the changing ways in which their mother responds
What are the evolutionary developments that have enabled to them and, more importantly, how each is likely to respond
expansion of the affectional system deployed in mother–infant to the other. This knowledge steadily becomes organized in the
bonding to incorporate other contexts of social/affiliative rela- form of internal working models of self and mother, encom-
tionships? In considering primates, strong interrelations are passing an understanding of both her moods and intentions.
beginning to emerge between the two important features noted Building on this early knowledge provides the infant with an
earlier, neocortical expansion, and matrilineal inheritance. ability to simulate happenings in an expanding world of rela-
These are common to all mammals that exhibit complex social tionships. Forward planning may occur with the advantages
organizations. The development of a larger neocortex has of foresight and security. Because these working models are
enabled motivated behavior to occur at will, such that maternal in constant daily use, their influence on thought, feeling, and
affiliation may take place without pregnancy and parturition behavior becomes routine and all pervasive.
(Keverne, 2013; Carter, 2014). This unique development in Deploying our brains in the construction of internalized
primate evolution has matched parturient females with non- working models of self and others would be difficult, if not
parturient females in sustaining the behavioral potential for impossible, without an attachment figure, and we would
infant caregiving (Hrdy, 2009). Such an emancipation of further argue, that attachment figures would be extremely
The Neurobiology of Social Affiliation and Pair Bonding 137

scarce without maternal bonding. Those parts of the brain that may help to explain the exceptionally long period of time
are characteristically enlarged in humans (neocortex and stria- necessary to produce a fully developed human nervous system.
tum) develop in the postnatal period (neotany) when infants The slow maturation of the nervous system also increases the
are forming their internal working models. Important in this developmental significance of social interactions and long-
context is the availability of exclusive access to mother and term attachments.
her undivided attention. Biology has assisted this possibility It has been shown that apoptosis can be inhibited by
by extending the interbirth interval, aided by the contraceptive oxytocin, and especially by the ‘fetal,’ extended form of oxytocin
effects of infant suckling. There can surely be no better way of (Jankowski et al., 2016). Oxytocin also has been shown to
increasing maternal investment than by reducing the number reduce apoptosis in adult animals and to promote adult neuro-
of infants in which the mother invests. genesis (Leuner et al., 2012). High levels of oxytocin might
facilitate neocortical growth, by inhibiting the programmed
destruction of brain cells, thus sculpting the cortex, and
1.04.11.2 Genomic Imprinting, Social Behavior, and Cortical
synchronizing neocortical development to the physical
Growth
demands of mammalian reproduction (Carter, 2014).
Oxytocin may have both direct and indirect effects on neocor- It is also important to note that in these early years, when
tical development. Live birth puts restrictions on the physical the neocortex is forming and making its connections and asso-
size of an infant and especially the head. A large baby with ciations with other parts of the brain, the limbic emotional
an expanded neocortex is a physical burden for the mother. brain is well developed. The large emotional repertoire of
In primates, infants are gestated, nursed, and carried for young infants bears testimony to this. Understanding
months or years. Reproductive restrictions are further increased emotions, curbing these emotions, and channeling these
in bipedal primates, since mothers must give birth through emotions for beneficial purposes must represent an important
a pelvic girdle adapted for upright locomotion. Thus, the phase in brain development. These cognitive abilities have
capacity of the mother to regulate offspring development and surely prospered from experiencing the dynamics of reciprocity
especially the size of the neocortex may be critical to both in the interplay between mother and child, while the subse-
her survival and reproductive success (Keverne, 2013). At the quent expansion of social relationships, based on secure attach-
same time the father’s genome may be better served by larger ment, provides a second cushion for the emotional turmoil of
offspring. puberty. The child’s need for an attachment figure and the
Through a process known as genomic imprinting, the mother’s predisposition to bond provides an optimal social
expression of a subset of genes, of particularly relevant impor- environment in which the human brain can develop. Weaning
tance to growth and development, can be determined by one and expansion of the child’s social world have not only coin-
parent versus the other. This is accomplished by selective cided, but are made easier by having a secure base from which
silencing of one of a pair of alleles for a given gene, allowing to explore.
the other allele to dominate. In theory the mother seeks to
produce a smaller infant with a smaller brain. Research in
1.04.11.3 Oxytocin and Love
mice (Keverne, 2013) showed that the matrilinear germ line
contains cells that will become the neocortex, while progenitor Among the most interesting behaviors and emotional states in
cells that will become the hypothalamus originate in the which oxytocin has been implicated are those that humans call
father’s genome. The primary source of oxytocin is from the ‘love’ (Carter, 1998; Carter and Porges, 2013; Fisher, 2016). Key
hypothalamus, in neurons that are regulated by paternally to loving relationships are selective social behaviors and recip-
expressed genes that are susceptible to genomic imprinting. rocal social bonds. Studies originally conducted in socially
One paternally expressed gene, known as Peg3, plays a critical monogamous prairie voles revealed that oxytocin was capable
role in the development of the hypothalamus, as well as the of facilitating social contact, as well as selective social prefer-
placenta (Broad et al., 2009; Champagne et al., 2009). Evidence ences in both sexes. Research in prairie voles showed that
for the importance of Peg3 comes from experiments in which mating facilitated the onset of pair bonding (Williams et al.,
the Peg3 gene was inactivated. In the absence of Peg3 expres- 1992), a behavior that was later shown to be dependent on
sion, females had a reduced number of hypothalamic oxytocin oxytocin (Williams et al., 1994). In this context, it is important
neuron, lower reproductive success, and specific reductions in to note that in the prairie vole model, access to both oxytocin
the growth of the offspring that survive. Peg3 mutant mothers and vasopressin receptors is necessary for pair bonding to
also were less attentive to their young, showing increased indi- emerge, while either oxytocin or vasopressin alone can facilitate
cations of anxiety and aggression, all suggestive of reductions in nonselective sociality (Cho et al., 1999). However, as explained
the functions of oxytocin. Genomic imprinting or silencing of here, it is likely that the quality of the biological experiences
the paternal allele of the Peg3 gene allows the maternal that are influenced by oxytocin and vasopressin differ (Carter,
genome to dominate the development of her fetus. 1998).
Thus, although the genes that produce oxytocin may origi- Research in animals, including humans, has confirmed and
nate in the father, it is the mother who determines the expres- extended many of the general conclusions drawn from that
sion of these genes, and thus the size of the infants. By research (Carter, 1998), leaving no doubt that oxytocin plays
genomically silencing genes from the father and allowing her a central role in the social behavior that lie at the heart of the
own to dominate, the mother’s genome can regulate the growth human experience of love. Oxytocin is released in response
of the brain and body of her offspring (Keverne, 2013). The to a variety of circumstances, both positive and negative
capacity of the mother to regulate cortical development also (Carter, 1992; Feldman, 2012; Dai et al., 2012). However,
138 The Neurobiology of Social Affiliation and Pair Bonding

whether human social behavior and attachments can be also serve to coordinate the autonomic and endocrine
formed in the absence of oxytocin is not known. consequences of positive social experiences with behavioral
states that support the formation and maintenance of social
bonds.
1.04.11.4 Oxytocin and Development
Unique actions of social behavior and of oxytocin may
The importance of oxytocin is especially appreciated in the have been necessary for encephalization, by facilitating birth,
context of the development of the human nervous system. lactation, maternal behavior, genetic regulation of the
The human brain is two to three times larger than that of other growth of the neocortex, and the maintenance of the blood
related primates (Somel et al., 2013). This difference is due supply to the cortex (Keverne, 2013; Carter, 2014).
primarily to increases in cortical tissue, especially neurons, Peptide-facilitated attachment also allows the extended
located in association areas such as the prefrontal cortex. The periods of nurture necessary for the emergence and optimiza-
elaboration of the neocortex was critical to the evolution of tion of human intellectual development. In general, oxytocin
novel features of human behavior, including cognition and acts to encourage a sense of safety, and the high levels of
speech. The origins of the neocortex are traced to the reptilian social sensitivity and attunement necessary for human soci-
ancestor of early mammals. These reptiles are believed to ality and for rearing a human child. Oxytocin dynamically
have had a thin sheet of cortical cells, which by the time of moderates the autonomic nervous system, and effects of
the evolution of the earliest mammals has been estimated to oxytocin on vagal pathways, as well as the antioxidant and
be only about six cell layers in thickness (Rakic, 2009). During anti-inflammatory effects of this peptide, help to explain
development, the cells of the neocortex differentiate, migrate, the pervasive adaptive consequences of social behavior for
enlarge, and in some cases undergo cell death (including emotional and physical health.
apoptosis). Initially subtle developmental events may have
allowed the evolution of the human neocortex. For example,
it has been estimated by Rakic (2009) that “fewer than 7 extra Acknowledgments
rounds of cell division in the progenitor cells at an early embry-
onic stage would be sufficient to create the 1000 fold difference We gratefully acknowledge many scientists, including our collaborators,
in total cortical surface area that differentiates the brains of whose work has contributed to the understanding of this subject.
mice from those of humans.” Studies on maternal bonding in sheep and primate behavior were
In rats, maternal oxytocin can act as a signaling mechanism sponsored by grants to EBK from the BBSRC and the Medical Research
between the mother and fetus (Kenkel, 2014). Of particular Council of the United Kingdom. Studies on pair bonding in prairie
importance to cortical and hippocampal functioning is voles were sponsored by grants to CSC from the United States National
GABA. Maternal oxytocin released during birth triggers a switch Institutes of Health (NICHD and NIMH) and National Science Foun-
dation, and National Alliance for Autism Research. Studies of human
in GABA signaling in the fetal brain from excitatory to inhibi-
breast feeding were sponsored by a grant from the United States
tory. In vivo administration of an oxytocin antagonist before
Department of Defense. We are particularly grateful for conceptual
delivery prevented this switch of GABA activity in fetal neurons guidance regarding the role of the autonomic nervous system in social
and aggravated the severity of hypoxic episodes. Maternal behavior from Stephen Porges.
oxytocin apparently inhibits fetal neurons and concurrently
increases their resistance to hypoxia, which can serve to protect
cortical tissue during birth. The birth-related surge in oxytocin
References
also helps to regulate the synchronization of the fetal hippo-
campal neurons, possibly allowing the transition from prenatal Acher, R., Chauvet, J., Chauvet, M.-T., Michel, G., 1997. Principles in protein
to postnatal life (Tyzio et al., 2006; Mazzuca et al., 2011). Such hormone evolution: the neurohypophysial peptides as avian evolutionary tracers.
changes have long-term consequences for both emotional and Poult. Avian Biol. Rev. 8, 33–51.
cognitive functions, including the capacity to exhibit positive Albers, H.E., 2015. Species, sex and individual differences in the vasotocin/vaso-
pressin system: relationship to neurochemical signaling in the social behavior
social behaviors and form social bonds.
neural network. Front. Neuroendocrinol. 36, 49–71.
Altemus, M., Deuster, P.A., Gallivan, E., Carter, C.S., Gold, P.W., 1995. Suppression
of hypothalamic-pituitary-adrenal responses to exercise stress in lactating women.
1.04.12 Summary J. Clin. Endocrinol. Metab. 80, 2954–2959.
Amico, J.A., Mantella, R.C., Vollmer, R.R., Li, X., 2004. Anxiety and stress responses
in female oxytocin deficient mice. J. Neuroendocrinol. 16, 319–324.
Pair bonding is an evolved trait and may play a critical role Aragona, B.J., Wang, Z., 2009. Dopamine regulation of social choice in a monoga-
in reproduction, as well as in individual and species mous rodent species. Front. Behav. Neurosci. 3, 15.
survival. The neurobiology of pair bonding or other forms Aragona, B.J., Liu, Y., Curtis, J.T., Stephan, F.K., Wang, Z., 2003. A critical role for
of social affiliation are most readily understood in this nucleus accumbens dopamine in partner-preference formation in male prairie
voles. J. Neurosci. 23, 3483–3490.
context. The proximate mechanisms underlying various
Aragona, B.J., Liu, Y., Yu, Y.J., Curtis, J.T., et al., 2006. Nucleus accumbens
forms of positive social behaviors, including pair bonding dopamine differentially mediates the formation and maintenance of monogamous
and maternal–infant behavior, rely on common neural and pair bonds. Nat. Neurosci. 9, 33–39.
endocrine systems. At the heart of pair bonding are neural Bales, K.L., Lewis-Reese, A.D., Pfeifer, L.A., Kramer, K.M., Carter, C.S., 2007a. Early
systems dependent on peptides, including oxytocin, vaso- experience affects the traits of monogamy in a sexually dimorphic manner. Dev.
Psychobiol. 49, 335–342.
pressin, opioids, CRF, and related hormones. Steroid Bales, K.L., Plotsky, P.M., Young, L.J., et al., 2007b. Neonatal oxytocin manipulations
hormones, although not directly essential for pair bonding, have long-lasting, sexually dimorphic effects on vasopressin receptors. Neurosci-
may facilitate and modulate these behaviors. Neuropeptides ence 144, 38–45.
The Neurobiology of Social Affiliation and Pair Bonding 139

Barberis, C., Tribollet, E., 1996. Vasopressin and oxytocin receptors in the central Carter, C.S., Witt, D.M., Kolb, B., Whishaw, I.Q., 1982. Neonatal decortication and
nervous system. Crit. Rev. Neurobiol. 10, 119–154. adult female sexual behavior. Physiol. Behav. 29, 763–766.
Bielsky, I.F., Young, L.J., 2004. Oxytocin, vasopressin, and social recognition in Chacko, R.C., Corbin, M.A., Harper, R.B., 2000. Acquired obsessive-compulsive
mammals. Peptides 25, 1565–1574. disorder associated with basal ganglia lesions. J. Neuropsychol. Clin. N. Am.
Bielsky, I.F., Hu, S.B., Ren, X., Terwilliger, E.F., Young, L.J., 2005. The V1a vaso- 12, 269–272.
pressin receptor is necessary and sufficient for normal social recognition: a gene Champagne, F.A., Chretien, P., Stevenson, C.W., Zhang, T.Y., Gratton, A.,
replacement study. Neuron 47, 503–513. Meaney, M.J., 2004. Variations in nucleus accumbens dopamine associated with
Boer, G.J., 1993. Chronic oxytocin treatment during late gestation and lactation individual differences in maternal behaviour in the rat. J. Neurosci. 24,
impairs development of rat offspring. Neurotoxicol. Teratol. 15, 383–389. 4113–4123.
Bouvard, M.P., Leboyer, M., Launay, J.-M., et al., 1995. Low-dose naltrexone effects Champagne, F.A., Curley, J.P., Swaney, W.T., Hasen, N.S., Keverne, E.B., 2009.
on plasma chemistries and clinical symptoms in autism: a double-blind, placebo- Paternal influence on female behavior: the role of Peg3 in exploration, olfaction and
controlled study. Psychiatry Res. 58, 191–201. neuroendocrine regulation of maternal behavior of female mice. Behav. Neurosci.
Bosch, O.J., Neumann, I.D., 2012. Both oxytocin and vasopressin are mediators of 123, 469–480.
maternal care and aggression in rodents: from central release to sites of action. Chiodera, P., Salvarani, C., Bacchi-Modena, et al., 1991. Relationship between plasma
Horm. Behav. 61, 293–303. profiles of oxytocin and adrenocorticotropic hormone during suckling or breast
Bosch, O.J., Dabrowska, J., Modi, M.E., et al., 2016. Oxytocin in the nucleus stimulation in women. Horm. Res. 35, 119–123.
accumbens shell reverses CRFR2-evoked passive stress-coping after partner loss Chirino, R., González-Mariscal, G., 2015. Changes in responsiveness to kit odors
in monogamous male prairie voles. Psychoneuroendocrinology 64, 66–78. across pregnancy: relevance for the onset of maternal behavior. World Rabbit Sci.
Bowlby, J., 1969. Attachment and loss. In: Attachment, vol. 1. Hogarth Press, 23, 103–109.
London. Cho, M.M., DeVries, A.C., Williams, J.R., Carter, C.S., 1999. The effects of oxytocin
Bowler, C.M., Cushing, B.S., Carter, C.S., 2002. Social factors regulate female-female and vasopressin on partner preferences in male and female prairie voles (Microtus
aggression and affiliation in prairie voles. Physiol. Behav. 76, 559–566. ochrogaster). Behav. Neurosci. 5, 1071–1080.
Broad, K.D., Kendrick, K.M., Sirinathsinghji, D.J.S., Keverne, E.B., 1993. Changes in Creutz, L.M., Kritzer, M.F., 2004. Mesostriatal and mesolimbic projections of midbrain
oxytocin immunoreactivity and messenger-RNA expression in the sheep brain neurons immunoreactive for estrogen receptor beta or androgen receptors in rats.
during pregnancy, parturition and lactation and in response to estrogen and J. Comp. Neurol. 476, 348–362.
progesterone. J. Neuroendocrinol. 5, 435–444. Da Costa, A., Guevara-Guzman, R., Ohkura, S., Goode, J., Kendrick, K.M., 1996. The
Broad, K.D., Hinton, M.R., Keverne, E.B., Kendrick, K.M., 2002. Involvement of the role of the paraventricular nucleus in the control of maternal behaviour in sheep.
medial prefrontal cortex in mediating behavioural responses to odour cues rather J. Neuroendocrinol. 8, 163–177.
than olfactory recognition memory. Neuroscience 114, 715–729. Dai, L., Carter, C.S., Ying, J., Bellugi, U., Pournajafi-Nazarloo, H., Korenberg, J.R.,
Broad, K.D., Curley, J.P., Keverne, E.B., 2009. Increased apoptosis during brain 2012. Oxytocin and vasopressin are dysregulated in Williams Syndrome, a genetic
development underlies the adult behavioral deficits seen in mice lacking a func- disorder affecting social behavior. PLoS One 7 (6), e38513.
tional paternally expressed gene 3 (Peg3). Dev. Neurobiol. 69, 314–325. DeVries, A.C., Carter, C.S., 1999. Sex differences in temporal parameters of
Brunton, P.J., Sabatier, N., Leng, G., Russell, J.A., 2006. Suppressed oxytocin neuron partner preference in prairie voles (Microtus ochrogaster). Can. J. Zool. 77,
responses to immune challenge in late pregnant rats: a role for endogenous 885–889.
opioids. Eur. J. Neurosci. 23, 1241–1247. DeVries, A.C., DeVries, M.B., Taymans, S.E., Carter, C.S., 1995. The modulation of
Brunton, P.J., Russell, J.A., 2010. Endocrine induced changes in brain function during pair bonding by corticosterone in female prairie voles (Microtus ochrogaster). Proc.
pregnancy. Brain Res. 1364, 198–215. Natl. Acad. Sci. U.S.A. 92, 7744–7748.
Burkett, J.P., Spiegel, L.L., Inoue, K., Murphy, A.Z., Young, L.J., 2011. Acti- DeVries, A.C., DeVries, M.B., Taymans, S.E., Carter, C.S., 1996. The effects of stress
vation of m-opioid receptors in the dorsal striatum is necessary for adult on social preferences are sexually dimorphic in prairie voles. Proc. Natl. Acad. Sci.
social attachment in monogamous prairie voles. Neuropsychopharmacology U.S.A. 93, 11980–11984.
36, 2200–2210. DeVries, A.C., Guptaa, T., Cardillo, S., Cho, M., Carter, C.S., 2002. Corticotropin-
Cacioppo, J.T., Hughes, M.E., Waite, L.J., Hawkley, L.C., Thisted, R.A., 2006. releasing factor induced social preferences in male prairie voles. Psychoneur-
Loneliness as a specific risk factor for depressive symptoms: cross-sectional and oendocrinology 27, 705–714.
longitudinal analysis. Psychol. Aging 21, 140–151. DeVries, A.C., Johnson, C.L., Carter, C.S., 1997. Familiarity and gender influence
Carmichael, M.S., Humbert, R., Dixen, J., Parmisano, G., Greenleaf, W., Davidson, J., social preferences in prairie voles (Microtus ochrogaster). Can. J. Zool. 75,
1987. Plasma oxytocin increases in the human sexual response. J. Clin. Endo- 295–301.
crinol. Metab. 64, 27–31. De Vries, G.J., Panzica, G.C., 2006. Sexual differentiation of central vasopressin and
Carter, C.S., 1992. Oxytocin and sexual behavior. Neurosci. Biobehav. Rev. 16, vasotocin systems in vertebrates: different mechanisms, similar endpoints.
131–144. Neuroscience 138, 947–955.
Carter, C.S., 1998. Neuroendocrine perspectives on social attachment and love. Dharmadhikari, A., Lee, Y.S., Roberts, R.L., Carter, C.S., 1997. Exploratory behavior
Psychoneuroendocrinology 23, 779–818. correlates with social organization and is responsive to peptide injections in prairie
Carter, C.S., 2003. Developmental consequences of oxytocin. Physiol. Behav. 79, voles. Proc. Natl. Acad. Sci. U.S.A. 807, 610–612.
383–397. Diergaarde, L., Gerrits, M.A., Brouwers, J.P., van Ree, J.M., 2005. Early
Carter, C.S., 2007. Sex differences in oxytocin and vasopressin: implications for autism amygdala damage disrupts performance on medial prefrontal cortex-related
spectrum disorders? Behav. Brain Res. 176, 170–186. tasks but spares spatial learning and memory in the rat. Neuroscience
Carter, C.S., 2014. Oxytocin pathways and the evolution of human behavior. Annu. 130, 581–590.
Rev. Psychol. 65, 17–39. Dunbar, R.I.M., 2009. The social brain hypothesis and its implication for social
Carter, C.S., Altemus, M., 1997. Integrative functions of lactational hormones in social evolution. Ann. Hum. Biol. 36, 562–572.
behavior and stress management. Ann. N.Y. Acad. Sci. 807, 164–174. Engelmann, M., Wotjak, C.T., Neumann, I., Ludwig, M., Landgraf, R., 1996. Behavioral
Carter, C.S., Boone, E.M., Pournajafi-Nazarloo, H., Bales, K.L., 2009. The conse- consequences of intracerebral vasopressin and oxytocin: focus on learning and
quences of early experiences and exposure to oxytocin and vasopressin are memory. Neurosci. Biobehav. Rev. 20, 341–358.
sexually-dimorphic. Dev. Neurosci. 31, 332–341. Fahrbach, S.E., Morrell, J.I., Pfaff, D.W., 1985. Possible role for endogenous oxytocin
Carter, C.S., DeVries, A.C., Getz, L.L., 1995. Physiological substrates of mammalian in estrogen-facilitated maternal behavior in rats. Neuroendocrinology 40,
monogamy: the prairie vole model. Neurosci. Biobehav. Rev. 19, 303–314. 526–532.
Carter, C.S., Getz, L.L., 1993. Monogamy and the prairie vole. Sci. Am. 268, Febo, M., Numan, M., Ferris, C.F., 2005. Functional magnetic resonance imaging
1000–1106. shows oxytocin activates brain regions associated with mother-pup bonding during
Carter, C.S., Lederhendler II, , Kirkpatrick, B. (Eds.), 1997. The Integrative Neurobi- suckling. J. Neurosci. 25, 11637–11644.
ology of Affiliation. Annals of the New York Academy of Sciences, vol. 807. Feldman, R., 2012. Oxytocin and social affiliation in humans. Horm. Behav. 61,
Carter, C.S., Porges, S.W., 2013. The biochemistry of love: an oxytocin hypothesis. 380–391.
EMBO Rep. 14, 12–16. Feldman, R., Weller, A., Zagoory-Sharon, O., Levine, A., 2007. Evidence for
Carter, C.S., Roberts, R.L., 1997. The psychobiological basis of cooperative breeding. a neuroendocrinological foundation of human affiliation: plasma oxytocin levels
In: Solomon, N.G., French, J.A. (Eds.), Cooperative Breeding in Mammals. Cam- across pregnancy and the postpartum period predict mother-infant bonding.
bridge Press, New York, pp. 231–266. Psychol. Sci. 18, 965–970.
140 The Neurobiology of Social Affiliation and Pair Bonding

Ferguson, J.N., Young, L.J., Hearn, E.F., Insel, T.R., Winslow, J.T., 2000. Social Grippo, A.J., Trahanas, D.M., Zimmerman II, R.R., Porges, S.W., Carter, C.S., 2009.
amnesia in mice lacking the oxytocin gene. Nat. Genet. 25, 284–288. Oxytocin protects against negative behavioral and autonomic consequences of
Ferreira, A., Dahlof, L.G., Hansen, S., 1987. Olfactory mechanisms in the control of long-term social isolation. Psychoneuroendocrinology 34, 1542–1553.
maternal aggression, appetite, and fearfulness: effects of lesions to olfactory Grippo, A.J., Pournajafi-Nazarloo, H., Sanzenbacher, L., et al., 2012. Peripheral
receptors, mediodorsal thalamic nucleus, and insular prefrontal cortex. Behav. oxytocin administration buffers autonomic but not behavioral responses to envi-
Neurosci. 101, 709–717. ronmental stressors in isolated prairie voles. Stress 15, 149–161.
Ferris, C.F., Kulkani, P., Sullivan, J.M., Harder, J.A., Messenger, T.L., Febo, M., 2005. Guastella, A.J., Hickie, I.B., 2016. Oxytocin treatment, circuitry, and autism: A critical
Pup suckling is more rewarding than cocaine: evidence from functional magnetic review of the literature placing oxytocin into the autism context. Biol. Psychiatry 79,
resonance imaging and three dimensional computational analysis. J. Neurosci. 25, 234–242.
149–156. Gutkowska, J., Jankowski, M., 2012. Oxytocin revisited: its role in cardiovascular
Ferris, C.F., Yee, J., Kenkel, W., et al., September 17, 2015. Distinct BOLD activation regulation. J. Neuroendocrinol. 24, 599–608.
profiles following central and peripheral oxytocin administration in awake rats. Hammock, E.A.D., Young, L.J., 2002. Variation in vasopressin V1a receptor promoter
Front. Behav. Neurosci. 9, 245. http://dx.doi.org/10.3389/fnbeh.2015.00245. and expression: implications for inter- and intraspecific variation in social behavior.
Fink, S., Excoffier, L., Heckel, G., 2006. Mammalian monogamy is not controlled by Eur. J. Neurosci. 16, 399–402.
a single gene. Proc. Natl. Acad. Sci. U.S.A. 103, 10956–10960. Hammock, E.A.D., Young, L.J., 2004. Functional microsatellite polymorphisms associated
Firestone, K.B., Thompson, K.V., Carter, C.S., 1991. Behavioral correlates of intra- with divergent social structure in vole species. Mol. Biol. Evol. 21, 1057–1063.
female reproductive suppression in prairie voles, Microtus ochrogaster. Behav. Hammock, E.A.D., Young, L.J., 2005. Microsatellite instability generates diversity in
Neural Biol. 55, 31–41. brain and sociobehavioral traits. Science 308, 1630–1634.
Fisher, H., 2016. Anatomy of Love. WW Norton, New York, NY. Harlow, H.F., 1971. Learning to Love. Albion Publishing Co, San Francisco, CA.
Fisher, H., Aron, A., Brown, L.L., 2015. Romantic love: an fMRI study of a neural Harris, J.C., Carter, C.S., 2013. Therapeutic interventions with oxytocin: current status
mechanism for mate choice. J. Comp. Neurol. 593, 58–62. and concerns. J. Am. Acad. Child Adolesc. Psychiatry 52, 998–1000.
Fleming, A.S., Cheung, U., Myhal, N., Kessler, Z., 1989. Effects of maternal hormones Hazen, C., Shaver, P.R., 1987. Romantic love conceptualized as an attachment.
on ‘timidity’ and attraction to pup-related odors in female rats. Physiol. Behav. 46, J. Pers. Soc. Psychol. 52, 511–524.
449–453. Hennessy, M.B., 1997. Hypothalamic-pituitary-adrenal responses to brief social
Franceschini, R., Venturini, P.L., Cataldi, A., Barrecca, T., Ragni, N., Rolandi, E.E., separation. Neurosci. Biobehav. Rev. 21, 11–29.
1989. Plasma beta-endorphin concentrations during suckling in lactating women. Henry, J.P., Wang, S., 1998. Effects of early stress on adult affiliative behavior.
Br. J. Obstet. Gynaecol. 96, 711–713. Psychoneuroendocrinology 23, 863–876.
Francis, D.D., Young, L.J., Meaney, M.J., Insel, T.R., 2002. Naturally occurring differ- Herman, B.H., Panksepp, J., 1978. Effects of morphine and naloxone on separation
ences in maternal care are associated with the expression of oxytocin and vaso- distress and approach attachment: evidence of opiate mediation of social effect.
pressin (V1a) receptors: gender differences. J. Neuroendocrinol. 14, 349–353. Pharmacol. Biochem. Behav. 9, 213–220.
Freeman, S.M., Young, L.J., 2016. Comparative perspectives on oxytocin and vaso- Hillegaart, V., Alster, P., Uvnas-Moberg, K., Ahlenius, S., 1998. Sexual motivation
pressin receptor research in rodents and primates. J. Neuroendocrinol. 28 (4) promotes oxytocin secretion in male rats. Peptides 19, 39–45.
http://dx.doi.org/10.111/jne.123.82. Hofer, M.A., 1987. Early social relationships: a psychobiologist’s view. Child. Dev. 58,
Gainer, H., 2012. Cell-type specific expression of oxytocin and vasopressin genes: an 633–647.
experimental odyssey. J. Neuroendocrinol. 24, 528–538. Hornak, J., Rolls, E.T., Wade, D., 1996. Face and voice expression identification in
Garrison, J.L., Macosko, E.Z., Bernstein, S., Pokala, N., Albrecht, D.R., patients with emotional and behavioural changes following ventral frontal lobe
Bargmann, C.I., 2012. Oxytocin/vasopressin-related peptides have an ancient role damage. Neuropsychologia 34, 247–261.
in reproductive behavior. Science 338, 540–543. Hrdy, S.B., 2009. Mothers and Others: The Evolutionary Origins of Mutual Under-
Getz, L.L., Carter, C.S., Gavish, L., 1981. The mating system of the prairie vole standing. Belknap Press, Harvard University Press, Cambridge, MA.
Microtus ochrogaster: field and laboratory evidence for pair-bonding. Behav. Ecol. Hurlemann, R., Patin, A., Onur, O.A., et al., 2010. Oxytocin enhances amygdala-
Sociobiol. 8, 189–194. dependent, socially reinforced learning and emotional empathy in humans.
Getz, L.L., McGuire, B., Pizzuto, T., Hormann, J.E., Frase, B., 1993. Social organi- J. Neurosci. 30, 4999–5007.
zation of the prairie vole (Microtus ochrogaster). J. Mammal. 74, 44–48. Insel, T.R., 1997. A neurobiological basis of social attachment. Am. J. Psychiatry 154,
Gingrich, B.S., Liu, Y., Cascio, C., Wang, Z., Insel, T.R., 2000. Dopamine D2 receptors 726–735.
in the nucleus accumbens are important for social attachment in female prairie Insel, T.R., Hulihan, T.J., 1995. A gender-specific mechanism for pair bonding:
voles (Microtus ochrogaster). Behav. Neurosci. 114, 173–183. oxytocin and partner preference formation in monogamous voles. Behav. Neurosci.
Goland, R.S., Wardlaw, S.L., Stark, R.I., Frantz, A.G., 1981. Human plasma beta- 109, 782–789.
endorphin during pregnancy, labor, and delivery. J. Clin. Endocrinol. Metab. 52, Insel, T.R., Preston, S., Winslow, J.T., 1995. Mating in the monogamous male:
74–78. behavioral consequences. Physiol. Behav. 57, 615–627.
Gouin, J.P., Carter, C.S., Pournajafi-Nazarloo, H., et al., 2010. Martial behavior, oxytocin, Insel, T.R., Shapiro, L.E., 1992. Oxytocin receptor distribution reflects social organi-
vasopressin and wound healing. Psychoneuroendocrinology 35, 1082–1090. zation in monogamous and polygamous voles. Proc. Natl. Acad. Sci. U.S.A. 89,
Greenberg, J.L., Lewis, S.E., Dodd, D.K., 1999. Overlapping addictions and self- 5981–5985.
esteem among college men and women. Addict. Behav. 24, 562–571. Insel, T.R., Wang, Z., Ferris, C.F., 1994. Patterns of brain vasopressin receptor
Gregory, S.G., Connelly, J.J., Towers, A.J., et al., 2009. Genomic and epigenetic distribution associated with social organization in microtine rodents. J. Neurosci.
evidence for oxytocin receptor deficiency in autism. BMC Med. 7, 62. 14, 5381–5392.
Grewen, K.M., Light, K.C., 2011. Plasma oxytocin is related to lower cardiovascular Insel, T.R., Winslow, J.T., 1991. Central administration of oxytocin modulates the
and sympathetic reactivity to stress. Biol. Psychol. 87, 340–349. infant rat’s response to social isolation. Eur. J. Pharmacol. 203, 149–152.
Grinevich, V., Knobloch-Bollmann, H.S., Eliava, M., Busnelli, M., Chini, B., 2016. Insel, T.R., Young, L., Wang, Z., 1997. Molecular aspects of monogamy. Ann. N.Y.
Assembling the puzzle: pathways of oxytocin signaling in the brain. Biol. Psychiatry Acad. Sci. 807, 302–316.
79, 155–164. Jack, A., Connelly, J.J., Morris, J.P., 2012. DNA methylation of the oxytocin receptor gene
Grippo, A.J., Cushing, B.S., Carter, C.S., 2007a. Depression-like behavior and predicts neural response to ambiguous social stimuli. Front. Hum. Neurosci. 6, 280.
stressor-induced neuroendocrine activation in female prairie voles exposed to Jacob, S., Brune, C.W., Carter, C.S., Leventhal, B., Lord, C., Cook Jr., E.H., 2007.
chronic social isolation. Psychosom. Med. 69, 149–157. Association of the oxytocin receptor gene (OXTR) in Caucasian children and
Grippo, A.J., Gerena, D., Huang, J., et al., 2007b. Social isolation induces behavioral adolescents with autism. Neurosci. Lett. 417, 6–9.
and neuroendocrine disturbances relevant to depression in female and male prairie Jankowski, M., Broderick, T.L., Gutkowska, J., 2016. Oxytocin and cardioprotection in
voles. Psychoneuroendocrinology 32, 966–980. diabetes and obesity. BMC Endocr. Disord. 16 (1), 34. http://dx.doi.org/10.1186/
Grippo, A.J., Lamb, D.G., Carter, C.S., Porges, S.W., 2007c. Social isolation disrupts s12902-016-0110-1.
autonomic regulation of the heart and influences negative affective behaviors. Biol. Jiménez, A., Young, L.J., Triana-Del Río, R., La Prairie, J.L., González-Mariscal, G., 2015.
Psychiatry 62, 1162–1170. Neuroanatomical distribution of oxytocin receptor binding in the female rabbit forebrain:
Grippo, A.J., Wu, K.D., Hassan, I., Carter, C.S., 2008. Social isolation in prairie voles variations across the reproductive cycle. Brain Res. 1629, 329–339.
induces behaviors relevant to negative affect: toward the development of a rodent Kanthak, M.K., Chen, F.S., Kumsta, R., Hill, L.K., Thayer, J.F., Heinrichs, M., 2016.
model focused on co-occurring depression and anxiety. Depress. Anxiety 25, Oxytocin receptor gene polymorphism modulates the effects of social support on
E17–E26. heart rate variability. Biol. Psychol. 117, 43–49.
The Neurobiology of Social Affiliation and Pair Bonding 141

Karelina, K., DeVries, A.C., 2011. Modeling social influences on human health. Psy- Landgraf, R., Neumann, I.D., 2004. Vasopressin and oxytocin release within the brain:
chosom. Med. 73, 67–74. a dynamic concept of multiple and variable modes of neuropeptide communication.
Kendrick, K.M., 2000. Oxytocin, motherhood and bonding. Exp. Physiol. 85S, Front. Neuroendocrinol. 25, 150–176.
111S–124S. Landgraf, R., Neumann, I., Pittman, Q.J., 1991. Septal and hippocampal release of
Kenkel, W., Paredes, J., Yee, J.R., Pournajafi-Nazarloo, H., Bales, K.L., Carter, C.S., vasopressin and oxytocin during late pregnancy and parturition in the rat.
2012. Exposure to an infant releases oxytocin and facilitates pair-bonding in male Neuroendocrinology 54, 378–383.
prairie voles. J. Neuroendocrinol. 24, 874–886. Leckman, J.F., Goodman, W.K., North, et al., 1994. The role of central oxytocin in
Kenkel, W.M., Paredes, J., Lewis, G.F., Yee, J.R., Pournajafi-Nazarloo, H., obsessive compulsive disorder and related normal behavior. Psychoneur-
Grippo, A.J., Porges, S.W., Carter, C.S., 2013. Autonomic substrates of the oendocrinology 19, 723–749.
response to pups in male prairie voles. PLoS One 8 (8), e69965. http://dx.doi.org/ LeSaux, M., DiPaolo, T., 2005. Chronic estrogenic drug treatment increases pre-
10.1371/journal.pone.0069965. proenkephalin mRNA levels in the rat striatum and nucleus accumbens. Psycho-
Kenkel, W.M., Yee, J.R., Carter, C.S., 2014. Is oxytocin a maternal foetal signaling neuroendocrinology 30, 251–260.
molecule at birth? Implications for development. J. Neuroendocrinol. 26, 739–749. Leuner, B., Caponiti, J.M., Gould, E., 2012. Oxytocin stimulates adult neurogenesis
Kendrick, K.M., Keverne, E.B., 1989. Effects of intracerebroventricular infusions of even under conditions of stress and elevated glucocorticoids. Hippocampus 22,
naltrexone and phentolamine on central and peripheral oxytocin release and on 861–868.
maternal behaviour induced by vaginocervical stimulation in the ewe. Brain Res. Levin, M.C., Sawchenko, P.E., 1993. Neuropeptide co-expression in the magnocellular
505, 329–332. neurosecretory system of the female rat: evidence for differential modulation by
Kendrick, K.M., Keverne, E.B., 1991. Importance of progesterone and estrogen estrogen. Neuroscience 54, 1001–1018.
priming for the induction of maternal behavior by vaginocervical stimulation in Lévy, F., Kendrick, K.M., Keverne, E.B., Piketty, V., Poindron, P., 1992. Intra-
sheep: effects of maternal experience. Physiol. Behav. 49, 745–750. cerebral oxytocin is important for the onset of maternal behavior in inexpe-
Kendrick, K.M., Keverne, E.B., 1992. Control of synthesis and release of oxytocin in rienced ewes delivered under peridural anaesthesia. Behav. Neurosci. 106,
the sheep brain. Ann. N.Y. Acad. Sci. 652, 102–121. 427–432.
Kendrick, K.M., Keverne, E.B., Baldwin, B.A., 1987. Intracerebroventricular oxytocin Li, Q., Levy, A.D., Cabrera, T.M., Brownfield, M.S., Battaglia, G., Van de Kar, L.D.,
stimulates maternal behaviour in the sheep. Neuroendocrinology 46, 56–61. 1993. Long-term fluoxetine, but not desipramine, inhibits the ACTH and oxytocin
Kendrick, K.M., Keverne, E.B., Hinton, M.R., Goode, J.A., 1986. Cerebrospinal fluid responses to the 5-HT1a agonist, 8-OH-DPAT, in male rats. Brain Res. 630,
levels of acetylcholinesterase, monoamines and oxytocin during labour, parturition, 148–156.
vaginocervical stimulation, lamb separation and suckling in sheep. Neuroendocri- Lidow, M.S., Wang, F., Cao, Y., Goldman-Rakic, P.S., 1998. Layer V neurons bear the
nology 44, 149–156. majority of mRNAs encoding the five distinct dopamine receptor subtypes in the
Kendrick, K.M., Levy, F., Keverne, E.B., 1992. Changes in the sensory processing of primate prefrontal cortex. Synapse 28, 10–20.
olfactory signals induced by birth in sheep. Science 256, 833–836. Lim, M.M., Young, L.J., 2004. Vasopressin-dependent neural circuits underlying pair
Keverne, E.B., 2013. Significance of epigenetics for understanding brain development, bond formation in the monogamous prairie vole. Neuroscience 125, 35–45.
brain evolution and behaviour. Neuroscience 264, 207–217. Lim, M.M., Wang, Z., Olazábal, D.E., Ren, X., Terwilliger, E.F., Young, L.J., 2004.
Keverne, E.B., Kendrick, K.M., 1991. Morphine and corticotropin releasing factor Enhanced partner preference in a promiscuous species by manipulating the
potentiate maternal acceptance in multiparous ewes after vaginocervical stimula- expression of a single gene. Nature 429, 754–757.
tion. Brain Res. 540, 55–62. Liu, Y., Wang, Z.X., 2003. Nucleus accumbens OT and DA interact to regulate pair
Keverne, E.B., Kendrick, K.M., 1992. Oxytocin facilitation of maternal behavior. Ann. bond formation in female prairie voles. Neuroscience 121, 537–544.
N.Y. Acad. Sci. 652, 83–101. Liu, Y., Curtis, J.T., Wang, Z., 2001. Vasopressin in the lateral septum regulates pair
Keverne, E.B., Lévy, F., Poindron, P., Lindsay, D.R., 1983. Vaginal stimulation: an bond formation in male prairie voles (Microtus ochrogaster). Behav. Neurosci. 115,
important determinant of maternal bonding in sheep. Science 219, 81–83. 910–919.
Keverne, E.B., Fundele, R., Narashima, M., Barton, S., Surani, M.A., 1996a. Genomic Lorberbaum, J.P., Newman, J.D., Horwitz, A.R., et al., 2002. A potential role for
imprinting and the differential roles of parental genomes in brain development. Dev. thalamocingulate circuitry in human maternal behavior. Biol. Psychiatry 51,
Brain Res. 92, 91–100. 431–445.
Keverne, E.B., Martel, F.L., Nevison, C.M., 1996b. Primate brain evolution: genetic and Luzzati, V., Mateu, L., Marquez, G., Borgo, M., 1999. Structural and electrophys-
functional considerations. Proc. Biol. Sci. 262, 689–696. iological effects of local anesthetics and of low temperature on myelinated
Keverne, E.B., Nevison, C.M., Martel, F.L., 1997. Early learning and the social bond. nerves: implication of the lipid chains in nerve excitability. J. Mol. Biol. 286,
Ann. N.Y. Acad. Sci. 807, 329–339. 1389–1402.
Kim, P., Strathearn, L., Swain, J.E., 2016. The maternal brain and its plasticity in Lyons, D.M., Schatzberg, A.F., 2003. Early maternal availability and prefrontal
humans. Horm. Behav. 77, 113–123. correlates of reward-related memory. Neurobiol. Learn. Mem. 80, 97–104.
Kirkpatrick, B., 1997. Affiliation and neuropsychiatric disorders: the deficit syndrome of Mansour, S.L., Thomas, K.R., Capecchi, M.R., 1988. Disruption of the proto-oncogene
schizophrenia. Ann. N.Y. Acad. Sci. 807, 455–468. int-2 in mouse embryo-derived stem cells: a general strategy for targeting
Klaus, M.H., Kennel, J.H., Klaus, P.H., 1995. In: Bonding. Addison Wesley, mutations to non-selectable genes. Nature 336, 348–352.
Reading, MA. Martel, F.L., Nevison, C.M., Rayment, F.D., Simpson, M.D.A., Keverne, E.B., 1993.
Kleiman, D., 1977. Monogamy in mammals. Q. Rev. Biol. 52, 39–69. Opioid receptor blockade reduces maternal affect and social grooming in rhesus
Kob, B.E., Fredrickson, B.L., 2010. Upward spirals of the heart: autonomic flexibility as monkeys. Psychoneuroendocrinology 18, 307–321.
indexed by vagal tone, reciprocally and prospectively predicts positive emotions and Martel, F.L., Nevison, C.M., Simpson, M.J.A., Keverne, E.B., 1995. Effects of opioid
social connectedness. Biol. Psychol. 85, 432–436. receptor blockade on the social behavior of rhesus monkeys living in large family
Koob, G.F., Le Moal, M., 1997. Drug abuse: hedonic homeostatic dysregulation. groups. Dev. Psychobiol. 28, 71–84.
Science 278, 52–58. Mason, W.A., Mendoza, S.P., 1998. Generic aspects of primate attachments: parents,
Kosfeld, M., Heinrichs, M., Zak, P.J., Fischbacher, U., Fehr, E., 2005. Oxytocin offspring and mates. Psychoneuroendocrinology 23, 765–778.
increases trust in humans. Nature 435, 673–676. Matsumoto, K., Tanaka, K., 2004. The role of the medial prefrontal cortex in achieving
Kovacs, G.L., Sarnyai, Z., Szabo, G., 1998. Oxytocin and addiction: a review. Psy- goals. Curr. Opin. Neurobiol. 14, 178–185.
choneuroendocrinology 23, 945–962. Mayes, L.C., 1995. Substance abuse and parenting. In: Bornstein, M.H. (Ed.),
Kraemer, G.W., 1992. A psychobiological theory of attachment. Behav. Brain Sci. 15, Handbook of Parenting: Applied and Practical Parenting, vol. 4. Lawrence Erlbaum
493–511. Associates, New Jersey, pp. 101–126.
Kramer, K.M., Cushing, B.S., Carter, C.S., Wu, J., Ottinger, M.A., 2004. Sex and Mazzuca, M., Minlebaev, M., Shakirzyanova, A., et al., 2011. Newborn analgesia
species differences in plasma oxytocin using an enzyme immunoassay. Can. J. mediated by oxytocin during delivery. Front. Cell. Neurosci. 12, 5.
Zool. 82, 1194–1200. McCarthy, M.M., Kow, L.M., Pfaff, D.W., 1992. Speculations concerning the physi-
Labad, J., Menchon, J.M., Alonso, P., Segalas, C., Jimenez, S., Vallejo, J., 2005. ological significance of central oxytocin in maternal behavior. Ann. N.Y. Acad. Sci.
Female reproductive cycle and obsessive-compulsive disorder. J. Clin. Psychiatry 652, 70–82.
66, 428–435. McNeilly, A.S., Robinson, I.C.A.F., Houston, M.J., Howie, P.W., 1983. Release of
Lancaster, K., Carter, C.S., Pournajafi-Nazarloo, H., et al., 2015. Plasma oxytocin oxytocin and prolactin in response to suckling. Br. Med. J. 286, 207–209.
explains individual differences in neural substrates of social perception. Front. Mendoza, S.P., Mason, W.A., 1997. Attachment relationships in New World primates.
Hum. Neurosci. 17 (9), 132. http://dx.doi.org/10.3389/fnhuman.2015.00132. Ann. N.Y. Acad. Sci. 807, 203–209.
142 The Neurobiology of Social Affiliation and Pair Bonding

Meyer-Lindenberg, A., Domes, G., Kirsch, P., Heinrichs, M., 2011. Oxytocin and Rault, J.-L., Carter, C.S., Garner, J.P., Marchant-Forde, J.N., Richert, B.T.,
vasopressin the human brain: social neuropeptides for translational medicine. Nat. Lay Jr., D.C., 2013. Repeated intranasal oxytocin administration in early life
Rev. Neurosci. 12, 524–538. dysregulates the HPA axis and alters social behavior. Physiol. Behav. 112-113,
Mitchell, J.P., Neil-Macrae, C., Banaji, M.R., 2005. Forming impressions of people 40–48.
versus inanimate objects: social-cognitive processing in the medial prefrontal Reite, M., Boccia, M.L., 1994. Physiological aspects of adult attachment. In:
cortex. Neuroimage 26, 251–257. Sperling, M.B., Berman, W.H. (Eds.), Attachment in Adults. Guilford Press, New
Modahl, C., Green, L.-A., Fein, D., Morris, M., Waterhouse, L., Feinstein, C., Levin, H., York, pp. 98–127.
1998. Plasma oxytocin levels in autistic children. Biol. Psychiatry 43, 270–277. Ridderinkhof, K.R., van den Wildenberg, W.P., Segalowitz, S.J., Carter, C.S., 2004.
Money, J., 1980. Love & Love Sickness. Johns Hopkins Press, Baltimore, MD. Neurocognitive mechanisms of cognitive control: the role of prefrontal cortex in
Nelson, E., Panksepp, J., 1996. Oxytocin mediates acquisition of maternally associated action selection, response inhibition, performance monitoring, and reward-based
odor preferences in preweanling rat pups. Behav. Neurosci. 110, 583–592. learning. Brain Cogn. 56, 129–140.
Nelson, E., Panksepp, J., 1998. Brain substrates of infant-mother attachment: Rilling, J.K., DeMarco, A.C., Hackett, P.D., Thompson, R., Ditzen, B., Patel, R.,
contributions of opioids, oxytocin and norepinephrine. Neurosci. Biobehav. Rev. 22, Pagnoni, G., 2012. Effects of intranasal oxytocin and vasopressin on cooperative
437–452. behavior and associated brain activity in men. Psychoneuroendocrinology 37,
Neumann, I.D., Landgraf, R., 2012. Balance of brain oxytocin and vasopressin: 447–461.
implications for anxiety, depression and social behaviors. Trends Neurosci. 35, Roberts, R.L., Zullo, A.S., Carter, C.S., 1997. Sexual differentiation in prairie voles: the
649–659. effects of corticosterone and testosterone. Physiol. Behav. 62, 1379–1383.
Nishimori, K., Young, L.J., Guo, Q., Wang, Z., Insel, T.R., Matzuk, M.M., 1996. Rogers, R.D., Ramnani, N., Mackay, C., et al., 2004. Distinct portions of anterior
Oxytocin is required for nursing but is not essential for parturition or reproductive cingulate cortex and medial prefrontal cortex are activated by reward processing in
behavior. Proc. Natl. Acad. Sci. U.S.A. 93, 11699–11704. separable phases of decision-making cognition. Biol. Psychiatry 55, 594–602.
Norman, G.J., Cacioppo, J.T., Morris, J.S., Malarkey, W.B., Berntsen, G.G., Rubin, L.H., Carter, C.S., Drogos, L., et al., 2011. Sex-specific associations between
DeVries, A.C., 2011. Oxytocin increases autonomic cardiac control: moderation by peripheral oxytocin and positive emotion perception in schizophrenia. Schizophr.
loneliness. Biol. Psychol. 86, 174–180. Res. 30, 266–270.
Okhovat, M., Berrio, A., Wallace, G., Ophir, A.G., Phelps, S.M., 2015. Sexual fidelity Ruscio, M.G., Sweeny, T., Hazelton, J., Suppatkul, P., Carter, C.S., 2007. Social
trade-offs promote regulation variation in the prairie vole brain. Science 350, environment regulates corticotropin releasing factor, corticosterone and vaso-
1371–1374. pressin in juvenile prairie voles. Horm. Behav. 51, 54–61.
Ortiz-Miranda, S., Dayanithi, G., Custer, E., Treistman, S.N., Lemos, J.R., 2005. Ruscio, M.G., Sweeny, T.D., Hazelton, J.L., Suppatkul, P., Booth, E., Carter, C.S.,
Micro-opioid receptor preferentially inhibits oxytocin release from neurohypophysial 2008. Pup exposure elicits hippocampal cell proliferation in the prairie vole. Behav.
terminals by blocking R-type Ca2þ channels. J. Neuroendocrinol. 17, 583–590. Brain Res. 187, 9–16.
Ostrowski, N.L., 1998. Oxytocin receptor mRNA expression in rat brain: implications Russell, J.A., Leng, G., 1998. Sex, parturition and motherhood without oxytocin?
for behavioral integration and reproductive success. Psychoneuroendocrinology 23, J. Endocrinol. 157, 343–359.
989–1004. Ryff, C.S., Singer, B., 1998. The concept of positive human health. Psychol. Inq. 9,
Panksepp, J., Nelson, E., Bekkedal, M., 1997. Brain systems for the mediation of 1–19.
social separation-distress and social-reward. Ann. N.Y. Acad. Sci. 807, 78–100. Sachser, N., Durschlag, M., Hirzel, D., 1998. Social relationships and the management
Pedersen, C.A., Ascher, J.A., Monroe, J.A., Prange Jr., A.J., 1982. Oxytocin induces of stress. Psychoneuroendocrinology 23, 891–904.
maternal behavior in virgin female rats. Science 216, 648–650. Schulkin, J., 1999. Corticotropin-releasing hormone signals adversity in both the
Pedersen, C.A., Prange Jr., A.J., 1979. Induction of maternal behavior in virgin rats placenta and the brain: regulation by glucocorticoids and allostatic overload.
after intracerebroventricular administration of oxytocin. Proc. Natl. Acad. Sci. U.S.A. J. Endocrinol. 161, 349–356.
76, 6661–6665. Schultz, W., 2007. Multiple dopamine functions at different time course. Annu. Rev.
Pedersen, C.A., Boccia, M.L., 2002. Oxytocin links mothering received, mothering Neurosci. 30, 259–288.
bestowed and adult stress responses. Stress 5, 259–267. Schultz, W., Dickinson, A., 2000. Neuronal coding of prediction errors. Annu. Rev.
Petersson, M., Lundeberg, T., Uvnas-Moberg, K., 1999. Short-term increase and long- Neurosci. 23, 473–500.
term decrease of blood pressure in response to oxytocin-potentiating effect of Schumacher, M., Coirini, H., Pfaff, D.W., McEwen, B.S., 1990. Behavioral effects of
female steroid hormones. J. Cardiovasc. Pharmacol. 33, 102–108. progesterone associated with rapid modulation of oxytocin receptors. Science 250,
Phelps, S.M., Young, L.J., 2003. Extraordinary diversity in vasopressin (V1a) receptor 691–694.
distributions among wild prairie voles (Microtus ochrogaster): patterns of variation Shamay-Tsoory, S.G., Tomer, R., Aharon-Peretz, J., 2005. The neuroanatomical basis
and covariation. J. Comp. Neurol. 466, 564–576. of understanding sarcasm and its relationship to social cognition. Neuropsychology
Poindron, P., Le Neindre, P., 1980. Endocrine and sensory regulation of maternal 19, 288–300.
behavior in the ewe. Adv. Study Behav. 11, 75–119. Singh, P.J., Hofer, M.A., 1978. Oxytocin reinstates maternal olfactory cues for nipple
Poindron, P., Lévy, F., Keller, M., 2007. Maternal responsiveness and maternal orientation and attachment in rat pups. Physiol. Behav. 20, 385–389.
selectivity in domestic sheep and goats: the two facets of maternal attachment. Sofroniew, M.W., 1983. Vasopressin and oxytocin in the mammalian brain and spinal
Dev. Psychobiol. 49, 54–70. cord. Trends Neurosci. 6, 467–472.
Porges, S.W., 1998. Love: an emergent property of the mammalian autonomic nervous Solomon, N.G., French, J. (Eds.), 1997. Cooperative Breeding in Mammals. Cambridge
system. Psychoneuroendocrinology 23, 837–862. University Press, New York.
Porges, S.W., 2007. The polyvagal perspective. Biol. Psychol. 74, 116–143. Somel, M., Liu, X., Khaitovich, P., 2013. Human brain evolution: transcripts, metab-
Porges, S.W., 2011. The Polyvagal Theory: Neurophysiological Foundations of olites and their regulators. Nat. Rev. Neurosci. 14, 112–127.
Emotions, Attachment, Communication and Self-regulation. WW Norton and Co, Sowell, E.R., Thompson, P.M., Tessner, K.D., Toga, A.W., 2001. Mapping continued
New York, NY. brain growth and gray matter density reduction in dorsal frontal cortex: inverse
Poulain, P., Pittman, Q., 1993. Oxytocin pretreatment enhances arginine vasopressin- relationships during postadolescent brain maturation. J. Neurosci. 21,
induced motor disturbances and arginine vasopressin-induced phosphoinositol 8819–8829.
hydrolysis in rat septum: a cross-sensitization phenomenon. J. Neuroendocrinol. 5, Spencer, S.J., Buller, K.M., Day, T.A., 2005. Medial prefrontal cortex control of
33–39. the paraventricular hypothalamic nucleus response to psychological stress:
Pournajafi-Nazarloo, H., Kenkel, W., Mohsenpour, S.R., Sanzenbacher, L., Saadat, H., possible role of the bed nucleus of the stria terminalis. J. Comp. Neurol.
Partoo, L., Yee, J., Azizi, F., Carter, C.S., 2013. Exposure to chronic isolation 481, 363–376.
modulates receptors mRNAs for oxytocin and vasopressin in the hypothalamus and Stoop, R., 2012. Neuromodulation by oxytocin and vasopressin. Neuron 76,
heart. Peptides 43, 20–26. 142–159.
Rakic, P., 2009. Evolution of the neocortex: a perspective from developmental biology. Stribley, J.M., Carter, C.S., 1999. Developmental exposure to vasopressin increases
Nat. Rev. Neurosci. 10, 724–735. aggression in adult prairie voles. Proc. Natl. Acad. Sci. U.S.A. 96, 12601–12604.
Ranote, S., Elliott, R., Abel, K.M., Mitchell, R., Deakin, J.F., Appleby, L., 2004. The Sun, P., Smith, A.S., Lei, K., Liu, Y., Wang, Z., 2014. Breaking bonds in male prairie
neural basis of maternal responsiveness to infants: an fMRI study. Neuroreport 15, voles: long-term effects on emotional and social behavior, physiology, and
1825–1929. neurochemistry. Behav. Brain Res. 265, 22–31.
Rauch, S.L., 2000. Neuroimaging research and the neurobiology of obsessive- Swaab, D.F., Boer, G.J., 1994. Neuropeptides and brain development: current perils
compulsive disorder: where do we go from here? Biol. Psychiatry. 47, 168–170. and future potential. J. Dev. Physiol. 5, 67–75.
The Neurobiology of Social Affiliation and Pair Bonding 143

Taylor, S.E., Klein, L.C., Lewis, B.P., Gruenewald, T.L., Gurung, R.A., Updegraff, J.A., Williams, J.R., Catania, K.C., Carter, C.S., 1992. Development of partner preferences
2000. Biobehavioral responses to stress in females: tend-and-befriend, not fight- in female prairie voles (Microtus ochrogaster): the role of social and sexual
or-flight. Psychol. Rev. 107, 411–429. experience. Horm. Behav. 26, 339–349.
Taylor, S.E., Gonzaga, G.C., Klein, L.C., Hu, P., Greendale, G.A., Seeman, T.E., 2006. Williams, J.R., Insel, T.R., Harbaugh, C.R., Carter, C.S., 1994. Oxytocin centrally
Relation of oxytocin to psychological stress responses and hypothalamic–pituitary– administered facilitates formation of a partner preference in female prairie voles
adrenocortical axis activity in older women. Psychosom. Med. 68, 238–245. (Microtus ochrogaster). J. Neuroendocrinol. 6, 247–250.
Taylor, S.E., Saphire-Bernstein, S., Seeman, T.E., 2010. Are plasma oxytocin in Williams, K.E., Koran, L.M., 1997. Obsessive-compulsive disorder in preg-
women and plasma vasopressin men biomarkers of distressed pair-bond rela- nancy, the puerperium, and the premenstruum. J. Clin. Psychiatry 58,
tionships? Psychol. Sci. 21, 3–7. 330–334.
Tost, H., Kolachana, B., Hakimi, S., et al., 2010. A common allele in the oxytocin Winslow, J.T., Hastings, N., Carter, C.S., Harbaugh, C.R., Insel, T.R., 1993. A role for
receptor gene (OXTR) impacts prosocial temperament and human hypothalamic- central vasopressin in pair bonding in monogamous prairie voles. Nature 365,
limbic structure and function. Proc. Natl. Acad. Sci. U.S.A. 107, 13936–13941. 545–548.
Tribollet, E., Dubois-Dauphin, M., Dreifuss, J.J., Barberis, C., Jard, S., 1992. Oxytocin Winslow, J.T., Hearn, E.F., Ferguson, J., Young, L.J., Matzuk, M.M., Insel, T.R., 2000.
receptors in the central nervous system: distribution, development, and species Infant vocalization, adult aggression, and fear behavior of an oxytocin null mutant
differences. Ann. N.Y. Acad. Sci. 652, 29–38. mouse. Horm. Behav. 37, 145–155.
Tyzio, R., Cossart, R., Khalilov, I., et al., 2006. Maternal oxytocin triggers a transient Winslow, J.T., Noble, P.L., Lyons, C.K., Sterk, S.M., Insel, T.R., 2003. Rearing effects
inhibitory switch in GABA signaling in the fetal brain during delivery. Science 314, of cerebrospinal fluid oxytocin concentration and social buffering in rhesus
1788–1792. monkeys. Neuropsychopharmacology 28, 910–918.
Uvnas-Moberg, K., 1994. Role of efferent and afferent vagal nerve activity during Winslow, J.T., Insel, T.R., 1991. Endogenous opioids: do they modulate the rat pup’s
reproduction: integrating function of oxytocin on metabolism and behavior. Psy- response to social isolation? Behav. Neurosci. 105, 253–263.
choneuroendocrinology 19, 687–695. Winslow, J.T., Insel, T.R., 1993. Effects of central vasopressin administration to infant
Uvnas-Moberg, K., 1997. Physiological and endocrine effects of social contact. Ann. rats. Eur. J. Pharmacol. 233, 101–107.
N.Y. Acad. Sci. 807, 146–163. Witt, D.M., 1997. Regulatory mechanisms of oxytocin-mediated sociosexual behavior.
Uvnas-Moberg, K., 1998. Oxytocin may mediate the benefits of positive social inter- Ann. N.Y. Acad. Sci. 807, 22–41.
action and emotions. Psychoneuroendocrinology 23, 819–836. Witt, D.M., Carter, C.S., Insel, T.R., 1991. Oxytocin receptor binding in female prairie
Uvnas-Moberg, K., Bjorkstrand, E., Hillegaart, V., Ahlenius, S., 1999. Oxytocin as voles: endogenous and exogenous oestradiol stimulation. J. Neuroendocrinol. 3,
a possible mediator of SSRI-induced antidepressant effects. Psychopharmacology 155–161.
142, 95–101. Witt, D.M., Carter, C.S., Chayer, R., Adams, K., 1989. Patterns of behaviour during
Van Kesteren, R.E., Smit, A.B., Dirkds, R.W., Dewith, N.D., Deraerts, W.P.M., postpartum oestrus in prairie voles, Microtus ochrogaster. Anim. Behav. 39,
Joosse, J., 1992. Evolution of the vasopressin/oxytocin superfamily: character- 528–534.
ization of a cDNA encoding a vasopressin-related precursor, preproconopressin, Witt, D.M., Carter, C.S., Walton, D., 1990. Central and peripheral effects of oxytocin
from the mollusc Lymnaea stagnalis. Proc. Natl. Acad. Sci. U.S.A. 89, administration in prairie voles (Microtus ochrogaster). Pharmacol. Biochem. Behav.
4593–4597. 37, 63–69.
Wang, Z., 1995. Species differences in the vasopressin-immunoreactive pathways in Witt, D.M., Winslow, J.T., Insel, T.R., 1992. Enhanced social interactions in rats
the bed nucleus of the stria terminalis and medial amygdaloid nucleus in prairie following chronic, centrally infused oxytocin. Pharmacol. Biochem. Behav. 43,
voles (Microtus ochrogaster) and meadow voles (Microtus pennsylvanicus). Behav. 855–861.
Neurosci. 109, 305–311. Yamamoto, Y., Cushing, B.S., Kramer, K.M., Epperson, P.D., Hoffman, G.E.,
Wang, Z., Zhou, L., Hulihan, T., Insel, T.R., 1996. Immunoreactivity of central vaso- Carter, C.S., 2004. Neonatal manipulations of oxytocin alter expression of
pressin and oxytocin pathways in microtine rodents: a quantitative comparative oxytocin and vasopressin immunoreactive cells in the paraventricular nucleus
study. J. Comp. Neurol. 366, 726–737. of the hypothalamus in a gender-specific manner. Neuroscience 125,
Wang, Z., Young, L.J., Liu, Y., Insel, T.R., 1997. Species differences in vasopressin 947–955.
receptor binding are evident early in development: comparative anatomic studies in Yee, J.R., Kenkel, W.M., Frijling, J.I., et al., 2016. Oxytocin promotes functional
prairie and montane voles. J. Comp. Neurol. 378, 535–546. coupling between paraventricular nucleus and both sympathetic and para-
Wardlaw, S.L., Frantz, A.G., 1983. Brain b-endorphin during pregnancy, parturition, sympathetic cardioregulatory nuclei. Horm. Behav. 80, 82–91.
and the post-partum period. Endocrinology 113, 1664–1668. Young, L.J., Juot, B., Nilsen, R., Wang, Z., Insel, T.R., 1996. Species differences in
Weisman, O., Zagoory-Sharon, O., Schneiderman, I., Gordon, I., Feldman, R., 2013. central oxytocin receptor gene expression: comparative analysis of promoter
Plasma oxytocin distributions in a large cohort of women and men and their sequences. J. Neuroendocrinol. 8, 777–783.
gender-specific associations with anxiety. Psychoneuroendocrinology 38, Young, L.J., Lilsen, R., Waymire, K.G., MacGregor, G.R., Insel, T.R., 1999. Increased
694–701. affiliative response to vasopressin in mice expressing the V1a receptor from
Weiss, M., Baerge, E., Wiseboard, S., Temple, J., 1995. The influence of gonadal a monogamous vole. Nature 400, 766–768.
hormones on periodicity of obsessive-compulsive disorder. Can. J. Psychiatry 40, Young, L.J., Wang, Z., Insel, T.R., 1998. Neuroendocrine bases of monogamy. Trends
205–207. Neurosci. 21, 71–75.
This page intentionally left blank
1.05 Hormones and the Development and Expression of Aggressive Behavior
Brian C Trainor, University of California, Davis, CA, USA
Cheryl L Sisk, Michigan State University, East Lansing, MI, USA
Randy J Nelson, Neuroscience Research Institute, Ohio State University Wexner Medical Center, Columbus, OH, USA
Ó 2017 Elsevier Inc. All rights reserved.

1.05.1 Introduction 146


1.05.1.1 Categories of Aggression 146
1.05.2 Animal Models and Tests of Aggression 147
1.05.3 Endocrine Signals’ and Receptors’ Contribution to Aggression 148
1.05.3.1 Steroid Hormones 148
1.05.3.1.1 Androgens 149
1.05.3.1.2 Estrogens 151
1.05.3.1.3 Glucocorticoids 151
1.05.3.2 Anabolic Steroid Abuse and Aggression 151
1.05.4 Brain Regions Contributing to Aggression 152
1.05.4.1 Studies in Rodents 153
1.05.4.2 Nonhuman Primates and Humans 154
1.05.5 Neurotransmitters, Hormones, and Aggression 155
1.05.5.1 Serotonin 155
1.05.5.2 Arginine Vasopressin 156
1.05.5.3 Monoamine Oxidase 156
1.05.5.4 Nitric Oxide 157
1.05.6 Development of Aggression 158
1.05.6.1 Rough-and-Tumble Play as an Antecedent to Aggressive Behavior 158
1.05.6.2 Endocrine Contributions to the Development of Aggressive Behavior: Perinatal Organizational Effects 159
1.05.6.3 Endocrine Contributions to the Development of Aggressive Behavior: Pubertal Organizational Effects 160
1.05.6.4 Endocrine Contributions to the Development of Aggressive Behavior: Pubertal Activational Effects 161
1.05.6.5 Social Experience and the Development of Aggressive Behavior 162
1.05.6.6 Conditioned Defeat 162
1.05.6.7 Aggression in Aged Individuals 162
1.05.7 Reciprocal Effects of Aggression on Steroid Hormones 163
1.05.7.1 Gene–Environment Interactions 163
1.05.8 Integration 165
References 166

Glossary
Aggression A form of social interaction that includes Lifetime History of Aggression (LHA) Scale An interview-
threat, attack, and fighting. based scale that is used by mental health workers to assess
Hormone response element Sequences of DNA in general aggressive tendencies in humans. Interviews can
promoter regions that are bound by hormone receptors. be supplemented with other sources such as clinical
Binding of the receptor complex promotes transcription. records.
Immediate early gene A gene that is expressed rapidly and Organizational/activational hypothesis The proposal that
transiently in response to various cellular stimuli. Several of early androgen exposure permanently organizes the
these genes are used by neuroscientists as indirect markers nervous system of mammals in a male-like manner. After
of neuronal activity because they are expressed when pre- or perinatal organization by androgens, these
neurons fire action potentials. hormones more readily activate male-typical postpubertal
Intermittent explosive disorder A disorder characterized behaviors by acting upon the organized structures.
by repeated episodes of aggressive, violent behavior that is Piloerection The erection of hair on the skin, used as
grossly out of proportion to the situation: thought to affect a threatening display by many animals.
as many as 7.3% of adults in the United States.

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00005-5 145


146 Hormones and the Development and Expression of Aggressive Behavior

1.05.1 Introduction question of whether neuroendocrine mechanisms of aggres-


sion were evolutionarily conserved. However, improved exper-
Aggression may be defined as overt behavior with the intention imental design and methodologies have provided new insights.
of inflicting harm or the threat of harm upon another indi- For example, androgens can be synthesized de novo within the
vidual. Aggressive behaviors range from lethal to subtle. The brain (Pradhan et al., 2010), so removal of testes does not
possibility for aggressive behavior exists whenever the interests necessarily remove all sources of testosterone. In addition,
of two or more individuals conflict (Svare, 1983). Conflicts rapid changes in testosterone (rather than baseline levels) are
typically arise over limited resources including territories, now known to have important effects on aggression (Gleason
mates, and food. Among social species, conflicts may arise et al., 2009). Improved experimental designs have revealed
over social status and limited resources. A social interaction that pairing testosterone measurements with competitive tasks
decides which individual gains status or access to the contested are more effective at revealing hormone-behavior relationships
resource. In many cases, a submissive posture or gesture from in humans (Section 1.05.4.2). Hormones affect aggression by
one individual avoids actual combat. Individuals may also acting on neural circuits in the brain, and these circuits have
participate in threat displays or ritualized combat in which been identified using lesions, immediate early gene studies,
dominance is determined, but no physical damage is inflicted. neuropharmacology studies, and more recently optical stimu-
Species- and situation-specific rules exist to regulate aggression. lation. Within these neural circuits neurotransmitters such as
When aggressive behaviors break these rules or when the serotonin have important modulatory effects on aggression.
aggression is excessive to situational norms, then it may be Finally, experience has important effects on how these neural
considered to be pathological or violent (Haller and Kruk, circuits respond in competitive contexts. These effects of expe-
2006; Miczek et al., 2007). Pathological or violent aggression rience occur across development, both early in life and in the
causes much personal and societal suffering and thus is an adult.
important topic for study. Treatment for violent or pathological
aggressive behavior remains primitive. Essentially, no effective
1.05.1.1 Categories of Aggression
interventions exist for violent humans – the most common
‘treatment’ for violent people is incarceration (Eastman and Several categories of aggression are generally recognized, and
Campbell, 2006). Remarkably, a death sentence is the sole the different types of aggression have different neuroendocrine
treatment for an aggressive dog that bites a person. Thus, bases. For example, there are important differences in the
understanding the pathophysiology of aggression remains an neuroendocrine mechanisms of maternal aggression versus
important, yet understudied endeavor. territorial aggression, but there is also a significant degree of
Indeed, in common with categorizing human behavior, it is overlap. Androgens (Bouissou, 1983) and estrogens (Laredo
often difficult for observers to draw the line between adaptive et al., 2014b) have important modulatory effects on intermale
aggression and pathological aggression in nonhuman animals aggression, especially in territorial contexts. Hormonal changes
(Sluyter et al., 2003). An important strategy for improving our associated with the pregnancy, including high levels of estro-
ability to draw this line is to understand the mechanistic basis gens, have an important role for inducing maternal aggression
for both species-specific aggression and pathological aggres- (Gammie et al., 2007). In the most widely studied rodent
sion. This review will examine the role of neural circuits, devel- species (domestic mice and rats), maternal aggression is the
opment, and neuroendocrine mechanisms in the regulation of main form of aggression in females. However, in other rodent
aggression, primarily in species-specific aggression. Under species such as Syrian hamsters intrafemale aggression is more
certain conditions, aggression can deviate from normal prevalent (Gutzler et al., 2010; Staffend and Meisel, 2012),
patterns (Miczek et al., 2013). Although these models are which provides opportunities for examining neuroendocrine
beginning to provide important insights into potential mecha- mechanism of female aggression outside of the context of
nisms of violence, it is necessary to first understand the basis for reproduction. Another type of agonistic behavior commonly
normative forms of aggression. studied in the laboratory has been called fear-induced aggres-
Mechanistic studies of aggression have historically focused sion, but this is more correctly termed ‘defense’ and is not
on aggression in nonpathological states, and in this state strongly modulated by estrogens or androgens (Blanchard
gonadal hormones play an important role. Across most species, and Blanchard, 1989). Recently there has been increased
males are more aggressive than females, and this difference interest in abnormal forms of aggression (or escalated aggres-
usually emerges at puberty, which coincides with important sion) that might resemble aspects of human violence.
sex differences in gonadal hormone secretion. These patterns Abnormal aggression has been characterized using quantitative
have contributed to a historic focus on the role of gonadal or qualitative criteria (Natarajan and Caramaschi, 2010; Miczek
hormones such as testosterone in the regulation of aggressive et al., 2013). Aggression can differ quantitatively such as induc-
behaviors. Initial studies using hormone ablation and replace- tion with low provocation or occur at higher intensity. Alterna-
ment approaches showed a clear role for androgens such as tively, aggression can differ qualitatively such as through
testosterone in many species (Vandenbergh, 1971; Leshner attacks directed at vulnerable regions or through a lack of
and Moyer, 1975; Soma, 2006). However, exceptional cases responding to submissive displays that normally trigger cessa-
appeared in which removal of testes had no obvious effect on tion of aggressive behavior. Abnormal aggression can be
aggression (Demas et al., 1999; Trainor and Marler, 2001). induced by very low glucocorticoid levels induced by adrenal-
Furthermore, for many years the link between testosterone ectomy (Haller et al., 2001). Intriguingly, in this context aggres-
and aggression in humans appeared very weak (Archer, sion occurs in the absence of autonomic activation, which is
1991). These apparently inconsistent observations raised the observed in some forms of human violence.
Hormones and the Development and Expression of Aggressive Behavior 147

Whereas specific aggressive behaviors in animals are typi- Here we will focus on questions 2–4. First we will review
cally highly stereotyped (wrestling, chasing, bites), aggression animal models and tests of aggression, because much of what
behaviors in humans can take many forms (e.g., physical versus is known about neurobiological mechanisms and development
verbal). Quantifying aggression in humans is a challenge. Early of aggression is based on animal models.
efforts to study aggression relied primarily on self-reports of
aggression that estimated general aggressive tendencies. These
measures generally correlate poorly with neuroendocrine 1.05.2 Animal Models and Tests of Aggression
measures such as testosterone (Archer, 2006). However,
a variety of tasks have been developed to induce aggressive The rigorous quantification of aggressive behaviors is an essen-
behavior under controlled conditions (e.g., point subtraction tial requirement for identifying underlying neuroendocrine
task). Aggressive behavior in these tasks is more closely linked mechanisms. The most common behavior test for quantifying
to testosterone levels, which are rapidly modulated during overt aggressive behaviors is the resident–intruder test
aggressive interactions (Carré et al., 2011). These tests also (Koolhaas et al., 2013). In a typical test the focal animal is
induce sympathetic nervous system activation (Gerra et al., housed in a home cage for at least 3 days. This allows the focal
2007). Increased arousal and anger are main components of animal to become familiar with home cage and adopt it as
what has been termed reactive aggression (Vitiello and Stoff, a territory. Next an unfamiliar intruder is introduced into the
1997), which is considered to be more impulsive yet can lead cage. The introduction of the intruder usually prompts the initi-
to sustained aggressive responses. It has been thought that reac- ation of anogenital sniffing by the resident, which results in the
tive aggression accounts for most societal problems related to detection of nonvolatile pheromones. Depending on the
aggression (Blair et al., 2006). Reactive aggression is usually species or strain, aggressive threats such as tail rattles or vocal-
associated with impulse control and low serotonergic signaling izations may precede overt aggressive behaviors (Nelson and
(Mehlman et al., 1994; Krakowski, 2003). In contrast instru- Chiavegatto, 2000). In rats and mice, attacks are usually
mental aggression occurs in the absence of physiological directed toward the flanks which have thick skin that prevent
arousal and is considered to be a more goal-oriented behavior. wounding. Each occurrence of these behavioral elements can
High-profile incidents (e.g., mass killings, genocides, or assassi- be measured in terms of frequency, as well as the onset and
nations) are likely to reflect instrumental mechanisms of termination of specific behaviors from both live observations
aggression. Attacking or otherwise bullying your neighbors to and video records. An advantage of the resident–intruder test
intimidate them is another example of instrumental aggres- is that it can be used in several different contexts, which can
sion. The controlled–instrumental subtype of aggression is allow the study of several forms of aggression.
thought to be regulated by higher cortical systems and less In many cases androgens either promote aggressive
dependent on the hypothalamic and limbic systems that are behavior in the resident–intruder test (Vandenbergh, 1971;
known to mediate impulsive aggression (Viding et al., 2007; Lima and Spinelli de Oliveira, 2014) or are secreted in response
and see below), and likely less dependent on hormones than to engaging in aggression (Marler et al., 2005). These contexts
other types of aggression. appear to correspond to reactive aggression in humans, which
Mental disorders such as intermittent explosive disorder is more likely to be linked with increases in testosterone (Carré
and posttraumatic stress disorder are associated with increased et al., 2011). Under these conditions residents typically reduce
autonomic arousal, which can contribute to sudden and aggression in response to the intruder engaging in submissive
uncontrolled reactive aggression (Blair et al., 2006; Viding postures. However, when adrenalectomized rats are tested as
et al., 2007). In contrast, individuals who are diagnosed with residents in this test, submissive displays by the intruder are
conduct disorder or antisocial personality disorder show ignored resulting in escalated aggression (Haller et al., 2001).
unusually low autonomic responsiveness (Viding et al., It is thought that aggression under these conditions more
2007), which can contribute to increased instrumental aggres- closely resembles the instrumental aggression associated with
sion by blunting the typical emotional responses (Raine, conditions such as conduct disorder (Haller and Kruk, 2006).
2002). Thus, exaggerated aggressive responses can be observed Aggression can also be observed in females using the resi-
in both high- and low-arousal states, with different biochem- dent–intruder test. Most often this is studied in lactating
ical, neuroanatomical, and neuroendocrine systems contrib- dams with male intruders. Maternal aggression under these
uting to behavior in each context. These results illustrate that contexts is considered defensive aggression (Lonstein and
more than one approach to studying neuroendocrine mecha- Gammie, 2002). It is characterized by short latency attacks of
nisms of aggression will be required. Animal model studies high intensity, mostly directed toward the head/neck region
in which testosterone has an important modulatory effect on of the opponent and usually without the introductory threat-
aggression will be more informative for reactive aggression ening behaviors typically displayed by male animals con-
whereas approaches examining aggression under low-arousal fronted with an intruder. In laboratory rats and mice, females
conditions may be more informative for understanding instru- are rarely aggressive toward other females. However, female–
mental aggression. female aggression is quite prevalent in other species. The Syrian
Aggressive behavior is a motivated behavior. In common hamster is a solitary species, and females are very aggressive
with other motivated behaviors, four types of questions arise: toward other females (Solomon et al., 2007; Staffend and
(1) What are the external factors that elicit aggressive behavior? Meisel, 2012). In the California mouse, males and females
(2) What neural circuitry mediates aggressive behaviors? form monogamous pairs, and females are also very aggressive
(3) How does aggression develop across ontogeny? (4) What toward other females (Davis and Marler, 2004; Silva et al.,
are the internal signals that mediate aggressive behaviors? 2010; Trainor et al., 2010b). These species allow for the ability
148 Hormones and the Development and Expression of Aggressive Behavior

to examine whether neuroendocrine mechanisms of aggression intruder can elicit different reactions from the resident. One
identified in males extend to females (Gutzler et al., 2010). way that this potential problem can be reduced, however, is
The resident–intruder test can be adapted to examine exag- to determine which group-housed intruders are not aggressive
gerated aggressive behavior following provocation. In this before the onset of the behavioral tests. An additional source of
protocol an intruder is first placed into the resident’s home variability can come from dominance relationships among
cage behind a protective barrier, which is then removed cage mates. For example, in the widely used C57Bl6 strain,
(Miczek et al., 2013). Elevated aggression using this protocol each cage of males has a dominance hierarchy. There are impor-
can be observed in both males (Fish et al., 1999) and females tant differences in behavior and brain function between the
(Potegal, 1991). This approach may provide insights into most dominant and most subordinate cage mate (Howerton
mechanisms contributing to exaggerated aggression following et al., 2008). For example, the lowest ranking cage mates had
perceived provocation. less corticotropin-releasing hormone 2 mRNA and estrogen
Under normal conditions, it is rare for male domestic mice receptor alpha in the bed nucleus of the stria terminalis than
and rats to direct aggression toward females. However among the most dominant cage mate (Greenberg et al., 2014). An
humans, women are often targets of male violence (Crowell easy solution for quantifying this variation is to run mice
and Burgess, 1996). Recently it was discovered that male rats through a ‘tube test,’ which provides an accurate estimate of
exposed to psychosocial stress during the peripubertal period dominance status.
were more aggressive with female breeding partners compared Experience is also important in the relationship between
to controls (Figure 1; Cordero et al., 2012). Intriguingly this hormones and aggressive behavior (Miczek and Fish, 2006).
effect was also observed in the male offspring of these pairs. Castration and hormone-replacement studies of males repre-
Female-directed aggression by males was reversed with mono- senting several species of reptiles, fish, and birds clearly demon-
amine oxidase inhibitor (Marquez et al., 2013). A recent strate reduced postcastration levels of aggression and
prospective study reported that individuals that were neglected restoration of aggression after testosterone treatment (e.g.,
as children were more likely to injure an adult intimate partner Crews and Moore, 1986; Wingfield et al., 1987). In mammals,
(Widom et al., 2014). However, the neurobiological mecha- the effects of androgens in supporting aggressive behavior
nisms contributing to this effect are unknown. Rodent peripu- depend largely on experience. Castrated mice and rats without
bertal stress models may facilitate the identification of prior aggressive experience rarely fight when tested with
physiological mechanisms contributing to intersex aggression. another male conspecific (Christie and Barfield, 1979). Individ-
Clearly, disrupting generational transmission of this form of uals that have won aggressive encounters before castration will
aggression is an important public health problem. exhibit aggression long after the testes have been removed (e.g.,
The resident–intruder test can provide insights into mecha- Christie and Barfield, 1979; DeBold and Miczek, 1981, 1984).
nisms of aggression in multiple contexts. However, several Winning aggressive encounters usually induces a temporary
factors need to be considered when interpreting the outcome surge in testosterone referred to as the challenge effect. The
of this test. For example, the behavior of the stimulus animal, combination of winning experience and testosterone has
or intruder, has an important impact on the behavior of the a powerful enhancing effect on aggressive behavior (Trainor
focal animal or resident. Previous experiences of the intruder et al., 2004; Fuxjager et al., 2011). Winning also increases the
can confound the results. Thus, a previously defeated or naïve expression of androgen receptors in motivational circuits
including the nucleus accumbens and ventral tegmental
area (Fuxjager et al., 2010). Interestingly, these motivational
* circuits are also altered by aggressive experience in female
50 rodents. Female Syrian hamsters also become very aggressive
Aggressive behavior (%)

Control with repeated aggressive encounters (Staffend and Meisel,


40
Stress 2012), and this behavioral change is mediated in part by gluta-
30 matergic synaptic plasticity in the nucleus accumbens (Been
et al., 2016).
20

10
1.05.3 Endocrine Signals’ and Receptors’
0 Contribution to Aggression
1.05.3.1 Steroid Hormones
0 10 20 30
Time (min) The role of steroid hormones has long been a focus of investi-
gators studying the neuroendocrine bases of aggressive
Figure 1 Peripubertal stress–induced abnormal aggressive behaviors behavior. Although testosterone is generally a key hormone
in resident–intruder tests performed in adulthood. Aggressive behaviors regulating aggression, either directly or by serving as a prohor-
of resident rats when intruders were similar to the resident rat in body
mone for DHT or estradiol, detailed experiments have demon-
weight (stress effect: F1, 29 ¼ 9.52, p ¼ 0.004; n ¼ 15–16 per group).
Reprinted from Marquez, C., Poirier, G.L., Cordero, M.I., Larsen, M.H.,
strated that the relationships between steroid hormones such as
Groner, A., Marquis, J., Magistretti, J., Trono, D., Sander, C., 2013. testosterone and aggression are complex. Other factors such as
Peripuberty stress leads to abnormal aggression, altered amygdala and steroid hormone synthesis in the brain, differential expression
orbitofrontal reactivity, and increased prefrontal MAOA gene expression. of steroid receptors, and environmental context have important
Transl. Psychiatry 3, e216 with permission from Nature. influences on the behavioral effects of circulating hormones.
Hormones and the Development and Expression of Aggressive Behavior 149

1.05.3.1.1 Androgens homogenization’ to reduce behavioral variability. This is an


The idea that hormones produced in the testes promote aggres- intriguing hypothesis to account for the disparate aggressive
sive behavior dates back to the mid-nineteenth century in the responses of males to different aggression-provoking stimuli,
classic experiments of Arnold Berthold (Quiring, 1944). In although further experiments are necessary to evaluate it fully.
these studies aggressive behavior in male chickens was abol- Although baseline testosterone concentrations regulate
ished by removal of testes and restored when donor testes aggression in many species, studies have identified several
were implanted. Subsequent castration and hormone-replace- species in which castration does not reduce male aggression
ment experiments have identified androgens as a key class of (Caldwell et al., 1984; Demas et al., 1999; Trainor and Marler,
hormones produced by the testes that facilitates aggression. 2001; Trainor et al., 2006a). Furthermore, dominance in more
Early studies focused on establishing correlations between complex social organizations may not be related to blood
plasma concentrations of androgens and aggression. Androgen concentrations of testosterone, especially in stable groups. For
levels are often increased during the breeding season when example, dominant dogs or squirrel monkeys can be castrated
males aggressively compete for breeding opportunities without affecting their position in the hierarchy (Dixson,
(Lincoln et al., 1972; Wingfield, 1984; Moore, 1986; Bales 1980). Also, treatment of low-ranking individuals with andro-
et al., 2006). Similarly, male aggressive behavior often increases gens does not change their status. The intuitive conclusion
at the time of puberty (Wallen et al., 1991; Pellis et al., 1997; from these results is that testosterone does not affect aggression
Delville et al., 2005), when testes mature and begin to secrete in these species. However, there are several ways in which
androgens. More definitive evidence that androgens facilitate aggressive behavior could be influenced by androgens indepen-
aggressive behavior comes from studies in which androgens dent of baseline testosterone concentrations.
are manipulated. Castration reduces male aggressive behavior Acute hormonal responses to the environment can have
in Syrian hamsters (Mesocricetus auratus) (Vandenbergh, different effects on behavior than the baseline hormonal state
1971), mice (Mus musculus) (Leshner and Moyer, 1975), pigs (Leshner, 1979). Winning aggressive encounters increases
(Sus scrofa) (Zamaratskaia et al., 2008), bulls (Bos taurus) male testosterone concentrations (‘challenge effect’) in birds
(Huxsoll et al., 1998), rats (Rattus rattus) (Albert et al., 1987), (Wingfield et al., 1990), fish (Oliveira et al., 2002), rodents
and red deer (Cervus elaphus) (Lincoln et al., 1972), whereas (Oyegible and Marler, 2005), nonhuman primates (Rose
testosterone replacement restores aggression in these species. et al., 1971), and humans (Mazur and Booth, 1998). Interest-
Similarly, elevated testosterone concentrations via implants ingly, the increase in testosterone after a winning experience
increase aggression in a variety of passerine birds (Wingfield is not observed unless the individual is in a familiar environ-
et al., 1987; Ketterson and & Nolan, 1992) and spiny lizards ment (Figure 2; Fuxjager et al., 2009). Initially, these rapid
(Marler and Moore, 1989). and transient responses were puzzling because it was thought
Strain differences certainly exist among house mice in the that the effects of steroid hormones such as testosterone
extent to which aggressive behaviors are expressed and in the required at least several hours for a behavioral effect to be
extent to which these aggressive behaviors are mediated by observed. It is now apparent, however, that these challenge
androgens. The effects of castration on predatory, shock- effects can influence behavior. Transient increases in testos-
induced, maternal, and isolation-induced aggression were terone may help crystallize the experience of winning an aggres-
studied in Swiss albino mice. Isolation-induced aggression sive encounter (Trainor et al., 2004). Several studies have
was generally reduced after castration; postgonadectomy treat- demonstrated that individuals that win aggressive encounters
ment with testosterone, 5a-dihydrotestosterone (DHT), or are more likely to win future encounters (Parmigiani and Brain,
estradiol restored this form of aggression (reviewed in Brain, 1983; Chase et al., 1994; Kudryavtseva, 2000), even when
1983). Castration increased intruder aggression toward variables such as intrinsic fighting ability are controlled
lactating females, and treatment with testosterone, DHT, or
estradiol reversed the elevated rate of aggressive responses in
this situation (Brain, 1983). These results imply that steroid Home cage Unfamiliar cage
2000
hormones do not merely ‘trigger’ aggression, but act to affect
testosterone (pg/ml)

the animal’s perception of and response to aggression- 1500


Post-encounter

provoking stimuli (Haug et al., 1986).


In another series of experiments (Whalen and Johnson, 1000
1987), male mice were pitted against either lactating females
or olfactory bulbectomized males (reviewed in Johnson and 500
Whalen, 1988). Gonad-intact males and castrated males
treated with testosterone attacked the olfactory bulbectomized 0
males, but did not attack lactating females. Untreated castrated 3 prior wins Handled controls
males tended to display tremendous individual differences in
Figure 2 Postencounter levels of testosterone in focal individuals.
aggressiveness, with some attacking either type of opponent,
The bars represent mean hormone levels and the error bars represent
others attacking only one type of opponent, and others display-
the standard error. The ) indicates significant differences between treat-
ing no attack behaviors (Johnson and Whalen, 1988). Because ment groups (p < 0.025). The data were log transformed for statistical
castration was associated with large individual variation in analysis. Reprinted from Fuxjager, M.J., Mast, G., Becker, E.A.,
aggressive responding, and because androgen treatment Marler, C.A., 2009. The ‘home advantage’ is necessary for a full winner
reduced that variation, Johnson and Whalen (1988) proposed effect and changes in post-encounter testosterone. Horm. Behav. 56,
that testicular steroid hormones act to induce ‘behavioral 214–219 with permission from Elsevier.
150 Hormones and the Development and Expression of Aggressive Behavior

(Oyegible and Marler, 2005). In addition, steroid hormones melatonin concentrations (Jasnow et al., 2002; Laredo et al.,
are now known to exert nongenomic effects which can occur 2014a). In hamsters, removal of the adrenal glands blocks
within seconds or minutes (Vasudevan and Pfaff, 2006). the aggression enhancing effects of melatonin (Figure 3).
Injections of testosterone can act within minutes to reduce This suggests that adrenal steroids are critical for sustaining
anxiety-like behavior in mice (Aikey et al., 2002), and an acute aggression outside of the breeding season when testosterone
injection of estradiol (a testosterone metabolite) can increase concentrations are low. However, in female hamsters, mela-
aggressive behavior in Peromyscus within 15 min (Nelson and tonin appears to work directly on adrenal function to elevate
Trainor, 2007; Trainor et al., 2007a). Testosterone can be con- DHEA. Short-day female hamsters are more aggressive than
verted to estradiol within the brain, a conversion mediated by their long-day counterparts; long-day hamsters provided with
the aromatase enzyme. Environmental factors can rapidly short-day melatonin doses also displayed increased aggression
convert testosterone to estrogens in the brain and affect aggres- and elevated DHEA concentrations (Rendon et al., 2015).
sive behaviors in a number of contexts (reviewed in Laredo Furthermore, melatonin increased DHEA secretion from
et al., 2014b). cultured adrenal glands. As noted above, testosterone some-
Steroid synthesis in the brain is not limited to the conver- times exerts behavioral effects indirectly through its conversion
sion of androgens to estrogens by the aromatase enzyme. to estrogens.
Many of the enzymes required for de novo steroid synthesis Several studies of human aggression in which psychological
have been identified in the brain (Baulieu and Robel, 1990; rating scales were used to quantify levels of aggressiveness or
Young et al., 1996; Soma, 2006), raising the possibility that hostility reported no relationship between blood or saliva
the brain may be producing androgens independently of the androgen concentrations and aggressiveness (Doering et al.,
testes. Dehydroepiandrosterone (DHEA) is produced in the 1975; Monti et al., 1977; Persky et al., 1977). However, rela-
adrenal gland but requires only two metabolic steps to convert tionships between blood testosterone concentrations and
to testosterone (Demas et al., 2007). Studies in hamsters behavior have been reported among aggressive, violent, and
(Demas et al., 2004; Figure 3) and song sparrows (Soma antisocial individuals, especially those incarcerated in prison
et al., 2002) suggest that adrenal steroids may promote aggres- (Kreuz and Rose, 1972; Ehrenkranz et al., 1974). Prison
sive behavior, especially under environmental conditions in inmates with high circulating testosterone concentrations,
which gonadal testosterone secretion is low. Most studied usually defined as the top 5% or 10% of the normal distribu-
rodents have elevated aggression levels under short-day photo- tion, had committed violent crimes (Ehrenkranz et al., 1974;
periods (Jasnow et al., 2000; Trainor et al., 2007b, 2008), an Dabbs et al., 1987, 1988), were more unruly in prison, and
effect that is mediated by extended periods of elevated were judged more harshly by their parole boards (Dabbs
et al., 1987, 1988). High testosterone concentrations have
also been associated with male juvenile delinquency (Olweus,
18 1983). Although some studies of criminal populations show
no association between plasma testosterone and violent
16
* behavior (for example, Matthews, 1979), the consensus is
that violence among prison inmates and blood androgen
14 concentrations are positively correlated. A similar relationship
Total duration of attacks (s)

was observed among female prison inmates (Dabbs and


12 Hargrove, 1997).
Two related hypotheses have been proposed to explain the
10
association between high androgen concentrations and human
antisocial behavior as observed in delinquent or criminal pop-
8
ulations: (1) androgens directly mediate the antisocial
6 activities and (2) androgens promote a constellation of traits,
including social dominance, competitiveness, and thrill-
4 seeking, that may be expressed either as antisocial or as proso-
cial behavior depending upon the individual’s resources and
2 background. To distinguish between these two possibilities,
a large sample of 4462 US military veterans was examined
0 beginning in 1985. Analyses of their psychological profiles
Sham ADx and saliva concentrations of testosterone suggested that andro-
Saline Melatonin gens directly mediate antisocial behavior in human males,
although socioeconomic status has a small moderating effect
Figure 3 Mean (SEM) total duration of attacks (s) in hamsters that (Dabbs and Morris, 1990).
received bilateral adrenalectomies (ADx) or sham operations (Sham) Few studies have addressed the role of androgens in aggres-
and subsequently treated with either melatonin or control (Saline) injec-
sive behavior in women; no consistent correlation between
tions. Significant differences between pairwise means are indicated by
an asterisk (*) if p < 0.05. Reprinted from Demas, G.E., Polacek, K.M.,
androgen concentrations and aggressive behavior has been
Durazzo, A., Jasnow, A.M., 2004. Adrenal hormones mediate reported for women (Persky et al., 1977; Dabbs et al., 1988;
melatonin-induced increases in aggression in male Siberian hamsters Dabbs and Hargrove, 1997). However, subtle effects of andro-
(Phodopus sungorus). Horm. Behav. 46:582–591 with permission from gens may influence aggression in women. Saliva testosterone
Elsevier. concentrations did not differ between female prison inmates
Hormones and the Development and Expression of Aggressive Behavior 151

and female college students. But further analyses discovered concentrations. Corticosterone acts rapidly to increase aggres-
that testosterone concentrations were highest in women pris- sion in rats (Mikics et al., 2004), hamsters (Hayden-Hixson
oners convicted of unprovoked violent crimes and lowest in and Ferris, 1991), and mice (Poole and Brain, 1974). The
women convicted of ‘defensive’ violent crimes, such as killing increase in aggression due to acute elevated corticosterone is
abusive husbands (Dabbs et al., 1988). particularly salient in challenge-situations, for example, when
confronted with unfamiliar opponents or other novel situa-
1.05.3.1.2 Estrogens tions (Mikics et al., 2007). In rough-skinned newts (Taricha
Often considered to be primarily a female class of hormones, granulosa) corticosterone acts rapidly to promote mating
estrogens have important effects on many male behaviors behavior (Moore and Miller, 1984), an effect that has been
including aggression. In most species that have been examined, linked to nongenomic hormone action (Orchinik et al.,
estrogens increase aggressive behavior. Blocking estrogen 1991). This raises the possibility that the effects of chronic
production with an aromatase inhibitor reduces aggression in elevation of glucocorticoids on aggressive behavior are medi-
Japanese quail (Schlinger and Callard, 1990) and song spar- ated by changes in gene expression (via activation of mineralo-
rows (Soma et al., 2000), whereas aromatase knockout mice corticoid and glucocorticoid receptors), whereas the effects of
display low aggression levels in resident–intruder tests (Toda a transient increases in glucocorticoids are mediated by nonge-
et al., 2001; Matsumoto et al., 2003). In CFW and CF-1 strains nomic responses of membrane receptors.
of mice, the negative effect of castration on aggression can be
reversed by treatment with estradiol (Simon and Whalen,
1.05.3.2 Anabolic Steroid Abuse and Aggression
1986). The effects of estrogens on aggression can be rapid in
both mammals and birds (reviewed in Heimovics et al., 2015). The use of anabolic steroid hormones (such as testosterone) as
Estrogens can bind to one of at least two estrogen receptor performance-enhancing drugs has become a high-profile topic
(ER) subtypes, a and b. Most of what is known about the effects in the news media, especially as several elite athletes have been
of these receptors on aggression comes from a series of studies disqualified or stripped of titles or medals. Anabolic steroids
on knockout mice. Male mice with targeted disruption of ERa are used because they stimulate the growth and development
display reduced aggression when tested with other males in of muscle tissue. Although anabolic steroids have certain ther-
a number of testing situations (Ogawa et al., 1997; Scordalakes apeutic uses (Ferrando and Wolfe, 2007), when used in excess
and Rissman, 2003, 2004). Curiously, male ERa knockout or abused they have many negative side effects such as infer-
mice are more likely than wild-type mice to attack female tility, suppressed immune function, and increased risk of
intruders. In male CD-1 mice, levels of aggression directed cardiovascular, liver, and kidney disease (Bahrke and Yesalis,
toward other males are positively correlated with the number 2004; Bonetti et al., 2008). In addition there is accumulating
of ERa immunopositive cells in the LAS, BNST, and AHA evidence that anabolic steroid abuse has adverse psychological
(Trainor et al., 2006a). The deletion of ERb is generally associ- effects, including aggression. Surveys have indicated that
ated with increased aggression (Ogawa et al., 1999; Nomura anabolic steroid abusers are more likely to engage in verbal
et al., 2006), although this effect appears to be context depen- aggression, fighting, violence toward women, and risk-taking
dent (Nomura et al., 2002). Deletion of both receptors is asso- behaviors (Choi and Pope, 1994; Pope and Katz, 1994;
ciated with increased male aggression (Ogawa et al., 2000). In Galligani et al., 1996). However, there are some inconsistencies
these knockout studies, the effects of ERa and ERb could be across studies, and study participants may not be forthcoming
organizational, activational, or both. Studies using ER-specific about their usage, especially because anabolic steroids are out-
ligands in adult animals have suggested that the directional lawed in many countries (McGinnis, 2004). To address
effects of these ERa and ERb may occur primarily during devel- whether many commonly abused anabolic steroids exert effects
opment (see Section 1.05.7.1), although additional studies are on aggression, researchers have developed animal model
needed to test this hypothesis. systems in which dosages and environmental variables are
controlled.
1.05.3.1.3 Glucocorticoids Studies in hamsters and rats have established that many
The effects of glucocorticoids on aggression are also variable, commonly abused anabolic steroids can influence aggressive
although the mechanistic basis for this is poorly understood. behaviors, although in some cases these effects depend on
Generally, chronic elevations in glucocorticoid concentrations testing conditions. Methyltestosterone, but not stanozolol
(usually associated with stress) inhibit aggressive behavior (a nonaromatizable androgen), increases male aggressive
(Leshner et al., 1980; Maestripieri et al., 1991; Summers behavior in castrated male rats compared to castrated rats
et al., 2005), whereas chronic deficiencies in glucocorticoid receiving oil injections (Clark and Barber, 1994). Other studies
secretion are associated with increased aggression (Haller in rats (Farrell and McGinnis, 2004) and mice (Martinez-
et al., 2001, 2004). Elevated baseline glucocorticoid concentra- Sanchis et al., 1996) have also reported that stanozolol does
tions inhibit testosterone secretion (Viau, 2002), increase sensi- not increase aggression (Figure 4). Nandrolone is another
tivity to serotonin (Meijer and de Kloet, 1998), and increase commonly abused androgen, but its effect on aggressive
glutamate neurotransmission in frontal cortex (Moghaddam, behavior in rodents is variable. One study reported that nan-
2002). All of these physiological responses could contribute drolone administration to male rats increased aggression
to reduced aggressive behavior and appear to do so in animals (Long et al., 1996), whereas two other studies observed that
subjected to chronic social defeat (Haller, 2014). However, the nandrolone administration had no effect on aggressive
immediate effect of a transient increase in glucocorticoids on behavior in rats (McGinnis et al., 2002a,b). It has been hypoth-
aggression is quite different than a chronic increase in baseline esized that the effects of nandrolone on aggression may depend
152 Hormones and the Development and Expression of Aggressive Behavior

(a) a forebrain region that facilitates male aggressive behavior.


20
There is also evidence that anabolic androgens can downregu-
Intact opponent
late GABAergic neurotransmission, thereby facilitating aggres-

Composite aggression score


15 Castrated opponent sive behavior (Henderson et al., 2006). The emerging picture
then is that anabolic androgens facilitate aggressive behaviors
by affecting several biochemical pathways and these neuro-
10 chemical changes are influenced by environmental factors.
Androgenic anabolic use is also a problem of adolescence.
According to the 2005 Monitoring the Future Survey,
5 over 3% of 12th-grade males in the United States report
* * having used anabolic androgens (http://www.nida.nih.gov/
ResearchReports/Steroids/AnabolicSteroids.html). Anecdot-
0
Test Nand Stan ally, anabolic steroid use by adolescents is associated with irri-
Control
tability and heightened aggression, but a causal link has not
(b)
20 been established. On the other hand, animal studies have
Home cage
provided compelling evidence for anabolic steroid-induced
Composite aggression score

Opponent cage aggression in adolescent males. Adolescent male hamsters


15 Neutral cage treated chronically with an anabolic steroid cocktail have
shorter attack latencies and a greater number of attacks and
* bites toward a male intruder compared with untreated males
10 (Melloni et al., 1997; Harrison et al., 2000), and anabolic
steroid-induced increases in aggression are more robust in
adolescent than in adults (Salas-Ramirez et al., 2008). These
5
effects are all the more striking considering that male–male
* * aggression in hamsters is not under strong activational influ-
* ences by endogenous testosterone. Similarly, a mild provoca-
0
Test Nand Stan Control tion (tail pinch) produces a persistent increase in aggression
in adolescent male rats treated with anabolic androgens,
Figure 4 Social discrimination is seen in aggression toward a gona- including aggression toward females (Cunningham and
dally intact versus a castrated male opponent (a). Environmental McGinnis, 2007). Importantly, adolescent exposure to
discrimination is depicted in (b). Aggression is shown toward a gona- anabolic androgens causes long-lasting changes in agonistic
dally intact male in the anabolic androgenic steroid (AAS) males’ home behavior (Grimes and Melloni, 2006), neurotransmitter
cage, the opponents’ home cage, or a neutral cage. The rats were
systems (Grimes and Melloni, 2006; Ricci et al., 2007), and
treated with testosterone (Test), nandrolone (Nand), Stanozolol (Stan),
synaptic organization (Cunningham and McGinnis, 2007)
or vehicle (Control). )p < 0.05. Reprinted from Farrell, S.F., McGinnis,
M.Y., 2003. Effects of pubertal anabolic-androgenic steroid (AAS) that persist even after the period of drug exposure. In light of
administration on reproductive and aggressive behaviors in male rats. the evidence that endogenous testosterone organizes aggressive
Behav. Neurosci. 117, 904–911, with permission from American behavior during puberty and adolescence, it seems likely that
Psychological Association. anabolic steroid use during adolescence would result in larger
magnitude or more enduring effects on the brain and aggres-
sive behavior than use in adulthood. Taken together, the
on experience or testing conditions (McGinnis, 2004). For evidence suggests significant aggression-promoting effects on
example, tail pinching can be used as a form of physical prov- aggressive behaviors (e.g., Oberlander and Henderson, 2012).
ocation (Miczek et al., 2004), and this can exaggerate the effects
of anabolic steroids on aggression (McGinnis et al., 2002a).
A further complicating factor is that many abusers of anabolic 1.05.4 Brain Regions Contributing to Aggression
androgens use more than one steroid simultaneously, a practice
also known as stacking (Trenton and Currier, 2005). Studies in A complicating factor in studying neural circuits affecting
hamsters show that activation of 5-HT1B receptors blocks the aggression is that many brain nuclei that regulate aggression
effects of an androgenic cocktail (testosterone cypionate, nor- also affect other social behaviors. For example, in rodents the
testosterone, and dihydroxytestosterone) on aggression in medial amygdala (MEA) is activated during both aggression
a resident–intruder test (Grimes and Melloni, 2005). Although and reproductive behavior (Kollack-Walker and Newman,
stanozolol treatment alone reduced aggressive behavior in 1995; Choi et al., 2005). Although there are sometimes subtle
male rats, stanozolol treatment with testosterone increased anatomical differences in activity as measured with immediate
aggression in aggression tests preceded by a tail pinch (Wesson early genes (Holt and Newman, 2004), more detailed analyses
and McGinnis, 2006). Anabolic androgens appear to affect will be necessary to sort out whether different cell types are acti-
aggressive behaviors by working at several biochemical and vated in different contexts, or whether different cellular
neurobiological levels. In hamsters, anabolic androgen responses result in different behavioral responses. Despite
administration increases arginine vasopressin (AVP) immuno- this uncertainty, it is clear that a common set of hypothalamic
reactivity (Grimes et al., 2007) and baseline c-fos immunoreac- and limbic brain areas play a role in regulating some form of
tivity (Ricci et al., 2007) in the anterior hypothalamus, aggressive behavior in a variety of species. Homologous brain
Hormones and the Development and Expression of Aggressive Behavior 153

structures appear to regulate social behaviors, including aggres- contexts (Swanson, 2000). The components of this network
sion, in both mammalian and nonmammalian species (Gregg have been identified over the years through increasingly precise
and Siegel, 2001; Crews, 2003; Goodson, 2005). Aggression is experimental methods. The involvement of the LAS, BNST,
a primitive, yet highly conserved vertebrate behavior, and it is AHA, VMH, and MEA in aggression was first established
reasonable to expect that the molecular mechanisms under- through lesion studies (Miczek et al., 1974; Annen and Fujita,
lying aggression are also similar (and possibly ancient) among 1985; Kruk, 1991; Figure 5(a)). Next, the use of electrical stim-
vertebrates. Species-specific features of aggression are likely the ulation of specific brain regions was found to induce aggressive
result of adaptive ‘co-opting’ of novel molecules as modulators behavior in regions such as the AHA (Kruk et al., 1984; Kruk,
that are ‘added to’ the primary neural circuits. These findings 2014). Both lesions and electrical stimulation can activate
support the hypothesis that at least some neurobiological fibers of passage, and so local infusion of GABAergic or gluta-
and neurochemical mechanisms governing the motivation to matergic agonists and antagonists were used to examine local
engage in aggressive behavior are evolutionarily conserved cell bodies (Haller et al., 1989; Roeling et al., 1993). Optoge-
(Scott, 1975). Thus, results from studies of aggression in netic stimulation of the ventrolateral VMH was also found to
rodents and other nonprimate species should provide insight increase aggression in male mice (Lin et al., 2011). The applica-
on the motivational circuits regulating aggression in other tions of immunostaining for immediate early gene products
species, including humans. such as c-fos largely confirmed that engaging in aggressive
behavior increases the activity of neurons in the LAS, BNST,
AHA, and MEA in both males (Kollack-Walker and Newman,
1.05.4.1 Studies in Rodents
1995; Delville et al., 2000) and females (Davis and Marler,
In rodents, sensory input from the olfactory bulbs (DaVanzo 2004; Hasen and Gammie, 2005; Gammie et al., 2007).
et al., 1983) is sent to the MEA and then relayed to the bed More recently, immunostaining for phosphorylated proteins
nucleus of the stria terminalis (BNST), medial preoptic area such as the transcription factor CREB has proved to be a useful
(MPOA), lateral septum (LAS), anterior hypothalamus complementary method for identifying changes in cellular
(AHA), ventral medial hypothalamus (VMH), and the peria- activity in the brain following aggressive encounters (Trainor
queductal gray region (PAG) (Wood and Newman, 1995; et al., 2010b; Heimovics et al., 2012). An important advantage
Delville et al., 2000; Figure 5(a)). This pathway has been of immediate early gene studies is the ability to identify cell
referred to as a social behavior network (Newman, 1999; types through multilabeling. For example subordinate male
Goodson, 2005), and it has also been hypothesized that mice had more AVP neurons in the caudal paraventricular
different subnuclei are more active in different contexts. For nucleus coexpressing fos compared to dominant mice (Ho
example, the posteroventral MEA and dorsomedial VMH are et al., 2010). Similarly both male and female California mice
thought to be more important for regulating aggression in had more AVP/c-fos-expressing cells in the caudal PVN
defensive contexts, whereas the posterodorsal MEA and ventro- following a resident–intruder test if they had previously experi-
lateral VMH are thought to be more important in offensive enced social defeat (Steinman et al., 2015). There is growing

(a) (b) Visual and auditory


social cues
Stress
Olfactory
social cues

OFC
Hippocampus
LAS
Olfactory OFC
bulb BNST BNST
PAG
AHA MeA
PVN

+
AHA PAG
MeA

Aggressive Aggressive
behavior behavior

Figure 5 Neuroanatomical pathways of aggression in rodents and nonhuman primates. In rodents (a), information from the olfactory bulb is pro-
cessed by the medial amygdala (MEA) and sent to the lateral septum (LAS), bed nucleus of the stria terminalis (BNST), and anterior hypothalamic
area (AHA). These brain areas are thought to prompt the periaqueductal gray (PAG) into promoting species-specific aggressive behaviors. Stress can
inhibit aggression via inhibitory inputs from the orbital frontal cortex (OFC), the hippocampus, and the paraventricular nucleus (PVN). In nonhuman
primates (b), aggression is typically evoked by vocal or visual signals. Activation of the MEA is thought to result in activation of the BNST and AHA,
which in turn activate the PAG. In general, the OFC appears to be important for the interpretation of social cues, and inhibitory inputs from the OFC
might inhibit aggression by reducing responsiveness in the amygdala. Thick arrows represent inputs and outputs to and from the brain; thin arrows
represent connections within the brain; dotted lines represent inhibitory connections. Reprinted from Nelson, R.J., Trainor, B.C., 2007 Neural mecha-
nisms of aggression. Nat. Rev. Neurosci. 8, 536–546 with permission from Nature.
154 Hormones and the Development and Expression of Aggressive Behavior

interest in understanding how specific cells types within the for these conflicting results is that studies that reported
social behavior network are activated under different social increased aggression reintroduced lesioned monkeys into
contexts. groups, whereas studies that reported decreased aggression
The development of viral gene transfer techniques has created tested monkeys in groups of two (Emery et al., 2001), which
new approaches for studying neural circuits of aggressive might be less threatening. Lesions of the OFC are generally
behavior. So far most studies have focused on the MEA and associated with reduced affiliative behavior such as grooming
VMH in mice. In one study aromatase-expressing neurons in or close contact (Butter et al., 1970; Machado and Bachevalier,
the MEA were inhibited using a designer receptor exclusively acti- 2006), whereas their effects on aggressive behavior depend on
vated by designer drugs (DREADD) (Unger et al., 2015). Inhibi- context. For example, OFC lesions produce increased aggres-
tion of MEA aromatase neurons in males increased the latency to sion in dominant, but not subordinate males (Machado and
engage in aggression without affecting the overall frequency of Bachevalier, 2006). In a different study, OFC lesions in domi-
attacks or threats. In females inhibition of MEA aromatase nant animals led to an initial increase in aggression that disap-
neurons increased the latency and decreased the frequency of peared after several months (Butter and Snyder, 1972). In
maternal aggression. The role of GABAergic and glutamatergic general, it seems that the OFC is important for the interpreta-
neurons in the MEA has been examined using optical stimula- tion of social cues and contributes to appropriate behavioral
tion. Activation of GABAergic neurons in the MEA was found responses in complex social situations.
to induce male–male aggressive behaviors while activation of A creative study used positron emission tomographic
glutamatergic neurons inhibited aggression (Hong et al., 2014). imaging to examine brain activity in rhesus macaques in the
In the ventrolateral VMH infusion of AAV expressing small context of mate competition (Rilling et al., 2004). Dominant
hairpin RNAs targeting ERa reduced male–male aggressive male monkeys witnessed a potential sexual interaction between
behavior (Sano et al., 2013). Intriguingly optical stimulation of a female they had been previously paired with and a subordi-
ERa neurons increased male aggression directed toward both nate male. This mate competition ‘challenge’ condition was
male and female intruders (Lee et al., 2014). Overall the results designed to model aspects of jealousy in humans. Males
of experiments using modern genetic approaches are largely exposed to this ‘challenge’ condition showed increased activa-
consistent with original reports while adding a new layer of tion in the right amygdala and right superior temporal sulcus
understanding of how specific cell types control behavior. compared to males exposed to the control condition in which
the subordinate male was absent. Interestingly, similar results
were observed in a functional MRI study on human partici-
1.05.4.2 Nonhuman Primates and Humans
pants. Brain activation was increased in the amygdala and
As in rodents, the hypothalamus seems to play a key role in hypothalamus when men read sentences depicting sexual infi-
regulating aggression in nonhuman primates (Figure 5(b)). delity compared to neutral sentences, whereas women showed
Electrical stimulation of the ventromedial hypothalamus increased activation in the posterior superior temporal sulcus
increases vocal threats and piloerection in male marmosets, in the same comparison (Takahashi et al., 2006). Other studies
Callithrix jacchus (Lipp and Hunsperger, 1978). Similarly, suggest that the superior temporal sulcus is activated when
lesions of the AHA and POA reduce vocal threats toward an assessing deception (Calarge et al., 2003), trustworthiness
intruder in male C. jacchus (Dixson and Lloyd, 1988). In rhesus (Winston et al., 2002), and violation of social norms (Greene
monkeys (Macaca mulatta), electrical stimulation of the AHA, et al., 2001). Thus, it appears that there are at least qualitative
BNST, or POA increases the frequency of aggressive vocaliza- similarities between human and nonhuman primate circuitries
tions (Robinson, 1967) and aggression toward subordinate that function during mate competition.
males (Alexander and Perachio, 1973). A more direct link between brain activation in humans and
Other studies have focused on the amygdala and orbital aggression was observed in imaging studies that reported an
frontal cortex (OFC). Lesions of the amygdala either increase inverse relationship between average baseline activity in the
(Machado and Bachevalier, 2006) or decrease intermale aggres- frontal cortex and measures of reactive aggression (Raine
sion (Emery et al., 2001) in rhesus monkeys. One explanation et al., 1994; Volkow et al., 1995; Soderstrom et al., 2000;

Right Left Right Left


1.2 1.2
Med-temp/whole brain
Prefrontal/whole brain

1.15 1.15
1.1 1.1
1.05 1.05
1 1
0.95 0.95
0.9 0.9
0.85 0.85
Nml Vlt Nml Vlt Nml Vlt Nml Vlt

Figure 6 PET scan indicating brain changes in violent people. Individual values for relative metabolism in right and left prefrontal and medial
temporal cortex of adult control volunteers (Nml) and violent patients (Vlt). Reprinted from Volkow, N.D., Tancredi, L.R., Grant, C., Gillespie, H.,
Valentine, A., Mullani, N., Wang, G.J., Hollister, L., 1995. Brain glucose metabolism in violent psychiatry patients: a preliminary study. Psychiatry
Res. 61, 243–253, with permission from Elsevier.
Hormones and the Development and Expression of Aggressive Behavior 155

Soloff et al., 2003; Figure 6). The frontal cortex provides inhib-
itory inputs to circuits in the hypothalamus and amygdala that
might promote aggression (Davidson et al., 2000), although
the role of these brain areas remains less well-established in
humans than in other animals. In one study, individuals that
had been diagnosed with intermittent explosive disorder
increased activation in the amygdala in response to angry faces
when compared to control participants, and amygdala activa-
tion across both groups was positively correlated with scores
on the Lifetime History of Aggression (LHA) scale (Coccaro
et al., 2007). Insights into brain areas that affect human aggres-
sive behavior also come from observing the behavioral effects
of brain injuries. Many studies have reported a link between
brain damage to the frontal cortex and increased aggressive
behavior (Grafman et al., 1996; Anderson et al., 1999). Brain
injury rarely causes selective damage to the hypothalamus or
amygdala. However, during a grim period in the mid-twentieth
century, electrolytic lesions of these brain regions were used to
treat what was deemed ‘excessive aggression’ (Heimburger
et al., 1966; Sasano et al., 1998). Although lesions of the hypo- Figure 7 Inverse correlation between the binding potential in the
thalamus and amygdala were reported to inhibit aggression, orbital prefrontal cortex and lifetime aggression score as assessed by
these conclusions are limited. Measurements of behavior in the Brown–Goodwin Scale. Open circles are males, filled circles are
these studies were usually crude and fail to account for the females. R ¼ 0.53, p ¼ 0.007 for the combined data. Reprinted from
complexities of human behavior (Blair, 2004; Cherek et al., Parsey, R.V., Oquendo, M.A., Simpson, N.R., Ogden, R.T., Van Heer-
2006; Scarpa and Raine, 2006; Trainor et al., 2006b). Addition- tum, R., Arango, V., Mann, J.J., 2002. Effects of sex, age, and aggres-
ally, electrolytic lesions damage fibers of passage as well as the sive traits in men on brain serotonin 5-HT1A receptor binding potential
target nuclei, and damage to the hypothalamus and amygdala measured by PET using [C-11]WAY-100635. Brain Res. 954, 173–182
with permission from Elsevier.
affect general arousal (Tonkonogy and Geller, 1992), not just
aggression. An intriguing experimental approach was devel-
oped by Carré and colleagues to examine the effects of testos-
terone on neural activation in response to threat-related aggression (Winslow and Insel, 1991; DeVries et al., 1997;
stimuli (Goetz et al., 2014). Healthy men were given a gonado- Winslow et al., 2000) and that oxytocin receptors are abundant
tropin-releasing hormone antagonist, which normalized T in the amygdala (Insel, 1992; Jiménez et al., 2015).
concentrations to low baseline levels. Then men were Thus oxytocin might reduce human aggressive responses in
randomly assigned to be treated with T or placebo and tested some contexts, although this should be tested pharmacologi-
in a face-matching task while undergoing fMRI. Men treated cally. These studies show that combining biochemical manipu-
with T had greater increases in reactivity in the amygdala and lations with a realistic social context can allow investigators to
hypothalamus to angry faces compared to men treated with ethically test hypotheses that been developed using animal
placebo. Several studies have taken an integrative approach to models.
studying the neurobiological circuits that influence aggression.
Previous studies reported reduced activation of the prefrontal
cortex in patients who were rated highly for impulsive 1.05.5 Neurotransmitters, Hormones, and
aggression and also showed that selective serotonin reuptake Aggression
inhibitors (SSRIs) reduced ratings of aggression (Coccaro and
1.05.5.1 Serotonin
Kavoussi, 1997); the effect of SSRIs on prefrontal cortex
(PFC) activity was examined in patients who had been Serotonin is the neurotransmitter most closely associated with
diagnosed with borderline personality disorder (these patients regulation of aggressive behavior. Nonetheless, there is no
score highly on measures of impulsive aggression). Twelve simple relationship between serotonin levels and aggressive
weeks of SSRI treatment increased baseline activation in the behavior overall (Takahashi et al., 2011). More granular anal-
PFC, and PFC activation was negatively correlated with ratings yses have established both subtle and not so subtle relation-
of aggression (New et al., 2004). In addition, positron emission ships between serotonin and aggression. For example, it is
tomography (PET) imaging studies using a selective serotonin now generally accepted that 5-HT1A and 5-HT1B receptor inhi-
receptor type 1A (5-HT1A) antagonist showed that scores on bition can reduce aggression (Morrison and Melloni, 2014),
the Lifetime History of Aggression (LHA) test were negatively although sex differences in 5-HT1A receptor activation have
correlated with 5-HT1A binding in the amygdala and been observed (Joppa et al., 1997). Rats display rapidly elicited
PFC (Parsey et al., 2002; Figure 7). Intranasal administration aggressive responses when an intruder is introduced into the
of the neuropeptide hormone oxytocin to human participants home cage and treatment with the 5-HT1A receptor agonist
reduced activation of the amygdala in response to (8-OH-DPAT) decreases offensive aggressive behaviors in resi-
fear-inducing pictures (e.g., sharks, snakes) (Kirsch et al., dent males (De Boer et al., 1999; De Boer and Koolhaas,
2005). Studies in animals indicate that oxytocin can reduce 2005). However, these drugs have no effect on aggression
156 Hormones and the Development and Expression of Aggressive Behavior

when administered to intruders. Additionally, highly aggressive Androgens, either acting directly or via estrogenic metabo-
male mice with depleted CNS 5-HT ameliorate agonistic lites, tend to facilitate aggression, whereas increased CNS
responses after 8-OH-DPAT treatment suggesting further that 5-HT tends to inhibit aggression. Androgens interact with
the 5-HT1A receptors inhibit aggressive responses. 5-HT in several ways to influence aggression. For example, peri-
Generally, activation of the 5-HT system dampens aggres- natal exposure influences the expression and distribution of 5-
sion in animals and violent behavior in humans. Impulsivity HT receptor subtypes (Simon et al., 1998; Sumner and Fink,
and high aggressiveness are correlated with low cerebrospinal 1998; Cologer-Clifford et al., 1999). Either testosterone or
fluid concentrations of 5-HT metabolite, 5-HIAA, in humans estradiol elevates 5-HT2A receptor mRNA expression and
and nonhuman primates, and reduced 5-HT levels or turnover binding site densities in male rat brains (Ferrari et al., 1999).
in the brain of laboratory animals (reviewed in Lesch and Importantly, both androgens and estrogens modulate 5-HT1A
Merschdorf, 2000). Pharmacological strategies of increasing and 5-HT1B receptor agonist effects on murine aggression
5-HT levels such as the use of 5-HT precursors, 5-HT reuptake (Simon, 2002). Thus, sex steroid hormones and 5-HT interact
inhibitors, as well as 5-HT1A and 5-HT1B receptor agonists are on several levels to influence the likelihood of aggression.
able to reduce aggressive behavior in rodents (reviewed in Several other classical neurotransmitters have also been
Manuck et al., 2006). Nonetheless, in models of excessive, linked to aggression. The most commonly related neurotrans-
proactive aggression, 5-HT is unable to modulate the abnormal mitters associated with aggressive behaviors are discussed
behavioral responses (Haller, 2013). This suggests that seroto- below.
nergic mechanisms are more important for species-typical
aggressive behaviors rather than models of abnormal or violent
1.05.5.2 Arginine Vasopressin
aggressive behavior.
Genetic evidence for a role of 5-HT in aggression comes AVP is another hormone that plays a critical role in aggression
from mice missing specific genes that either directly or indi- and other social behaviors (Goodson and Bass, 2001; Ferris,
rectly affect 5-HT concentrations or metabolism. The 5-HT1B 2006). AVP produced in the anterior hypothalamus (AH) can
receptor is expressed in a variety of brain regions, including modulate aggression through its effects on AVP V1a receptors,
the basal ganglia, periaqueductal gray, hippocampus, lateral 5-HT, and the serotonin receptors 5-HT1A and 5-HT1B. Treat-
septum, and raphe nuclei, either presynaptically inhibiting ment of male hamsters with a V1A antagonist increases attack
5-HT release or as a heteroreceptor modulating the release of latencies and decreases frequency of biting in a standard
other neurotransmitters (Bibancos et al., 2007). Male mice resident–intruder test (Ferris et al., 2006). Interestingly, V1a
that lack functional expression of the 5-HT1B receptor gene antagonist infusions into the AH of female hamsters increased
(5-HT1B /) are more aggressive than wild-type mice (Saudou aggression (Gutzler et al., 2010). Somewhat consistent with
et al., 1994). Lactating female 5-HT1B / mice also attack unfa- this effect, V1a antagonist infused into the lateral ventricle
miliar male mice more rapidly and violently than wild-type also enhanced maternal aggression in rat dams (Nephew and
females (Ramboz et al., 1996). Notably, administration of Bridges, 2008). These findings indicate that the V1a receptor
the nonselective 5-HT1B agonist eltoprazine (one of the so- has very different effects on aggression in males and females.
called ‘serenics’) significantly reduces aggressive behavior in Microinjections of AVP into the AH in combination with
both 5-HT1B knockout mice and WT mice, presumably by 5-HT1A or 5-HT1B receptor agonists revealed that only the
affecting 5-HT1A receptors (Ramboz et al., 1996). Although 5-HT1A receptor activation inhibited AVP-facilitated
the 5-HT1B receptor contributes to aggression, these results aggression (Ferris, 2006). 5-HT neurons project into the AH,
suggest that the 5-HT1B receptor subtype is not the sole 5-HT and 5-HT appears to inhibit AVP-facilitated offensive
receptor modulating aggressive behavior. Specifically, 5-HT1A aggression by activating 5-HT1A receptors (Ferris, 2006).
receptor activation, which is also induced by eltoprazine, can Several drugs that affect both AVP and the 5-HT systems
also influence aggressive behaviors. Although both 5-HT1A reportedly reduce aggression. For example, systemic treatment
and 5-HT1B receptors control the tone of 5-HT system, it seems of humans with nicotine increases blood AVP concentrations
likely that these two receptors contribute differently in partic- and central 5-HT release while reducing aggression (reviewed
ular brain areas modulating the postsynaptic 5-HT inhibitory in Morrison and Melloni, 2014). It appears that development
effects on aggression (Bibancos et al., 2007). The role of other can impact the effects of V1a receptors on aggression. V1A
5-HT receptor subtypes on aggression remains unspecified, but knockout mice have normal levels of aggression after
these receptors likely play a minor role (Montoya et al., 2012). isolation, whereas isolation of V1B knockout mice reduces
Long-term treatment with selective serotonin reuptake aggression (Wersinger et al., 2002; Morrison and Melloni,
inhibitors (SSRIs) reduces aggression in many species ranging 2014). This suggests that genetic deletion of V1a results in
from fish to primates (reviewed in Morrison and Melloni, a compensatory developmental effect in aggression circuits,
2014). Acute SSRI treatment blocks the pro-aggressive effects possibly a greater role for V1b receptor.
of several agents such as synthetic anabolic androgenic steroids
(AAS), testosterone, alcohol, and apomorphine (Morrison and
1.05.5.3 Monoamine Oxidase
Melloni, 2014). Taken together, the evidence suggests that sero-
tonin in the CNS tends to decrease aggressive behaviors Metabolic enzymes such as monoamine oxidase A (MAOA)
(Takahashi and Miczek, 2014). However, a recent meta- also influence aggression because they function to alter neuro-
analysis of the serotonin–aggression relationship in humans transmitter concentrations. MAOA is predominantly located in
only revealed a small effect size between central 5-HT and catecholaminergic neurons in the brain, but MAOA catalyzes
aggression, hostility, and anger (Duke et al., 2013). with high affinity the oxidative deamination of 5-HT,
Hormones and the Development and Expression of Aggressive Behavior 157

norepinephrine, and dopamine (Shih et al., 1999). Although aggressive interactions (Nelson et al., 1995). However, cas-
MAOA deficiency due to a point mutation in its coding gene trated nNOS/ mice indicate that testosterone is necessary, if
is correlated with impulsive aggression in several males from not sufficient, to maintain elevated aggression in these
a single Dutch family (Brunner et al., 1993), humans treated knockout mice (Kriegsfeld et al., 1997). Androgen replacement
with pharmacological MAO inhibitors for depression generally therapy restored the elevated levels of aggression to precastra-
display no change in impulsivity or aggression (Manuck et al., tion levels in both nNOS/ and wild-type mice.
2006; Buckholtz and Meyer-Lindenberg, 2008). Substantial Serotonin function was hypothesized to be disrupted in the
work has related low MAOA expression with genetic factors aggressive nNOS/ mice because of the inverse relation of
to influence aggressive behaviors in humans (reviewed in 5-HT system activity and aggression. Serotonin metabolism
Dorfman et al., 2014). was reduced in several brain regions involved in aggression
MAOA knockout mice display markedly increased aggres- (Chiavegatto et al., 2001). Changes in 5-HT turnover were
sive behaviors (Cases et al., 1995; Godar et al., 2011). Ablation due to increased concentrations of 5-HT with no changes in
of the MAOA gene in mice leads to high levels of offensive its metabolite in most brain regions studied (Figure 8). The
aggression despite elevated 5-HT concentrations (Cases et al., disturbed neurochemical profile appears specific to the 5-HT
1995); the metabolic disturbances caused by MAOA deficiency system, because norepinephrine, dopamine, and metabolites
throughout life likely account for the effects on aggression. were generally unaffected. As noted, monoamine oxidase has
Notably, the elevated aggression in humans and mice with been implicated in aggression; however, the relatively normal
MAOA gene disruption mostly affects males (Manuck et al., norepinephrine and dopamine values suggest that it is unlikely
2000; Buckholtz and Meyer-Lindenberg, 2008). MAOA activity that alterations in monoamine oxidase account for the 5-HT
is directly regulated by estrogen (Chakravorty and Halbreich, abnormalities in the nNOS knockout mice (Chiavegatto
1997), which may be mediated by hormone receptor response et al., 2001).
elements present in the MAOA promoter (Ou et al., 2006). In Gonadal hormones directly influence the expression of
humans, environmental risk factors interact with the MAOA nNOS in many regions within the hypothalamus and limbic
to produce elevated risk for aggression (Kim-Cohen et al., system (Panzica et al., 2006). The effects of sex steroid
2006). Interestingly, low MAOA gene expression interacts hormones have primarily been achieved after medium- or
with prenatal cigarette exposure to result in elevated conduct long-term treatments. However, significant changes occur in
disorder diagnoses later in life (Wakschlag et al., 2010). particular physiological conditions, for example, during the
estrous cycle. Changes are not uniform throughout the brain,
but vary in specific directions in different populations of
1.05.5.4 Nitric Oxide
neurons (Panzica et al., 2006). Estradiol treatment seems
Nitric oxide (NO) was initially identified as an endogenous to increase nNOS activity in the ventrolateral nucleus of
regulator of blood vessel tone, but is now recognized as guinea pigs (Warembourg et al., 1999) and in PVN (Sanchez
a neurotransmitter in both the central and the peripheral et al., 1998) and MPA (Okamura et al., 1994) of rats. These
nervous systems (Baranano and Snyder, 2001). Male mice discrepancies in the effects of gonadal hormones on nNOS
with targeted deletion of the gene encoding the neuronal
version of NOS (nNOS/ or NOS1/) displayed three to
four times more aggressive behaviors than wild-type mice in
the intruder–resident test (Nelson et al., 1995; Trainor et al., 60 5-HT *
5-HIAA
Change from WT values (%)

2007c; Bedrosian and Nelson, 2014). Nearly 90% of the aggres- 5-HIAA/5-HT
sive encounters were initiated by the nNOS/ animals. In all 40
test situations, male nNOS/ mice rarely displayed submissive *
* *
behaviors (Nelson et al., 1995). Behavioral studies of mice with 20 *
targeted deletion of specific genes suffer from the criticism that
the gene product is not only missing during the testing period, 0
but also missing throughout ontogeny when critical develop-
mental processes, including activation of compensatory mech- −20
anisms, could be affected (Nelson et al., 1997). Furthermore, * * *
differences in genetic background might also contribute to †
−40
the observed changes in behavior of knockout mice (Wolfer Cor Hyp Hipp Amy Mid Cer
et al., 2002). To address these criticisms, mice were treated
with 7-nitroindazole (7-NI) to specifically inhibit nNOS Figure 8 Serotonin (5-HT), 5-HIAA, and 5-HT turnover (5-HIAA/5-HT)
formation in vivo (Demas et al., 1997). Isolated mice treated ratios are reduced in mice lacking the gene for nNOS. Determination of
with 7-NI displayed substantially increased aggression in two 5-HIAA/5-HT ratio was made by HPLC in the cerebral cortex (Cor),
different tests of aggression compared to control animals hypothalamus (Hyp), hippocampus (Hipp), amygdala (Amy), midbrain
(Mid), and cerebellum (Cer) of nNOS/ as compared to WT mice. Data
(Demas et al., 1997). The combination of the traditional phar-
are percent change in relation to WT mice SEM; )p < 0.05. Reproduced
macological approach and a targeted gene disruption approach from Chiavegatto, S., Dawson, V.L., Mamounas, L.A., Koliatsos, V.E.,
to studies of aggression enhances the strengths and minimizes Dawson, T.M., Nelson, R.J., 2001. Brain serotonin dysfunction
the weaknesses of each single approach. accounts for aggression in male mice lacking neuronal nitric oxide syn-
Plasma androgen concentrations in mice do not differ thase. Proc. Natl. Acad. Sci. U.S.A. 98, 1277–1281 with permission
between wild types and nNOS/ either before or after from the National Academy of Sciences.
158 Hormones and the Development and Expression of Aggressive Behavior

could reflect a combination of factors including species The gradual replacement of play fighting by ‘serious’
differences, methodology, regional specificity, or assays of fighting over the course of ontogeny, coupled with the male
mRNA compared to protein (Panzica et al., 2006). Further bias in rough-and-tumble play, invites the conclusion that
work is necessary to understand the relationship among sex play fighting and adult fighting are a developmental
steroid hormones, 5-HT, and NO in mediating aggression. continuum in which play fighting is the immature form of
Additional work is necessary to understand the relationship adult aggression. However, based on several lines of evidence
among sex steroid hormones, 5-HT, and NO in mediating gleaned primarily from studies in rats and hamsters, Pellis
aggression. and colleagues contend that play fighting and adult aggression
Interestingly the effects of nNOS on aggression are mediated are distinct behaviors, and that play fighting is not practice for
by the social environment. Initial studies examining effects of adult fighting skills, but instead is practice for general social
nNOS inactivation on aggression were conducted on singly skills and competence (Pellis and Pellis, 1988, 1997, 1998,
housed males. Effects of nNOS inactivation through gene 2007). First, play fighting and adult fighting have different
knockout or inhibition with 3BrN were blunted in males that topographies. In play fighting, attacks are initiated toward the
were housed in pairs (Trainor et al., 2007c). Thus the effects head and nape, whereas in serious fighting, attacks are initiated
of nNOS on aggression are dependent on the social environ- toward the rump (Pellis and Pellis, 1988; Wommack et al.,
ment. Social isolation decreases 5-HT1A receptors (Rilke et al., 2003; Taravosh-Lahn and Delville, 2004). Defensive maneu-
1998) and increases V1a receptors (Albers et al., 2006), which vers during play and adult fighting differ as well. To evade an
could potentially mediate the impact of nNOS on aggression. attack, juvenile male rats rotate their bodies fully to a supine
Interestingly nNOS inactivation reduced motivation to position, but adults rotate only partially so that their hind
approach unfamiliar individuals or social odors. The combina- feet remain on the ground (Pellis, 2002). Play fighting is also
tion of increased aggression and reduced social interaction has characterized by frequent role reversals; for example, the chasee
also been observed in rhesus monkeys with the low activity may suddenly turn around and become the chaser, or a larger
form of the serotonin transporter promoter and raised in animal may voluntarily self-handicap, letting a smaller animal
peer-only groups. In monkeys raised by their mothers, no pin or chase the larger one (reviewed in Pellis and Pellis, 2007).
differences between serotonin transporter genotypes were Such role reversals are uncommon in adult aggression. Further-
observed (Barr et al., 2003). more, infant (preweaning) rats display adultlike defensive
Two recent studies implicate a role of NO in human aggres- tactics, which are then replaced by the juvenile tactics (Pellis
sion as well. For example, a NOS1 promoter repeat length vari- and Pellis, 1997). Thus, it does not appear that the specific
ation (NOS1 Ex1f variable number tandem repeat (VNTR)) motor patterns of juvenile play fighting are immature or
was identified, and the short repeat variant is associated more simpler forms of adult fighting. Second, although the frequency
frequently with adult ADHD, personality disorder, and aggres- of play fighting decreases over the course of pubertal matura-
sion. The short variants lead to reduced transcriptional activity tion, play fighting is not unique to the juvenile period, and
of the NOS1 exon 1f promoter (Reif et al., 2009). Adverse envi- both play fighting and adult fighting can and do occur in adult-
ronmental stressors can interact with the short variant to hood (Pellis and Pellis, 1988). When play fighting occurs
promote elevated impulsive behaviors, including impulsive among adult animals, however, it is more likely to escalate to
aggressive behavior, in children aged 9–18 (Reif et al., 2011). adult fighting, presumably because the adults have decreased
Taken together, these studies suggest a role of NO in mediation tolerance for one another (Pellis and Pellis, 1988). Third,
of human impulsive aggression. play fighting and adult fighting appear to have opposite
valences. In anticipation of play and during play, rats emit
50 kHz ultrasonic vocalizations, which are associated with
1.05.6 Development of Aggression rewarding stimuli and positive social affect. In contrast, during
threatening situations, including intermale fighting, rats emit
1.05.6.1 Rough-and-Tumble Play as an Antecedent
22 kHz vocalizations, which are associated with aversive
to Aggressive Behavior
stimuli and negative social affect (Knutson et al., 1998;
Juveniles of most species engage in agonistic behaviors that at reviewed in Knutson et al., 2002). Thus, play fighting and adult
least superficially resemble adult aggression. These behaviors aggression appear to involve different psychological states.
are referred to as rough-and-tumble play or play fighting. In Fourth, neurochemical correlates of male rat juvenile play
common with adult aggression, juvenile rough-and-tumble and adult aggression are not identical. Specifically, juvenile
play comprises both offensive and defensive maneuvers in play is associated with a decrease in hypothalamic levels of
which animals attack, bite, pin, wrestle, roll over, and flee. cholecystokinin (CCK), whereas submission during adult
Unlike adult aggression, juvenile rough-and-tumble play does aggressive encounters is not (Burgdorf et al., 2006). This
not involve competition for resources, territory, or mates. In finding supports the notion that juvenile play has positive
most species, including humans, juvenile males engage in valence in light of the fact that elevated levels of CCK in cortex
more rough-and-tumble play than females. Rough-and-tumble are associated with submissive behavior during adult aggres-
play predominates during social interactions in prepubertal sion and negative affective states (Knutson et al., 2002;
and juvenile animals and gradually declines over the course Panksepp et al., 2004). Finally, opportunities to engage in
of pubertal maturation. In general, overt aggression is relatively rough-and-tumble play during the juvenile period appear to
uncommon prepubertally and increases concomitantly with facilitate development of social skills and competency and, if
reproductive maturation and the associated rise in circulating anything, lead to reduced overt aggressive encounters in adult-
concentrations of gonadal steroids. hood (Pellis and Pellis, 2007; Cooke and Shukla, 2011;
Hormones and the Development and Expression of Aggressive Behavior 159

Panksepp and Scott, 2012). For example, play-deprived adult investigations have examined the role of gonadal steroids in
rats do not appear to have learned the behavioral strategies the sexual differentiation of these behaviors. Overall, sexual
that signal submission to a dominant rat, and this lack of social differentiation of play fighting conforms to the classical model
competency only invites more aggression (Von Frijtag et al., in which the presence of testosterone prenatally (nonhuman
2002). Together, these lines of evidence do not support the primates) or during the first few days after birth (rodents)
idea that play fighting is a practice for adult aggression. masculinizes play fighting, and in the absence of testosterone
The relationship between play fighting and adult aggression or in the presence or absence of the ovaries, a female-typical
is viewed differently by Delville and colleagues, who maintain level of play fighting is observed (reviewed in Wallen, 1996;
that they are the same behaviors expressed during different Pellis, 2002; Cooke and Shukla, 2011). In addition, human
stages of development (Delville et al., 2003; Cervantes et al., females with congenital adrenal hyperplasia, who experience
2007; Wommack and Delville, 2007). Based on their extensive relatively high levels of adrenal androgens in utero, display
studies on the development of aggression in Syrian hamsters, higher levels of rough-and-tumble play and ‘tomboyism’ relative
they argue that play fighting attacks are similar to adult attacks to unaffected siblings (reviewed in Collaer and Hines, 1995).
in intent, even though the body part that is the target of the Perinatal masculinization of play behavior most likely
attack is different at the two ages (head vs rear), because both involves activation of both androgen and estrogen receptors
juveniles and adults flank mark during agonistic interactions in the nervous system. The androgen receptor blocker fluta-
as a means of communicating dominant/subordinate status. mide disrupts masculinization of play behavior, either when
Furthermore, because the selective serotonin reuptake inhibitor given to rat dams during the last half of pregnancy (Casto
fluoxetine inhibits both juvenile play fighting and adult aggres- et al., 2003) or when given to male pups over the first
sion, there appears to be a common underlying neurobiology 10 days of life (Meaney et al., 1983). Other experiments
(Delville et al., 2003). The key to resolving these opposing provide evidence that perinatal estrogen receptor activation
viewpoints about whether play fighting is an immature form also contributes to the masculinization of play behavior. Two
of adult aggression may lie in the different methodologies of these experiments investigated play fighting in tfm rats, in
used to evaluate agonistic interactions. Pellis and colleagues which a mutation in the androgen receptor gene renders the
have studied play fighting almost exclusively among group- receptor protein nonfunctional and the rats androgen-
housed siblings or familiar males in familiar environments, insensitive. Therefore, effects of testosterone, which is synthe-
whereas Delville and colleagues have studied play fighting sized and secreted by Tfm rats, are presumably due to estrogen
almost exclusively using a resident–intruder paradigm in which receptor activation after aromatization of testosterone to estra-
the resident has been socially isolated since weaning and the diol. The two investigations of play fighting in tfm rats are
intruder is a younger and smaller animal. The latter conditions somewhat contradictory. One of them reports similar levels
create competition and favor aggressive responses by an advan- of play fighting in tfm and wild-type male rats, suggesting
taged resident. Therefore, agonistic interactions between juve- that estrogen receptor-mediated mechanisms are sufficient to
nile males under these circumstances may in fact be adultlike masculinize the behavior (Field et al., 2006). The other one
aggression in defense of territory, despite the animal’s young reports that levels of play fighting are less in tfm than in wild-
age and immature reproductive status, and different topog- type males, supporting a role for androgen receptor-mediated
raphy of aggressive behavior. Thus, the distinction between mechanisms (Meaney et al., 1983). However, play behavior
play fighting and adult fighting may not rest so much on the was measured in different social contexts in the two studies,
age of the animal as it does on whether or not stakes are and while this may make the results not directly comparable,
involved. together they implicate both androgenic and estrogenic action
As a case in point, sibling rivalry between spotted hyena cub in the sexual differentiation of play behavior.
twins involves overt aggression and can result in siblicide Hormone-mediated masculinization of play behavior may
(Frank et al., 1991; Wahaj et al., 2007). Sibling aggression involve pathways apart from the classical mechanism of
within the first year of life in hyenas establishes a rank relation- hormone activation of its cognate receptor. The first of these
ship within the litter and is primarily over competition for milk is ligand-independent activation of the estrogen receptor.
and food. Sibling aggression is more intense when local prey is Administration of a dopamine D1 receptor agonist to neonatal
scarce and tends to be higher within litters of low-ranking female rats masculinizes their play fighting behavior, and this
females, who are disadvantaged for access to resources within effect can be blocked by prior treatment with the estrogen
the clan (Wahaj and Holekamp, 2006). Thus, siblicide in receptor antagonist tamoxifen (Olesen et al., 2005). The second
hyenas is not obligate, as once proposed, but instead is rela- of these involves epigenetic regulation of gene transcription,
tively uncommon and facultative, occurring when maternal specifically DNA methylation and repression of gene expres-
resources are insufficient to sustain two cubs (Smale et al., sion in the amygdala. Testosterone delivered directly to the
1999). This example reinforces the idea that the distinction amygdala of neonatal female rats is sufficient to induce male-
between play fighting and aggression is not age per se, but typical levels of play fighting (Meaney and McEwen, 1986),
rather whether competition for resources is involved. suggesting that testosterone or a biologically active metabolite
acts within the amygdala to organize sex differences in play
behavior. If the methyl-binding protein MeCP2 is silenced
1.05.6.2 Endocrine Contributions to the Development of
within the amygdala of newborn male rats, juvenile play
Aggressive Behavior: Perinatal Organizational Effects
fighting is reduced to female-typical levels (Kurian et al.,
Given that levels of both play fighting and adult aggression are 2008); this effect on behavior is similar to what one would
higher in males than in females of most species, numerous predict if testosterone action within the amygdala of newborn
160 Hormones and the Development and Expression of Aggressive Behavior

males were blocked. Overall, it appears that sexual differentia- decline. These behaviors are replaced by flank marking as
tion of play fighting involves multiple hormones and multiple a means to more peacefully maintain the relationship. If male
mechanisms. hamsters are castrated prepubertally, then testosterone replace-
Similar principles apply to the perinatal sexual differentia- ment in adulthood fails to activate flank marking behavior, as
tion of adult aggression. That is, the transient elevation in it normally does if hormone replacement is given to hamsters
testosterone in male neonates leads to higher levels of aggres- that are castrated in adulthood (Schulz et al., 2006). Further-
sion in adulthood (compared with females), and surgical or more, male hamsters deprived of testosterone during puberty
pharmacological castration of neonatal males leads to reduced fail to replace overt aggression with flank marking and resort
levels of aggression in adulthood (Bronson and Desjardins, to fighting again when reintroduced to each other after having
1969). Conversely, treatment of neonatal females with testos- established a dominant–subordinate relationship during earlier
terone masculinizes their levels of adult aggression (Bronson encounters (De Lorme and Sisk, 2013). Similarly, territorial
and Desjardins, 1970). Prenatal androgens also appear to scent marking in tree shrews is organized by the pubertal rise
masculinize aggression in humans, as a study found that girls in testosterone, since castration prior to puberty prevents activa-
with CAH are not only rated as more aggressive than unaffected tion of this behavior by testosterone in adulthood (Eichmann
girls, but also are as aggressive as boys. A fascinating variation and Holst, 1999).
on this theme occurs in spotted hyenas, in which higher social Testicular hormones during puberty also program the level
rank of females within the clan is associated with higher of aggression displayed by adult hamsters. In one study (Schulz
maternal androgens during late gestation (Dloniak et al., et al., 2006), males were castrated either before or after puberty,
2006). These higher gestational concentrations of androgen and then 6 weeks later were treated with either vehicle or testos-
lead to higher levels of aggression in the offspring. Thus, terone. One week after hormone replacement, agonistic behav-
maternal androgens not only organize aggressive behavior, iors were assessed in a resident–intruder test (Figure 9).
but they also are a mechanism through which social status
traits are epigenetically transferred from mother to daughter.
At least some of the masculinizing effects of perinatal testos-
terone on adult aggression are due to estrogenic action
(Martinez-Sanchis et al., 1996). As described elsewhere in this
chapter, males have both an androgen and an estrogen-sensi-
tive circuitry that underlies hormone-facilitated aggression.
Work by Simon and colleagues in mice has demonstrated
that estradiol, presumably derived from aromatized testos-
terone, masculinizes the estrogen-sensitive circuit, while
masculinization of the androgen-sensitive circuit is due to
direct androgenic action during early postnatal development
(Martinez-Sanchis et al., 1996).

1.05.6.3 Endocrine Contributions to the Development of


Aggressive Behavior: Pubertal Organizational Effects
Another period of hormone-dependent organization of social
behaviors, including aggression, occurs during puberty, when
testicular hormone concentrations are once again elevated in
males and when ovarian hormone cycles commence in females
(reviewed in Schulz et al., 2009). An organizational role for
pubertal hormones has been demonstrated by experiments in
which gonadectomy performed after the perinatal period of
sexual differentiation but before the onset of puberty results
in long-lasting alterations in agonistic interactions. Prepubertal Figure 9 Aggressive and submissive behaviors expressed by male
castration prevents the normal transition from complete to Syrian hamsters in a 10-min resident–intruder test. Subjects were cas-
partial rotations in male play fighting defensive behaviors. trated either prepubertally or in adulthood, and 6 weeks later treated for
Interestingly, ovarian hormones appear to suppress in females 1 week with either placebo or testosterone. When endogenous testos-
the pubertal emergence of a male-typical increase in roughness terone was absent during adolescent development (prepubertal castra-
of play fighting (Pellis, 2002). tion group), behavior in adulthood was characterized by fewer attacks
Testicular hormones during puberty program agonistic and more escapes compared to when endogenous testosterone was
present during adolescent development. Thus, testicular hormones,
behaviors in adult hamsters. One agonistic behavior commonly
acting during puberty, program higher levels of aggression in adult-
observed in male–male encounters is flank marking, in which
hood, even though testosterone does not exert activational effects on
flank gland secretions are rubbed onto objects in the environ- these behaviors in adulthood in this species. Reprinted from Schulz,
ment as a means of communicating dominant/subordinate K.M., Sisk, C.L., 2006. Pubertal hormones, the adolescent brain, and
status. Male hamsters initially use overt aggression and submis- the maturation of social behaviors: lessons from the Syrian hamster.
sion to quickly establish a dominant–subordinate relationship. Mol. Cellular Endocrinol. 254–255, 120–126, with permission from
In subsequent encounters, aggressive and submissive behaviors Elsevier.
Hormones and the Development and Expression of Aggressive Behavior 161

Figure 10 Aggressive and submissive behaviors expressed by male Syrian hamsters in a 10-min resident–intruder test. Subjects were castrated
either prepubertally or in adulthood, and 6 weeks later treated for 1 week with either placebo or testosterone. When endogenous testosterone was
absent during adolescent development (prepubertal castration group), behavior in adulthood was characterized by fewer attacks and more escapes
compared to when endogenous testosterone was present during adolescent development. Thus, testicular hormones, acting during puberty, program
higher levels of aggression in adulthood, even though testosterone does not exert activational effects on these behaviors in adulthood in this species.
Reproduced from Schulz, K.M., Sisk, C.L., 2006. Pubertal hormones, the adolescent brain, and the maturation of social behaviors: lessons from the
Syrian hamster. Mol. Cellular Endocrinol. 254–255, 120–126, with permission from Elsevier

Irrespective of testosterone or vehicle treatment in adulthood, differentiation of neural circuits to result in sex-typical expres-
males castrated prior to puberty did not attack the intruder sion of aggressive behavior in adulthood (Figure 10).
and displayed high levels of submissive behaviors. In contrast,
males that were castrated after puberty attacked the intruder
1.05.6.4 Endocrine Contributions to the Development of
and rarely displayed submissive behaviors. Organizational
Aggressive Behavior: Pubertal Activational Effects
effects of adolescent hormones on male aggression have also
been reported in other species, as evidenced by long-lasting Generally speaking, levels of aggression increase over the
changes in aggressive behavior when hormones are manipu- course of puberty as an animal achieves reproductive fertility
lated during the pubertal period. Male DBA/1Bg mice are nor- and faces the responsibility of obtaining its own food and
mally very aggressive, but the absence of gonadal hormones shelter, fending for itself, finding a mate, and potentially caring
during adolescence prevents activation of aggressive behavior for offspring. Because the pubertal increase in aggression
by testosterone in adulthood (Shrenker et al., 1985). Similarly, temporally coincides with the pubertal rise in gonadal and
adult testosterone treatment only partially restores aggressive adrenal steroid hormones, it is logical to conclude that pubertal
behavior in prepubertally castrated male gerbils (Lumia et al., hormones activate the behavior in particular social contexts.
1977), indicating that pubertal hormones program behavioral Indeed, as detailed above, there is strong evidence for andro-
responses to hormones in adulthood. genic and estrogenic activation of adult aggressive behavior in
Agonistic behaviors in female rodents may also be orga- many rodent species, with the caveats that the causal relation-
nized during adolescence. If female mice are ovariectomized ship between hormones and aggression is often a two-way
at the onset of puberty (30 days of age), treated with testos- street, and that effects of hormones on aggression are modu-
terone for 3 weeks during adolescent development, and then lated by genetic background, experience, and complex interac-
tested 6 weeks after discontinuation of testosterone treatment, tions between the two. Evidence for activational effects of
the levels of aggressive behavior toward another female in hormones on aggression in adulthood notwithstanding, the
a neutral arena are much higher than in females treated with expression of agonistic behaviors over ontogeny is not gov-
vehicle (Edwards, 1970). Thus, adolescent exposure to erned by hormones in all cases. Play fighting during the prepu-
androgen has long-term effects on aggression in female mice, bertal and juvenile periods of life is clearly not under gonadal
and the nervous system remains sensitive to organizing influ- hormone control, because hormone levels are at their nadir at
ences of gonadal steroid hormones well into postnatal life. this point in development. However, during early puberty,
However, the adolescent brain appears to be less sensitive initiation of play is activated by testicular hormones, as evi-
than the neonatal brain to organizational effects because denced by a reduction in play in male rats gonadectomized
more testosterone and longer duration of treatment are just prior to puberty (Cooke and Woolley, 2009). In Syrian
required to masculinize aggression during puberty than hamsters, pubertal increases in intermale aggression proceed
on PND1. similarly in both gonad-intact males and males castrated
Overall, sexual differentiation of play fighting and adult a few days after weaning (Whitsett and Vanderbergh, 1975).
aggression is a two-stage process involving gonadal hormone Thus, the pubertal increase in testosterone is not an absolute
action in the nervous system during perinatal and pubertal requirement for the expression of adult aggression.
periods of development. Perinatally, testicular hormones, via Normal pubertal changes in play fighting involve both
both androgenic and estrogenic action, drive the initial mascu- androgen and estrogen receptor-mediated processes as revealed
linization and defeminization of circuits underlying juvenile by an examination of play fighting in tfm rats. Tfm rats do not
play and adult aggression. Pubertally, both testicular and show the typical decrease in play fighting with age and are more
ovarian hormones reinforce and refine the sexual likely than wild-type rats to show the juvenile-typical defensive
162 Hormones and the Development and Expression of Aggressive Behavior

complete rotation in adulthood (Field et al., 2006). On the easily lead to the prediction that the presence of testicular
other hand, tfm males do show normal age-related changes in hormones during adolescence would result in conditioned
the use of partial rotations and upright postures. Thus, func- defeat responses to subjugation.
tional androgen receptors appear to be necessary for some,
but not all, developmental changes in the quantity and quality
1.05.6.6 Conditioned Defeat
of play fighting. The transition from play fighting (attacks
toward head) to adult aggression (attacks toward rear) in After defeat in the home cage of an aggressive conspecific, male
hamsters appears to be due to increasing corticosteroid concen- hamsters (Mesocricetus auratus) will subsequently fail to defend
trations during puberty (Wommack and Delville, 2007). their home territory even if the intruder is a smaller, nonaggres-
sive male (Huhman et al., 2003). This phenomenon has been
called conditioned defeat and appears to evoke a stress
1.05.6.5 Social Experience and the Development of
response via fear conditioning (Huhman and Jasnow, 2005).
Aggressive Behavior
The physiological effects of defeat include elevated HPA axis
In both humans and animals, early life social experience and activity such as increased plasma ACTH, b-endorphin, cortisol,
opportunities for juvenile play fighting influence the expression and corticosterone concentrations, as well as decreased plasma
of aggression in adulthood (reviewed in Cooke and Shukla, testosterone and prolactin concentrations (Huhman et al.,
2011; Veenema, 2012; Haller et al., 2014). For example, male 1990, 1991). This endocrine profile is observed among previ-
rats that are singly housed from 3 to 5 weeks of age (during ously defeated hamsters upon reexposure to another animal
the prepubertal/adolescent period) show less submissive – even when the new opponent is blocked by a physical barrier
behavior during territorial aggression by a resident male (Huhman et al., 1992). This latter response suggests that this
compared with rats that are group-housed during adolescence change in endocrine profile is in response to a psychological
(van den Berg et al., 1999). Similarly, isolation rearing from stressor and not to the pain or anxiety of the combat itself.
3 to 7 weeks of age leads to increased shock-induced defensive Social defeat also affects immune responses (Fleshner et al.,
aggression, an effect that is ameliorated by daily play fighting 1989; Jasnow et al., 2001). The physiological and behavioral
experience during the period of social isolation (Potegal and consequences of conditioned social defeat persist for at least
Einon, 1989). Thus, social interactions in the form of juvenile 33 days (Huhman et al., 2003), and perhaps throughout adult-
play appear to buffer against heightened aggression in response hood (Delville et al., 1998). Few female hamsters exhibit
to provocation. conditioned social defeat, although ACTH concentrations
Research using the Syrian hamster to examine the effects of were reduced in those females that displayed low levels of
social subjugation on subsequent expression of aggression illus- submissive/defensive behavior (Huhman et al., 2003). In
trates the importance of two types of interaction that influence contrast to males, the conditioned defeat response did not
the development of aggressive behavior. First is the interaction persist beyond the first test among female hamsters. These
between social experience and context. If prepubertal male results suggest that in male hamsters conditioned defeat is
hamsters are socially subjugated by experiencing repeated defeat a profound, persistent behavioral change characterized by
in male–male social encounters, then they subsequently show a total absence of territorial aggression and by the frequent
enhanced aggression toward a smaller and younger intruder, display of submissive and defensive behaviors (Huhman and
whereas they show reduced offensive responses toward an Jasnow, 2005).
intruder of similar age and size (Delville et al., 1998). Thus,
subjugation can lead to heightened or reduced aggression,
1.05.6.7 Aggression in Aged Individuals
depending on social context. Prepubertal subjugation also accel-
erates the transition from play fighting to adult aggression and At the other end of the life span, heightened aggressive and
increases aggression in adulthood, which may be mediated by sexually offending behaviors by aged individuals can pose diffi-
an increase in adrenal glucocorticoid secretion brought about culties for themselves, their caretakers, family members, and
by the stress of defeat (Wommack et al., 2003). Second, the fellow elderly residents living in assisted care facilities (Pulsford
effect of subjugation on aggression depends on the age at which and Duxbury, 2006). Some clinical studies have assessed
subjugation occurs. In contrast to prepubertal subjugation, hostility as a ‘proxy’ for human aggression. Despite its impreci-
subjugation in adulthood leads to complete suppression of sion, hostility has proven to be a useful construct in studies of
aggressive behavior in male hamsters, a phenomenon known the influences of hormones among aggressive elderly people.
as conditioned defeat (see below Huhman et al., 2003). Subju- For example, postmenopausal women using HRT scored lower
gation of hamsters after mid-puberty leads to an adult-typical on hostility scales than women that did not use HRT (Steffen
response to subjugation, i.e., decreased aggression toward an et al., 1999; Olson et al., 2004). These reports suggest estrogens
intruder (Delville et al., 2003), suggesting that the develop- can influence the expression of aggressive behavior. Studies of
mental switch responsible for the different responses to subju- men and women who have been diagnosed with dementia and
gation observed in juvenile and adult hamsters occurs shortly display physical or verbal aggression suggest a positive correla-
after the pubertal rise in testosterone. However, it is not clear tion with circulating testosterone and a negative correlation
that testosterone either triggers the switch or is part of the with circulating estradiol (Orengo et al., 2002). Treatment of
switch, because as described above, the absence of testicular patients with dementia with estrogens reduced aggression
hormones during adolescent brain development renders male and sexually offending behaviors (Kyomen et al., 1991,
hamsters less aggressive and more submissive during male– 1999). Despite the significant problems associated with height-
male encounters (Schulz et al., 2006). This finding does not ened aggression among some elderly patients, especially those
Hormones and the Development and Expression of Aggressive Behavior 163

with moderate-to-severe dementia, there has been remarkably behavior. The short allele of the 5-HTT gene is associated with
little animal research on this topic. Mice with mutated human reduced expression of 5-HTT in the brain and inefficient reup-
amyloid precursor protein (APP) and presenilin (PS1) genes take of 5-HT from the synapse (Greenberg et al., 1999). The
display shorter latencies to attack and increased numbers of interaction between stress and 5-HTT genotype was examined
attacks, suggesting that the plaques and tangles associated in men and women who were instructed to administer shocks
with dementia may contribute to aggression (Minkeviciene to a confederate as punishment for incorrect responses in
et al., 2004; Alexander et al., 2011). a memory task (no shocks were actually delivered) (Verona
et al., 2006). Half of the participants were subjected to a phys-
ical stressor (unpredictable air blasts to the throat) whereas the
1.05.7 Reciprocal Effects of Aggression on Steroid other half were not. Men, but not women, who were homozy-
Hormones gous for the short allele were more likely to administer shocks
under the stressed condition, whereas there were no genotype
Hormones influence aggressive behaviors, but it should be differences in the control condition. This interaction could be
emphasized that aggressive behavior can feed back and affect mediated by differences in threat perception, as individuals
hormone concentrations. Male mice and Syrian hamsters carrying the short allele have increased activation in the amyg-
reduce circulating androgen concentrations if they lost a fight dala in response to fear-inducing pictures (Hariri et al., 2002).
in paired aggressive encounters (Lloyd, 1971; Huhman and A recent meta-analysis of 18 studies also found that the short
Jasnow, 2005). This endocrine suppression lasted for several allele was significantly associated with increased antisocial
days postdefeat. Similarly, rhesus monkeys that were defeated behavior (Ficks and Waldman, 2014). Interestingly, there was
by a higher-ranking male had dramatically reduced testos- some evidence for publication bias such that studies with
terone concentrations for several weeks postdefeat. In contrast, smaller sample size tended to report large effect sizes. In addi-
winning males’ circulating testosterone concentrations quadru- tion, as with MAOA, there are many unanswered questions on
pled within 24 h of victory (Bernstein et al., 1977). how the environment interacts with 5-HTT function to modu-
late aggression.
In rodents several studies have demonstrated that parental
1.05.7.1 Gene–Environment Interactions
behaviors can influence the effects of genes on aggression.
Studies of aggression are typically conducted under a single set Males of the NZB strain are more aggressive the CBA/H strain
of environmental conditions. However, mechanisms of aggres- (Roubertoux and Carlier, 1988). If male NZB mice are crossed
sive behavior have evolved in fluctuating physical and social with females of the CBA/H strain, then the resulting male
environments. Perhaps not surprisingly, several neurochemical offspring are more aggressive, but only if raised by CBA/H
pathways of aggression function differently depending on the dams. However, if hybrid pups are cross-fostered to hybrid
environment. mothers, then the pups are no more aggressive as adults
One of the most repeatable observations in studies of than male CBA/H strains (Carlier et al., 1991). In these studies
human aggression is the interaction between early life experi- the specific differences in maternal care were not identified.
ence and genotype for the human MAOA gene. Caspi et al. More detailed studies have observed the effects of parental
(2003) reported that the effect of child abuse on behavior care on aggression in Peromyscus. Male Peromyscus californicus
was significantly stronger if the child carried alleles associated are more aggressive than male Peromyscus leucopus (Bester-
with low MAOA activity (Figure 11(a)). Abused children Meredith et al., 1999), but if male P. californicus are cross-
with low MAOA activity had increased antisocial behavior, fostered to P. leucopus parents, then this species difference in
greater prevalence of conduct disorder, and a higher likelihood aggression disappears (Bester-Meredith and Marler, 2001).
of convictions of violent offenses than abused children with Correlational analyses suggested that parental retrieving
high MAOA activity. In children who were not abused, the behavior was a critical factor (Bester-Meredith and Marler,
polymorphism had no effect on these measures of behavior. 2003; Marler et al., 2003), and a subsequent study showed that
This gene–environment interaction has been replicated in experimentally increasing retrieval behavior in P. californicus
some studies (Foley et al., 2004; Kim-Cohen et al., 2006), increased aggression in male and female offspring (Frazier
but not others (Huizinga et al., 2006; Young et al., 2006). An et al., 2006), possibly by inducing increased T secretion in
initial meta-analysis indicated that on average, children with pups (Becker et al., 2010). Interestingly, the absence of
genotypes for low MAOA activity have elevated rates of antiso- a California mouse father has a major impact on the develop-
cial behavior when exposed to parental maltreatment (Kim- ment of aggression. Female but not male California mice
Cohen et al., 2006). A more recent meta-analysis of 31 studies raised without the father were significantly more aggressive
(including both community-based and clinically referred and had reduced the sensitivity of the frontal cortex to dopa-
samples) also found that the allele for low MAOA activity mine (Bambico et al., 2015).
was significantly associated with antisocial behavior and The context in which mice are tested can also have impor-
aggression (Ficks and Waldman, 2014). However, variability tant effects on behavior. Male mice are usually more aggressive
in effect size was quite large across studies and this variability in resident–intruder tests, when intruders are introduced into
was not associated with sample or study characteristic. Thus a residents’ home cage compared to neutral tests when two
it is still unclear how the environment interacts with MAOA mice are introduced into a neutral arena. Patterns of aggression
function to modulate antisocial behavior and aggression. in one context do not necessarily transfer to a different context.
There is also growing evidence that genetic variation in sero- For example, the correlation between mossy fibers in the
tonin transporter (5-HTT) is linked to aggression and antisocial hippocampus and aggression is positive if male mice are tested
164 Hormones and the Development and Expression of Aggressive Behavior

Figure 11 Gene–environment interactions in humans and mice. The interaction between the monoamine oxidase A (MAOA) genotype and the rear-
ing environment affects aggressive behavior (a). Although they have not been replicated in every study, most data suggest that children carrying the
short form of the MAOA promoter gene, which confers decreased MAOA activity, are more likely to develop conduct disorders and increased antiso-
cial behavior when exposed to abusive home environments. This environmental effect is less prevalent in individuals carrying the long form of the
promoter. Photoperiod determines the directional effects of estrogens on aggressive behavior in beach mice (Peromyscus polionotus) (b). Peromyscus
polionotus are more aggressive when exposed to short days (shown in the left graph) than when exposed to long days (shown in the right graph).
Treatment with the estrogen synthesis inhibitor fadrozole (fad) decreases aggression if beach mice are tested in short days, but increases aggression
if tested in long days. The effects of fad are reversed with cotreatment with estradiol (E2). This does not appear to be mediated by differences in
receptor expression, because the drugs PPT (propylpyrazole-triol, an estrogen receptor (ER)-a agonist) and DPN (diarylpropionitrile, an ERb agonist)
both increase aggression on short days and decrease aggression on long days. Photoperiod apparently regulates the molecular actions of estrogens,
acting rapidly on short days (presumably nongenomically) and more slowly on long days (presumably genomically). Reprinted from Nelson, R.J.,
Trainor, B.C., 2007. Neural mechanisms of aggression. Nat. Rev. Neurosci. 8, 536–546 with permission from Nature.

in a resident–intruder test (Guillot et al., 1994), but is absent if neuroscience studies using male rodents compared to studies
mice are tested in a neutral arena (Roubertoux et al., 1999). It using females, even when estrous cycle is not controlled for
has also become clear that social status may be a critically over- (Prendergast et al., 2014). Finally, the outcomes of resident–
looked variable in studies using domestic mice. For example, intruder tests also depend on whether the intruders used are
male C57 mice are typically housed four to five mice per a different genotype than the test mice (Maxson et al., 1989).
cage. However, each cage typically has one dominant mouse For example, genetic variation in the steroid sulfatase gene
that directs aggression at its cage mates (Howerton et al., (Sts) affects male aggressive behavior when there is no risk of
2008). This alters not only behavior, but brain circuits that the opponent retaliating (such as when males are olfactory bul-
modulate aggression (Greenberg et al., 2014). This is likely bectomized), but has no effect when there is a risk of injury
an important factor contributing to increased variability in from the opponent (Maxson et al., 2001).
Hormones and the Development and Expression of Aggressive Behavior 165

In mice of the genus Peromyscus, photoperiod determines Saline Fadrozole


the mechanisms through which estrogens control male aggres-
sive behavior (Laredo et al., 2014b; Figure 11(b)). Similar to 20
hamsters, three species of Peromyscus are more aggressive **
when exposed to short days than when exposed to long days 15

Bites (freq)
(Peromyscus maniculatus and Peromyscus polionotus (Trainor
et al., 2007b); P. californicus (Nelson and Trainor, 2007; Trainor 10
et al., 2007b)). In P. polionotus, estrogens decrease aggression
when mice are housed in long days, but increase aggression if 5
mice are housed in short days (Trainor et al., 2007a). Hormone
manipulation studies showed that the ERa agonist PPT and the 0
ERb agonist DPN increased aggression in ‘short-day’ mice and Carefresh Corncob
decreased aggression in ‘long-day’ mice. These data suggested
Figure 12 The effects of estrogens on aggression depend on cage
that photoperiod regulates processes that occur after estrogens
bedding. Aromatase inhibition with fadrozole increased the number of
bind their cognate receptors. Steroids can affect physiological
bites on Carefresh bedding but not on corncob bedding. **, P < 0.01,
and behavioral processes via genomic or nongenomic path- planned comparison for effect of fadrozole; n ¼ 10–14 per group.
ways (Vasudevan and Pfaff, 2006). Classical genomic action Reprinted from Villalon Landeros, R., Yoo, H.J., Morisseau, C., Fu, S.,
occurs when ligand-bound receptors bind to hormone Hammock, B.D., Trainor, B.C., 2012. Corncob bedding reverses the
response elements that facilitate transcription. This process effects of estrogens on aggressive behavior and reduces estrogen
typically takes hours or days. Nongenomic action can occur receptor alpha expression in the brain. Endocrinology 153, 949–953
through several pathways including phosphorylation of with permission from the Endocrine Society.
cellular signaling pathways and changes in intracellular
calcium. Nongenomic effects can occur within seconds of estro-
gens binding receptors. Gene chip analyses of P. polionotus indi- pERK immunoreactivity in BNST and MEA is increased
cated that estrogen-dependent gene expression was increased in during resident–intruder tests when aggression is induced
the BNST of long-day mice compared to short-day mice, sug- (Trainor et al., 2010a,b). Further study is needed to
gesting estrogens might act via nongenomic pathways in mice determine how environmental estrogens modulate the rapid
exposed to short days. In P. polionotus estradiol injections acted effects of endogenous estrogens on aggression.
rapidly (15 min) to increase aggression in short-, but not long-
day mice, suggesting that estradiol increases aggression via
nongenomic action (Trainor et al., 2007a). This same result 1.05.8 Integration
was also observed in P. californicus (Trainor et al., 2008). The
rapid effects of estradiol on aggression in short-day mice were Neurochemical and neuroanatomical pathways of aggression
not blocked by pretreatment with a protein synthesis inhibitor, have been investigated in various species, and it is apparent
consistent with nongenomic action (Laredo et al., 2013). that some pathways are common to humans and nonhuman
However, these follow-up studies identified an unexpected animals. Increasing serotonergic activity decreases reactive
twist. Whether estradiol increased or decreased aggression aggression in humans and also reduces aggression in a mouse
was dependent on type of bedding used to line the cages. resident–intruder test, probably by decreasing impulsivity.
In the initial studies, cages were lined with corncob bedding A more challenging task is determining how murine behavior
and estrogens increased aggression under short days (Trainor in a resident–intruder test relates to reactive or instrumental
et al., 2007a, 2008). However, when a cardboard-based aggression in humans. Aggression researchers have been strug-
bedding was used, estradiol decreased aggression under short gling with this question, and a comprehensible answer has not
days (Laredo et al., 2013). Importantly, the rapid effects of yet emerged. This may be because there is no unambiguous
estradiol on aggression were absent under long days. When answer. In humans, reactive aggression appears to be governed
the two beddings were directly compared, the aromatase inhib- more by serotonergic pathways, whereas the motivated charac-
itor fadrozole increased aggression in mice housed with card- teristics of instrumental aggression suggest a role for dopami-
board-based bedding and decreased aggression in mice nergic pathways. Given the enormous differences in biology
housed with corncob bedding (Figure 12; Villalon Landeros and social structure, it is unlikely that mouse and human
et al., 2012). Mice housed on corncob bedding had elevated aggression can be classified into homologous categories.
blood levels of tetrahydrofuran diols (THF-diols), which are However, it is clear that many neurochemical systems (such
estrogen-like compounds that can alter estrogen-dependent as the serotonergic system) have coevolved in mice and
behavior such as lordosis (Markaverich et al., 2002). Corncob humans to regulate species-specific aggressive behaviors.
bedding has been found to reduce anxiety-like behavior and Thus, although aggressive behavior is expressed in different
decrease sex differences in stress responses. Although the contexts with different behavioral outputs in mice and
exact mechanism through which corncob bedding alters humans, similar neurochemical and neuroanatomical path-
estrogen-dependent behaviors is unknown, one possibility is ways are activated. Difficult questions remain to be answered.
the regulation of extracellular signal regulated kinase (ERK). For example, to what extent does an impoverished background
California mice housed on corncob bedding had greatly influence the development of these neurochemical and neuro-
reduced pERK immunoreactivity in the BNST, MPOA, MEA, anatomical pathways, and to what extent are they activated by
and VMH. The effects of bedding are significant because observing aggression? Considerable debate ensues on the
166 Hormones and the Development and Expression of Aggressive Behavior

effects of violence in the media on aggression, and myriad con- effective strategy for dealing with uncontrolled aggressive
founding factors make it difficult to study these putative effects. behavior may lie in a combination of biological and behavioral
Aggression is increased in animals that observe conflicts among approaches, especially for instrumental aggression.
other individuals (Earley and Dugatkin, 2002; Peake et al.,
2002). Generally overlooked by mental health researchers,
these data show that vicarious experiences have important bio- References
logical effects. Sports fans respond to watching their team win
Aikey, J.L., Nyby, J.G., Anmuth, D.M., James, P.J., 2002. Testosterone rapidly
or lose with corresponding increases or decreases in testos-
reduces anxiety in male house mice (Mus musculus). Horm. Behav. 42, 448–460.
terone levels (Bernhardt et al., 1998; Figure 13). Children play- Albers, H.E., Dean, A., Karom, M.C., Smith, D., Huhman, K.L., 2006. Role of V1a
ing violent video games show reduced activation of brain areas vasopressin receptors in the control of aggression in Syrian hamsters. Brain Res.
involved in affect, such as the amygdala and the anterior cingu- 1073–1074, 425–430.
late cortex (Mathiak and Weber, 2006). Reduced brain activity Albert, D.J., Walsh, M.L., Gorzalka, B.B., Siemens, Y., Louie, H., 1987. Testosterone
removal in rats results in a decrease in social aggression and a loss of social
in frontal areas has also been reported in children with high dominance. Physiol. Behav. 36, 401–407.
exposure to violent video games and television programs Alexander, G., Hanna, A., Serna, V., Younkin, L., Younkin, S., Janus, C., 2011.
(Mathews et al., 2005). Although it is not clear whether these Increased aggression in males in transgenic Tg2576 mouse model of Alzheimer’s
experiences have long-term behavioral effects, it is clear that disease. Behav. Brain Res. 216, 77–83.
Alexander, M., Perachio, A.A., 1973. The influence of target sex and dominance on
vicarious experiences have consistent short-term influences
evoked attack in rhesus monkeys. Am. J. Phys. Anthropol. 38, 543–548.
on brain activity. It is perhaps unsettling that these patterns Anderson, S.W., Bechara, A., Damasio, H., Tranel, D., Damasio, A.R., 1999.
resemble those identified in individuals with dysregulated Impairment of social and moral behavior related to early damage in human
aggression (Volkow et al., 1995; Soloff et al., 2003). Biology- prefrontal cortex. Nat. Neurosci. 2, 1032–1037.
based approaches to examine the effects of observing violence Annen, Y., Fujita, O., 1985. Septal lesions and biting attacks in rats bidirectionally
selected for emotionality. Behav. Neural Biol. 43, 132–142.
on aggressive behavior, if they are conducted in realistic social Archer, J., 1991. The influence of testosterone on human aggression. Br. J. Psychol.
contexts (in addition to questionnaires and other pencil-and- 82, 1–28.
paper approaches), have potential because they allow more Archer, J., 2006. Testosterone and human aggression: an evaluation of the challenge
precise measurements of the neural circuits that influence hypothesis. Neurosci. Biobehav. Rev. 30, 319–345.
Bahrke, M.S., Yesalis, C.E., 2004. Abuse of anabolic androgenic steroids and related
aggressive behaviors. Another issue of concern to clinicians is
substances in sport and exercise. Curr. Opin. Pharmacol. 4, 614–620.
how to treat uncontrolled aggression. This is a complicated Bales, K.L., French, J.A., McWilliams, J., Lake, R.A., Dietz, J.M., 2006. Effects of
issue because, although it is agreed that unchecked aggression social status, age, and season on androgen and cortisol levels in wild male golden
has negative consequences, a certain amount of human aggres- lion tamarins (Leontopithecus rosalia). Horm. Behav. 49, 88–95.
sion is probably necessary to succeed in life. Clinical trials have Bambico, F.R., Lacoste, B., Hattan, P.R., Gobbi, G., 2015. Father absence in the
monogamous California mouse impairs social behavior and modifies dopamine and
investigated many treatments aimed at reducing elevated glutamate synapses in the medial prefrontal cortex. Cereb. Cortex 25, 1163–1175.
aggression that is associated with mental disorders, but treat- Baranano, D.E., Snyder, S.H., 2001. Neural roles for heme oxygenase: contrasts to
ments that can ameliorate excessive aggression have unwanted nitric oxide synthase. Proc. Natl. Acad. Sci. U.S.A. 98, 10996–11002.
side effects on processes such as arousal (Cherek et al., 2006). Barr, C.S., Newman, T.K., Becker, M.L., Parker, C.C., Champoux, M., Lesch, K.P.,
Goldman, D., Suomi, S.J., Higley, J.D., 2003. The utility of the non-human
Although further advances in drug development may lead to
primate; model for studying gene by environment interactions in behavioral
additional improvements in the treatment of pathological research. Genes Brain Behav. 2, 336–340.
aggression, the complexity of aggressive behavior suggests Baulieu, E.-E., Robel, P., 1990. Neurosteroids: a new brain function? J. Steroid
that it might not be possible to control aggression. A more Biochem. Mol. Biol. 37, 395–403.
Becker, E.A., Moore, B.M., Auger, C., Marler, C.A., 2010. Paternal behavior increases
testosterone levels in offspring of the California mouse. Horm. Behav. 58,
385–389.
9 Bedrosian, T.A., Nelson, R.J., 2014. Nitric oxide and serotonin interactions and
aggression. Curr. Top. Behav. Neurosci. 17, 131–142.
Been, L.E., Moore, K.M., Kennedy, B.C., Meisel, R.L., 2016. Metabotropic glutamate
Salivary testosterone (ng dl−1)

8 Winning receptor and fragile X signaling in a female model of escalated aggression. Biol.
fans Psychiatry 79.
Bernhardt, P.C., Dabbs, J.M., Fielden, J.A., 1998. Testosterone changes during
7 vicarious experiences of winning and losing among fans at sporting events. Physiol.
Behav. 65, 59–62.
Bernstein, I.S., Rose, R.M., Gordon, T.P., 1977. Behavioral and environmental events
6 influencing primate testosterone levels. J. Hum. Evol. 6, 517–525.
Bester-Meredith, J.K., Marler, C.A., 2001. Vasopressin and aggression in cross-
Losing fostered California mice (Peromyscus californicus) and white-footed mice
5 fans (Peromyscus leucopus). Horm. Behav. 40, 51–64.
Bester-Meredith, J.K., Marler, C.A., 2003. The association between male offspring
aggression and paternal and maternal behavior of Peromyscus mice. Ethology 109,
4
Before After 797–808.
Bester-Meredith, J.K., Young, L.J., Marler, C.A., 1999. Species differences in paternal
College basketball game
behavior and aggression in Peromyscus and their associations with vasopressin
immunoreactivity and receptors. Horm. Behav. 36, 25–38.
Figure 13 Basketball fans’ testosterone levels before and after their Bibancos, T., Jardim, D.L., Aneas, I., Chiavegatto, S., 2007. Social isolation and
team has won or lost. Reprinted from Bernhardt, P.C., Dabbs, J.M., expression of serotonergic neurotransmission-related genes in several brain areas
Fielden, J.A., 1998. Testosterone changes during vicarious experiences of male mice. Genes Brain Behav. 6, 529–539.
of winning and losing among fans at sporting events. Physiol. Behavior. Blair, R.J.R., 2004. The neurobiology of aggression. In: Charney, D.S., Nestler, E.J. (Eds.),
65, 59–62, with permission from Elsevier. Neurobiology of Mental Illness. Oxford University Press, Oxford, pp. 1076–1085.
Hormones and the Development and Expression of Aggressive Behavior 167

Blair, R.J.R., Peschardt, K.S., Budhani, S., Pine, D.S., 2006. Neurobiology of Choi, P.Y., Pope, H.G.J., 1994. Violence toward women and elicit androgenic-anabolic
aggression in children. In: Nelson, R.J. (Ed.), Biology of Aggression. Oxford steroid use. Ann. Clin. Psychiatry 6, 21–25.
University Press, Oxford, pp. 351–368. Christie, M.H., Barfield, R.J., 1979. Effects of castration and home cage residency on
Blanchard, R.J., Blanchard, D.C., 1989. Attack and defense in rodents as ethoex- aggressive behavior in rats. Horm. Behav. 13, 85–91.
perimental models for the study of emotion. Prog. Neuropsychopharmacol. Biol. Clark, A.S., Barber, D.M., 1994. Anabolic-androgenic steroids and aggression in
Psychiatry 13, S3–S14. castrated male rats. Physiol. Behav. 56, 1107–1113.
Bonetti, A., Tirelli, F., Catapano, A., Dazzi, D., Dei Cas, A., Solito, F., Ceda, G., Coccaro, E.F., Kavoussi, R.J., 1997. Fluoxetine and impulsive aggressive behavior in
Reverberi, C., Monica, C., Pipitone, S., Elia, G., Spattini, M., Magnati, G., 2008. personality-disordered subjects. Arch. Gen. Psychiatry 54, 1081–1088.
Side effects of anabolic androgenic steroids abuse. Int. J. Sports Med. 29, Coccaro, E.F., McCloskey, M.S., Fitzgerald, D.A., Phan, K.L., 2007. Amygdala and
679–687. orbitofrontal reactivity to social threat in individuals with impulsive aggression. Biol.
Bouissou, M.F., 1983. Androgens, aggressive behaviour and social relationships in Psychiatry 62, 168–178.
higher mammals. Horm. Res. 18, 43–61. Collaer, M.L., Hines, M., 1995. Human behavioral sex differences: a role for gonadal
Brain, P.F., 1983. Pituitary-gonadal influences on. social aggression. In: Svare, B. hormones during early development? Psychol. Bull. 118, 55–107.
(Ed.), Hormones and Aggressive Behavior. Plenum, New York, pp. 3–25. Cologer-Clifford, A., Simon, N.G., Richter, M.L., Smoluk, S.A., Lu, S., 1999. Andro-
Bronson, F.H., Desjardins, C., 1969. Aggressive behavior and seminal vesicle function gens and estrogens modulate 5-HT1A and 5-HT1B agonist effects on aggression.
in mice: differential sensitivity to androgen given neonatally. Endocrinology 85, Physiol. Behav. 65, 823–828.
971–974. Cooke, B.M., Shukla, D., 2011. Double helix: reciprocity between juvenile play and
Bronson, F.H., Desjardins, C., 1970. Neonatal androgen administration and adult brain development. Dev. Cogn. Neurosci. 1, 459–470.
aggressiveness in female mice. Gen. Comp. Endocrinol. 15, 320–325. Cooke, B.M., Woolley, C.S., 2009. Effects of prepubertal gonadectomy on a male-
Brunner, H.G., Nelen, M., Breakefield, X.O., Ropers, H.H., van Oost, B.A., 1993. typical behavior and excitatory synaptic transmission in the amygdala. Dev.
Abnormal behavior associated with a point mutation in the structural gene for Neurobiol. 69, 141–152.
monoamine oxidase A. Science 262, 578–580. Cordero, M.I., Poirier, G.L., Marquez, C., Veenit, V., Fontana, X., Salehi, B.,
Buckholtz, J.W., Meyer-Lindenberg, A., 2008. MAOA and the neurogenetic archi- Ansermet, F., Sandi, C., 2012. Evidence for biological roots in the transgenera-
tecture of human aggression. Trends Neurosci. 31, 120–129. tional transmission of intimate partner violence. Transl. Psychiatry 2, e106.
Burgdorf, J., Panksepp, J., Beinfeld, M.C., Kroes, R.A., Moskal, J.R., 2006. Regional Crews, D., 2003. The development of phenotypic plasticity: where biology and
brain cholecystokinin changes as a function of rough-and-tumble play behavior in psychology meet. Dev. Psychobiol. 43, 1–10.
adolescent rats. Peptides 27, 172–177. Crews, D., Moore, M.C., 1986. Evolution of mechanisms controlling mating behavior.
Butter, C.M., Snyder, D.R., 1972. Alterations in aversive and aggressive behaviors Science 231, 121–125.
following orbital frontal lesions in rhesus monkeys. Acta Neurobiol. Exp. 32, Crowell, N.A., Burgess, A.W., 1996. Understanding Violence Against Women. National
525–565. Academies Press, Washington, DC.
Butter, C.M., Snyder, D.R., McDonald, J.A., 1970. Effects of orbital frontal lesions on Cunningham, R.L., McGinnis, M.Y., 2007. Factors influencing aggression toward
aversive and aggressive behaviors in rhesus monkeys. J. Comp. Physiol. Psychol. females by male rats exposed to anabolic androgenic steroids during puberty.
72, 132–144. Horm. Behav. 51, 135–141.
van den Berg, C.L., Hol, T., Van Ree, J.M., Spruijt, B.M., Everts, H., Koolhaas, J.M., Dabbs Jr., J.M., Frady, R.L., Carr, T.S., Besch, N.F., 1987. Saliva testosterone and
1999. Play is indispensable for an adequate development of coping with social criminal violence in young adult prison inmates. Psychosom. Med. 49, 174–182.
challenges in the rat. Dev. Psychobiol. 34, 129–138. Dabbs Jr., J.M., Hargrove, M.F., 1997. Age, testosterone, and behavior among female
Calarge, C., Andreasen, N.C., O’Leary, D.S., 2003. Visualizing how one brain prison inmates. Psychosom. Med. 59, 477–480.
understands another: a PET study of theory of mind. Am. J. Psychiatry 160, Dabbs Jr., J.M., Morris, R., 1990. Testosterone, social class, and antisocial behavior in
1954–1964. a sample of 4,462 men. Psychol. Sci. 1, 209–211.
Caldwell, G.S., Glickman, S.E., Smith, E.R., 1984. Seasonal aggression independent of Dabbs, J.M., Ruback, R.B., Frady, R.L., Hopper, C.H., Sgoutas, D.S., 1988. Saliva
seasonal testosterone in wood rats. Proc. Natl. Acad. Sci. U.S.A. 81, 5255–5257. testosterone and criminal violence among women. Pers. Individ. Differ. 9,
Carlier, M., Roubertoux, P.L., Pastoret, C., 1991. The Y chromosome effect on 269–275.
intermale aggression in mice depends on the maternal environment. Genetics 129, DaVanzo, J.P., Sydow, M., Garris, D.R., 1983. Influence of isolation and training on
231–236. fighting in mice with olfactory bulb lesions. Physiol. Behav. 31, 857–860.
Carré, J.M., McCormick, C.M., Hariri, A.R., 2011. The social neuroendocrinology of Davidson, R.J., Putnam, K.M., Larson, C.L., 2000. Dysfunction in the neural
human aggression. Psychoneuroendocrinology 36, 935–944. circuitry of emotion regulation- a possible prelude to violence. Science 289,
Cases, O., Seif, I., Grimsby, J., Caspar, P., Chen, K., Pourin, S., Muller, U., Aguet, M., 591–594.
Babinet, C., Shih, J.C., DeMaeyer, E., 1995. Aggressive behavior and altered Davis, E.S., Marler, C.A., 2004. C-fos changes following an aggressive encounter in
amounts of brain serotonin and norepinephrine in mice lacking MAOA. Science female California mice: a synthesis of behavior, hormone changes and neural
268, 1763–1766. activity. Neuroscience 127, 611–624.
Caspi, A., McClay, J., Moffitt, T.E., Mill, J., Martin, J., Craig, I.W., Taylor, A., De Boer, S.F., Lesourd, M., Mocaer, E., Koolhaas, J., 1999. Selective antiaggressive
Poulton, R., 2003. Role of genotype in the cycle of violence in maltreated children. effects of alnespirone in resident-intruder test are mediated via
Science 297, 851–854. 5-hydroxytryptamine 1A receptors: a comparative, pharmacological study with
Casto, J.M., Ward, O.B., Bartke, A., 2003. Play, copulation, anatomy, and testosterone 8-hydroxy-2-dipropylaminotetralin, ipsapirone, buspirone, eltoprazine, and
in gonadally intact male rats prenatally exposed to flutamide. Physiol. Behav. 79, WAY-100635. J. Pharmacol. Exp. Ther. 288, 1125–1133.
633–641. De Lorme, K.C., Sisk, C.L., 2013. Pubertal testosterone programs context-appropriate
Cervantes, M.C., Taravosh-Lahn, K., Wommack, J.C., Delville, Y., 2007. Character- agonistic behavior and associated neural activation patterns in male Syrian
ization of offensive responses during the maturation of play-fighting into aggression hamsters. Physiol. Behav. 112–113, 1–7.
in male golden hamsters. Dev. Psychobiol. 49, 87–97. De Boer, S.F., Koolhaas, J., 2005. 5HT1A and 5HT1B receptor agonists and
Chakravorty, S.G., Halbreich, U., 1997. The influence of estrogen on monoamine aggression: a pharmacological challenge of the serotonin deficiency hypothesis.
oxidase activity. Psychopharmacol. Bull. 33, 229–233. Eur. J. Pharmacol. 526, 125–139.
Chase, I.D., Bartolomeo, C., Dugatkin, L.A., 1994. Aggressive interactions and inter- DeBold, J.F., Miczek, K.A., 1981. Sexual dimorphism in the hormonal control of
contest interval-how long do winners keep winning? Anim. Behav. 48, 393–400. aggressive behavior of rats. Pharmacol. Biochem. Behav. 14 (Suppl. 1), 89–93.
Cherek, D.R., Tcheremissine, O.V., Lane, S.D., 2006. Psychopharmacology of human DeBold, J.F., Miczek, K.A., 1984. Aggression persists after ovariectomy in female rats.
aggression. In: Nelson, R.J. (Ed.), Biology of Aggression. Oxford University Press, Horm. Behav. 18, 177–190.
Oxford, pp. 424–446. Delville, Y., David, J.T., Taravosh-Lahn, K., Wommack, J.C., 2003. Stress and the
Chiavegatto, S., Dawson, V.L., Mamounas, L.A., Koliatsos, V.E., Dawson, T.M., development of agonistic behavior in golden hamsters. Horm. Behav. 44, 263–270.
Nelson, R.J., 2001. Brain serotonin dysfunction accounts for aggression in male Delville, Y., De Vries, G.J., Ferris, C.F., 2000. Neural connections of the anterior
mice lacking neuronal nitric oxide synthase. Proc. Natl. Acad. Sci. U.S.A. 98, hypothalamus and agonistic behavior in golden hamsters. Brain Behav. Evol. 55,
1277–1281. 53–76.
Choi, G., Dong, H., Murphy, A., Valenzuela, D., Yancopoulos, G., Swanson, L., Delville, Y., Melloni Jr., R.H., Ferris, C.F., 1998. Behavioral and neurobiological
Anderson, D., 2005. Lhx6 delineates a pathway mediating innate reproductive consequences of social subjugation during puberty in golden hamsters.
behaviors from the amygdala to the hypothalamus. Neuron 46, 647–660. J. Neurosci. 18, 2667–2672.
168 Hormones and the Development and Expression of Aggressive Behavior

Delville, Y., Newman, M.L., Wommack, J.C., Taravosh-Lahn, K., Cervantes, M.C., Frank, L.G., Glickman, S.E., Licht, P., 1991. Fatal sibling aggression, precocial
2005. Developlment of aggression. In: Nelson, R.J. (Ed.), Biology of Aggression. development, and androgens in neonatal spotted hyenas. Science 252, 702–704.
Oxford University Press, New York, pp. 327–350. Frazier, C.R.M., Trainor, B.C., Cravens, C.J., Whitney, T.K., Marler, C.A., 2006.
Demas, G.E., Cooper, M.A., Albers, H.E., Soma, K.K., 2007. Novel mechanisms Paternal behavior influences development of aggression and vasopressin expres-
underlying neuroendocrine regulation of aggression: a synthesis of rodent, avian, sion in male California mouse offspring. Horm. Behav. 50, 699–707.
and primate studies. In: Blaustein (Ed.), Handbook of Neurochemistry and Fuxjager, M.J., Forbes-Lorman, R.M., Coss, D.J., Auger, C.J., Auger, A.P.,
Molecular Neurobiology. Springer, New York, pp. 338–372. Marler, C.A., 2010. Winning territorial disputes selectively enhances androgen
Demas, G.E., Eliasson, M.J., Dawson, T.M., Dawson, V.L., Kriegsfeld, L.J., sensitivity in neural pathways related to motivation and social aggression. Proc.
Nelson, R.J., Snyder, S.H., 1997. Inhibition of neuronal nitric oxide synthase Natl. Acad. Sci. U.S.A. 107, 12393–12398.
increases aggressive behavior in mice. Mol. Med. 3, 610–616. Fuxjager, M.J., Mast, G., Becker, E.A., Marler, C.A., 2009. The ‘home advantage’ is
Demas, G.E., Moffatt, C.A., Drazen, D.L., Nelson, R.J., 1999. Castration does not necessary for a full winner effect and changes in post-encounter testosterone.
inhibit aggressive behavior in adult male prairie voles (Microtus ochrogaster). Horm. Behav. 56, 214–219.
Physiol. Behav. 66, 59–62. Fuxjager, M.J., Oyegbile, T.O., Marler, C.A., 2011. Independent and additive contri-
Demas, G.E., Polacek, K.M., Durazzo, A., Jasnow, A.M., 2004. Adrenal hormones butions of postvictory testosterone and social experience to the development of the
mediate melatonin-induced increases in aggression in male Siberian hamsters winner effect. Endocrinology 152, 3422–3429.
(Phodopus sungorus). Horm. Behav. 46, 582–591. Galligani, N., Renick, A., Hansen, S., 1996. Personality profile of mean using anabolic
DeVries, A.C., Young, S.W., Nelson, R.J., 1997. Reduced aggressive behavior in mice androgenic steroids. Horm. Behav. 30, 170–175.
with targeted disruption of the oxytocin gene. J. Neuroendocrinol. 9, 363–368. Gammie, S.C., Auger, A.P., Jessen, H.M., Vanzo, R.J., Awad, T.A., Stevenson, S.A.,
Dixson, A.F., 1980. Androgens and aggressive-behavior in primates – review. 2007. Altered gene expression in mice selected for high maternal aggression.
Aggress. Behav. 6, 37–67. Genes Brain Behav. 6, 432–443.
Dixson, A.F., Lloyd, S.A.C., 1988. Effects of hypothalamic lesions upon sexual and Gerra, G., Zaimovic, A., Raggi, M.A., Moi, G., Branchi, B., Moroni, M., Brambilla, F.,
social behaviour of the male common marmoset (Callithrix jacchus). Brain Res. 2007. Experimentally induced aggressiveness in heroin-dependent patients treated
463, 317–329. with buprenorphine: comparison of patients receiving methadone and healthy
Dloniak, S.M., French, J.A., Holekamp, K.E., 2006. Rank-related maternal effects of subjects. Psychiatry Res. 149, 201–213.
androgens on behaviour in wild spotted hyaenas. Nature 440, 1190–1193. Gleason, E.D., Fuxjager, M.J., Oyegbile, T.O., Marler, C.A., 2009. Testosterone release
Doering, C.H., Kraemer, H.C., Brodie, H.K., Hamburg, D.A., 1975. A cycle of plasma and social context: when it occurs and why. Front. Neuroendocrinol. 30, 460–469.
testosterone in the human male. J. Clin. Endocrinol. Metab. 40, 492–500. Godar, S.C., Bortolato, M., Frau, R., Dousti, M., Chen, K., Shih, J., 2011. Maladaptive
Dorfman, H.M., Meyer-Lindenberg, A., Buckholtz, J.W., 2014. Neurobiological defensive behaviours in monoamine oxidase A-deficient mice. Int. J.
mechanisms for impulsive-aggression: the role of MAOA. Curr. Top. Behav. Neuropsychopharmacol. 14, 1195–1207.
Neurosci. 17, 297–313. Goetz, S.M.M., Tang, L., Thomason, M.E., Diamond, M.P., Hariri, A.H., Carré, J.M.,
Duke, A.A., Begue, L., Bell, R., Eisenlohr-Moul, T., 2013. Revising the serotonin- 2014. Testosterone rapidly increases neural reactivity to threat in healthy men:
aggression relation in humans: a meta-analysis. Psychol. Bull. 139, 1148–1172. a novel two-step pharmacological challenge paradigm. Biol. Psychiatry 76,
Earley, R.L., Dugatkin, L.A., 2002. Eavesdropping on visual cues in green swordtail 324–331.
(Xiphophorus helleri ) fights: a case for networking. Proc. R. Soc. Lond. Ser. B Goodson, J.L., 2005. The vertebrate social behavior network: evolutionary themes and
269, 943–952. variations. Horm. Behav. 48, 11–22.
Eastman, N., Campbell, C., 2006. Neuroscience and legal determination of criminal Goodson, J.L., Bass, A.H., 2001. Social behavior functions and related anatomical
responsibility. Nat. Rev. Neurosci. 7, 311–318. characteristics of vasotocin/vasopressin systems in vertebrates. Brain Res. Brain
Edwards, D.A., 1970. Post-neonatal androgenization and adult aggressive behavior in Res. Rev. 35, 246–265.
female mice. Physiol. Behav. 5, 465–467. Grafman, J., Schwab, K., Warden, D., Pridgen, A., Brown, H.R., Salazar, A.M., 1996.
Ehrenkranz, J., Bliss, E., Sheard, M.H., 1974. Plasma testosterone: correlation with Frontal lobe injuries, violence, and aggression: a report of the Vietnam head injury
aggressive behavior and social dominance in man. Psychosom. Med. 36, study. Neurology 46, 1231–1238.
469–475. Greenberg, B.D., Tolliver, T.J., Huang, S.J., Li, Q., Bengel, D., Murphy, D.L., 1999.
Eichmann, F., Holst, D.V., 1999. Organization of territorial marking behavior by Genetic variation in the serotonin transporter promoter region affects serotonin
testosterone during puberty in male tree shrews. Physiol. Behav. 65, 785–791. uptake in human blood platelets. Am. J. Med. Genet. 88, 83–87.
Emery, N.J., Capitanio, J.P., Mason, W.A., Machado, C.J., Mendoza, S.P., Greenberg, G.D., Howerton, C.L., Trainor, B.C., 2014. Fighting in the home cage:
Amaral, D.G., 2001. The effects of bilateral lesions of the amygdala on dyadic agonistic encounters and effects on neurobiological markers within the social
social interactions in rhesus monkeys (Macaca mulatta). Behav. Neurosci. 115, decision-making network of house mice (Mus musculus). Neurosci. Lett. 566,
515–544. 151–155.
Farrell, S.F., McGinnis, M.Y., 2004. Long-term effects of pubertal anabolic-androgenic Greene, J.D., Sommerville, R.B., Nystrom, L.E., Darley, J.M., Cohen, J.D., 2001. An
steroid exposure on reproductive and aggressive behaviors in male rats. Horm. fMRI investigation of emotional engagement in moral judgement. Science 293,
Behav. 46, 193–203. 2105–2108.
Ferrando, A.A., Wolfe, R.R., 2007. Restoration of hormonal action and muscle protein. Gregg, T.R., Siegel, A., 2001. Brain structures and neurotransmitters regulating
Crit. Care Med. 35, S630–S634. aggression in cats: implications for human aggression. Prog. Neuro-
Ferrari, P.F., Lowther, S., Tidbury, H., Greengrass, P., Wilson, C.A., Horton, R.W., psychopharmacol. Biol. Psychiatry 25, 91–140.
1999. The influence of gender and age on neonatal rat hypothalamic 5-HT1A and Grimes, J.M., Melloni Jr., R.H., 2006. Prolonged alterations in the serotonin neural
5-HT2A receptors. Cell. Mol. Neurobiol. 19, 775–784. system following the cessation of adolescent anabolic-androgenic steroid exposure
Ferris, C.F., 2006. Neuroplasticity and aggression: an interaction between vasopressin in hamsters (Mesocricetus auratus). Behav. Neurosci. 120, 1242–1251.
and serotonin. In: Nelson, R.J. (Ed.), Biology of Aggression. Oxford University Press, Grimes, J.M., Melloni, R.H.J., 2005. Serotonin-1B receptor activity and expression
Oxford, pp. 163–175. modulate the aggression-stimulating effects of adolescent anabolic steroid expo-
Ficks, C.A., Waldman, I.D., 2014. Candidate genes for aggression and antisocial sure in hamsters. Behav. Neurosci. 119, 1184–1194.
behavior: a meta-analysis of association studies of the 5HTTLPR and MAOA- Grimes, J.M., Ricci, L.A., Melloni, R.H.J., 2007. Alterations in anterior hypothalamic
uVNTR. Behav. Genet. 44, 427–444. vasopressin, but not serotonin, correlate with the temporal onset of aggressive
Field, E.F., Whishaw, I.Q., Pellis, S.M., Watson, N.V., 2006. Play fighting in androgen- behavior during adolescent anabolic-androgenic steroid exposure in hamsters
insensitive tfm rats: evidence that androgen receptors are necessary for the (Mesocricetus auratus). Behav. Neurosci. 121, 941–948.
development of adult playful attack and defense. Dev. Psychobiol. 48, 111–120. Guillot, P.-V., Roubertoux, P.L., Crusio, W.E., 1994. Hippocampal mossy fiber distri-
Fish, E.W., Faccidomo, S., Miczek, K.A., 1999. Aggression heightened by alcohol or butions and intermale aggression in seven inbred mouse strains. Brain Res. 660,
social instigation in mice: reduction by the 5-HT1B receptor agonist CP-94,253. 167–169.
Psychopharmacology 146, 391–399. Gutzler, S.J., Karom, M., Erwin, W.D., Albers, H.E., 2010. Arginine-vasopressin and
Fleshner, M., Laudenslager, M.L., Simons, L., Maier, S.F., 1989. Reduced serum the regulation of aggression in female Syrian hamsters (Mesocricetus auratus). Eur.
antibodies associated with social defeat in rats. Physiol. Behav. 45, 1183–1187. J. Neurosci. 31, 1655–1663.
Foley, D.L., Eaves, L.J., Wormley, B., Silberg, J.L., Maes, H.H., Kuhn, J., Riley, B., Haller, J., 2013. The neurobiology of abnormal manifestations of aggressionda review
2004. Childhood adversity, monoamine oxidase a genotype, and risk for conduct of hypothalamic mechanisms in cats, rodents, and humans. Brain Res. Bull. 93,
disorder. Arch. Gen. Psychiatry 61, 738–744. 97–109.
Hormones and the Development and Expression of Aggressive Behavior 169

Haller, J., 2014. The glucocorticoid/aggression relationship in animals and humans: an Huxsoll, C.C., Price, E.O., Adams, T.E., 1998. Testis function, carcass traits, and
analysis sensitive to behavioral characteristics, glucocorticoid secretion patterns, aggressive behavior of beef bulls actively immunized against gonadotropin-releasing
and neural mechanisms. Curr. Top. Behav. Neurosci. 17, 73–109. hormone. J. Anim. Sci. 76, 1760–1766.
Haller, J., Abraham, I., Zelena, D., Juhasz, G., Makara, G.B., Kruk, M.R., 1989. Insel, T.R., 1992. Oxytocin- a neuropeptide for affiliation: evidence from behavioral, receptor
Aggressive experience affects the sensitivity of neurons towards pharmacological autoradiographic, and comparative studies. Psychoneuroendocrinology 17, 3–35.
treatment in the hypothalamic attack area. Behav. Pharmacol. 9, 469–475. Jasnow, A.M., Drazen, D.L., Huhman, K.L., Nelson, R.J., Demas, G.E., 2001. Acute
Haller, J., Halasz, J., Mikics, E., Kruk, M.R., 2004. Chronic glucocorticoid deficiency- and chronic social defeat suppresses humoral immunity of male Syrian hamsters
induced abnormal aggression, autonomic hypoarousal, and social deficit in rats. (Mesocricetus auratus). Horm. Behav. 40, 428–433.
J. Neuroendocrinol. 16, 550–557. Jasnow, A.M., Huhman, K.L., Bartness, T.J., Demas, G.E., 2000. Short-day increases
Haller, J., Harold, G., Sandi, C., Neumann, I.D., 2014. Effects of adverse early-life in aggression are inversely related to circulating testosterone concentrations in
events on aggression and anti-social behaviours in animals and humans. male siberian hamsters (Phodopus sungorus). Horm. Behav. 38, 102–110.
J. Neuroendocrinol. 26, 724–738. Jasnow, A.M., Huhman, K.L., Bartness, T.J., Demas, G.E., 2002. Short days and
Haller, J., Kruk, M.R., 2006. Normal and abnormal aggression: human disorders and exogenous melatonin increase aggression of male Syrian hamsters (Mesocricetus
novel laboratory models. Neurosci. Biobehav. Rev. 30, 292–303. auratus). Horm. Behav. 42, 13–20.
Haller, J., van de Schraaf, J., Kruk, M.R., 2001. Deviant forms of aggression in Jiménez, A., Young, L.J., Rio, R.T., LaPrairie, J.L., Gonzalez-Mariscal, G., 2015.
glucocorticoid hyporeactive rats: a model for ’pathological’ aggression? J. Neuro- Neuroanatomical distribution of oxytocin receptor binding in the female rabbit
endocrinol. 13, 102–107. forebrain: variations across the reproductive cycle. Brain Res. 1629, 329–339.
Hariri, A.R., Mattay, V.S., Tessitore, A., Kolachana, B., Fera, F., Goldman, D., Johnson, F., Whalen, R.E., 1988. Testicular hormones reduce individual differences in
Egan, M.F., Weinberger, D.R., 2002. Serotonin transporter genetic variation and the aggressive behavior of male mice: a theory of hormone action. Neurosci.
the response of the human amygdala. Science 297, 400–403. Biobehav. Rev. 12, 93–99.
Harrison, R.J., Connor, D.F., Nowak, C., Nash, K., Melloni Jr., R.H., 2000. Chronic Joppa, M.A., Rowe, R.K., Meisel, R.L., 1997. Effects of serotonin 1A or 1B receptor
anabolic-androgenic steroid treatment during adolescence increases anterior agonists on social aggression in male and female Syrian hamsters. Pharmacol.
hypothalamic vasopressin and aggression in intact hamsters. Psychoneur- Biochem. Behav. 58, 349–353.
oendocrinology 25, 317–338. Ketterson, E.D., Nolan, V., 1992. Hormones and life histories: an integrative approach.
Hasen, N.S., Gammie, S.C., 2005. Differential fos activation in virgin and lactating Am. Nat. 140, S33–S62.
mice in response to an intruder. Physiol. Behav. 84, 681–695. Kim-Cohen, J., Caspi, A., Taylor, A., Williams, B., Newcombe, R., Craig, I.W.,
Haug, M., Spetz, J.F., Ouss-Schlegel, M.L., Benton, D., Brain, P.F., 1986. Effects of Moffitt, T.E., 2006. MAOA, maltreatment, and gene-environment interaction pre-
gender, gonadectomy and social status on attack directed towards female intruders dicting children’s mental health: new evidence and a meta-analysis. Mol.
by resident mice. Physiol. Behav. 37, 533–537. Psychiatry 11, 903–913.
Hayden-Hixson, D.M., Ferris, C.F., 1991. Steroid-specific regulation of agonistic Kirsch, P., Esslinger, C., Chen, Q., Mier, D., Lis, S., Siddhanti, S., Gruppe, H.,
responding in the anterior hypothalamus of male hamsters. Physiol. Behav. 50, Mattay, V.S., Gallhofer, B., Meyer-Lindenberg, A., 2005. Oxytocin modulates
793–799. neural circuitry for social cognition and fear in humans. J. Neurosci. 25, 11489–
Heimburger, R.F., Whillock, C.C., Kalsbeck, J.E., 1966. Stereotaxic amygdalotomy for 11493.
epilepsy with aggressive behavior. J. Am. Med. Assoc. 198, 741–745. Knutson, B., Burgdorf, J., Panksepp, J., 1998. Anticipation of play elicits high-
Heimovics, S.A., Prior, N.H., Maddison, C.J., Soma, K.K., 2012. Rapid and widespread frequency ultrasonic vocalizations in young rats. J. Comp. Psychol. 112, 65–73.
effects of 17b-estradiol on intracellular signaling in the male songbird brain: Knutson, B., Burgdorf, J., Panksepp, J., 2002. Ultrasonic vocalizations as indices of
a seasonal comparison. Endocrinology 153, 1364–1376. affective states in rats. Psychol. Bull. 128, 961–977.
Heimovics, S.A., Trainor, B.C., Soma, K.K., 2015. Rapid effects of estradiol on aggression Kollack-Walker, S., Newman, S.W., 1995. Mating and agonistic behavior produce
in birds and mice: the fast and the furious. Integr. Comp. Biol. 55, 281–293. different patters of Fos immunolabeling in the male Syrian hamster brain.
Henderson, L.P., Penatti, C.A.A., Jones, B.L., Yang, P., Clark, A.S., 2006. Anabolic Neuroscience 66, 721–736.
androgenic steroids and forebrain GABAergic transmission. Neuroscience 138, Koolhaas, J.M., Coppens, C.M., de Boer, S.F., Buwalda, B., Meerlo, P.,
793–799. Timmermans, P.J., 2013. The resident-intruder paradigm: a standardized test for
Ho, J.M., Murray, J.H., Demas, G.E., Goodson, J.L., 2010. Vasopressin cell groups aggression, violence and social stress. J. Vis. Exp. 77, e4367.
exhibit strongly divergent responses to copulation and male-male interactions in Krakowski, M., 2003. Violence and serotonin: influence of impulse control, affect
mice. Horm. Behav. 58, 368–377. regulation, and social functioning. J. Neuropsychiatry Clin. Neurosci. 15, 294–305.
Holt, A.G., Newman, S.W., 2004. Distribution of methionine and leucine enkephalin Kreuz, L.E., Rose, R.M., 1972. Assessment of aggressive behavior and plasma
neurons within the social behavior circuitry of the male Syrian hamster brain. Brain testosterone in a young criminal population. Psychosom. Med. 34, 321–332.
Res. 1030, 28–48. Kriegsfeld, L.J., Dawson, T.M., Dawson, V.L., Nelson, R.J., Snyder, S.H., 1997.
Hong, W., Kim, D-w, Anderson, D.J., 2014. Antagonistic control of social versus Aggressive behavior in male mice lacking the gene for neuronal nitric oxide syn-
repetitive self-grooming behaviors by separable amygdala neuronal subsets. Cell thase requires testosterone. Brain Res. 769, 66–70.
158, 1348–1361. Kruk, M.R., 1991. Ethology and pharmacology of hypothalamic aggression in the rat.
Howerton, C.L., Garner, J.P., Mench, J.A., 2008. Effects of a running wheel-igloo Neurosci. Biobehav. Rev. 15, 527–538.
enrichment on aggression, hierarchy linearity, and stereotypy in group-housed Kruk, M.R., 2014. Hypothalamic attack: a wonderful artifact or a useful perspective on
male CD-1 (ICR) mice. Appl. Anim. Behav. Sci. 115, 90–103. escalation and pathology in aggression? A viewpoint. Curr. Top. Behav. Neurosci.
Huhman, K.L., Bunnell, B.N., Mougey, E.H., Meyerhoff, J.L., 1990. Effects of social 17, 143–188.
conflict on POMC-derived peptides and glucocorticoids in male golden hamsters. Kruk, M.R., Van der Lann, C.E., Mos, J., Van der Poel, A.M., Meelis, W., Olivier, B.,
Physiol. Behav. 47, 949–956. 1984. Comparison of aggressive behaviour induced by electrical stimulation in the
Huhman, K.L., Jasnow, A.M., 2005. Conditioned defeat. In: Nelson, R.J. (Ed.), Biology hypothalamus of male and female rats. Prog. Brain Res. 61, 303–314.
of Aggression. Oxford University Press, Oxford. Kudryavtseva, N.N., 2000. Agonistic behavior: a model, experimental studies, and
Huhman, K.L., Moore, T.O., Ferris, C.F., Mougey, E.H., Meyerhoff, J.L., 1991. Acute perspectives. Aggress. Behav. 26, 241–256.
and repeated exposure to social conflict in male golden hamsters: increases in Kurian, J.R., Bychowski, M.E., Forbes-Lorman, R.M., Auger, C.J., Auger, A.P., 2008.
plasma POMC-peptides and cortisol and decreases in plasma testosterone. Horm. Mecp2 organizes juvenile social behavior in a sex-specific manner. J. Neurosci. 28,
Behav. 25, 206–216. 7137–7142.
Huhman, K.L., Moore, T.O., Mougey, E.H., Meyerhoff, J.L., 1992. Hormonal responses Kyomen, H.H., Nobel, K.W., Wei, J.Y., 1991. The use of estrogen to decrease
to fighting in hamsters: separation of physical and psychological causes. Physiol. aggressive physical behavior in elderly men with dementia. J. Am. Geriatr. Soc. 39,
Behav. 51, 1083–1086. 1110–1112.
Huhman, K.L., Solomon, M.B., Janicki, M., Harmon, A.C., Lin, S.M., Israel, J.E., Kyomen, H.H., Satlin, A., Hennen, J., Wei, J.Y., 1999. Estrogen therapy and aggressive
Jasnow, A.M., 2003. Conditioned defeat in male and female Syrian hamsters. behavior in elderly patients with moderate-to-severe dementia: results from a short-
Horm. Behav. 44, 293–299. term, randomized, double-blind trial. J. Am. Geriatr. Soc. 7, 339–348.
Huizinga, D., Haberstick, B.C., Smolen, A., Menard, S., Young, S.E., Corley, R.P., Laredo, S.A., Orr, V.N., McMackin, M.Z., Trainor, B.C., 2014a. The effects of exog-
Stallings, M.C., Grotpeter, J., Hewitt, J.K., 2006. Childhood maltreatment, enous melatonin and melatonin receptor blockade on aggression and estrogen-
subsequent antisocial behavior, and the role of monoamine oxidase A genotype. dependent gene expression in male California mice (Peromyscus californicus).
Biol. Psychiatry 60, 677–683. Physiol. Behav. 128, 86–91.
170 Hormones and the Development and Expression of Aggressive Behavior

Laredo, S.A., Villalon Landeros, R., Trainor, B.C., 2014b. Rapid effects of estrogens on Martinez-Sanchis, S., Brain, P.F., Salvador, A., Simon, V.M., 1996. Long-term chronic
behavior: environmental modulation and molecular mechanisms. Front. Neuro- treatment with stanozolol lacks significant effects on aggression and activity in
endocrinol. 35, 447–458. young and adult male laboratory mice. Gen. Pharmacol. 27, 293–298.
Laredo, S.A., Villalon Landeros, R., Dooley, J.C., Steinman, M.Q., Orr, V.N., Silva, A.L., Mathews, V.P., Kronenbeerger, W.G., Wang, Y., Lurito, J.T., Lowe, M.J., Dunn, D.W.,
Crean, K.K., Robles, C.F., Trainor, B.C., 2013. Nongenomic effects of estradiol on 2005. Media violence exposure and frontal lobe activation measured by functional
aggression under short day photoperiods. Horm. Behav. 64, 557–565. magnetic resonance imaging in aggressive and nonaggressive adolescents.
Lee, H., Kim, D.-W., Remedios, R., Anthony, T.E., Chang, A., Madisen, L., Zeng, H., J. Comput. Assist. Tomogr. 29, 287–292.
Anderson, D.J., 2014. Scalable control of mounting and attack by Esr1þ neurons Mathiak, K., Weber, R., 2006. Toward brain correlates of natural behavior: fMRI during
in the ventromedial hypothalamus. Nature 509, 627–632. violent video games. Hum. Brain Mapp. 27, 948–956.
Lesch, K.P., Merschdorf, U., 2000. Impulsivity, aggression, and serotonin: a molecular Matsumoto, T., Honda, S., Harada, N., 2003. Alteration in sex-specific behaviors in
psychobiological perspective. Behav. Sci. Law 18, 581–604. male mice lacking the aromatase gene. Neuroendocrinology 77, 416–424.
Leshner, A., Moyer, J., 1975. Androgens and agnoistic behavior in mice: relevance Matthews, R., 1979. Testosterone levels in aggressive offenders. In: Sandler, M. (Ed.),
to aggression and irrelevance to avoidance-of-attack. Physiol. Behav. 15, Psychopharmacology of Aggression. Raven Press, New York, pp. 123–130.
695–699. Maxson, S.C., Didier-Erickson, A., Ogawa, S., 1989. The Y chromosome, social
Leshner, A.I., 1979. Kinds of hormonal effects on behavior: a new view. Neurosci. signals, and offense in mice. Behav. Neural Biol. 52, 251–259.
Biobehav. Rev. 3, 69–73. Maxson, S.C., Roubertoux, P.L., Guillot, P.-V., Goldman, D., 2001. The genetics of
Leshner, A.I., Korn, S.J., Mixon, J.F., Rosenthal, C., Besser, A.K., 1980. Effects of aggression: from mice to men. In: Martinez, M. (Ed.), Prevention and Control of
corticosterone on submissiveness in mice: some temporal and theoretical Aggression and the Impact on Its Victims. Kluwer Academic, New York, pp. 71–81.
considerations. Physiol. Behav. 24, 283–288. Mazur, A., Booth, A., 1998. Testosterone and dominance in men. Behav. Brain Sci.
Lima, F.B., Spinelli de Oliveira, E., 2014. What is the impact of low testosterone levels 21, 353–397.
on the anatomical and behavioral repertoire of long-term enriched housing of male McGinnis, M.Y., 2004. Anabolic androgenic steroids and aggression: studies using
mice? Behav. Process. 108, 57–64. animal models. Ann. N.Y. Acad. Sci. 1036, 399–415.
Lin, D.Y., Boyle, M.P., Dollar, P., Lee, H., Lein, E.S., Perona, P., Anderson, D.J., 2011. McGinnis, M.Y., Lumia, A.R., Breuer, M.E., Possidente, B., 2002a. Physical provo-
Functional identification of an aggressive locus in the mouse hypothalamus. Nature cation potentiates aggression in male rats receiving anabolic androgenic steroids.
470, 221–226. Horm. Behav. 41, 101–110.
Lincoln, G.A., Guinness, F., Short, R.V., 1972. The way in which testosterone controls McGinnis, M.Y., Lumia, A.R., Possidente, B.P., 2002b. Effects of withdrawal from
the social and sexual behavior of the Red Deer stag (Cervus Elaphus). Horm. anabolic androgenic steroids on aggression in adult male rats. Physiol. Behav. 75,
Behav. 3, 375–396. 541–549.
Lipp, H.P., Hunsperger, R.W., 1978. Threat, attack, and flight elicited by electrical Meaney, M.J., McEwen, B.S., 1986. Testosterone implants into the amygdala during
stimulation of the ventromedial hypothalamus of the marmoset monkey Callithrix the neonatal period masculinize the social play of juvenile female rats. Brain Res.
jacchus. Brain Behav. Evol. 15, 260–293. 398, 324–328.
Lloyd, J.A., 1971. Weights of testes, thymi, and accessory reproductive glands in Meaney, M.J., Stewart, J., Poulin, P., McEwen, B.S., 1983. Sexual differentiation of
relation to rank in paired and grouped house mice (Mus musculus). Proc. Soc. Exp. social play in rat pups is mediated by the neonatal androgen-receptor system.
Biol. Med. 137, 19–22. Neuroendocrinology 37, 85–90.
Long, S.F., Wilson, M.C., Sufka, K.J., Davis, W.M., 1996. The effects of cocaine and Mehlman, P.T., Higley, J.D., Faucher, I., Lilly, A.A., Taub, D.M., Vickers, J.,
nandrolone co-administration on aggression in male rats. Prog. Neuro- Suomi, S.J., Linnoila, M., 1994. Low CSF 5-HIAA concentrations and severe
psychopharmacol. Biol. Psychiatry 20, 839–856. aggression and impaired impulse control in nonhuman primates. Am. J. Psychiatry
Lonstein, J.S., Gammie, S.C., 2002. Sensory, hormonal, and neural control of maternal 151, 1485–1491.
aggression in laboratory rodents. Neurosci. Biobehav. Rev. 26, 869–888. Meijer, O.C., de Kloet, E.R., 1998. Corticosterone and serotonergic neurotransmission
Lumia, A.R., Raskin, L.A., Eckhert, S., 1977. Effects of androgen on marking and in the hippocampus: functional implications of central corticosteroid receptor
aggressive behavior of neonatally and prepubertally bulbectomized and castrated diversity. Crit. Rev. Neurobiol. 12, 1–20.
male gerbils. J. Comp. Physiol. Psychol. 91, 1377–1389. Melloni Jr., R.H., Connor, D.F., Hang, P.T., Harrison, R.J., Ferris, C.F., 1997. Anabolic-
Machado, C.J., Bachevalier, J., 2006. The impact of selective amygdala, orbital androgenic steroid exposure during adolescence and aggressive behavior in golden
frontal cortex, or hippocampal formation lesions on established social rela- hamsters. Physiol. Behav. 61, 359–364.
tionships in rhesus monkeys (Macaca mulatta). Behav. Neurosci. 120, Miczek, K.A., Brykczynski, T., Grossman, S.P., 1974. Differential effects of lesions in
761–786. the amygdala, periamygdaloid cortex, and stria terminalis on aggressive behaviors
Maestripieri, D., Badiani, A., Puglisi-Allegra, S., 1991. Prepartal chronic stress in rats. J. Comp. Physiol. Psychol. 87, 760–771.
increases anxiety and decreases aggression in lactating female mice. Behav. Miczek, K.A., Covington, H.E., Nikulina, E.M., Hammer, R.P., 2004. Aggression and
Neurosci. 105, 663–668. defeat: persistent effects on cocaine self-administration and gene expression in
Manuck, S.B., Flory, J.D., Ferrell, R.E., Mann, J.J., Muldoon, M.F., 2000. A regulatory peptidergic and aminergic mesocorticolimbic circuits. Neurosci. Biobehav. Rev. 27,
polymorphism of the monoamine oxidase-A gene may be associated with variability 787–802.
in aggression, impulsivity, and central nervous system serotonergic responsivity. Miczek, K.A., de Almeida, R.M., Kravitz, E.A., Rissman, E.F., de Boer, S.F., Raine, A.,
Psychiatry Res. 95, 9–23. 2007. Neurobiology of escalated aggression and violence. J. Neurosci. 27,
Manuck, S.B., Kaplan, J.R., Lotrich, F.E., 2006. Brain serotonin and aggressive 11803–11806.
disposition in humans and nonhuman primates. In: Nelson, R.J. (Ed.), Biology of Miczek, K.A., De Boer, S.F., Haller, J., 2013. Excessive aggresion as model of
Aggression. Oxford University Press, Oxford, pp. 65–113. violence: a critical evaluation of current preclinical methods. Psychopharmacology
Markaverich, B.M., Mani, S., Alejandro, M.A., Mitchell, A., Markaverich, D., Brown, T., 226, 445–458.
Velez-Trippe, C., Murchison, C., O’Malley, B., Faith, R., 2002. A novel endocrine- Miczek, K.A., Fish, E.W., 2006. Monoamines, GABA, glutamate, and aggression. In:
disrupting agent in corn with mitogenic activity in human breast and prostatic Nelson, R.J. (Ed.), Biology of Aggression. Oxford University Press, Oxford,
cancer cells. Environ. Health Perspect. 110, 169–177. pp. 114–149.
Marler, C.A., Bester-Meredith, J.K., Trainor, B.C., 2003. Paternal behavior and Mikics, E., Barsy, B., Haller, J., 2007. The effect of glucocorticoids on aggressiveness
aggression: endocrine mechanisms and nongenomic transmission of behavior. in established colonies of rats. Psychoneuroendocrinology 32, 160–170.
Adv. Study Behav. 32, 263–323. Mikics, E., Kruk, M.R., Haller, J., 2004. Genomic and non-genomic effects of
Marler, C.A., Moore, M.C., 1989. Time and energy costs of aggression in testosterone- glucocorticoids on aggressive behavior in male rats. Psychoneuroendocrinology 29,
implanted free-living male mountain spiny lizards (Sceloporus jarrovi). Physiol. 618–635.
Zool. 62, 1334–1350. Minkeviciene, R., Banjeree, R., Tanila, H., 2004. Memantine improves spatial learning
Marler, C.A., Oyegbile, T., Plavicki, J., Trainor, B.C., 2005. Response to wingfield in a transgenic mouse model of Alzheimer’s disease. J. Pharmacol. Exp. Ther.
commentary on “a continuing saga: the role of testosterone in aggression”. Horm. 311, 677–682.
Behav. 48, 256–258. Moghaddam, B., 2002. Stress activation of glutamate neurotransmission in the
Marquez, C., Poirier, G.L., Cordero, M.I., Larsen, M.H., Groner, A., Marquis, J., prefrontal cortex: implications for dopamine-associated psychiatric disorders. Biol.
Magistretti, J., Trono, D., Sander, C., 2013. Peripuberty stress leads to abnormal Psychiatry 51, 775–787.
aggression, altered amygdala and orbitofrontal reactivity and increased prefrontal Monti, P.M., Brown, W.A., Corriveau, D.P., 1977. Testosterone and components of
MAOA gene expression. Transl. Psychiatry 3, e216. aggressive and sexual behavior in man. Am. J. Psychiatry 134, 692–694.
Hormones and the Development and Expression of Aggressive Behavior 171

Montoya, E.R., Terburg, D., Bos, P.A., van Honk, J., 2012. Testosterone, cortisol, and Ou, X.M., Chen, K., Shih, J.C., 2006. Glucocorticoid and androgen activation of
serotonin as key regulators of social aggression: a review and theoretical monoamine oxidase A is regulated differently by R1 and Sp1. J. Biol. Chem. 281,
perspective. Motivation Emot. 36, 65–73. 21512–21525.
Moore, F.L., Miller, L.J., 1984. Stress-induced inhibition of sexual behavior: cortico- Oyegible, T.O., Marler, C.A., 2005. Winning fights elevates testosterone levels in
sterone inhibits courtship behaviors of a male amphibian (Taricha granulosa). California mice and enhance future ability to win fights. Horm. Behav. 48, 259–267.
Horm. Behav. 18, 400–410. Panksepp, J., Burgdorf, J., Beinfeld, M.C., Kroes, R.A., Moskal, J.R., 2004. Regional
Moore, M.C., 1986. Elevated testosterone levels during nonbreeding season territo- brain cholecystokinin changes as a function of friendly and aggressive social
riality in a fall-breeding lizard, Sceloporus jarrovi. J. Comp. Physiol. A 158, interactions in rats. Brain Res. 1025, 75–84.
159–163. Panksepp, J., Scott, E.L., 2012. Reflections on rough and tumble play, social
Morrison, T.R., Melloni, R.H., 2014. The role of serotonin, vasopressin, and serotonin/ development, and attention-deficit hyperactivity disorders. In: Meyer, A.L. (Ed.),
vasopressin interactions in aggressive behavior. Curr. Top. Behav. Neurosci. 17, Physical Activity across the Life Span. Springer, New York, pp. 23–40.
189–228. Panzica, G.C., Viglietti-Panzica, C., Sica, M., Gotti, S., Martini, M., Pinos, H.,
Natarajan, D., Caramaschi, D., 2010. Animal violence demystified. Front. Behav. Carrillo, B., Collado, P., 2006. Effects of gonadal hormones on central nitric oxide
Neurosci. 4, 9. producing systems. Neuroscience 138, 987–995.
Nelson, R.J., Chiavegatto, S., 2000. Aggression in knockout mice. ILAR J. 41, Parmigiani, S., Brain, P.F., 1983. Effects of residence, aggressive experience and
153–162. intruder familiarity on attack shown by male mice. Behav. Process. 8, 45–58.
Nelson, R.J., Demas, G.E., Huang, P.L., Fishman, M.C., Dawson, V.L., Dawson, T.M., Parsey, R.V., Oquendo, M.A., Simpson, N.R., Ogden, R.T., Van Heertum, R.,
Snyder, S.H., 1995. Behavioural abnormalities in male mice lacking neuronal nitric Arango, V., Mann, J.J., 2002. Effects of sex, age,and aggressive traits in men on
oxide synthase. Nature 378, 383–386. brain serotonin 5-HT1A receptor binding potential measured by PET using [C-11]
Nelson, R.J., Kriegsfeld, L.J., Dawson, V.L., Dawson, T.M., 1997. Effects of nitric WAY-100635. Brain Res. 954, 173–182.
oxide on neuroendocrine function and behavior. Front. Neuroendocrinol. 18, Peake, T.M., Terry, A.M.R., McGregor, P.K., Dabelsteen, T., 2002. Do great tits assess
463–491. rivals by combining direct experience with information gathered by eavesdropping?
Nelson, R.J., Trainor, B.C., 2007. Neural mechanisms of aggression. Nat. Rev. Proc. R. Soc. Lond. B 269, 1925–1929.
Neurosci. 8, 536–546. Pellis, S.M., 2002. Sex differences in play fighting revisited: traditional and nontra-
Nephew, B.C., Bridges, R.S., 2008. Central actions of arginine vasopressin and a V1a ditional mechanisms of sexual differentiation in rats. Arch. Sex. Behav. 31, 17–26.
receptor antagonist on maternal aggression, maternal behavior,and grooming in Pellis, S.M., Field, E.F., Smith, L.K., Pellis, V.C., 1997. Multiple differences in the play
lactating rats. Pharmacol. Biochem. Behav. 91, 77–83. fighting of male and female rats. Implications for the causes and functions of play.
New, A.S., Buchsbaum, M.S., Hazlett, E.A., Goodman, M., Koenigsberg, H.W., Lo, J., Neurosci. Biobehav. Rev. 21, 105–120.
Iskander, L., Newmark, R., Brand, J., O’Flynn, K., Siever, L.J., 2004. Fluoxetine Pellis, S.M., Pellis, V.C., 1988. Play-fighting in the Syrian golden hamster Meso-
increases relative metabolic rate in the prefrontal cortex in impulsive aggression. cricetus auratus waterhouse, and its relationship to serious fighting during post-
Psychopharmacology 176, 451–458. weaning development. Dev. Psychobiol. 21, 323–337.
Newman, S., 1999. The medial extended amygdala in male reproductive behavior. Pellis, S.M., Pellis, V.C., 1997. The prejuvenile onset of play fighting in laboratory rats
A node in the mammalian social behavior network. Ann. N.Y. Acad. Sci. 877, (Rattus norvegicus). Dev. Psychobiol. 31, 193–205.
242–257. Pellis, S.M., Pellis, V.C., 1998. Play fighting of rats in comparative perspective:
Nomura, M., Andersson, S., Korach, K., Gustafsson, J., Pfaff, D., Ogawa, S., 2006. a schema for neurobehavioral analyses. Neurosci. Biobehav. Rev. 23, 87–101.
Estrogen receptor-beta gene disruption potentiates estrogen-inducible aggression Pellis, S.M., Pellis, V.C., 2007. Rough-and-tumble play and the development of the
but not sexual behaviour in male mice. Eur. J. Neurosci. 23, 1860–1868. social brain. Curr. Dir. Psychol. Sci. 16, 95–98.
Nomura, M., Durbak, I., Chan, J., Gustafsson, J.A., Smithies, O., Korach, K.S., Persky, H., O’Brien, C.P., Fine, E., Howard, W.J., Khan, M.A., Beck, R.W., 1977. The
Pfaff, D.W., Ogawa, S., 2002. Genotype/age interactions on aggressive behavior in effect of alcohol and smoking on testosterone function and aggression in chronic
gonadally intact estrogen receptor beta knockout (bERKO) male mice. Horm. alcoholics. Am. J. Psychiatry 134, 621–625.
Behav. 41, 288–296. Poole, A.E., Brain, P., 1974. Effects of adrenalectomy and treatments with ACTH and
Oberlander, J.G., Henderson, L.P., 2012. The sturm und drang of anabolic steroid use: glucocorticoids on isolation-induced aggressive behavior in male albino mice. Prog.
angst, anxiety, and aggression. Trends Neurosci. 35, 382–392. Brain Res. 41, 465–472.
Ogawa, S., Chan, J., Chester, A.E., Gustafsson, J., Korach, K.S., Pfaff, D.W., 1999. Pope, H.G.J., Katz, D.L., 1994. Psychiatric and medical effects of anabolic-androgenic
Survival of reproductive behaviors in estrogen receptor beta gene-deficient (bERKO) steroid use: a controlled study of 160 athletes. Arch. Gen. Psychiatry 51, 375–382.
male and female mice. Proc. Natl. Acad. Sci. U.S.A. 96, 12887–12892. Potegal, M., 1991. Attack priming and satiation in female golden hamsters: tests of
Ogawa, S., Chester, A.E., Hewitt, S.C., Walker, V.R., Gustafsson, J., Smithies, O., some alternatives to the aggression arousal interpretation. Aggress. Behav. 17,
Korach, K.S., Pfaff, D.W., 2000. Abolition of male sexual behaviors in mice 327–335.
lacking estrogen receptors a and b (abERKO). Proc. Natl. Acad. Sci. U.S.A. 97, Potegal, M., Einon, D., 1989. Aggressive behaviors in adult rats deprived of play-
14737–14741. fighting experience as juveniles. Dev. Psychobiol. 22, 159–172.
Ogawa, S., Lubahn, D.B., Korach, K.S., Pfaff, D.W., 1997. Behavioral effects of estrogen Pradhan, D.S., Newman, A.E., Wacker, D.W., Wingfield, J.C., Schlinger, B.A.,
receptor gene disruption in male mice. Proc. Natl. Acad. Sci. U.S.A. 94, 1476–1481. Soma, K.K., 2010. Aggressive interactions rapidly increase androgen synthesis in
Okamura, H., Yokosuka, M., Hayashi, S., 1994. Estrogenic induction of NADPH- the brain during the non-breeding season. Horm. Behav. 57, 381–389.
diaphorase activity in the preoptic neurons containing estrogen receptor Prendergast, B.J., Onishi, K.G., Zucker, I., 2014. Female mice liberated for inclusion in
immunoreactivity in the female rat. J. Neuroendocrinol. 6, 597–601. neuroscience and biomedical research. Neurosci. Biobehav. Rev. 40, 1–5.
Olesen, K.M., Jessen, H.M., Auger, C.J., Auger, A.P., 2005. Dopaminergic activation Pulsford, D., Duxbury, J., 2006. Aggressive behaviour by people with dementia in
of estrogen receptors in neonatal brain alters progestin receptor expression and residential care settings: a review. J. Psychiatry Ment. Health Nurs. 13, 611–618.
juvenile social play behavior. Endocrinology 146, 3705–3712. Quiring, D.P., 1944. The transplantation of testes (by A.A. Berthold). Bull. Hist. Med.
Oliveira, R.F., Hirschenhauser, K., Carneiro, L.A., Canario, A.V.M., 2002. Social 16, 399–401.
modulation of androgen levels in male teleost fish. Comp. Biochem. Physiol. B 132, Raine, A., 2002. Annotation: the role of prefrontal deficits, low autonomic arousal, and
203–215. early health factors in the development of antisocial and aggressive behavior in
Olson, M.B., Bairey Merz, C.N., Shaw, L.J., Mankad, S., Reis, S.E., Pohost, G.M., children. J. Child Psychol. Psychiatry 43, 417–434.
Smith, K.M., McGorray, S.P., Cornell, C.E., Kelsey, S.F., 2004. Hormone Raine, A., Buchsbaum, M.S., Stanley, J., Lottenberg, S., Abel, L., Stoddard, J., 1994.
replacement, race, and psychological health in women: a report from the NHLBI- Selective reductions in prefrontal glucose metabolism in murderers. Biol. Psychiatry
Sponsored WISE Study. J. Womens Health 13, 325–332. 36, 365–373.
Olweus, D., 1983. Low school achievement and aggressive behavior in adolescent Ramboz, S., Saudou, F., Amara, D.A., Belzung, C., Segu, L., Misslin, R., Buhot, M.C.,
boys. In: Magnusson, D. (Ed.), Human Development: An Interactional Perspective. Hen, R., 1996. 5-HT1B receptor knock out–behavioral consequences. Behav.
Academic, New York, pp. 353–365. Brain Res. 73, 305–312.
Orchinik, M., Murray, T.F., Moore, F.L., 1991. A corticosteroid receptor in neuronal Reif, A., Jacob, C.P., Rujescu, D., Herterich, S., Lang, S., Gutknecht, L., Baehne, C.G.,
membranes. Science 252, 1848–1851. Strobel, A., Freitag, C.M., Giegling, I., Romanos, M., Hartmann, A., Rösler, M.,
Orengo, C., Kunik, M.E., Molinari, V., Wristers, K., Yudofsky, S.C., 2002. Do testos- Renner, T.J., Fallgatter, A.J., Retz, W., Ehlis, A.C., Lesch, K.P., 2009. Influence of
terone levels relate to aggression in elderly men with dementia? J. Neuropsychiatry functional variant of neuronal nitric oxide synthase on impulsive behaviors in
Clin. Neurosci. 14, 161–166. humans. Arch. Gen. Psychiatry 66, 41–50.
172 Hormones and the Development and Expression of Aggressive Behavior

Reif, A., Kiive, E., Kurrikoff, T., Paaver, M., Herterich, S., Konstabel, K., Tulviste, T., Simon, N.G., Whalen, R.E., 1986. Hormonal regulation of aggression: evidence for
Lesch, K.-P., Harro, J., 2011. A functional NOS1 promoter polymorphism interacts a relationship among genotype, receptor binding, and behavioral sensitivity to
with adverse environment on functional and dysfunctional impulsivity. Psycho- androgen and estrogen. Aggress. Behav. 12, 255–266.
pharmacology 214, 239–248. Sluyter, F., Arseneault, L., Moffitt, T.E., Veenema, A.H., de Boer, S., Koolhaas, J.M.,
Rendon, N.M., Rudolph, L.M., Sengelaub, Dr, Demas, G.E., 2015. The agonistic 2003. Toward an animal model for antisocial behavior: parallels between mice and
adrenal: melatonin elicits female aggression via regulation of adrenal androgens. humans. Behav. Genet. 33, 563–574.
Proc. R. Soc. Lond. Ser. B 282, 20152080. Smale, L., Holekamp, K.E., White, P.A., 1999. Siblicide revisited in the spotted hyaena:
Ricci, L.A., Grimes, J.M., Melloni Jr., R.H., 2007. Lasting changes in neuronal acti- does it conform to obligate or facultative models? Anim. Behav. 58, 545–551.
vation patterns in select forebrain regions of aggressive, adolescent anabolic/ Soderstrom, H., Tullberg, M., Wikkelso, C., Ekholm, S., Forsman, A., 2000. Reduced
androgenic steroid-treated hamsters. Behav. Brain Res. 176, 344–352. regional cerebral blood flow in non-psychotic violent offenders. Psychiatry Res. 98,
Rilke, O., Freier, D., Jahkel, M., Oehler, J., 1998. Dynamic alterations of serotonergic 29–41.
metabolism and receptors during social isolation of low- and high-active mice. Soloff, P.H., Meltzer, C.C., Becker, C., Greer, P.J., Kelly, T.M., Constantine, D., 2003.
Pharmacol. Biochem. Behav. 59, 891–896. Impulsivity and prefrontal hypometabolism in borderline personality disorder.
Rilling, J.K., Winslow, J.T., Kilts, C.D., 2004. The neural correlates of mate competition Psychiatry Res. 123, 153–163.
in dominant male rhesus macaques. Biol. Psychiatry 56, 364–375. Solomon, M.B., Karom, M.C., Huhman, K.L., 2007. Sex and estrous cycle differences in
Robinson, B.W., 1967. Vocalization evoked from forebrain in Macaca mulatta. Physiol. the display of conditioned defeat in Syrian hamsters. Horm. Behav. 52, 211–219.
Behav. 2, 345–354. Soma, K.K., 2006. Testosterone and aggression: Berthold, birds, and beyond.
Roeling, T.A., Kruk, M.R., Schuurmans, R., Veening, J.G., 1993. Behavioural J. Neuroendocrinol. 18, 543–551.
responses of bicucculline methiodide injections into the ventral hypothalamus of Soma, K.K., Tramontin, A.D., Wingfield, J.C., 2000. Oestrogen regulates male
freely moving, socially interacting rats. Brain Res. 615, 121–127. aggression in the non-breeding season. Proc. R. Soc. Lond. Ser. B Biol. Sci. 267,
Rose, R.M., Holaday, J.W., Bernstein, I.S., 1971. Plasma testosterone, dominance 1089–1096.
rank and aggressive behaviour in male rhesus monkeys. Nature 231, 366–368. Soma, K.K., Wissman, A.M., Brenowitz, E.A., Wingfield, J.C., 2002. Dehydroepian-
Roubertoux, P.L., Carlier, M., 1988. Differences between CBA/H and NZB mice on drosterone (DHEA) increases territorial song and the size of an associated brain
intermale aggression. II. Maternal effects. Behav. Genet. 18, 175–183. region in a male songbird. Horm. Behav. 41, 203–212.
Roubertoux, P.L., Le Roy, I., Mortaud, S., Perez-Diaz, F., Tordjman, S., 1999. Measuring Staffend, N.A., Meisel, R.L., 2012. Aggressive experience increases dendritic spine
mouse aggression. In: Crusio, W.E. (Ed.), Handbook of Molecular Genetic Techniques density within the nucleus accumbens core in female syrian hamsters. Neurosci-
for Brain and Behavioral Research. Elsevier, Amsterdam, pp. 696–709. ence 227, 163–169.
Salas-Ramirez, K.Y., Montalto, P.R., Sisk, C.L., 2008. Anabolic androgenic steroids Steffen, A.M., Thompson, L.W., Gallagher-Thompson, D., Koin, D., 1999. Physical and
differentially affect social behaviors in adolescent and adult male Syrian hamsters. psychosocial correlates of hormone replacement therapy with chronically stressed
Horm. Behav. 53, 378–385. postmenopausal women. J. Aging Health 11, 3–26.
Sanchez, F., Martinez, M.E., Rubio, M., Carretero, J., Moreno, M.N., Vazquez, R., Steinman, M.Q., Laredo, S.A., Lopez, E.M., Manning, C.E., Hao, R.C., Doig, I.E.,
1998. Reduced nicotinamide adenine dinucleotide phosphate-diaphorase activity in Campi, K.L., Flowers, A.E., Knight, J.K., Trainor, B.C., 2015. Hypothalamic
the paraventricular nucleus of the rat hypothalamus is modulated by estradiol. vasopressin systems are more sensitive to social defeat in males versus females.
Neurosci. Lett. 253, 75–78. Psychoneuroendocrinology 51, 122–134.
Sano, K., Tsuda, M.C., Musatov, S., Sakamoto, T., Ogawa, S., 2013. Differential Summers, C.H., Watt, M.J., Ling, T.L., Forster, G.L., Carpenter, R.E., Korzan, W.J.,
effects of site-specific knockdown of estrogen receptor a in the medial amygdala, Lukkes, J.L., Overli, O., 2005. Glucocorticoid interaction with aggression in non-
medial pre-optic area, and ventromedial nucleus of the hypothalamus on sexual mammalian vertebrates: reciprocal action. Eur. J. Neurosci. 526, 21–35.
and aggressive behavior in male mice. Eur. J. Neurosci. 37, 1308–1319. Sumner, B.E., Fink, G., 1998. Testosterone as well as estrogen increases serotonin2A
Sasano, H., Takashashi, K., Satoh, F., Nagura, H., Harada, N., 1998. Aromatase in the receptor mRNA and binding site densities in the male rat brain. Brain Res. Mol.
human central nervous system. Clin. Endocrinol. 48, 325–329. Brain Res. 59, 205–214.
Saudou, F., Amara, D.A., Dierich, A., LeMeur, M., Ramboz, S., Segu, L., Buhot, M.C., Svare, B., 1983. Hormones and Aggressive Behavior. Plenum, New York.
Hen, R., 1994. Enhanced aggressive behavior in mice lacking 5-HT1B receptor. Swanson, L.W., 2000. Cerebral hemisphere regulation of motivated behavior. Brain
Science 265, 1875–1878. Res. 886, 113–164.
Scarpa, A., Raine, A., 2006. The psychophysiology of human antisocial behavior. In: Takahashi, A., Miczek, K.A., 2014. Neurogenetics of aggressive behavior: studies in
Nelson, R.J. (Ed.), Biology of Aggression. Oxford University Press, Oxford, pp. 447–461. rodents. Curr. Top. Behav. Neurosci. 17, 3–44.
Schlinger, B.A., Callard, G.V., 1990. Aromatization mediates aggressive behavior in Takahashi, A., Quadros, I.M., de Almeida, R.M.M., Miczek, K.A., 2011. Brain serotonin
quail. General Comp. Endocrinol. 79, 39–53. receptors and transporters: initiation vs. termination of escalated aggression.
Schulz, K.M., Menard, T.A., Smith, D.A., Albers, H.E., Sisk, C.L., 2006. Testicular Psychopharmacology 213, 183–212.
hormone exposure during adolescence organizes flank-marking behavior and Takahashi, H., Matsuura, M., Yahata, N., Koeda, M., Suhara, T., Okubo, Y., 2006. Men
vasopressin receptor binding in the lateral septum. Horm. Behav. 50, 477–483. and women show distinct brain activations during imagery of sexual and emotional
Schulz, K.M., Molenda-Figueira, H.A., Sisk, C.L., 2009. Back to the future: the infidelity. Neuroimage 32, 1299–1307.
organizational activational hypothesis adapted to puberty and adolescence. Horm. Taravosh-Lahn, K., Delville, Y., 2004. Aggressive behavior in female golden hamsters:
Behav. 55, 597–604. development and the effect of repeated social stress. Horm. Behav. 46, 428–435.
Scordalakes, E.M., Rissman, E.F., 2003. Aggression in male mice lacking functional Toda, K., Saibara, T., Okada, T., Onishi, S., Shizuta, Y., 2001. A loss of aggressive
estrogen receptor a. Behav. Neurosci. 117, 38–45. behaviour and its reinstatement by oestrogen in mice lacking the aromatase gene
Scordalakes, E.M., Rissman, E.F., 2004. Aggression and arginine vasopressin (Cyp19). J. Endocrinol. 168, 217–220.
immunoreactivity regulation by androgen receptor and estrogen receptor a. Genes Tonkonogy, J.M., Geller, J.L., 1992. Hypothalamic lesions and intermittent explosive
Brain Behav. 3, 20–26. disorder. J. Neuropsychiatry Clin. Neurosci. 4, 45–50.
Scott, J.P., 1975. Aggression. University of Chicago Press, Chicago. Trainor, B.C., Bird, I.M., Marler, C.A., 2004. Opposing hormonal mechanisms of
Shih, J.C., Chen, K., Ridd, M.J., 1999. Monoamine oxidase: from genes to behavior. aggression revealed through short-lived testosterone manipulations and multiple
Annu. Rev. Neurosci. 22, 197–217. winning experiences. Horm. Behav. 45, 115–121.
Shrenker, P., Maxson, S.C., Ginsburg, B.E., 1985. The role of postnatal testosterone in Trainor, B.C., Crean, K.K., Fry, W.H.D., Sweeney, C., 2010a. Activation of extracellular
the development of sexually dimorphic behaviors in DBA/1Bg mice. Physiol. Behav. signal-regulated kinases in social behavior circuits during resident-intruder
35, 757–762. aggression tests. Neuroscience 165, 325–336.
Silva, A.L., Fry, W.H., Sweeney, C., Trainor, B.C., 2010. Effects of photoperiod and Trainor, B.C., Finy, M.S., Nelson, R.J., 2008. Rapid effects of estradiol on male
experience on aggressive behavior in female California mice. Behav. Brain Res. aggression depend on photoperiod in reproductively non-responsive mice. Horm.
208, 528–534. Behav. 53, 192–199.
Simon, N.G., 2002. Hormonal processes in the development and expression of Trainor, B.C., Greiwe, K.M., Nelson, R.J., 2006a. Individual differences in estrogen
aggressive behavior. In: Pfaff, D.W. (Ed.), Hormones Brain and Behavior. Academic receptor a in select brain nuclei are associated with individual differences in
Press, San Diego. aggression. Horm. Behav. 50, 338–345.
Simon, N.G., Cologer-Clifford, A., Lu, S.F., McKenna, S.E., Hu, S., 1998. Testosterone Trainor, B.C., Kyomen, H.H., Marler, C.A., 2006b. Estrogenic encounters: how inter-
and its metabolites modulate 5HT1A and 5HT1B agonist effects on intermale actions between aromatase and the environment modulate aggression. Front.
aggression. Neurosci. Biobehav. Rev. 23, 325–336. Neuroendocrinol. 27, 170–179.
Hormones and the Development and Expression of Aggressive Behavior 173

Trainor, B.C., Lin, S., Finy, M.S., Rowland, M.R., Nelson, R.J., 2007a. Photoperiod Wallen, K., Eisler, J.A., Tannenbaum, P.L., Nagell, K.M., Mann, D.R., 1991. Antide
reverses the effects of estrogens on male aggression via genomic and non- (NAL-LYS GnRH antagonist) suppression of pituitary-testicular function and sexual
genomic pathways. Proc. Natl. Acad. Sci. U.S.A. 104, 9840–9845. behavior in group-living rhesus monkeys. Physiol. Behav. 50, 429–435.
Trainor, B.C., Marler, C.A., 2001. Testosterone, paternal behavior, and aggression in Warembourg, M., Leroy, D., Jolivet, A., 1999. Nitric oxide synthase in the guinea pig
the monogamous California mouse (Peromyscus californicus). Horm. Behav. 40, preoptic area and hypothalamus: distribution, effect of estrogen, and colocalization
32–42. with progesterone receptor. J. Comp. Neurol. 407, 207–227.
Trainor, B.C., Rowland, M.R., Nelson, R.J., 2007b. Photoperiod affects estrogen Wersinger, S.R., Ginns, E.I., O’Carroll, A.M., Lolait, S.J., Young, W.S.I., 2002.
receptor alpha, estrogen receptor beta, and aggressive behavior. Eur. J. Neurosci. Vasopressin V1b receptor knockout reduces aggressive behavior in male mice.
26, 207–218. Mol. Psychiatry 7, 975–984.
Trainor, B.C., Takahashi, E.Y., Silva, A.L., Crean, K.K., Hostetler, C., 2010b. Sex Wesson, D.W., McGinnis, M.Y., 2006. Stacking anabolic androgenic steroids (AAS)
differences in hormonal responses to social conflict in the monogamous California during puberty in rats: a neuroendocrine and behavioral assessment. Pharmacol.
mouse. Horm. Behav. 58, 506–512. Biochem. Behav. 83, 410–419.
Trainor, B.C., Workman, J.L., Jessen, R., Nelson, R.J., 2007c. Impaired nitric oxide Whalen, R.E., Johnson, F., 1987. Individual differences in the attack behavior of male
synthase signaling dissociates social investigation and aggression. Behav. mice: a function of attack stimulus and hormonal state. Horm. Behav. 21, 223–233.
Neurosci. 121, 362–369. Whitsett, J.M., Vanderbergh, J.G., 1975. Influence of testosterone propionate
Trenton, A.J., Currier, G.W., 2005. Behavioural manifestations of anabolic steroid use. administered neonatally on puberty and bisexual behavior in female hamsters.
CNS Drugs 19, 571–595. J. Comp. Physiol. Psychol. 88, 248–255.
Unger, E.K., Burke, K.J.J., Yang, C.F., Bender, K.J., Fuller, P.M., Shah, N.M., 2015. Widom, C.S., Czaja, S., Dutton, M.A., 2014. Child abuse and neglect and intimate
Medial amygdalar aromatase neurons regulate aggression in both sexes. Cell Rep. partner violence victimization and perpetration: a prospective investigation. Child
10, 453–462. Abuse Negl. 38, 650–663.
Vandenbergh, J.G., 1971. The effects of gonadal hormones on the aggressive Wingfield, J.C., 1984. Environmental and endocrine control of reproduction in the song
behaviour of adult golden hamsters (Mesocricetus auratus). Anim. Behav. 19, sparrow, Melospiza melodia. I. Temporal organization of the breeding cycle.
589–594. General Comp. Endocrinol. 56, 406–416.
Vasudevan, N., Pfaff, D.W., 2006. Membrane initiated actions of estrogens in Wingfield, J.C., Ball, G.F., Dufty, A.M., Hegner, R.E., Ramenofsky, M., 1987.
neuroendocrinology: emerging principles. Endocr. Rev. 28, 1–19. Testosterone and aggression in birds. Am. Sci. 75, 602–608.
Veenema, A.H., 2012. Toward understanding how early-life social experiences alter Wingfield, J.C., Hegner, R.E., Dufty, J.,A.M., Ball, G.F., 1990. The ’challenge
oxytocin- and vasopressin-regulated social behaviors. Horm. Behav. 61, 304–312. hypothesis’: theoretical implications for patterns of testosterone secretion, mating
Verona, E., Joiner, T.E., Johnson, F., Bender, T.W., 2006. Gender specific gene- systems, and breeding strategies. Am. Nat. 136, 829–846.
environment interactions on laboratory-assessed aggression. Biol. Psychol. 71, Winslow, J.T., Hearn, E.F., Ferguson, J.F., Young, L.J., Matzuk, M.M., Insel, T.R.,
33–41. 2000. Infant vocalization, adult aggression, and fear behavior of an oxytocin null
Viau, V., 2002. Functional cross-talk between the hypothalamic-pituitary-gonadal and mutant mouse. Horm. Behav. 37, 145–155.
-adrenal axes. J. Neuroendocrinol. 14, 506–513. Winslow, J.T., Insel, T.R., 1991. Social status in pairs of male squirrel monkeys
Viding, E., Frick, P.J., Plomin, R., 2007. Aetiology of the relationship between callous- determines the behavioral response to central oxytocin administration. J. Neurosci.
unemotional traits and conduct problems in childhood. Br. J. Psychiatry Suppl. 49, 11, 2032–2038.
s33–s38. Winston, J.S., Strange, B.A., O’Doherty, J., Dolan, R.J., 2002. Automatic and
Villalon Landeros, R., Yoo, H.J., Morisseau, C., Fu, S., Hammock, B.D., Trainor, B.C., intentional brain responses during evaluation of trustworthiness of faces. Nat.
2012. Corncob bedding reverses the effects of estrogens on aggressive behavior Neurosci. 5, 277–283.
and reduces estrogen receptor alpha expression in the brain. Endocrinology 153, Wolfer, D.P., Crusio, W.E., Lipp, H.P., 2002. Knockout mice: simple solutions to the
949–953. problems of genetic background and flanking genes. Trends Neurosci. 25,
Vitiello, B., Stoff, D.M., 1997. Subtypes of aggression and their relevance to child 336–340.
psychiatry. J. Am. Acad. Child Adolesc. Psychiatry 36, 307–315. Wommack, J.C., Delville, Y., 2007. Stress, aggression, and puberty: neuroendocrine
Volkow, N.D., Tancredi, L.R., Grant, C., Gillespie, H., Valentine, A., Mullani, N., correlates of the development of agonistic behavior in golden hamsters. Brain
Wang, G.J., Hollister, L., 1995. Brain glucose metabolism in violent psychiatric Behav. Evol. 70, 267–273.
patients: a preliminary study. Psychiatry Res. 61, 243–253. Wommack, J.C., Taravosh-Lahn, K., David, J.T., Delville, Y., 2003. Repeated exposure
Von Frijtag, J.C., Schot, M., van den Bos, R., Spruijt, B.M., 2002. Individual housing to social stress alters the development of agonistic behavior in male golden
during the play period results in changed responses to and consequences of hamsters. Horm. Behav. 43, 229–236.
a psychosocial stress situation in rats. Dev. Psychobiol. 41, 58–69. Wood, R.I., Newman, S.W., 1995. Hormonal influence on neurons of mating behavior
Wahaj, S.A., Holekamp, K.E., 2006. Functions of sibling aggression in the spotted pathway in male hamsters. In: Micevych, P.E. (Ed.), Neurobiological Effects of Sex
hyaena, Crocuta crocuta. Anim. Behav. 71, 1401–1409. Steroid Hormones. Cambridge University Press, New York, pp. 3–39.
Wahaj, S.A., Place, N.J., Weldele, M.L., Glickman, S.E., Holekamp, K.E., 2007. Young, J., Corpechot, C., Perche, F., Eychenne, B., Haug, M., Baulieu, E.-E., Robel, P.,
Siblicide in the spotted hyena: analysis with ultrasonic examination of wild and 1996. Neurosteroids in the mouse brain: behavioral and pharmacological effects of
captive individuals. Behav. Ecol. 18, 974–984. a 3 beta-hydroxysteroid dehydrogenase inhibitor. Steroids 61, 144–149.
Wakschlag, L.S., Kistner, E.O., Pine, D.S., Biesecker, G., Pickett, K.E., Skol, A.D., Young, S.I., Smolen, A., Hewitt, J., Haberstick, M., Stallings, M., Corley, R.P.,
Dukic, V., Blair, R.J.R., Leventhal, B.L., Cox, N.J., Burns, J.L., Kasza, K.E., Crowley, T.J., 2006. Interaction between MAO-A genotype and maltreatment in the
Wright, R.J., Cook Jr., E.H., 2010. Interaction of prenatal exposure to cigarettes risk for conduct disorder: failure to confirm in adolescent patients. Am. J.
and MAOA genotype in pathways to youth antisocial behavior. Mol. Psychiatry 15, Psychiatry 163, 1019–1025.
928–937. Zamaratskaia, G., Rydhmer, L., Andersson, H.K., Chen, G., Lowagie, S.,
Wallen, K., 1996. Nature needs nurture: the interaction of hormonal and social Andersson, K., Lundstrom, K., 2008. Long-term effect of vaccination against
influences on the development of behavioral sex differences in rhesus monkeys. gonadotropin-releasing hormone, using Improvac, on hormonal profile and
Horm. Behav. 30, 364–378. behaviour of male pigs. Anim. Reprod. Sci. 108, 37–48.
This page intentionally left blank
1.06 Food Intake and Its Control by Signaling Molecules
Francisco Vázquez-Cuevas, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
Raúl Aguilar-Roblero, Instituto de Fisiología Celular, Ciudad Universitaria, UNAM, México, Mexico
Elvira Arellanes-Licea, Instituto Potosino de Investigación Científica y Tecnológica, San Luis Potosí, Mexico
Yazmı́n Macotela and Olivia Vázquez-Martı́nez, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
Iván Villanueva, Escuela Nacional de Ciencias Biológicas, IPN, México, Mexico
Mauricio Dı́az-Muñoz, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
Ó 2017 Elsevier Inc. All rights reserved.

1.06.1 Preface 177


1.06.2 Hunger–Satiety Cycle–Historical View and Modern Perspective 177
1.06.2.1 Energy Input: Food Intake and Its Physiological Control 178
1.06.2.2 Not All Feeding Is Related to Energy 178
1.06.2.3 Study of the Physiological Control of Food Intake 179
1.06.2.4 Main Concepts in the Study of Food Intake Control and Their Historical Development 179
1.06.2.5 Energy: A Compulsory Element 180
1.06.2.6 Quest for the Signals 180
1.06.3 Bioenergetics of the Fasting–Feeding Cycle 183
1.06.4 Hypothalamus and Feeding Control 184
1.06.4.1 General Perspective 184
1.06.4.2 Neuronal Circuits 184
1.06.4.3 Feeding Circuits in the Brain 185
1.06.4.3.1 Homeostatic Circuits 185
1.06.5 Chronostasis: Physiological Regulation across Time 187
1.06.6 Neuropeptides 188
1.06.6.1 Neuropeptides Regulating Feeding Behavior 188
1.06.6.1.1 Orexins 188
1.06.6.1.2 Orexins and Feeding Control 189
1.06.6.1.3 Orexins and Energy Regulation 189
1.06.6.2 Endocannabinoid System 190
1.06.6.2.1 Endocannabinoids and Feeding Control 190
1.06.6.3 Melanocortin System 190
1.06.6.3.1 POMC-Derived Peptide Family 190
1.06.6.3.2 Aguoti-Related Peptide (AgRP), a Natural Antagonist of the Melanocortin System 191
1.06.6.3.3 Receptors of Melanocortin 191
1.06.6.3.4 Melanocortins and Feeding Control 191
1.06.6.4 Neuropeptide Y 192
1.06.6.4.1 NPY and Feeding Control 192
1.06.7 Endocrine Systems: Hormones in Fasting and in the Feeding Response 193
1.06.7.1 Ghrelin and Leptin Biology and Their Role in Food-Intake Regulation 193
1.06.7.1.1 Biology: Synthesis, Secretion, Physiology, and Signaling 193
1.06.7.1.2 Leptin 194
1.06.7.1.3 Food-Intake Regulation by Ghrelin and Leptin 195
1.06.8 Other Adipokines Regulating Food Intake 196
1.06.8.1 Adiponectin 196
1.06.8.2 Resistin 197
1.06.8.3 Nesfatin 197
1.06.8.4 Apelin 198
1.06.9 Pancreas and Feeding Control 198
1.06.10 Glucocorticoids and the Control of Food Intake 199
1.06.10.1 Glucocorticoids and the Stress System 200
1.06.11 Concluding Remarks 200
Acknowledgments 201
References 201

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00006-7 175


176 Food Intake and Its Control by Signaling Molecules

Abbreviations
11-b HSD 11-b-hydroxysteroid dehydrogenase LH Luteinizing hormone
ACTH Adrenocorticotropic hormone LHA Lateral hypothalamic area
AG Acylated ghrelin b-LPH b-Lipotropin
AGRP Agouti-related peptide LR Leptin receptor
AMPK AMP-dependent kinase MAPK Mitogen-activated protein kinase
ANS Autonomic nervous system MCR Melanocortin receptor
APJ Apelin receptor MR Mineralocorticoid receptors
ARC Arcuate nucleus mRNA Messenger RNA
ATP Adenosine triphosphate a-MSH a-Melanocyte-stimulating hormone
BBB Blood–brain barrier N-POC N-terminal peptide proopiomelanocortin
cAMP/PKA Protein kinase A activated by cAMP NADPD Nicotinamide adenine dinucleotide phosphate,
CART Cocaine- and amphetamine-regulated transcript oxidized form
CB1 Cannabinoid receptor one NADPH Nicotinamide adenine dinucleotide phosphate,
CCK Cholecystokinin reduced form
cDNA Complementary DNA NE Norepinephrine
CLIP Corticotropin-like intermediate lobe peptide NEFA/NUCB2 Nesfatin or nonesterified fatty acid/
CNR1 Endocanabinoids receptor 1 nucleobindin2
CNR2 Endocanabinoids receptor 2 NPY Neuropeptide Y
CPE Carboxypeptidase E NS Nervous system
CRF Corticotropin-releasing factor NTS Nucleus of the solitary tract
CRH Corticotropin-releasing hormone OXR Orexin receptor
CSF Cerebrospinal fluid PAM Peptidyl a-amidating mono-oxygenase
DAG Diacylglycerol PBMC Blood mononuclear cells
DMH Dorsomedial hypothalamic nucleus PBN Parabrachial nucleus
DREADD Designer receptors exclusively activated by PC Posterior commissure
designer drugs PEPCK Phosphoenol pyruvate carboxykinase
DVC Dorsal vagal complex PI3K Phosphatidylinositol 3-kinase
b-end b-Endorphin PKA Protein kinase A
FAS Fatty acid synthase PLC Phospholipase C
GABA a-Aminobutyric acid POMC Proopiomelanocortin
GABAA GABA receptor type A PP Pancreatic polypeptide
GH Growth hormone PPARs Peroxisome proliferator-activated receptors
GHRL Ghrelin/obestatin prepropeptide PPOX Prepro-orexin
GHRS Ghrelin receptors PVH Paraventricular nucleus of the hypothalamus
GHSR Growth hormone secretagogue receptor PVN Paraventricular thalamic nucleus
GLP-1 Glucagon-like peptide-1 PYY Peptide tyrosine tyrosine
GOAT Ghrelin O-acyltransferase RSTN Resistin
GPCR G protein-coupled receptor SF1 Steroidogenic factor 1
GR Glucocorticoid receptors SOC3 Suppressor of cytokine signaling
H6PDH Hexose-6-phosphate dehydrogenase SON Supraoptic nucleus
HCl Clorhydric acid STAT3/5 Signal transducers and activators of transcription
HCRTR Hypocretin (orexin) receptor TNF-a Tumor necrosis factor-a
HPA Hypothalamic–pituitary–adrenal axis TRH Thyrotropin-releasing hormone
IML Intermediolateral column of the spinal cord TZDs Thiazolidinediones
InsR Insulin receptors UAG Unacylated ghrelin
IP3 1,4,5-trisphosphate VLDL Very low density lipoproteins
Jak2 Janus kinase family VMH Ventromedial hypothalamus
JP Joining peptide WAT White adipose tissue
LC Locus caeruleus
Food Intake and Its Control by Signaling Molecules 177

1.06.1 Preface calorie is defined as the amount of heat required to raise the
temperature of 1 g of water by 1  C. Since for many purposes,
Frequently, feeding implies moving within a subject’s habitat calorie (cal) is too small, kilocalorie (kcal) is often used;
in search for food, perhaps engaged in intense motor activity 1 kcal ¼ 1000 cal. In many scientific studies, calorie is replaced
to suppress the sensation of hunger. When the animal has by joule (J) as the unit of energy; 4.184 J ¼ 1 cal and
secured its food it continues to the actual process of feeding, 4184 J ¼ 1 kcal (Franklin et al., 2010).
which will eventually finish as hunger vanishes or the food This chapter is focused on the underlying mechanisms that
has been consumed. This oversimplified view of feeding enable the physiological activity of food intake in animals,
already lets us grasp the complexity of the physiological endothermic organisms with heterotrophic nutrition. The first
processes involved in the regulation and execution of feeding section explains feeding as a complex function with regulatory
behavior, such as interoception involving neuronal networks, principles that can be studied using homeostatic, rheostatic,
behavioral state control, motivation, selective attention, spatial and chronostatic criteria. This section will illustrate the concept
and temporal memory, motor coordination, secretions from that eating should be visualized as a rhythmic function (with
exocrine and endocrine glands (associated with the gastrointes- ultradian or circadian periods) within the chronobiological
tinal tract) involved in digestion and absorption of nutrients, regulation of the organism’s physiology.
and biochemical processes of intermediate metabolism inte- In animals, food intake is a behavior comprising volitional
grated mainly, but not exclusively, at the hepatic level. In the and reflexive activities of more than 30 nerves and muscles.
case of humans, beyond the already complex system outlined However, feeding involves several steps, such as searching for
above, there is an additional level of regulatory interactions food, food ingestion, assimilation of nutrients, the restoration
arising from the fact that we can disregard satiety signals and of energetic deposits (hepatic glycogen and adipose lipids), and
continue eating when we are not hungry. In the present chapter, eventually the energy consumption that empties those
we will review some aspects of the system regulating feeding deposits and returns the system to the first step. Underlying
behavior. this fasting–feeding cycle is the equilibrium between satiety
The term feeding is linked to the concept of nourishment, and hunger, the physiological responses to the energetic state
which is the process by which nutrients are obtained. As chem- of the organism. The second section of this chapter will review
ical entities, nutrients are a set of molecules with an energetic the principal theories that have been postulated to explain the
value for living beings. Living entities, from single cells to hunger–satiety cycle. Some of them have historical significance,
multicellular fungi, plants, and animals, are thermodynami- but others continue to provide the foundation for an active
cally open systems formed by dissipative structures specialized field of biomedical research.
in exchanging matter and energy with the environment. All of Eating involves a display of behavioral patterns that vary
these systems are complex, dynamic, nonlinear, and far from with environmental conditions. Most of the time food intake
being at equilibrium. Indeed, living beings fulfill the Second is driven by sensory inputs, such as tastes and odors, as well
Law of Thermodynamics of increasing global entropy while as by affective or emotional states. Indeed, it has been postu-
presenting emerging properties that are characteristic of biolog- lated that eating behavior is based on genetics, but recently
ical organization (Mrosovsky, 1990). this behavior has also been postulated to be partially attribut-
According to physics, energy is defined as the ability to able to an environmental parameter called behavioral suscepti-
execute work. In animals energy is assimilated through the bility (Carnell and Wardle, 2008).
degradation of dietary carbohydrates, lipids, and proteins. The chapter also contains a section dealing with signaling
Within the cellular milieu, the principal forms of energy are molecules that enable coordination among organs and tissues
those with the capacity to allow chemical (biochemical reac- during feeding activity. We will discuss the principal hypotha-
tions within metabolic networks), electrical (maintaining lamic peptides and factors (purines and endocannabinoids
membrane potential in excitable and nonexcitable tissues), (ECs)) associated with hunger and satiety as well as peripheral
and mechanical work (assembly and disassembly of cytoskel- endocrine molecules with regard to their site of synthesis,
etal components). Energy is, therefore, essential to maintain modulation of secretory events, types of receptors, and mecha-
life processes such as metabolism, growth, reproduction, and nisms of signal transduction. Each messenger molecule will be
ambulatory activity. In particular, life on our planet is depen- dealt with in the context of its role under fasted or fed condi-
dent on radiant solar energy and its subsequent fixation by tions. Finally the chapter will end with some concluding
photosynthetic mechanisms and conversion by green plants remarks that highlight the most recent concepts in the control
and algae into biochemical energy (i.e., carbohydrates). Carbo- of feeding activity.
hydrates and other biomolecules are an energy source for
animals that have the ability to oxidize them through the
process of metabolic respiration. For example, when an animal 1.06.2 Hunger–Satiety Cycle–Historical View
releases the potential energy of glucose during mitochondrial and Modern Perspective
respiration, approximately two-thirds of it is converted into
chemical and mechanical energy to be used for work (metabo- Energy is acquired by organisms in a number of ways. Prokary-
lism and growth), and one-third is dissipated as heat (Jequier otic and plant cells employ diverse strategies to obtain energy
et al., 1987). from their environment. Virtually all animal cells are heterotro-
Energy is usually expressed in terms of heat units, since all phic, which means that they depend on the energy captured
forms of energy are convertible and eventually turn into heat and stored by other living species to capture and store the
energy. The basic heat unit normally used is the calorie. One energy they need in the form of carbon present in specific
178 Food Intake and Its Control by Signaling Molecules

organic molecules. Consequently, animals must feed on these pathways, hormones, and other messengers, brain centers,
energy-rich molecules synthesized by other living organisms. and subcellular enzymatic processes in a number of effector
Energy availability often becomes an environmental tissues) have an enormously complex organization and are
constraint for the perpetuation and growth of animal popula- grouped together under the concept of ‘physiological
tions. Animals have developed a series of mechanisms to control of food intake.’
improve the balance between energy acquisition and expendi- 2. In order to assure energy availability, food intake should be
ture, so that proportionally more energy is available for their physiologically adjusted to ensure energy balance. A nega-
internal processes. This explains the exquisite specialization tive energy balance (i.e., expenses larger than input) would
and efficiency of the physiological mechanisms dedicated to elicit a strong physiological response promoting food search
food acquisition, processing, and storing, along with the and consumption, which we call ‘hunger’; a positive balance
notable strategies for optimizing internal energy use. Birds (i.e., more energy available than needed) would cause the
and mammals must also face the energetic cost of endothermy, weakening of this drive and the cessation of feeding, which
which implies a several-fold increase of the continuous energy we call ‘satiety.’ A cybernetic model of this relationship is
requirements of their body mass and thus, a still greater ener- presented in Figure 1.
getic constraint. As a whole, these particular conditions result
in a set of physiological mechanisms that finely control the
acquisition and incorporation of energetic molecules, the 1.06.2.2 Not All Feeding Is Related to Energy
internal management of metabolic fuels, and the distribution
The formation and maintenance of animal tissues also
of the energy-consuming processes.
requires a series of exogenous elements whose primary role
is structural and their functions go beyond providing only
1.06.2.1 Energy Input: Food Intake and Its Physiological energy; instead, they have specific roles as biochemical constit-
Control uents and catalytic factors. Some of these elements are so crit-
ical that their availability is maintained by specialized
The ingestion of food is the only way to fulfill the elevated
physiological mechanisms specifically controlling their acqui-
energy demands derived from the high metabolic rate in
sition and elimination. Though independent from energy,
mammals. As such, food intake has the primary function of
these special requirements are met primarily by the same route
providing metabolic fuels in accord with the energy require-
as energy, i.e., by ingestion. This creates additional drives that
ments of the organism. The recognition of this basic relation-
are combined together in a single food-intake response. In
ship results in two logical principles:
some cases, such as water, the particular requirements generate
1. Energy availability is a critical variable that requires physi- a special repertoire of physiological and behavioral traits
ological mechanisms devoted to its maintenance. These (thirst/drinking) that is clearly distinguishable from that
mechanisms (involving a diversity of sensory cells, nerve devoted to energy (hunger/feeding). In most cases, however,

Figure 1 The control of hunger in the maintenance of energy availability. Energy is consumed continually in biological work and must be provided
from the environment. The energy availability to tissues results from the balance between energy acquisition and expenditure. The only significant
form of energy acquisition in mammals is food intake. The energy availability is critical and it requires a constant measurement of the variable by the
sensor and the processing of this information in an integration center. If too low or too high, the integration center ‘corrects’ the energy availability
by adjusting the processes determining it (feedback loops). In mammals, the correction is made essentially on the energy intake (i.e., feeding) while
expenditure is only modestly modified, so that feeding becomes a major determinant of the energy status of the organism. A positive energy balance
(increased input relative to expenditure; downward dotted arrow) results in excessive accumulation and obesity. A negative balance (increased expen-
diture; upward dotted arrow) implies fat reserve consumption. The physiological control of food intake refers to the mechanisms of adjustment of
food intake according to the energy status of the organism (dark frame in the figure). Based on Cabanac, M., 2001. Regulation and the ponderostat.
Int. J. Obesity Relat. Metab. Disord. 25 (Suppl. 5), S7–S12.
Food Intake and Its Control by Signaling Molecules 179

Figure 2 Feeding behavior is the resultant of a decision-making process aimed at maintaining energy availability. The process recruits information
from diverse sources, integrates it, and generates the pertinent outputs. The outputs include corrective adjustments in both energy income and
expenditure. Metabolic energy expenditure does not change greatly, so that most adjustments are made on the energy input by food intake.

the special requirements become manifest as subtle prefer- to recover a certain ideal state. Ingestive behavior (and the
ences for a certain flavor or food type, and they are called resulting food intake) is a main mechanism to meet that
‘specific appetites.’ These specific drives can produce clearly purpose. A schematic representation of the integrative
directed preferences such as for sodium (salt appetite), but decision-making process and its behavioral output is presented
in many cases (as for specific amino acids) an expected prefer- in Figure 2. From this perspective it emerges that feeding
ence behavior does not manifest itself clearly, probably behavior results from the integration of physiological signals
because it is overridden by another preference or because related mostly, but not exclusively, to energy availability.
such a mechanism simply does not exist. Either way, the turn- Over a time span of more than 100 years, diverse research
over rate of energy is much larger than that of the nonenergetic groups have tried to extract a logical principle describing the
nutriments, which means that the energetic molecules are physiological control of food intake. The motivation could
depleted and need to be replenished at a much higher rate have been an interest to manipulate food intake for therapeutic
than the nonenergetic ones. Consequently, the molecules (such as obesity treatment) or economical (such as cattle
potentially yielding metabolic energy are consumed in larger growing) purposes, but in most cases the motivation seems
quantities, for which they are called ‘macronutrients.’ In to be rooted in the human wish to understand biological
contrast, nonenergetic molecules, except water, generally complexity. The study of the physiological control of food
have long residence times within the organism and low elim- intake is represented by a succession of theories, enriched
ination rates, making their need quantitatively low (hence over time by previous experience, so that a progression is noted
‘micronutrients’). The consequence is that the physiological toward more accurate models. Let us make a brief review of this
control of food intake is centered on energy availability, while process.
the contribution of the specific appetites to feeding behavior is
controversial.
1.06.2.4 Main Concepts in the Study of Food Intake Control
and Their Historical Development
1.06.2.3 Study of the Physiological Control of Food Intake
As noted before, a negative energy balance is the main motiva-
Food intake can be envisaged as an array of behavioral mecha- tion for food intake. Energy deficit generates a series of physio-
nisms devoted to the acquisition of energy from the environ- logical signals that result in enhanced feeding behavior. This is
ment in accord with the particular requirements of the called hunger. The inhibition of these signals by the ingestion
organism. The behavioral events that make up the food- of food, with the consequent cessation of feeding, is called
intake pattern are typically divided in two categories: the satiety. In simple terms, feeding behavior can be visualized as
appetitive (related to craving and search of food) and the the alternation of periods of hunger and satiety. According to
consummatory (related to actual ingestion: biting, chewing, this, the first approach to understanding food-intake control
and swallowing). Each behavioral trait can be considered as is to elucidate the physiological origin of hunger.
the execution of a series of operative commands (i.e., an In everyone’s personal experience, the most evident and
output) produced by a physiological decision-making process. first-hand manifestation of hunger is a characteristic sensation
In order to match the actual body requirements, the decision- similar to a painful contraction of the stomach, known as an
making process recruits and integrates a vast amount of infor- epigastric pang. The relation between this pang and the physi-
mation from different sources, either directly related or not to ological process of hunger is intuitive, as the epigastric pang
the energy needs. The decision-making process consists of originates after a certain time of food deprivation, it increases
composing a comprehensive picture of the energetic state of in intensity with continued deprivation, ceases shortly after
the organism and determining the necessary actions, if any, food is ingested, and is clearly distinguishable from any other
180 Food Intake and Its Control by Signaling Molecules

gastric ache. The first attempt to establish a physiological mech- 1.06.2.6 Quest for the Signals
anism of hunger was the demonstration by Cannon and
Based on the concept that the main purpose of feeding is to
Washburn (1912) that the epigastric pang coincides with the
provide energy-rich substrates, the study of food intake
periodic contractions of the empty stomach. On this basis,
encountered a very complex set of facts to disentangle. Meta-
Carlson (1916) explained hunger as a set of sensations that
bolic energy has multiple forms that acquire different configu-
originated from contractions of the stomach when empty and
rations in different tissues; it is sensitive to different signals, it
ceased as food distended the gastric wall. This can be consid-
changes over time, and it has different functional meanings.
ered the first model proposed to explain the physiological
For instance, of the three main metabolic substrates, glucose
origin of hunger and satiety, and it is remarkable that it is
enters and exits the blood pool at a high rate but, unlike fatty
centered in the stomach as a major determinant of the feeding
acids and amino acids, it is subjected to a strict regulatory
response. In fact, experimental evidence demonstrates that
mechanism that severely opposes change. Fatty acids provide
gastric stimulation strongly influences feeding behavior. For
c.2.4 times more energy per gram than carbohydrates, but
instance, permanent gastric filling by means of a balloon
energy extraction in this case requires an additional metabolic
suppresses feeding in rats to the point of causing death by star-
apparatus based on mitochondrial oxidation, so that this
vation, but many aspects of feeding behavior cannot be
energy is inaccessible for some tissues. The estimated size of
explained in terms of gastric processes. In particular, this
the energy stores varies strikingly between the different forms;
‘gastric theory’ of hunger disregards the theoretical relationship
fats exceed by several hundred-fold the stores of glucose. This
between food intake and metabolic energy. Over the following
implies that the utilization of a certain quantity of stored
years the perceived role of the stomach in the control of food
energy impacts very differently the size of each deposit. Lastly,
intake has changed and it is currently visualized as a major
certain organs (e.g., the brain) are very sensitive to changes in
element of a feeding-control submechanism committed specif-
energy availability, for which they make use of vigorous
ically to the digestive process.
compensatory responses. Other organs (e.g., the liver) easily
handle marked reductions in metabolic energy eliciting,
1.06.2.5 Energy: A Compulsory Element instead, moderate routine responses. As the understanding of
In parallel with the motivational analysis of feeding behavior, food-intake control developed, it took the form of different
the problem of the physiological control of body weight was hypotheses that focused on certain processes or variables
examined experimentally. For several decades, it was debated considered to be indicative of the animal’s general energy
whether body weight is a regulated variable or a passive result status. Still, the subject was hunger, and the question was how?
of the processes on which it depends, i.e., energy intake and Glucose is the only substrate from which energy can be
expenditure. On the one hand, strong evidence indicated extracted in the absence of mitochondria and/or oxygen.
that body weight variability over time is much less than would Tissues in hypoxia or lacking mitochondria (e.g., erythrocytes)
be expected from a passive outcome; yet, human obesity sug- obtain energy exclusively from glucose or certain closely related
gested a lack of tight physiological control of fat accumula- compounds. Moreover, the enormous operating cost of
tion. The controversy motivated a number of studies nervous tissue (c.20% of the daily energy expenditure of the
focused on the physiology of body weight control, indirectly whole body for a tissue that represents only 2% of the body
including food intake as a factor. In this context a body mass) is supplied almost exclusively by glucose, because
weight-centered hypothesis emerged that guided much of nervous tissue does not oxidize fatty acids. Glucose reserves
the subsequent research in the field: the lipostatic theory. are relatively small (hepatic glycogen lasts for less than 24 h
Proposed by Gordon Kennedy (1953), the lipostatic theory of fasting), and when they have been exhausted the availability
establishes that a certain ‘desirable’ amount of fat stores is is maintained by hepatic de novo glucose synthesis at the
to be kept by adjusting the underlying processes. This mecha- expense of body proteins. It is evident that glucose is a critical
nism assumes that fat stores are constantly being energy substrate that also impacts more than any other energy
measured and that the information is sent to a regulatory source on brain metabolism. These premises point to glucose
center that ‘monitors’ the stores and determines the appro- as a reliable index of energy availability and thus, it can act
priate corrective actions if needed. This center must have as a feasible indicator of the need for food. The glucostatic
outputs to effector structures that execute the corrective theory proposed by Jean Mayer in 1953 states that blood
actions, such as adjustments in food intake. glucose is the signal determining hunger: the blood glucose
The lipostatic theory was originally proposed as a predictive pool is replenished after feeding and the subsequent extraction
model meaning that although some specific mechanisms, gradually reduces the size of the pool. The demonstration of
structures, and messengers were unknown at the time, their brain neurons sensitive to high and to low glucose concentra-
discovery was expected. This was particularly the case for the tions completed the scheme. Sensed by brain glucoreceptors,
mechanism of fat measurement, central to the theory, for which the reduction in blood glucose would be interpreted as an
a number of hypotheses were issued but not convincingly energy deficit, triggering the motivation to feed, i.e., provoking
solved until leptin was discovered in 1994 (see below). The hunger.
lipostatic theory is centered on the control of adipose mass The glucostatic theory was amply accepted as an explana-
and not directly on feeding; however, it must be taken as tion of hunger, probably because of its logical basis. However,
a crucial antecedent for the study of food intake as it offered there are some practical observations that conflict with it. First,
the first comprehensive model linking the ingestive response glycemia is a regulated variable, which means that changes in
to the broad energy status of the organism. glucose availability are physiologically compensated for, so
Food Intake and Its Control by Signaling Molecules 181

that blood glucose is expected to vary minimally. In fact, The theoretical models of hunger became gradually more
a significant hunger-triggering drop in blood glucose has inclusive as they were enriched with the concepts of previous
been hard to demonstrate. Modern measuring techniques theories and with new experimental evidence. In 1963
have revealed modest drops of 10%, at best, preceding sponta- a comprehensive model was issued that explained hunger in
neous food intake (Campbell, 1991). Also, brain tissue is rela- terms of the glucose reserves instead of the glucose concentra-
tively protected from variations in glucose availability because tion in blood. Glucose is a critical energy source, and metabo-
of the blood–brain barrier, which allows the cerebrospinal lism is clearly tuned to preserve glucose availability. Glucose
fluid to buffer the variations in plasma composition. However, cannot be stored in large quantities because glycogen retains
the strongest argument against the glucostatic theory is feeding water osmotically, and this effect multiplies the volume and
behavior in the diabetic state. Both types of diabetes mellitus the weight of the reserve. Virtually all cell types keep a glycogen
reduce insulin functionality and elevate blood glucose levels. reserve under these conditions to be used by the same cell.
Glucose entry into neural tissue is constitutive and not Only the liver maintains a glucose reserve (the hepatic
insulin-dependent, so that diabetic hyperglycemia implies glycogen) intended to provide glucose to other tissues during
excessive glucose provision to the brain. The ingestive response the nonfeeding periods. This reserve is replenished from the
predicted by the glucostatic theory would be permanent satiety, intestinal blood after feeding, and the glucose is gradually
but instead, the diabetic state is characterized by overfeeding. returned to the circulation as the fasting period progresses. In
The glucostatic theory was too simple to encompass all the fact, the liver is irrigated by a special arm of the circulation,
complexity involved in the physiology of hunger. Blood the hepatic-portal system that conveys the intestinal blood
glucose is a signal to be taken into account, but evidently it directly through the hepatic tissue before draining into the
does not suffice per se to explain feeding behavior. The gluco- general circulation. Hepatocytes express diverse enzymatic
static theory was not discarded, but in subsequent years mechanisms that can destroy, store, or transform metabolites
a number of research groups sought a model that could during their transit through the liver. Thus, the portal system
complement or substitute satisfactorily for the glucostatic funnels all absorbed nutriments from the intestine first to the
idea. In fact, the quest was for a signal that could simulta- liver, which can sense and modify blood composition before
neously indicate metabolic state of the organism and match it reaches other tissues. The liver is also the main site of glucose
the known relationships with feeding behavior. The different synthesis and export when glycogen reserves are exhausted. It is
approaches shared the premise that food intake was part of strategically situated to be a key element in energy metabolism
a mechanism intended to keep constant some other critical, controlling the availability not only of glucose, but of all major
still undetermined, variable which then became the signal energy substrates in the blood.
determining the feeding responses. Since the models were The hepatostatic theory, proposed by Mauricio Russek in
based on maintaining a certain variable, the suffix -static 1981, states that the control of hunger relies on the size of
(implying steadiness) was used to name the concepts. Hence, the hepatic glycogen store, sensed by glucosensitive cells in
glucostatic theory implies the control of feeding according to hepatic tissue. This information is conveyed to the brain
the glucose stability requirements. mainly by the hepatic branch of the vagus nerve and it is inte-
Over the following decades a series of hypotheses were grated at specific brain regions to produce the proper response,
proposed to explain hunger and satiety in the terms described hunger or satiety. Satiety is maintained as a function of the
above. The aminostatic theory proposed by Sherman hepatic glycogen, and when this reserve is consumed beyond
Mellinkoff et al. (1956) was based on the satiating power of a certain threshold, hunger begins to develop as a stimulus to
dietary protein, which is much stronger than that of carbohy- replenish it. At first the hunger sensation is mild but gradually
drates. In this case a supposed regulation of amino acid avail- becomes more intense as glycogen wanes. Hepatic glycogen
ability would be the main drive for feeding. Yet, this theory lasts for several hours; its consumption is initially rapid but it
did not resolve well the difficulties already posed against the gradually slows down as glucose is spared at the expense of
glucose-centered model and it did not gain further acceptance other fuels, mainly fat. The glycogen reserve is completely
as a general model for the control of feeding. depleted after c.18 h of fast. The time frame of these events
The thermostatic theory (Brobeck, 1948) had a different roughly matches the chronology of spontaneous feeding.
rationale. Feeding is accompanied by an increase in body The hepatostatic theory includes the central elements of the
temperature (prandial thermogenesis) originated mainly in glucostatic theory while it solves the theoretical difficulties.
visceral tissues involved in the digestion and absorption of Glycogen varies in a consistent way over time and it is
food. This warming is a consequence of both an increase in a much better indicator of glucose availability than blood
the rate of metabolic reactions and the reflex activation of the glucose itself. Most importantly, in the diabetic state, glycemia
sympathetic nervous system. Artificial warming of some is elevated at the expense of glycogen and amino acid reserves;
visceral organs, particularly the liver, has been shown to reduce hepatic glycogen is persistently low in this condition, which is
spontaneous feeding in rats, presumably to avoid excessive consistent with a persistent craving for food. Finally, the liver is
warming. From this, it was proposed that food intake is a key factor for the dynamics of other metabolites such as fats
controlled by a visceral temperature-centered mechanism and amino acids. Information on the supply of other fuels can
based on the capacity of food to stimulate thermogenesis and thus be incorporated into the feeding response by the same
on the impact of this effect on the thermal balance of homeo- nerves from the liver to the brain; this possibility is excluded
thermic animals. Again, this thermostatic principle does not if the brain itself acquires the information from blood glucose.
seem to operate consistently in all situations and cannot, there- The most controversial aspect of the hepatostatic theory has
fore, be considered a general mechanism of feeding control. been the central role it attributes to the liver and its innervation.
182 Food Intake and Its Control by Signaling Molecules

Previous observations had established the importance of other organs in providing information for central integration and
organs besides the liver in determining feeding behavior, for even in coordinating general responses for which the brain
example the stomach, the adipose mass, and the brain. It was was only one of many effectors (a kind of ‘peripheralism’). In
even debated how patients who suffered liver transplants this latter view, a major peripheral administrator was the liver,
showed hunger–satiety cycles if the liver innervation had much in the way proposed in Russek’s hepatostatic theory.
been completely sectioned. Subsequent experiments favored The subsequent incorporation of experimental evidence
the view that the liver is a major controller of metabolism and the corresponding evolution of ideas led to the ischymetric
and a strong determinant of feeding behavior, but it also sup- theory, proposed by Stylianos Nicolaïdis in 1987. This theory
ported the integration of information from other tissues to proposes that the essential factor in the control of food intake
form a comprehensive picture of the metabolic needs of the is the potential contribution that the different fuel forms in the
body. The prevailing view was that the metabolic information body can make to the general energy budget. Conversion of the
controlling food intake should come from different sources, chemical energy contained in the fuel molecule to cell-usable
involving structures both inside (central) and outside (periph- ATP (i.e., the ‘power’ of the fuel) is then the critical variable
eral) the central nervous system. to be measured and maintained with little regard for the loca-
In the 1950s stereotaxic surgery, an experimental technique tion of the fuel. This theory still holds the liver as a prime
developed 10 years earlier, began to be applied to explore element, but it broadens the scope of the model to include
experimentally the role of the brain in controlling feeding in other reserve structures, particularly the adipose tissue. As can
laboratory animals. Stereotaxic preparations make it possible be seen, the ischymetric theory did not resolve the central
to manipulate a specific brain region while sparing neighboring versus peripheral controversy, but it incorporated and
zones. The selective destruction of a certain cell group creates condensed the main ideas at that time about the different
a permanent condition allowing the effects of such lesions to aspects involved in the physiology of hunger.
be thoroughly studied. Stereotaxic destruction of different The central versus peripheral controversy was never
brain regions demonstrated that a group of hypothalamic resolved. The debate was abandoned in 1994 when the newly
neurons, the so-called ventromedial hypothalamus (VMH) available molecular biology tools facilitated the discovery of
was implicated in the production of satiety, for animals lacking leptin. The next step was to find the main site of leptin action,
it ate beyond normal limits and developed obesity. The loss of i.e., the location of leptin receptors, which turned out to be the
appetite, even leading to death, was observed after the destruc- brain. From that time on, a series of studies gradually unveiled
tion of a nearby region known as the lateral hypothalamic area the details of the mechanism by which the brain receives
(LHA) (Anand and Brobeck, 1951). These results were inter- peripheral information carried by hormones. A number of
preted as evidence that the brain contains a group of neurons previously unknown hormones have been described that
dedicated to causing hunger (a ‘hunger center’) and another constitute an elaborate network of metabolic signals involved
one to causing satiety (the ‘satiety center’). In the following not only in the control of food intake but also in the long-
years a great deal of work was dedicated to collecting additional term maintenance of energy balance and body weight and
data reinforcing the concept. A model was then proposed in how they are coordinated with massive, energy-consuming
which hunger and satiety originate by the alternating inhibition processes such as growth and reproduction. The discovery of
of these two antagonistic centers by each other. Moreover, the leptin brought about the key element missing in Kennedy’s
model proposes that the centers themselves collect information lipostatic theory, i.e., a signal indicating the size of the fat
about the general metabolic state by means of glucosensitive deposit. The discovery of the hunger hormone ghrelin, secreted
neurons measuring glucose concentration directly from blood by the stomach, paradoxically reintroduced gastric fullness as
or cerebrospinal fluid, in much the same way proposed by an important determinant of hunger in a way similar to that
the glucostatic theory. This model is known as the dual-center conceived by Cannon’s gastrostatic concept.
hypothesis. Understandably, the points of conflict previously The new data have been incorporated into a modern model
noted in the glucostatic theory were also present in the dual- of the control of food intake. This model relies on the activity of
center hypothesis. In particular, this hypothesis again two neuron groups inside the hypothalamic arcuate nucleus
conceived the control of feeding as based mainly, if not exclu- (ARC) at the base of the brain. These neurons receive hormonal
sively, on glucose availability to the brain, thus neglecting all information from the blood and nervous information from
the possible contributions made by the peripheral organs. other areas of the central nervous system. One of the neuron
This conflicted with the main views at that time and initiated groups initiates a nerve pathway that has the overall effect of
a debate that motivated research in the field. The dual-center promoting food intake (hence orexigenic group), and the other
hypothesis soon became widespread in spite of its various reduces food intake by a similar route (hence anorexigenic
weaknesses, probably because it offered a simple solution to group). The orexigenic group inhibits the anorexigenic one.
the problem of the origin of hunger and because it assumed The output of this integration is conveyed to other hypotha-
a brain-centralized control of the physiological functions of lamic areas, including the VMH and the LHA as second-order
the body in the way a man-designed mechanism would be centers. Integration successively involves other brain areas
organized. and terminates in hormonal, nervous, and behavioral outputs
The controversy extended for years and derived basically that determine the compensatory responses executed by the
from two opposite perspectives: one asserting brain self- effectors. These responses include the appetitive and the
sufficiency for collecting all pertinent information and gener- consummatory phases of food intake. This new model has
ating the corresponding responses (a kind of ‘centralism’) been extensively adopted as an appropriate substitute for all
and another supporting the critical involvement of peripheral previous ones.
Food Intake and Its Control by Signaling Molecules 183

In some aspects, the modern model can be envisaged as a re- action of bile acids and lipases, peptidases, and amylases,
adaptation of the dual-center hypothesis. The antagonistic producing mono- and disaccharides, free fatty acids, and glyc-
hunger and satiety centers have been replaced by the orexigenic erol, as well as free amino acids and short peptides.
and anorexigenic neuron groups, though with the addition of Assimilation of nutrients into the ‘internal medium’ of the
a sensory mechanism that gathers diverse peripheral informa- body occurs in two phases: triacylglycerides are packed by intes-
tion. However, the participation of the peripheral organs is tinal cells into lipoproteins called chylomicrons which are
again underestimated. The contributions made by the vagal directed to lymphatic vessels. Eventually, chylomicrons reach
afferents and the brainstem integration are barely considered, the blood circulation via the subclavian vein. All the other
and the possibility of extra-brain control of feeding is simply nutrients are directed from duodenum to the liver by the portal
beyond the scope of the model. Clearly, the current model artery. In the liver, nutrients are first in contact with a special-
for the control of food intake still needs to be improved. ized population of hepatocytes called ‘periportal’ hepatocytes.
The implicit purpose in constructing a model for the phys- This part of the liver is rich in metabolic pathways that need
iology of hunger is to create a comprehensive structure that a high concentration of O2, such as b-oxidation of fatty acids,
includes all known factors affecting feeding behavior, describes gluconeogenesis, and ureagenesis. Periportal hepatocytes also
their interrelations, and predicts their effects (Figure 2). In the show more numerous mitochondria and higher xenobiotic
past, the evolution of ideas progressed from theories based on processing as well as glutamine synthesis activity. Some meta-
specific structures or signals to complex models encompassing bolic pathways such as glycogen synthesis do not show hepatic
many elements. Involvement and interrelationship seem to be ‘zonation.’
fundamental features of physiological mechanisms, so there is When food is available, the anabolic reactions are favored
no reason to expect that the mechanism of food-intake control and hence, pathways such as those for the synthesis of
should reproduce the straightforward design of a man-made glycogen, fatty acids, triacylglycerols, cholesterol, and the
machine. The way we now explain feeding is predicted to pentose phosphate pathway, are activated. This anabolic
grow in complexity and inclusiveness over time. ‘wave’ is coordinated by a set of endocrine messengers, among
which insulin plays a prominent, but not unique, role. Other
hormones that are secreted in response to feeding are the intes-
1.06.3 Bioenergetics of the Fasting–Feeding Cycle tinal peptides tyrosine tyrosine (PYY) and cholecystokinin
(CCK), adipose tissue leptin, pancreatic polypeptide (PP),
All heterotrophic organisms are subject to a metabolic rhyth- gastric obestatin, etc. Overall, the endocrine response to
micity dictated by the fasting–feeding cycle. Even in particular feeding promotes the synthesis of biomolecules and their poly-
circumstances, such as animals in a vivarium or on the farm mers, cellular growth and maturation, storage of caloric fuels,
with nourishment available all the time, or many humans in and the message of satiety for the hypothalamus. Levels of gly-
modern societies with food constantly accessible, the organ- cemia are increased.
isms display a clear dichotomy in their behavior: a period of Within the liver, triacylglycerides combine with other
resting/fasting and a period of activity/feeding. This rhyth- components to form lipoproteins, in this case, very low density
micity is more pronounced in wild animals and in nonindustri- lipoproteins (VLDL) which contain a high proportion of lipidic
alized human cultures where meal availability is uncertain on material (more than 90%), and are exported to the circulation
a daily basis. From a metabolic and endocrine point of view, for transport to other peripheral tissues and organs.
these two periods are characterized by specific adaptations The definition of fasting varies. For some authors, fasting
that have important consequences for the bioenergetic status starts as soon as the organism stops eating; for others, fasting
of the biological systems. Recognizing that the fasting–feeding exists until the anabolic response associated with feeding
cycle is continuous, we will start by describing the adaptations culminates and the metabolic fuels, glycogen and triacylglycer-
associated with food ingestion. ols, are replenished. Most organisms are subject to a period of
Feeding triggers a complex set of coordinated responses that fasting when they are sleeping/resting. This natural or physio-
recruit different organs as the digestive process progresses. logical fasting is usually enough to deplete the carbohydrate
Digestion allows the complex nutrients ingested to be broken reserves that form the hepatic glycogen deposits, releasing
down into biomolecules (carbohydrates, lipids, and amino glucose units to keep blood glucose at normal levels. Coordi-
acids). The steps involved in this process take place in different nated with the depletion of glycogen, the liver responds by
sections of the digestive track: (1) in the mouth the food is activating the expression of gluconeogenic enzymes (phosphe-
macerated, and a set of amylase enzymes start to hydrolyze nolpyruvate kinase, PEPCK, glucose 6-phosphatase, G6Pase,
polysaccharides, mainly starches; (2) from the mouth the pyruvate carboxylase, PC, and others) to allow the conversion
food is swallowed and transits to the gastric chamber through of nonglycosidic molecules (glycerol, alanine, lactate) to
the esophagus; (3) digestive juice secreted into the stomach glucose that is mainly released into the blood but can also be
contains zymogens that are activated by the acidic environ- incorporated into glycogen depots. These hepatic adaptations
ment, unmasking their hydrolytic activity toward proteins are coordinated by endocrine regulators, mainly pancreatic
and peptides; (4) the assimilative part of the digestive process glucagon and the glucocorticoids produced in the suprarenal
occurs in the first section of the small intestine called the gland cortex.
duodenum. In this segment the pancreatic and bile ducts After the glycogen reserve is depleted, the next fasting adap-
release pancreatic juice and bile via the ampulla of Vater in tation that occurs is the mobilization of lipid deposits in the
an action controlled by the sphincter of Oddi. It is in the adipose tissue. Activation of hormone-sensitive lipase activity
duodenum where nutrients are simplified by the coordinated promotes the hydrolysis of triacylglycerides into fatty acids
184 Food Intake and Its Control by Signaling Molecules

and glycerol. Both molecules leave the adipocytes, enter the The ARC has a major role in the control of feeding. In this
circulation, and are captured by gluconeogenic tissues (mainly hypothalamic region there are two populations of neurons
liver) and cardiac muscle. In the liver, fatty acids are oxidized by that integrate nutritional information: one expresses neuropep-
the coordinated action of mitochondria and peroxisomes to tide Y (NPY) and the Agouti-related peptide (AgRP), both
yield energy substrates that fuel the Krebs cycle. In addition, orexigenic factors; the other population expresses anorexigenic
modified forms of fatty acids act as ligands of transcriptional peptides such as proopiomelanocortin (POMC) and the
factors such as peroxisome proliferator-activated receptors cocaine- and amphetamine-regulated transcript (CART). All
(PPARs) to orchestrate an adaptive fasting response. In the these neurons are differentially sensitive to circulating
liver, the excess oxidized fatty acids are channeled to the gener- hormones such as leptin, insulin, peptide tyrosine tyrosine
ation of ketone bodies (acetoacetate and b-hydroxybutyrate; (PYY), and ghrelin, and to metabolites such as glucose, fatty
dimethyl ketone is formed by spontaneous decarboxylation acids, and amino acids (Blouet and Schwartz, 2010).
of acetoacetate). As a consequence, ketonemia levels increase
in the fasting condition. Ketone bodies serve as oxidizable
1.06.4.2 Neuronal Circuits
substrates, being incorporated into the Krebs cycle in the brain,
heart, kidney, skeletal and cardiac muscles, and the lactating Anatomical regions participating in feeding behavior can be
mammary gland. viewed from different degrees of complexity, for example: as
Peripheral mechanisms for regulating appetite include the a set of organs forming the digestive system; as individual
motor functions of the stomach, such as the rate of emptying organs like the liver, with all its diverse and coordinated cellular
and accommodation, which convey symptoms of satiation to types; or as a set of specialized cells communicating with each
the brain. The rich repertoire of peripherally released peptides other, sending and receiving chemical messages. Additionally,
and hormones provides feedback from the arrival of nutrients we can introduce the concept of assemblies of neuronal cells,
in different regions of the gut from where they are released to i.e., neuronal circuits that establish dynamic connections to
exert effects on satiation or to regulate metabolism through regulate diverse physiological responses such as feeding.
their incretin effects. Ultimately, these peripheral factors Considered simplistically, such neuronal circuits seem
provide input to the highly organized hypothalamic circuitry linear, but in reality they form overlapping and interconnected
and vagal complex of nuclei to bring about cessation of energy populations of neurons. Importantly, the neuronal resting
intake during a meal and the return of appetite and hunger after membrane potential and the action potential firing frequency
fasting. Understanding these mechanisms is key to the physio- of the active circuits controlling feeding behavior are under
logical control of feeding and the derangements that occur in a dual regulation: (1) by the intrinsic biophysical properties
obesity and other pathological conditions related to feeding. of each neuron and (2) by the integrated synaptic activity in
response to inputs from other neuronal circuits and from
peripheral signaling molecules such as leptin, insulin, CCK,
1.06.4 Hypothalamus and Feeding Control and others. Overall, the set of these communicating networks
integrate neuronal and endocrine stimuli, relevant to the
1.06.4.1 General Perspective
eating response, such as (1) satiety hormones from the gut,
The hypothalamus is a diencephalic structure formed by (2) homeostatic sensors of energetic demands, (3) hedonic
a variety of nuclei (around 15 according to neuroanatomy text- and motivational drives, (4) taste and smell clues, and
books) which play a dominant role in maintaining the internal (5) retrieved memory of previous experiences (Hirayama and
milieu. It is the anatomical substrate that controls important Gillette, 2012).
physiological activities such as body temperature, thirst, sleep, The principal circuits that regulate feeding and energy
and circadian rhythmicity. Moreover, the hypothalamus is metabolism are located in the hypothalamus and the
a key element in regulating hunger and the energetic state of midbrain. Among the best-characterized chemical messengers
the body (Lechan and Toni, 2013). Located in the tuberal in these neuronal circuits are the orexigenic peptides NPY/
region, the ARC plays a preeminent role in the driving of AgRP, which are secreted by neurons in the ARC to target nuclei
feeding and the ventromedial nucleus is one of the major brain in other brain areas and the spinal cord. The anorexigenic factor
regions controlling satiety. The tuberomammillary nucleus a-MSH (a-melanocyte stimulating hormone) is synthesized in
(TMN), situated in the posterior hypothalamus, also partici- POMC neurons (within the arcute nucleus) and secreted
pates in feeding and the sensing of energy balance. Nuclei in into the paraventricular nucleus of the hypothalamus (PVH),
the anterior region that participate in the control of food intake parabrachial nucleus (PBN), dorsal vagal complex (DVC),
are the lateral nucleus, which contains orexigenic neurons that and intermediolateral column of the spinal cord (IML) (Valassi
project throughout the brain and spinal cord, and the supra- et al., 2008).
chiasmatic nucleus (SCN), which is the major pacemaker that Recent studies have begun to demonstrate a role for non-
controls circadian physiology (Buijs et al., 2006). neuronal cell types in the regulation of energy homeostasis.
Classical ablation experiments demonstrated that bilateral In particular, the potential importance of different glial cell
lesions of the extreme lateral part of the ventromedial nucleus types is increasingly being recognized. A number of studies
impaired food intake. In contrast, stimulation of this zone of have described changes in the activity of hypothalamic macro-
the nucleus promoted increased feeding. The medial part of glia (principally astrocytes and tanycytes) in response to states
the ventromedial nucleus was also shown to be important in of positive and negative energy balance, such as obesity and
feeding since its bilateral lesion was associated with hyper- fasting (Buckman and Ellacott, 2014). For example, endoge-
phagia, eventually leading to obesity (Swaab et al., 1993). nous proteins with ‘benzodiazepine-like’ activity known as
Food Intake and Its Control by Signaling Molecules 185

endozepines are downregulated in fasting conditions, whereas were localized in the hindbrain (Ritter et al., 1981), in catechol-
cerebral administration of benzodiazepines reduces food aminergic nuclei A1/C1 C2–C3, and in surrounding nuclei
intake in rodents (Lanfray et al., 2013). Receptors for endoze- such as the reuniens (Ritter et al., 1998). In addition, the area
pines are not well characterized. They have been reported to postrema (AP) and the solitary tract nucleus (NTS) vagal sensi-
bind with high affinity to the a-aminobutyric acid receptor tive neurons are necessary to integrate the glycoprivic-induced
type A (GABAA) receptor as well as to recognize a mitochondrial feeding (Ritter and Taylor, 1990). NTS/PVT projects to the
protein with translocation activity (Christian and Huguenard, PBN in the pons and the paraventricular nucleus (PVN), the
2013). Further studies are necessary to clarify the physiological ARC, and other areas of the hypothalamus (Juárez and
role of this pathway in the regulation of energy homeostasis. Tongju-Koh, 1978). The PB also projects to the PVN, ARC,
VMH, and dorsomedial hypothalamic nucleus (DMH) (Saper
and Loewy, 1980). These connections indicate that hindbrain
1.06.4.3 Feeding Circuits in the Brain
primary relay nuclei connect to hypothalamic primary relay
Neuronal circuits involved in sensing peripheral signals of neurons and also to some of the secondary relay hypothalamic
energy balance from the body, and the regulation of feeding neurons.
itself are mainly located in the hypothalamus and the hind- In the ARC a set of neurons coexpress AgRP/NPY, which
brain (Drazen and Woods, 2003; Blouet and Schwartz, initiate feeding (Ollmann et al., 1997; Hahn et al., 1998; Gropp
2010). Neurons from these areas are characterized by the et al., 2005; Luquet et al., 2005; Aponte et al., 2011). Another
expression of receptors for adiposity signals such as leptin neuronal group coexpresses POMC/CART which stops feeding
and insulin, hunger signals such as ghrelin or hypoglycemia, (Koylu et al., 1997; Schwartz et al., 1997; Kristensen et al.,
or to respond to postprandial increase of circulating levels of 1998; Elias et al., 1999; Chen et al., 2015). AGRP/NPY neurons
nutrients or release of gastrointestinal peptides such as CCK in the ARC also contain GABA (Ovesjo et al., 2001; Meister,
or NPY (Drazen and Woods, 2003; Wynne et al., 2005; 2007), while GABAA receptors have been located both in
Kleinridders et al., 2009; Berthoud et al., 2012). Additionally, AGRP/NPY neurons in the ventromedial ARC and POMC/
other neuronal systems, like the corticolimbic and the reward CART neurons in the ventrolateral ARC. It has been proposed
systems of the brain, participate in a nonhomeostatic regula- that GABA release from ARGP/NPY neurons into POMC/
tion of feeding (Woods, 1991; Berthoud, 2004, 2011). These CART neurons in the ARC is part of the regulatory mechanism
circuits are shown in Figure 3. of feeding (Cowley et al., 2001).
ARC neuronal populations containing AgRP/NPY or
1.06.4.3.1 Homeostatic Circuits POMC/CART express the growth hormone secretagogue
Primary relay nuclei that receive peripheral signals for hunger receptor (GHSR) (Guan et al., 1997; Willesen et al., 1999)
and satiation are located in the hindbrain and the ARC in the and the orexin A receptor (OXI1-R) (Guan et al., 2002; Suzuki
mediobasal hypothalamus (Leibowitz and Wortley, 2004; et al., 2002). In response to gherlin or orexin, AgRP/NPY
Sanchez-Lasheras et al., 2010). Neurons responding to gluco- neurons show pacemaker bursts of action potentials driven
privation after intraventricular administration of 5-thioglucose by low-threshold calcium conductances and a transient

Figure 3 Schematic representation of a sagittal view of rodent neural networks involved in the control and regulation of feeding behavior. Inputs or
primary relay nuclei are shown in red; integration or secondary relay nuclei are shown in light blue; efferent or third order relay nuclei are shown in
green. Modulatory neural networks involved in nonhomeostatic regulation of food intake are shown in brown. Arrows extending beyond the brain
represent peripheral inputs (red) and outputs (green), thin arrows represent neural projections while very thick arrows indicate hormonal signaling.
Abbreviations: Amy, amygdala; AP, area postrema; ARC, arcuate nucleus; DMH, dorsomedial hypothalamus; E, endocrine inputs/outputs; Hip, hippo-
campus; LA, lateral hypothalamus; NAc, nucleus accumbens; NTS, nucleus of the solitary tract; PBN, parabrachial nucleus; PFA, perifornical area;
PFC, prefrontal cortex; PS, parasympathetic system; PVN, paraventricular nucleus of the hypothalamus; PVT, thalamic paraventricular nucleus;
S, sympathetic system; SCN, suprachiasmatic nuclei; VMH, ventromedial hypothalamus; VTA, ventrotegmental area.
186 Food Intake and Its Control by Signaling Molecules

outwardly rectifying potassium conductance (van den Top PVN induced feeding and drinking behavior (Leibowitz, 1978).
et al., 2004). ARC neurons also express leptin receptor (OB- Similar but more potent effects were also found after PVN injec-
R) in both populations AgRP/NPY (Mercer et al., 1996; tions of NPY at picogram doses (Stanley and Leibowitz, 1985).
Schwartz et al., 1996a) and POMC/CART (Cheung et al., The current view points to a role of the PVN as part of the
1997; Elias et al., 1998a; Cowley et al., 2001). Leptin output pathway that regulates the autonomic and endocrine
administration inhibits electrical activity and mRNA influence on the control of feeding (Leibowitz and Wortley,
expression in AgRP/NPY arcuate neurons (Elias et al., 1999; 2004).
Higuchi et al., 2005) but increases electrical activity and Efferent or third-order relay nuclei that participate in the
mRNA expression of POMC/CART neurons (Wilson et al., output pathways for regulation of food intake and metabolic
1999; Cowley et al., 2001; Cone, 2005). Since leptin inhibits energy balance involve mainly the dorsal and ventral medulla
NPY/AgRP neurons in the ARC, the concomitant decrease in and AP/NTS nuclei (Adan et al., 2008), the hypothalamic
GABAergic input to the POMC/CART neurons would projections from PVN to the median eminence, as well as its
synergize leptin-induced activation in the latter. projections to the autonomic sympathetic system in the spinal
Secondary relay nuclei integrating hunger and satiety receive cord (Jobst et al., 2004; Leibowitz and Wortley, 2004; Cone,
projections from both NPY/AgRP and POMC/CART neuronal 2005). Dorsal and ventral medulla and AP/NTS nuclei are
populations in the ARC. These secondary relays include the capable of integrating feeding responses. This was shown in
lateral hypothalamus (LH), the perifornical area (PFA), the decerebrated animals, which discriminate taste (bitter from
VMH, the DMH, and the PVN. The LH was considered sweet), increase sucrose intake in response to 24 h food depri-
the neural ‘center’ controlling hunger but nowadays neurons vation or glucoprivation induced by 2-deoxyglucose or
synthesizing melanin-concentrating hormone (MCH) have 5-thioglucose administration, and show a reduced intake after
been described in the LH. Within the LH and PFA, neurons syn- direct sucrose injection into the stomach (Norgren and Grill,
thetizing orexin (OXI) have been described. ICV administra- 1982; Flynn and Grill, 1983; Ritter and Taylor, 1990; Ritter
tion of either peptide induces spontaneous feeding in et al., 1994; Andrew et al., 2007; Darling and Ritter, 2009). It
satiated animals (Qu et al., 1996; Sakurai et al., 1998). Both has been shown that GLUT2-expressing neurons from NTS
MCH and OXI neurons in the LH and PFA express melanocor- comprise a distinct population of hypoglycemia-activated
tin receptors MC3R-MC4R, stimulated by a-MSH and b-MSH neurons via increased AMP-activated protein kinase
(peptides produced by posttranslational processing of activity and closure of leak Kþ channels (Lamy et al., 2014).
POMC), and inhibited by AgRP (Sánchez-Lasheras et al., These receptors seem to be involved in the parasympathetic
2010). NPY receptors Y1 (NPY1-R) and Y5 (NPY5-R) are response leading to glucagon secretion. On the hand, the PVN
present in all secondary feeding relay nuclei (Eva et al., 2006; projects to the median eminence from CRH- and thyrotropin-
Higuchi, 2012). On the other hand, OXI neurons from the releasing factor (TRF)-containing neurons (Palkovits, 1987;
PFN project to the ARC, PVN, DMH (Elias et al., 1998b; Fliers et al., 1994) to control adrenocorticotropin (ACTH) and
Horvath et al., 1999; Suzuki et al., 2002), locus coeruleus TSH secretion from the pituitary in relation to energy balance
(LC), pedunculo pontine and laterodorsal tegmental pons (Smith and Ferguson, 2008). On the other hand, a PVN
(PPT/LDT), and the TMN to regulate arousal (Siegel, 2004; projection to the intermediolateral column of the spinal cord,
Saper et al., 2005; Harris and Aston-Jones, 2006) and its the NTS, and X nucleus in the medulla (Saper et al., 1976;
relation to energy balance (Yamanaka et al., 2003). LH Luiten et al., 1985) is involved in the parasympathetic and
neurons expressing orexin also project to the mesencephalic sympathetic regulation promoted by insulin and glucagon in
ventral tegmental area (VTA) and to the shell region of the relation to feeding (McCabe et al., 1984; Weiss and Leibowitz,
nucleus accumbens (NAc), which participate in the brain 1985; Thorens, 2015).
reward system, possibly involved in the effects of food as Other neural circuits involved in the regulation of feeding
a behavioral reinforcer. are the reward system of the brain, which assigns a hedonic
The VMH has been implicated in the regulation of food value to food (Alonso-Alonso et al., 2015) and the corticolim-
intake since the early 1940s; after a revision of its role on meta- bic regions, which regulate feeding in relation to food choice
bolic regulation recent new evidence also supports its role in and preferences (Berthoud, 2007). These areas include the orbi-
food intake. Similarly, DMH neurons have been implicated tofrontal, insular, and cingulate cortices, as well as the amyg-
in food intake and body mass regulation as its lesion produces dala and hippocampus (Berthoud, 2011; Parent et al., 2014).
hypophagia and hypodipsia and decreases body weight in indi- The reward system lies in the mesolimbic dopaminergic projec-
viduals with normal body composition. It has been suggested tion from the VTA to the NAc and the prefrontal cortex. In turn,
that the DMH functions as an interface between the LH and these nuclei form a circuit feeding-back among them and
the VMH which projects to the PVN, thus integrating informa- receiving inputs from the LH (Harris and Aston-Jones, 2006;
tion related to feeding behavior with the autonomic and endo- Adan et al., 2008). Furthermore, VTA neurons have insulin
crine regulation of metabolism (Bernardis and Bellinger, receptors (InsR) that assign a reward value to food intake by
1998). The PVN has also a role in the regulation of ingestive the release of dopamine into the NAc (Karatsoreos et al.,
behaviors; the results of early experiments in which VMH 2013). EC and opioid signaling at LH also participate in the
lesions deeply affected feeding have been reinterpreted and it assignment of hedonic value to food (Di Marzo and Matias,
is now known that such VMH lesions were disrupting the 2005; Di Marzo et al., 2009; Alonso-Alonso et al., 2015; Liu
ventral noradrenergic bundle that connects the hindbrain and Borgland, 2015). Corticolimbic areas are involved in the
noradrenergic nuclei with the PVN (Gold, 1973). Furthermore, cognitive and learning processes related to food recognition
injection of low doses of noradrenaline (less than 5 ng) in the and preferences which are particularly relevant for human
Food Intake and Its Control by Signaling Molecules 187

feeding behavior (Berthoud, 2004; Zheng et al., 2009). Thus throughout the day, in a coordinated manner within the whole
previous experience with food is represented in the orbitofron- organism. Chronostasis operates upon ongoing homeostatic
tal and insular cortices, while food- or feeding-related affective processes by updating the set point of physiological systems
information is processed and stored in the amygdala and the according to a timetable determined by the circadian clocks.
hippocampus (Zheng and Berthoud, 2007). A neural circuit In mammals the SCN is the only neural structure shown to
which may integrate homeostatic information along with the function as a biological clock, although the molecular oscilla-
hedonic and affective value assigned to food includes the para- tors driving the clock are present in most (if not all) cells in
ventricular thalamic nucleus (PVT), which is reciprocally con- the organism and are known as peripheral circadian oscillators
nected with the feeding areas of the brain stem and (Albrecht, 2012; Aguilar-Roblero et al., 2015). The set point has
hypothalamus and the NAc and amygdala (Li and Kirouac, been defined as the information used to compare the instant
2008; Kirouac, 2015; Lee et al., 2015). value of a variable by a regulatory system in order to generate
Finally, the SCN is the circadian clock controlling the an error signal to correct deviations from this value (Russek
expression of behavioral and physiological circadian and Cabanac, 1982). The coding of this information in the
rhythms in mammals (Morgado et al., 2015). The SCN system could result from the structure and dynamics of the
projects to hypothalamic nuclei involved in the regulation feedback loops in the regulatory system, such as endocrine or
of food intake and metabolism mainly the DMH, the subpar- neural networks. Alternatively, it could be represented in the
aventricular zone (SPvZ) and to the PVN (Watts and Swan- concentration of chemical signals (hormones or neurotrans-
son, 1987; Watts et al., 1987; Chou et al., 2003; mitters), the density of specific receptors or effectors, such as
Abrahamson and Moore, 2006; Kalsbeek et al., 2010). The ionic channels, or even the level of gene expression related to
SCN also projects to the sympathetic system via the ventro- all of the above.
lateral medulla A5 which, in turn, projects to the intermedio- The value of the set point for each physiological variable,
lateral column of the spinal cord and then to the adrenal along with its daily variations, has resulted from natural
medulla and other sympathetic outputs (Nagai et al., 1996; selection throughout thousands of years of evolution.
Buijs et al., 1999). SCN regulation of hypothalamic nuclei However, it is possible that nowadays such set point values
allows chronostatic regulation of feeding and metabolism, may be outdated, since during at least the last 1000 years
as discussed below. we have dramatically altered the environment, and adapta-
tion through natural selection occurs in a much larger time
scale. Frequent transoceanic flights, night work, and shift
1.06.5 Chronostasis: Physiological Regulation work schedules are good examples of altered chronostatic
across Time regulation. These alterations are due to outdated and time-
misaligned set points in relation to habits and demands of
Feeding behavior starts when we feel hungry and ends when we modern lifestyle and they induce physiological changes
feel satiated. If only it were as simple as that! But when do we leading to obesity and metabolic diseases (Bass and
feel hungry? The answer is quite complex, particularly in the Takahashi, 2010; Buxton et al., 2012).
case of humans and other domesticated animals, but when Chronostatic regulation of feeding involves SCN neural
food is easily available – even in the wilderness – feeding outputs to the hypothalamic and autonomic neural networks
behavior starts because it is time. Such time or, rather, times related to feeding and metabolism, which are responsible for
to hunt or collect and then to eat are initially selected consid- circadian rhythms in food intake, activity of digestive enzymes,
ering the status of energy deposits and circulating levels of gastrointestinal motility and absorption, and hormonal secre-
nutrients (homeostatic process) and adjusted on the basis tion of cortisol, insulin, and leptin (Kalsbeek et al., 2006;
of the effort involved in gathering food, the risk involved in Garaulet and Madrid, 2010; Kalsbeek et al., 2011). Further-
the process, and the preferences for a particular diet (nonho- more, restriction of food availability to 2–3 h each day
meostatic processes) (Lenard and Berthoud, 2008; Shin et al., entrains circadian rhythmicity of feeding behavior, endocrine
2009; Berthoud et al., 2012). The latter involve cognitive secretion, and liver metabolism (Escobar et al., 1998; Díaz-
processes such as motivation, learning, and memory related Muñoz et al., 2000; Escobar et al., 2000; Aceves et al., 2003;
to previous experience (Alonso-Alonso et al., 2015). But Ángeles-Castellanos et al., 2004; Martínez-Merlos et al.,
besides cognitive processes, the animal must be able to 2004; Báez-Ruiz et al., 2005), even in SCN-lesion animals
measure the time of day. Circadian rhythms are fluctuations (Stephan, 1983, 1989; Marchant and Mistlberger, 1997).
in physiological and behavioral parameters which are repeated Such evidence has supported the idea of a food entrainable
almost every 24 h; these rhythms are generated by endogenous oscillator whose location has been so far elusive
biological clocks in living organisms (Menaker et al., 1978). (Rosenwasser and Adler, 1986). This has lead us to propose
Circadian rhythms and their underlying clocks have evolved a distributed oscillator involving neural, endocrine, and
in relation to a cyclic environment and provide adaptive rele- gastrointestinal elements (Aguilar-Roblero and Díaz-Muñoz,
vance by synchronizing the individual to daily environmental 2010) where ghrelin inputs to the DMH may play a major
cycles such as light–dark, temperature, and food availability role as an interphase between peripheral and neural
(Albrecht, 2012). components (Mieda et al., 2004).
Circadian rhythms manifest themselves in physiological Recent advances in molecular genomics have shown that
regulation across time or chronostasis (Aguilar-Roblero and chronostatic regulation of feeding may occur by direct interac-
Díaz-Muñoz, 2010; Aguilar-Roblero, 2015). Thus each physio- tion among ‘clock genes’ and genes related to metabolism
logical system adjusts itself to different but optimal values which include per1, 2 and 3, cry1 and 2, dec1 and 2, rev-erb-a,
188 Food Intake and Its Control by Signaling Molecules

rora, bmal1 (also named arntl), and clock (Green et al., 2008). 1.06.6 Neuropeptides
Thus, mice with a mutation in the gen clock increase their food
1.06.6.1 Neuropeptides Regulating Feeding Behavior
intake during the day which decreases the amplitude of
feeding circadian rhythms. They also present increases in 1.06.6.1.1 Orexins
blood levels of glucose, triacylglycerides, cholesterol, and lep- The orexins, also named hypocretins are neuropeptides
tin and decreases in insulin; plus, they become obese (Turek described in the late 1990s. The family comprises two peptides:
et al., 2005). On the other hand, mice with a mutation of orexin A with 33 amino acids and orexin B with 28 amino
bmal-1 show a decreased insulin sensitivity throughout the acids. The sequence of orexin A contains four cysteines, and it
day and also become obese (Shi et al., 2013). These data has been suggested that it forms two different interchain disul-
clearly indicate a chronostatic regulation of metabolism fide bonds (Sakurai, 1998). Both orexins originate from the
which, if absent, leads to metabolic dysfunctions. Other exam- same precursor named prepro-orexin (PPOX), a peptide of
ples of chronostatic regulation by interaction among clock and 130 amino acids in rodents and 131 in humans. The gene
metabolic genes include (1) SIRT, a deacetylase which senses coding for this precursor is on chromosome 17 in humans
NADþ, and thus functions as a metabolic sensor; it binds to and on chromosome 10 in mice (De Lecea et al., 1998; Sakurai
Clock/Bmal1 to promote Per2 deacetylation and degradation et al., 1998). The PPOX sequence is highly conserved among
which, in turn, may amplify the oscillation in the transcription vertebrates; the human sequence shows 82% identity with
of other clock-controlled genes (which contain an E-BOX in that of mice, 95% with macaca, and 39% with zebrafish
their promoter region) (Nakahata et al., 2008; Grimaldi (Figure 4). Orexin function as a regulator of feeding has been
et al., 2009); (2) cyclic AMP-activated protein kinase maintained since the origin of vertebrates.
(AMPK) also functions as a nutrient sensor due to its The orexins were first described as the endogenous ligands
sensitivity to the AMP/ADP/ATP ratio; by promoting of an orphan G protein-coupled receptor (GPCR) named
phosphorylation and dephosphorylation of different OX1R (Sakurai et al., 1998). It is now known that orexins
proteins including CRY, it could function as an energy- have two specific receptors, OX1R and OX2R, also named
charge transducer to the circadian oscillator (Lamia et al., HCRTR1 and HCRTR2, both of which are preferentially
2009); and (3) the PPAR-1a (and also PPAR-b/d and PPAR- coupled to Gaq proteins and consequently to the PLC-IP3-
g) a nuclear receptor functioning as a transcription factor Ca2þ pathway (Voisin et al., 2003). Human OX1R and OX2R
that regulates metabolism. It has been recently shown that display 64% amino acid identity (Sakurai et al., 1998).
PPAR-1a binds PER while REV-ERBa is a target of PPAR-g The distribution of the mRNA encoding for OX1R and
thus promoting bmal-1 expression via the PPAR gamma OX2R in the rat brain showed marked differences; OX1R is
coactivator 1-alpha (PGC-1a) (Liu et al., 2007; Eckel-Mahan most abundant in the VMH nucleus, but it is also present in
and Sassone-Corsi, 2013). the tenia tecta, dorsal raphe, and nucleus coeuruleus. OX2R

Figure 4 Alignment (a) and percent identity (b) of pre-pro-orexin (PPOX) peptides from different species of mammals by the Clustal Wallis
algorithm.
Food Intake and Its Control by Signaling Molecules 189

is expressed most abundantly in the paraventricular nucleus, tetracycline is removed from the diet was recently applied to
and it is also expressed in the cerebral cortex, NAc, subthala- study the orexinergic system. These mice conditionally express
mic and paraventricular thalamic nucleus, and anterior pre- diptheria toxin A under the control of the PPOX locus. When
tectal nucleus (Trivedi et al., 1998). Interestingly, dogs and the toxic transgene is expressed after puberty, a rapid loss of
mice without a functional OX2R gene display a phenotype orexinergic neurons was observed (80% cell loss in 7 days).
similar to humans with narcolepsy (Lin et al., 1999; Chemelli In addition to the marked sleep alterations and narcolepsy,
et al., 1999). these mice were also obese and had a low rate of activity; their
Orexinergic neurons in the LHA project their axons to levels of leptin and insulin were higher than those of control
several regions of the cerebral cortex, limbic system, and brain animals but the levels of free fatty acids and glucose were not
stem (Nambu et al., 1999). The distribution of orexinergic affected (Tabuchi et al., 2014).
neurons had been analyzed in detail by immunohistochem-
istry in slices of the rat brain. The main immunoreactive 1.06.6.1.3 Orexins and Energy Regulation
neurons were detected in the hypothalamus, specifically in The actions of orexinergic neurons involve an intricate web of
the LHA, DMH, perifornical nucleus and posterior hypotha- interactions that are only partially known. The orexinergic
lamic area. The axons of these neurons are widely distributed terminals contact neurons relevant for feeding regulation.
in the brain. The orexinergic terminals have been identified Important interactions are those established with NPY- and
in several regions of the hypothalamus, two of which, the POMC-positive neurons of the ARC nucleus; thus, injection
ARC and the paraventricular hypothalamic nucleus, are of orexins into the ARC nucleus induces food intake. Moreover,
strongly related to feeding control. The extrahypothalamic in isolated NPY neurons, orexins induce a rise in intracellular
nuclei showing orexinergic terminals include the cerebral Ca2þ ([Ca2þ]i), and in POMC neurons they induce the oppo-
cortex, hippocampus, amygdala, brain stem, locus coeuruleus, site effect (Muroya et al., 2004). In agreement with this result,
and raphe nuclei (Nambu et al., 1999). food intake induced by intracerebroventricular injection of
orexin-A (10 nmol) is partially blocked by the administration
1.06.6.1.2 Orexins and Feeding Control of BIBO3304 (60 mg), an antagonist of the Y1 receptor for
The location of the orexinergic neurons in the LHA, a region NPY (Yamanaka et al., 2000). These pieces of evidence suggest
classically related to feeding that also contains other peptides that NPY- and POMC-expressing neurons are effectors of
associated with appetite regulation (Bernardis and Bellinger, orexins.
1993), suggests that the orexinergic neurons have a role in Orexinergic neurons are sensitive to nutritional status, since
this function. Other seminal observations support an essential fasting induces an increase in PPOX (Sakurai, 1998). Orexins
role for these peptides in feeding control: (1) the intracerebro- can also induce increases in glucose levels by interacting with
ventricular injection of orexin A or B in the light phase induces the sympathetic system (Yi et al., 2009). Indeed, orexinergic
an increase in food consumption (Sakurai et al., 1998) and neurons are able to sense glucose levels since, in isolated orex-
water intake (Kunii et al., 1999); (2) fasting for 48 h or acute inergic neurons, glucose application induces hyperpolarization
hypoglycemic manipulation induces an increase in the tran- and abolishes action potentials; conversely, glucose depriva-
script of PPOX (Cai et al., 1999); and (3) the intraperitoneal tion induces depolarization and increases the frequency of
injection of the selective antagonist for OX1R, SB-334867-A, action potentials (Yamanaka et al., 2003).
reduces food intake in rats (Haynes et al., 2000). Feeding regu- Leptin, a hormone secreted by adipocytes that negatively
lation by orexins has also been documented in bullfrog larvae regulates feeding, also interacts with the orexinergic system.
(Shimizu et al., 2014) and zebrafish (Yokobori et al., 2011). The intracerebroventricular administration of leptin reduces
Genetic approaches have been applied to the study of the hypothalamic expression of PPOX and of the OX1 receptor
orexin; a knockout (KO) mouse for PPOX has been developed; and also inhibits the fasting-dependent increase in PPOX
its most conspicuous phenotype was a severe sleep alteration mRNA (López et al., 2000). In agreement, leptin stimulation
very similar to narcolepsy in humans (Chemelli et al., 1999; of isolated orexinergic neurons, which express the leptin
Hara et al., 2001), revealing that orexinergic neurons display receptor (Håkansson et al., 1999), has the direct effect of
complex functions including feeding behavior and sleep reducing the electrical activity of these neurons (Yamanake
control. The ataxin-3 gene product was isolated from et al., 2003). Orexins can also regulate the activity of glucose-
a Machado-Joseph disease patient under the control of the responsive neurons, with effects opposite to those of leptin,
PPOX gene promoter; using gene ablation techniques it was particularly in neurons from the VMH (Muroya et al., 2004).
possible to reduce the number of orexinergic cells selectively. Experimental evidence indicates that the orexinergic system is
Like the PPOX/ mice, this transgenic mouse has sleep alter- part of a complex circuitry modulating feeding in an opposite
ations and narcolepsy, as well as metabolic alterations. way to leptin.
Although the transgenic mice show a decrease in food intake Recent studies of orexinergic neurons with a pharmacoge-
in the first weeks after birth, adult mice are obese. The authors netic technique named designer receptors exclusively activated by
explain that this apparent contradiction is the result of low designer drugs (DREADD), a method that makes possible the
motor activity and/or a low metabolic rate and that the loss selective activation or ablation of orexinergic neurons, have
of other secreted peptides regulating feeding might be partici- reinforced a role for orexins in feeding behavior. The pharma-
pating in the induction of this phenotype in the transgenic cogenetic stimulation of orexinergic neurons induced an
mice (Hara et al., 2001). increase in locomotor activity, food and water intake, and the
A new model with conditional genetic ablation employing blood glucose level. When ablation was induced, it was
the tet-off system, where the transgene is expressed when observed that the elimination of 73% of the orexinergic
190 Food Intake and Its Control by Signaling Molecules

neurons did not induce any changes; however, elimination of and glucose levels were normal. CNR1/ mice also display
83% of these same neurons resulted in decreased food and important changes in hypothalamic neuropeptides, for
water intake (Inutsuka et al., 2014). This study demonstrates example, high levels of corticotropin-releasing hormone
that the selective activation or elimination of orexinergic (CRH) and the CART, which is also related to feeding control
neurons has clear effects on feeding. (Cota et al., 2003).
Studies analyzing a second CB1/ mouse (Di Marzo et al.,
2001) indicate that, in response to 18 h of fasting, CB/
1.06.6.2 Endocannabinoid System
animals reduced their food consumption compared with
The ECs are chemical messengers derived from arachidonic acid normal littermates, in agreement with a role for the CB1
(AA). All are eicosanoids produced by the conjugation of AA receptor in this response. The administration of the CB1 antag-
with ethanolamide to form fatty acid amides (FAA), i.e., onist SR1416A reduced food intake in the wild type but not in
N-arachidonoylethanolamide (anandamide), or with glycerol the KO mice, also indicating a role for CB1 in the control of
to form monoacylglycerols (MAGS), the best known of which feeding. This study outlined a functional interaction between
is 2-arachidonoyl-glycerol (2-AG) (Devane et al., 1992; leptin, the main signal regulating the anorexigenic response,
Mechoulam et al., 1995; D’Addario et al., 2014). and the EC system; thus, the administration of leptin in normal
EC exert their action through binding to specific receptors rats induced a 50% reduction in the hypothalamic concentra-
that belong to the GPCR superfamily. Two subtypes of EC tion of anandamide and 2-AG; in agreement, obese Zucker
receptors have been described in vertebrates: CNR1 and rats deficient in leptin signaling displayed high levels of hypo-
CNR2 (originally named CB1 and CB2); they were cloned in thalamic ECs.
the early 1990s from mouse, rat, and human (Matsuda et al., On the other hand, an interaction of the EC system with
1990; Gerard et al., 1991; Munro et al., 1993). Both are coupled ghrelin has also been shown; thus, blocking the CB1 receptor
to proteins Gi/o, and consequently to the negative regulation of in the hypothalamic paraventricular nucleus (PVN) induces
adenylyl cyclase; their activation modulates the activity of an increase in the fasting-induced hyperphagia and potentiates
mitogen-activated protein kinase (MAPK), inward-rectifying the orexigenic effect of ghrelin. These observations suggested
potassium channels (Kir), A and D outward-rectifying potas- that the PVN participates in the integration of orexigenic–
sium channels, and N- and P/Q-type calcium channels (Mu anorexigenic signals (Soria-Gómez et al., 2014).
et al., 1999; Pertwee, 2005). In addition, CNR1 can also acti- A functional interaction between the cannabinoid and orex-
vate the Gs protein, thereby activating adenylyl cyclase (Chen inergic systems has emerged from evidence demonstrating that
et al., 2010). these two systems act synergistically to modulate feeding (for
CNR1 is the main cannabinoid receptor expressed in the detailed review see Flores et al., 2013). This notion arose
nervous system. It has been reported to be present in neuron from the fact that CB1, OX1R, and OX2R have similar expres-
terminals of the central and peripheral nervous system sion patterns in the central nervous system (Hervieu et al.,
(Egertová et al., 2003), but it is also expressed in some periph- 2001; Mackie, 2005), and it is reinforced by the fact that CB1
eral organs (reviewed in Maccarrone et al., 2014); CNR2 and OX1R can form dimers (Hilairet et al., 2003) that,
is mainly expressed in the immune system (Pertwee, 2005). regarding some functions, e.g., regulating energy balance,
In situ binding studies in brain slices of several mammalian constitute a functional unit (Crespo et al., 2008; Verty et al.,
species (rat, guinea pig, dog, rhesus monkey, and human) 2009; Cristino et al., 2013).
using a high-affinity ligand named [H3]CP55,940 showed It has long been established that POMC neurons promote
specific binding mainly in the basal ganglia (the sustantia nigra satiety (Yang et al., 2011); however, recent findings, obtained
pairs reticulata and globus palidus), the hippocampus, and the by applying DREADD to regulate the activity of specific
cerebellum (Herkenham et al., 1990). By in situ hybridization, POMC neurons, suggested that POMC neural activity is neces-
mRNA coding for CB1 was detected in neuronal populations of sary for the CB1-dependent activation of food intake in the
the rat dorsal ganglia (Hohmann and Herkenham, 1999). satiety state (Koch et al., 2015). These observations reveal
that the new experimental approaches open novel ways to
1.06.6.2.1 Endocannabinoids and Feeding Control study the details of feeding control circuits.
Early evidence demonstrated that D9-tetrahydrocannabinol
(Williams et al., 1998) or anandamide induces hyperphagia
1.06.6.3 Melanocortin System
(Williams and Kirkham, 1999). This role for cannabinoids is
conserved in chicken (Alizadeh et al., 2015). 1.06.6.3.1 POMC-Derived Peptide Family
There has been growing clinical interest in the EC system Existing evidence shows that the melanocortin system is
since the introduction of the CB antagonist SR141716A (rimo- strongly associated with feeding; the melanocortins (a-, b-
nabant or AcompliaÒ) by the pharmaceutical company Sanofi- and g-MSH), the ACTH, and b-endorphin (b-end) are a family
Aventis for the treatment of obesity. of peptides derived from one precursor named
The study of the CNR1 KO mouse (CNR1/) highlighted POMC (Figure 5; Switonski et al., 2013). The POMC gene is
the role of components of the cannabinoid system in feeding. located on chromosome 2p23; it consists of three exons and
Thus, the CNR1/ mice exhibit marker hypophagia, probably two introns, and its organization and processing have
induced by the disruption of the central control of appetite; been analyzed (Chang et al., 1980; Drouin and Goodman,
these mice also showed decreased body weight and reduced 1980). In humans, POMC is expressed in corticotrophs of
fat mass, as well as adipocyte dysfunction. In addition, CNR1 the hypophysis; however, in other vertebrates it is also
KO mice exhibit lower levels of plasma leptin, but insulin expressed in melanotrophs, cells that are vestigial in humans
Food Intake and Its Control by Signaling Molecules 191

Figure 5 Posttranslational processing of proopiomelanocortin (POMC) peptide. Modified from Coll, A.P., Tung, Y.C.L., 2009. Pro-opiomelanocortin
(POMC)-derived peptides and the regulation of energy homeostasis. Mol. Cell. Endocrinol. 300, 147–151.

(Raffin-Sanson et al., 2003). POMC is also produced by a well- hypothalamic neurons expressing AgRP are located specifically
defined neuronal cell population in the ARC nucleus (Civelli in the ARC nucleus, an essential structure for feeding regula-
et al., 1982). Other tissues expressing POMC are dermal micro- tion. These neurons are also positive for NPY and they inner-
vascular endothelial cells (Scholzen et al., 2000) and keratino- vate hypothalamic and extra-hypothalamic structures
cytes (Schauer et al., 1994). (Broberger et al., 1998; Hahn et al., 1998).
POMC by itself is inactive; it is posttranslationally processed
by subtilisin-related proprotein convertases PC1-3 and the 1.06.6.3.3 Receptors of Melanocortin
expression of these proteins in a specific tissue determines the The melanocortin family of peptides exerts its action by acting
presence of a specific, POMC-derived peptide. The processing on specific receptors. The first melanocortin receptor (MCR)
of POMC potentially generates various peptides of was cloned in the early 1990s and was designated the a-MSH
physiological importance, for example ACTH, a-MSH, receptor (Mountjoy et al., 1992; Gantz et al., 1993). Currently,
b-MSH, and b-end. Thus, in the anterior pituitary where PC1 the family of MCR contains five members (MC1R–MC5R) that
is expressed, the cleavage of POMC produces mainly ACTH; are encoded in autosomes by single-exon genes. All of them
in lower vertebrates the intermediate lobule of the pituitary belong to the GPCR super-family and they display differential
expresses PC2 and cleaves ACTH to produce a-MSH (Seidah affinity for the natural agonist and antagonists, giving speci-
and Chretien, 1992). ficity to the system (Rodrigues et al., 2015).
In the hypothalamus, the most important structure for In general, MCRs transduce their signals via the cAMP/PKA
feeding regulation, the 32-kDa precursor POMC is cleaved by system because they are coupled to Gs proteins. In addition,
PC1 to generate pro-ACTH and b-lipotropin (b-LPH), and MC3R is able to couple Go proteins and MC4R to Go and Gq
pro-ACTH is cleaved again by PC1 to form ACTH, joining proteins (Newman et al., 2006); signaling through MC4R
peptide (JP), and N-POC. After that, PC2 cleaves ACTH to also implicates activation of the PLC-IP3-Ca2þ pathway.
generate ACTH1–17 and corticotropin-like intermediate lobe The expression of melanocortin receptors is tissue specific:
peptide (CLIP), and it cleaves b-LPH to form a-LPH and MC1R is mainly expressed in melanocytes and leukocytes,
b-end; the processing of POMC in the hypothalamus is MC2R in the adrenal gland, MC3R and MC4R in the nervous
specific, because the hypophysis does not express PC2. system and MC5R in diverse tissues (Yang et al., 2011).
Carboxypeptidase E (CPE), peptidyl a-amidating mono- MC3R and MC4R are strongly related to feeding and energy
oxygenase (PAM), and N-acetyltransferase catalyze the balance and mutations in MC3R are associated with type 2 dia-
conversion of ACTH1-17 into mature a-MSH (Zhou et al., betes and pediatric obesity (Mountjoy, 2010; Feng et al., 2005).
1993; reviewed in Pritchard et al., 2002). MC4R mutations are linked to morbid obesity in humans
(Loos, 2011).
1.06.6.3.2 Aguoti-Related Peptide (AgRP), a Natural
Antagonist of the Melanocortin System 1.06.6.3.4 Melanocortins and Feeding Control
An outstanding characteristic of melanocortin system is the The first insight that POMC participated in feeding regulation
existence of natural antagonistic peptides; the most relevant was provided by the report of children lacking Pomc products.
of these for food-intake control is AgRP. This peptide of 132 These individuals developed different degrees of metabolic
amino acids was originally cloned by two groups (Ollman deficiencies including hypocortisolemia, hyperphagia, and
et al., 1997; Shutter et al., 1997). It displays a high similarity obesity (Krude et al., 1998).
with the Agouti protein present in the melanocytes that Animal models to study this deficiency were developed: KO
produce the agouti-colored coat in mice; AgRP inhibits the mice lacking all the POMC peptides have characteristics resem-
actions of a-MHS on MCR1 in skin cells. bling those observed in humans: hyperphagia, obesity, altered
AgRP is mainly expressed in the hypothalamus, and its pigmentation, and adrenal insufficiency (Yaswen et al., 1999;
main function is to act as an antagonist of the MC3R and Challis et al., 2004). Pomc/ mice also showed reduced
MC4R melanocortin receptors, thereby inhibiting the actions activity, low metabolic rate, and reduced activity of the
of a-MSH (Ollman et al., 1997; Yang et al., 1999). The hypothalamus–hypophysis–thyroid axis (Challis et al., 2004).
192 Food Intake and Its Control by Signaling Molecules

Taking into account that deletion of the Pomc gene resulted in mice lacking InsR expression in AgRP neurons had altered
the lack of several peptides, these mice were used to define hepatic glucose production (Könner et al., 2007), revealing
the importance of each one of the POMC-derived peptides that these neurons are elements of a long chain that participates
in the metabolic phenotype; thus, Pomc/ mice were treated in integrating information from the whole organism.
with intracerebroventricular injections of the different POMC-
derived peptides at the onset of the dark period, and the
1.06.6.4 Neuropeptide Y
anorexigenic effect was evaluated. a-MSH had the most
potent effect, reducing food intake by 65%; this peptide also The hypothalamic peptide named NPY was discovered in the
induced a decrease in body weight after 3 days of early 1980s, in extracts of porcine brain (Tatemoto et al.,
administration (Tung et al., 2006). These observations 1982). NPY, peptide YY (PYY), and PP belong to the same
showed that a-MSH is the main POMC-derived anorexigenic family; NPY is most abundant in the central and peripheral
peptide. On the other hand, a potent orexigenic effect has nervous system (Pedragosa-Badia et al., 2013) and is highly
been described for AgRP when it was administered by expressed in the brain stem, in the hypothalamus, and in the
intracerebroventricular injection into the paraventricular adenohypophysis. Hypothalamic neurons expressing NPY
nucleus (PVN) (Wirth and Giraudo, 2000) or ARC nucleus have been detected in the ARC and DMH nuclei (Chronwall
(Aponte et al., 2011; Krashes et al., 2011). et al., 1985; Bi et al., 2001).
Feeding control is the result of the integration of several NPY receptors (NPYR) belong to class A (rhodopsin-like) of
signals; importantly, the actions of hormones regulating the GPCR superfamily; in mammals five subtypes are currently
diverse aspects of feeding and energetic state by acting on hypo- known: Y1, Y2, Y4, Y5, and Y6; in humans Y6 have not been
thalamic neurons are now outlined. The functional interactions reported. All subtypes are coupled to Gi/o heterotrimeric
between POMC and AgRP hypothalamic neurons and leptin, G-proteins and, in consequence, their activity induces
ghrelin, and insulin have already been described. a decrease in the concentration of cAMP; through a Gbg
Starvation and inhibition of leptin signaling induce a greater dimer they regulate Ca2þ and Kþ ion channels (Pedragosa-
increase in hypothalamic AgRP cDNA than in POMC mRNA Badia et al., 2013).
(Thornton et al., 1997; Mizuno et al., 1998). In fact, POMC The NPYR are expressed in a tissue-specific pattern, and the
and AgRP neurons in the ARC nucleus express leptin receptors location and receptor subtype in the CNS determines their
and are the main target for the anorexinergic actions of this physiological function. In accord with their widespread expres-
hormone, since leptin induces depolarization of POMC sion, they participate in diverse processes, e.g., learning and
neurons (Cowley et al., 2001) and hyperpolarization of AgRP memory, anxiety, and circadian rhythms (Mercer et al.,
neurons (van den Top et al., 2004). 2011). The Y4 and Y5 receptors are noteworthy for the orexi-
Recent genetic restitution experiments in mice lacking the genic actions of NPY since their expression has been described
expression of leptin (Lepob/ob) or its receptor (Leprdb/db) in the ARC and VMN hypothalamic nuclei. Thus, Y5 transcript
showed that the recovery of leptin signaling specifically in was identified in the hippocampal dentate gyrus, in the CA1
hypothalamic AgRP neurons, but not in POMC or SF1 neurons, area, and in the ARC nucleus (Naveilhan et al., 1998); Y4 tran-
is sufficient to restore the regulatory actions of leptin on glucose script was found in the brain stem, solitary tract nucleus (NTS),
concentration (Gonçalves et al., 2014). This evidence demon- the vagal complex, and the ARC and VMN nuclei (Parker and
strates that the hypothalamic melanocortin system, specifically Herzog, 1999).
AgRP neurons, is essential to regulate the glucose levels in mice. The projections of NPY-ergic neurons connect some
Another important element in the machinery of feeding hypothalamic nuclei such as the PVN, VMN, LHA, and DMN
control is ghrelin, an orexigenic hormone secreted by a popula- (Mercer et al., 2011) to establish an informational web.
tion of enteroendocrine cells (Cummings et al., 2001). Impor-
tantly, the injection of ghrelin into the ARC nucleus induces 1.06.6.4.1 NPY and Feeding Control
a strong food-intake response (Nakazato et al., 2001; Wren A role for NPY in food intake has been demonstrated by diverse
et al., 2001). This hypothalamic structure expresses ghrelin experiments. Thus, the acute intracerebroventricular injection
receptors (GHRS), mainly in AgRP-positive neurons of NPY induces an increase in food intake (Clark et al.,
(Kamegai et al., 2000). These observations led to the 1985), and chronic administration of the peptide leads to
proposal that ghrelin actions are mediated, at least in part, by obesity (Zarjevski et al., 1993). In agreement, NPY levels are
AgRP hypothalamic neurons; this hypothesis was supported increased in the hypothalamus of obese rats (Dryden et al.,
by the fact that ghrelin modulates the electrical activity of 1995), and conditions of low energy, such as food restriction
AgRP neurons (Cowley et al., 2003). Recently, using directed or fasting, induce elevated hypothalamic NPY levels (Brady
genetic restitution in mice lacking GHRS expression, it was et al., 1990).
shown that AgRP neurons are sensitive to ghrelin signaling NPY exerts its orexigenic action by directly regulating the
(Wang et al., 2013). anorexigenic POMC neurons which express NPYR. Neurons
On the other hand, deletion of InsR in the brain was found expressing NPY in the hypothalamus also express AgRP and
to induce obesity and increase food intake of female mice GABA, and these neurons release NPY in the postsynaptic
(Brüning et al., 2000), raising the possibility that insulin acts POMC neurons, negatively regulating their electrical activity
directly on the CNS. This idea was tested by the development (Hahn et al., 1998; Cowley et al., 2001; Roseberry et al.,
of transgenic mice with selective inhibition of the InsR in 2004; Acuña-Goycolea et al., 2005).
POMC or AgRP hypothalamic neurons. Although no direct Although the first mice lacking NPY expression (NPY/)
effects on food intake or body weight were observed, the did not manifest altered feeding phenotypes such as obesity
Food Intake and Its Control by Signaling Molecules 193

or fat accumulation (Erickson et al., 1996), this genetic back- GHRL gene encodes six exons, four of which code for preprogh-
ground sensitized the pathway related to feeding. Thus, the relin mRNA products of 117 amino acids, which include the
cross between NPY/ with the leptin-deficient mice ob/ob peptides: AG, UAG (unacylated ghrelin), and obestatin (Seim
attenuated notably the obesity syndrome that resulted from et al., 2010; Wajnrajch et al., 2000); however, it also contains
the deletion of leptin expression (Erickson et al., 1996). transcriptional variants. The promotor region of the ghrelin
It is well known that the blood–brain barrier in the ARC gene has several consensus sequences for a number of transac-
nucleus is semipermeable, allowing actions of the feeding- tivating regulatory proteins (e.g., upstream stimulatory factor
control hormones leptin and insulin. Leptin inhibits NPY/ 1/2) and includes an E-box site (Kanamoto et al., 2004).
AgRP neurons of the ARC nucleus and stimulates POMC Finally, ghrelin gene expression in human tissue is
neurons. The result is a potent anorexigenic response (Ahima widespread in endocrine and nonendocrine tissues, with the
et al., 1996); similar findings had been described for insulin, highest ghrelin mRNA levels found in the stomach fundus
a hormone produced by b-pancreatic cells (Schwartz et al., (Gnanapavan et al., 2002).
1992). However, it is not possible to generalize the actions of Ghrelin is also present in the unacylated isoform or de-
hypothalamus NPY and AgRP neurons, since a study applying octanoylated (UAG). UAG comprises 70–90% of total plasma
genetic ablation techniques, using transgenic mice expressing ghrelin (AG plus UAG), which is partially explained by the
the diphtheria toxin receptor in the brain under the regulation short half-life (<8 min) of AG (Hosoda and Kangawa, 2004;
of the promoter of AgRP showed that the populations of AgRP Kojima et al., 1999). Ghrelin biosynthesis and secretion occur
and NPY neurons in the hypothalamus are not identical primarily in the stomach, specifically in endocrine cells of the
(Luquet et al., 2005), suggesting differential roles in feeding- oxyntic gland; however, it is also produced by other cells and
behavior regulation. organs such as gut, alpha and epsilon cells of pancreatic islets,
To define the contribution of the NPYR subtypes in feeding placenta, kidney, testis, lung, and the hypothalamic
regulation, some studies have used KO animals. Y4 KO mice ARC (Ghelardoni et al., 2006; Granata et al., 2010; Kanamoto
were crossed with ob/ob mice, but the obese phenotype of ob/ et al., 2004; Kojima et al., 1999). In rats, ghrelin-containing
ob was not affected (Sainsbury et al., 2002). Meanwhile, Y5 cells are localized in the large curvature of the pars glandularis
null mice develop mild late-obesity characterized by elevated and also in the small stomach curvature, while ghrelin-
food intake, adiposity, and body weight, unchanged re-feeding containing cells are distributed throughout the human
after fasting or sensitivity to leptin, and the absence of a response stomach, although mainly in the fundus (Li et al., 2011).
to intracerebroventricular injection of NPY (Marsh et al., 1998). The regulation of ghrelin secretion has not been completely
These results demonstrated that the Y5, but not the Y4, receptor elucidated. Fasting, CCK-octapeptide, and acetylcholine stimu-
mediates the actions of NPY on feeding. late total ghrelin release (Shrestha et al., 2009); in contrast,
A recent study showed that germline ablation of the inhibitory stimuli are feeding, hyperglycemia, insulin, and
expression of both the Y1 and Y5 receptors (Y1Y5/) results somatostatin (Sakata et al., 2012; Shrestha et al., 2009). In
in hypophagia. When the ablation was directed to the hypo- pathological conditions such as obesity, ghrelin circulating
thalamus (Y1Y5 (hyp/hyp)), despite hypophagia, the mice levels are reduced, whereas they are increased by malnutrition
showed increased body weight and fat accumulation possibly or cachexy (Veldhuis and Bowers, 2010). Basal plasma ghrelin
due to compensatory mechanisms; however, these results did levels are usually lower in obesity and higher in individuals
reveal a role for Y1 and Y5 in food-intake regulation (Nguyen suffering from anorexia nervosa (Germain et al., 2007; Yildiz
et al., 2012). et al., 2004).
Ghrelin is secreted in a pusatile pattern, and it is released in
larger quantities before meals, in agreement with its important
1.06.7 Endocrine Systems: Hormones in Fasting role for meal initiation (Cummings et al., 2001; Kojima and
and in the Feeding Response Kangawa, 2005). In rodents, the daily profile of circulating
AG or UAG is distinctly modulated in response to feeding para-
1.06.7.1 Ghrelin and Leptin Biology and Their Role in Food-
digms (Arellanes-Licea et al., 2014; Bodosi et al., 2004) and the
Intake Regulation
AG receptor participates in regulating food anticipatory activity
1.06.7.1.1 Biology: Synthesis, Secretion, Physiology, and under conditions of regularly scheduled feeding (Blum et al.,
Signaling 2009; LeSauter et al., 2009).
Ghrelin from the Proto-Indo-European language root ghre, Beyond their metabolic functions, both ghrelin isoforms
meaning grow, was purified in 1999 from rat and human stomach have pleiotropic ones, ranging from cell proliferation to cogni-
extracts with activity as a growth hormone- (GH-) releasing tive processes (Müller et al., 2015; Seim et al., 2010). Even
peptide. Moreover, it proved to be the endogenous ligand for though the UAG receptor(s) has not yet been characterized,
the GH secretagogue receptor (Kojima et al., 1999). Ghrelin is UAG downregulates lipogenic gene expression in the absence
a peptide hormone of 28 amino acids (molecular weight of of the GHSR in muscle and white adipose tissue (WAT) of
3.37 kDa) with a lipid moiety, an n-octanoyl group, on serine mice (Delhanty et al., 2010). In the digestive system, some of
three; it is designated acylated ghrelin (AG). This serine acylation the functions of AG and UAG are antagonistic. AG controls
is catalyzed by the ghrelin O-acyltransferase (GOAT) enzyme, gastric motility, augmenting its tone, while UAG decreases it;
a membrane-bound acyltransferase of the endoplasmic HCl secretion is stimulated by AG, but UAG, does the opposite.
reticulum (Yang et al., 2008). In pancreatic islets, AG modulates glucose metabolism and the
Ghrelin is encoded by the GHRL gene; in humans it is sensitivity to insulin secretion depending on its concentration
located on the short arm of chromosome 3 (3p25–26). The and metabolic state. AG, but not UAG, inhibits insulin
194 Food Intake and Its Control by Signaling Molecules

secretion and the latter reverts AG insulin-desensitizing effects 1994 as a protein hormone secreted by adipocytes (Halaas
(Delporte, 2013; Gauna et al., 2007; Salehi et al., 2004). In et al., 1995; Zhang et al., 1994). The discovery of leptin came
the cardiovascular system, the two ghrelin isoforms regulate in part from the existence of mutant obese rodents. One was
cell proliferation and blood pressure (Delporte, 2013; Li a mouse colony with an obese phenotype at the Jackson Labo-
et al., 2006). The third gene product of the ghrelin gene, obesta- ratory in 1950, designated ob/ob. Another strain had a pheno-
tin, might have effects opposite to those of AG, reducing food type similar to ob/ob but was also markedly diabetic and it was
intake; however, debate exists as it might not affect food-intake termed db/db; both mouse strains were infertile and lethargic
regulation (Murphy and Bloom, 2006). (Coleman, 1978; Friedman and Halaas, 1998). It was later
AG stimulates hypophyseal GH secretion directly or by found that the genes that were mutated in these animals were
modulating GH-releasing hormone (Feng et al., 2011; Kojima the ligand leptin (ob) and the long leptin receptor isoform
et al., 1999; LeRoith et al., 2001). Further, AG causes a positive (db) genes (Friedman and Mantzoros, 2015).
energy balance, i.e., a surplus of energy stores in the organism, The genes ob (obese) and Lep (leptin) are localized on chro-
that mechanistically could be explained by the adipogenic mosome 7 in humans and chromosome 6 in mice (Green et al.,
effects of AG, as it promotes the proliferation of adipocytes 1995). Leptin gene expression has been detected in WAT,
from WAT (Lanfranco et al., 2010; Veldhuis and Bowers, stomach, liver, heart, placenta, pituitary gland, and brain
2010; Wells, 2009). Another explanation is based on the (Denver et al., 2011; Green et al., 1995). The profile of leptin
relevant orexigenic role of AG in central feeding regulation gene expression in rat WAT is circadian, with a peak during
(Murphy and Bloom, 2006; Tschöp et al., 2001), explained in the dark phase (Saladin et al., 1995).
the following sections. Leptin is a protein with 167 amino acids, a molecular
Ghrelin was originally discovered as the natural ligand for weight of 16 kDa, and a crystal structure homologous to the
the GHSR, an orphan receptor known to bind drugs that members of the long-chain helical cytokine family with a 4-
increased the release of GH from the pituitary gland (Howard helix bundle (Zhang et al., 1997), proteins with a high degree
et al., 1996; Kojima et al., 1999). GHSR belongs to the G of conformational conservation among mammals. Leptin
protein-coupled receptor family, and it occurs as isoforms 1a expression is broad; its main site of biosynthesis is WAT,
and 1b. GHSR-1a is functional, possesses seven transmem- although it is also present in brown adipose tissue, skeletal
brane domains; it is highly expressed in somototrophs, the muscle, placenta, ovaries, bone marrow, gastric and mammary
ARC nucleus, and WAT and, to a lesser extent, in thyroid, epithelium, and brain (Ahima and Flier, 2000; Münzber and
pancreas, spleen, myocardium, adrenal gland, testis, ovary, Morrison, 2015). In epididymal WAT of rats, leptin is continu-
and liver. It has a Kd of 0.1 nM; however, tissue distribution ously synthesized and secreted from secretory vesicles;
in humans of mRNA for isoform 1b is broader than for 1a, however, de novo synthesis is required for a long-term leptin
although the former is a truncated, nonfunctional isoform secretion (Cammisotto et al., 2006). In this sense, constitutive
(Gnanapavan et al., 2002; Davenport et al., 2005; Wells, 2009). leptin secretion is the main secretory pathway (Bradley et al.,
GHSR-1a has several signaling mechanisms, including the 2001). Leptin synthesis is regulated by several factors, in a posi-
activation of distinct kinases and constitutive activity (Yin tive fashion by insulin, glucocorticoids, tumor necrosis factor-
et al., 2014). In pituitary and other targets, this receptor acti- a (TNF-a), and estrogens, and negatively by catecholamines,
vates phospholipase C (PLC) through the G protein G11a, androgens, and b-adrenergic agonists (Bradley et al., 2001;
producing inositol 1,4,5-trisphosphate (IP3) and diacylglycerol Cammisotto et al., 2006; Saladin et al., 1995). Moreover, leptin
(DAG). IP3 binding to the IP3 receptor causes Ca2þ release from production is sensitive to the size of the adipose depot in obese
intracellular Ca2þ stores, while DAG activates protein kinase C subjects; subcutaneous adipocytes are more responsive to
(Yin et al., 2014). In NPY-positive ARC neurons, the IP3 insulin than to glucocorticoids, but the reverse is true for
receptor activates cAMP/protein kinase A (PKA) through G omental adipocytes (Russell et al., 1998), which also present
protein Gas, leading to ion channel modulation that increases gender differences. Male adipose tissue responds only weakly
the intracellular Ca2þ concentration (Kohno et al., 2003). In to steroid hormones, as compared to female adipose tissue
the VMH nucleus region, AG regulates energy balance through (Casanueva and Diéguez, 1999).
AMPK (AMP-dependent kinase), inhibiting fatty acid Leptin circulates as a free form or bound to the soluble lep-
biosynthesis (López et al., 2008). tin receptor; in lean subjects up to 98% of total leptin is in the
Finally, a number of genetically altered mouse models have bound form while in obese ones the majority of leptin is in the
been developed that lack or overexpress either GOAT, GHSR, or free form (Sinha et al., 1996). The half-life of leptin in humans
ghrelin isoforms, and upon diverse feeding protocols, they is approximately 75 min (Hill et al., 1998). Leptin is expressed
have yielded useful information about the ghrelin system and secreted by mice or human adipocytes, regardless of their
(Kirchner et al., 2009; Sun et al., 2003; Zigman et al., 2005). location (retroperitoneal, omental, or subcutaneous); however,
Additionally, ghrelin mimetic drugs show promise as treat- there is more leptin synthesis in subcutaneous than in visceral
ments for GH deficiency or gastrointestinal disorders (Müller WAT (Casanueva and Diéguez, 1999; Ahima and Flier, 2000).
et al., 2015). The level of plasma leptin is positively correlated with body
mass index in lean and obese humans and in rodents (Maffei
1.06.7.1.2 Leptin et al., 1995), i.e., levels of this hormone are related to body
1.06.7.1.2.1 Biology: Biosynthesis, Secretion, Physiology, and fat, and its circulating levels are higher in obesity (Considine
Signaling et al., 1996). The circulating concentration of leptin is sexually
Leptin, from Greek leptos meaning thin, has anorexigenic and dimorphic; it is higher in females than in males with a similar
weight-diminishing effects; it was discovered and cloned in body fat mass, an effect attributed to regulation by sexual
Food Intake and Its Control by Signaling Molecules 195

hormones or to different proportions of subcutaneous fat Intracellularly, it signals through the Janus kinase family (Jak2)-
(Rosenbaum and Leibel, 1999; Ahima and Flier, 2000). signal transducers and activators of transcription (STAT3/5)
Leptin expression and circulating levels show circadian fluc- signaling pathway, among others (Myers et al., 2008; Münzberg
tuations, and they also change with nutritional state; in fact, and Morrison, 2015).
leptin circulating levels decrease in mice during short-term star- Ligand binding allows transphosphorylation and activation
vation (Saladin et al., 1995; Ahima et al., 1996). Plasma leptin of Jak2 which, in turn, phosphorylates Tyr residues in a complex
has a diurnal rhythm, with the highest concentrations at signal transduction phosphorylation pattern. In fact, mutations
midnight and entrained to meal time (Schoeller et al., 1997). of specific signaling sites on the leptin receptor in mice have
Leptin is an important player in energy homeostasis; the provided information about its in vivo phenotype and functions
mechanism for its essential role in decreasing appetite and aug- and also suggested a targeted therapy for obesity and type 2 dia-
menting energy expenditure will be explained in the following betes (Myers, 2010). LRb activates a number of downstream
section. In addition, leptin has numerous physiological roles in mechanisms, such as the ERK (extracellular regulated kinase)
metabolism as an adiposity signal, affecting reproductive and signaling pathway; STAT3 activation and nuclear translocation
immune function, hematopoiesis, and brain and bone devel- for regulation of transcription, including a feedback inhibitor
opment (Ahima and Flier, 2000; Friedman and Mantzoros, of this pathway, the SOC3 (suppressor of cytokine signaling
2015). In fact, leptin forms a linkage between nutritional status 3); STAT5 or PI3K (phosphatidylinositol 3-kinase) (Bates
and neuroendocrine and immune functions (La Cava and et al., 2003; Myers et al., 2008; Münzberg and Morrison,
Matarese, 2004). The anorexigenic role of leptin has been 2015). Hypothalamic neuropeptides that participate in food-
evolutionarily conserved in vertebrates, suggesting that its intake regulation and are modulated by LRb include POMC,
effects on energy balance regulation are ancient (Denver CART, AgRP, and NPY (Korner et al., 2003; Kristensen et al.,
et al., 2011). 1998; Schwartz et al., 1996b, 1997).
Leptin participates in fertility regulation and it is a permis- Leptin resistance is a condition in which hyperleptinemia
sive factor for puberty (Casanova and Dieguez, 1999). Leptin in obese subjects is unable to activate the leptin receptor,
also functions as an essential factor for brain development, suppress feeding, and prevent or mitigate obesity (Myers
promoting formation of hypothalamic pathways that later et al., 2008). Several mechanisms have been suggested to
convey leptin signals to brain regions regulating food intake explain this failure, such as the involvement of leptin transport
and energy consumption (Bouret et al., 2004). In the immune by the brain–blood barrier (BBB), alterations in the cellular
system, leptin is an immune mediator and is considered LRb signal, neurodevelopmental hypothalamic programming
a hormone/cytokine, affecting a number of immune functions; by leptin or even a population of ARC/LRb neurons with
for instance, this hormone modulates experimental autoim- increased sensitivity to circulating leptin prone to develop
mune diseases and regulates secretion of interleukin-1 and cellular leptin resistance (Bouret and Simerly, 2006;
tumor necrosis factor (La Cava and Matarese, 2004). El-Haschimi et al., 2000; Faouzi et al., 2007; Myers et al.,
Further, the central action of leptin and insulin on hypotha- 2008). Yet, this is still an unsolved issue.
lamic ARC POMC neurons in mice promotes the development
of beige adipocytes in WAT and weight loss (Dodd et al., 2015). 1.06.7.1.3 Food-Intake Regulation by Ghrelin and Leptin
Recently reviewed (Stieg et al., 2015), leptin has been associ- Although both ghrelin and leptin have been linked to the
ated with neuropathologies due to its neuroendocrine interac- cephalic-phase response in the anticipatory regulation of
tions. It has antidepressant effects and the decrease of leptin feeding (Power and Schulkin, 2008), one of the main actions
levels in dementia facilitates neurodegeneration. of AG and leptin in the counterregulation of food intake, is
The leptin receptor (LR) is encoded by the lepr gene, with 17 via the central NS, particularly through the modulation of
exons. In mice there are six LR isoforms, products of alternative ARC subpopulations of neurons with marker neuropeptides.
splicing, named LRa to LRf, that belong to the categories: On one side, a ventromedial population that coexpresses the
secreted, short and long, all sharing the leptin-binding domain. 36-amino acid peptide, NPY (Muroya et al., 1999; Tatemoto
The secreted forms may regulate the concentration of free leptin et al., 1982), and the 112-amino acid protein, AgRP (Gropp
and the transport of short leptin across blood–brain barrier et al., 2005; Luquet et al., 2005) that coexists with a GABAergic
(BBB). It is the long form, LRb (or LepRb), that is crucial for lep- system (Meister, 2007); on the other side, a ventrolateral pop-
tin signaling in db/db mice, as they only lack this isoform ulation that colocalizes with the 241-amino acid precursor
(Myers et al., 2008; Münzberg and Morrison, 2015). POMC that is cleaved to produce and secrete the melanocortin
The db gene for the leptin receptor was cloned in 1996 (Chen a-melanocyte-stimulating hormone (a-MSH) and the CART,
et al., 1996; Leibel, 2008). Human leptin long receptor has 1165 that coexists with a glutamatergic system (Meister, 2007;
amino acids, and its expression is highest in neurons within Millington, 2007; Figure 6).
nuclei of the basomedial hypothalamus, i.e., the ARC, dorsome- Anatomically, the ARC nucleus is outside of the BBB, and it
dial hypothalamic and VMH nuclei, and LHA. It is also present expresses receptors for both leptin and AG. Moreover, AG is
in other nervous system regions such as the VTA and the nucleus locally produced in neurons of the ARC nucleus, which central
of the solitary tract (NTS) and, to a lesser extent, in the liver, leptin can access by several potential mechanisms (Cowley
kidney, lung, stomach, pancreatic beta, and immune cells (Myers et al., 2003; Kojima et al., 1999; Münzberg and Morrison,
et al., 2008; Denver et al., 2011). LRb belongs to the class I family 2015). Central administration of AG or in vitro treatment of
of cytokine receptors and it possesses three functional domains: ARC or ARC slices with AG increases the activity and stimulates
an extracellular ligand-binding, a single transmembrane, and the synthesis and release of NPY/AgRP neurons, exerting orexi-
a cytoplasmic-signaling domain without intrinsic kinase activity. genic effects by the stimulation of feeding (Cowley et al., 2003;
196 Food Intake and Its Control by Signaling Molecules

Figure 6 Schematic overview of acylated ghrelin (AG) and leptin action on the nervous system to regulate food intake. Abbreviations: PVN, para-
ventricular nucleus; ARC, arcuate nucleus; LHA, lateral hypothalamic area; NTS, nucleus of the solitary tract; GHSR-1a, growth hormone secreta-
gogue receptor-1a; LepRb, leptin long receptor; NPY, neuropeptide Y; AgRP, agouti-related protein; MCR, melanocortin receptors; POMC,
proopiomelanocortin; a-MSH, a-melanocyte-stimulating hormone; CART, cocaine and amphetamine regulated transcript.

Nakazato et al., 2001). In contrast, leptin has dual mechanisms (Di Marzo et al., 2001) and by modulating the levels of neuro-
on ARC; first it activates POMC/CART neurons which, in turn, transmitters such as noradrenaline, dopamine, and serotonin
produce and release a-MSH, activating MC3 and MC4 melano- that also contribute to decrease food intake (Schwartz et al.,
cortin receptors on second-order neurons. Importantly, orexi- 2000). Additionally, the number of LRb in the ARC nucleus
genic AgRP is an endogenous antagonist of MC3 and MC4 constitutes 15–20% of the total number of LRb within the
receptors, further blocking this anorexigenic pathway CNS, so leptin signaling participates in a number of homeo-
(Ollmann et al., 1997). Secondly, leptin inhibits the expression static mechanisms, such as glucose homeostasis and hedonic
and secretion of NPY/AgRP neurons by blocking NPY gene feeding (Myers et al., 2008; Münzberg and Morrison, 2015).
expression, and it lowers GABA release (Cowley et al., 2001; Both AG and leptin interact with GLP-1 signaling via vagal
Morton et al., 2006; Murphy and Bloom, 2006; Schwartz afferent neurons; AG blocks translocation of GLP-1R to the
et al., 1996b; Woods et al., 1998). In this way, NPY/AgRP orexi- plasma membrane, whereas leptin interacts with GLP-1 to
genic neurons activated by AG and inhibited by leptin, stimu- induce satiation (Ronveaux et al., 2015).
late feeding and inhibit satiety, while anorexigenic POMC/
CART neurons, activated by leptin, stimulate satiety, block
feeding, and increase energy expenditure. 1.06.8 Other Adipokines Regulating Food Intake
The ARC nucleus is innervated bidirectionally by reciprocal
connections to second-order neurons in hypothalamic areas Adipose tissue is an endocrine organ that plays a central role in
such as the PVN (anorexigenic signaling) with catabolic effects, the regulation of whole-body metabolic homeostasis. As previ-
and in the LHA (orexigenic signaling) with anabolic effects. It is ously discussed, the most important adipocyte product regu-
also connected to other brain regions (e.g., the NTS), together lating food intake is leptin; however, adipose tissue produces
with which the ARC participates in the energy homeostasis a myriad of proteins that regulate many aspects of metabolism.
regulatory circuit (Meister, 2007; Münzberg and Morrison, To date, additional adipokines with well-documented roles in
2015; Murphy and Bloom, 2006; Schwartz et al., 2000). food intake are adiponectin, resistin, nesfatin, and apelin.
Although initial reports of murine models that lack either
ghrelin or its receptor showed that, under normal feeding,
1.06.8.1 Adiponectin
this hormone had a redundant or masked role in the regulation
of appetite, under diet-induced obesity AG participates in the Adiponectin was discovered 20 years ago by Scherer and
selection of the metabolic substrate for energy, favoring fat colleagues (Scherer et al., 1995), and 1 year later by three other
ingestion. Consequently, the lack of AG protects against obesity groups (Hu et al., 1996; Maeda et al., 1996; Nakano et al., 1996)
(Wortley et al., 2004, 2005; Sun et al., 2003; Zigman et al., as a highly adipocyte-specific secreted protein. It has a structure
2005). Moreover, leptin regulates food intake by reducing the similar to the complement C1q and the tumor necrosis factor
hypothalamic content of ECs with an orexigenic effect superfamilies of proteins (Shapiro and Scherer, 1998).
Food Intake and Its Control by Signaling Molecules 197

Adiponectin is a highly abundant circulating protein present in 1.06.8.2 Resistin


blood in micrograms per milliliter, and it is estimated that it
Resistin (RSTN) was first described in 2001 as an adipocyte-
accounts for 0.01% percent of total plasma proteins (Turer
derived protein downregulated by the insulin-sensitizing thia-
and Scherer, 2012). It is a pleiotropic adipokine exerting
zolidinediones (TZDs) drugs (Steppan et al., 2001). Its levels
a wide variety of actions in different tissues; it acts as an insulin
increase in obesity in humans (Savage et al., 2001; Degawa-
sensitizer, promotes healthy adipose tissue expansion, and is
Yamauchi et al., 2003) and in rodents, in which its neutraliza-
anti-inflammatory. Accordingly, adiponectin-deficient mice are
tion improves insulin sensitivity (Steppan et al., 2001). In
glucose intolerant and insulin resistant (Kubota et al., 2002;
agreement, lean mice treated with resistin become insulin resis-
Nawrocki et al., 2006). Adiponectin has received great attention
tant and glucose intolerant (Steppan et al., 2001). However, in
as a therapeutic factor due to its roles as an antidiabetic, antia-
humans, RSTN is not expressed in adipocytes but in blood
therogenic, and possibly anticarcinogenic factor (Ye and
mononuclear cells (PBMC), mainly macrophages (Fain et al.,
Scherer, 2013; Yamauchi and Kadowaki, 2013). Adiponectin
2003; Tomaru et al., 2009). Human RSTN is upregulated by
levels correlate inversely with obesity and insulin resistance
proinflamatory stimuli (TNFa, IL1b, IL6, LPS) (Kaser et al.,
(Arita et al., 1999; Turer et al., 2011; Gao et al., 2013) and
2003; Lehrke et al., 2004) and, as in rodents, it is downregu-
also with several types of cancer (Dalamaga et al., 2012). Adipo-
lated by TZDs (Patel et al., 2003). RSTN promotes insulin resis-
nectin actions initiate when the protein binds to its receptor
tance, inflammation, atherosclerosis, and heart failure, acting
species AdipoR1 and AdipoR2, which are seven transmembrane
on several cell types such as endothelium, smooth muscle,
domain proteins linked to different signaling molecules such as
and cardiomyocytes (Park and Ahima, 2013). The receptor
AMPK, Ca2þ, PPARa, PPARg, SIRT1, and ceramide/sphingoli-
mediating resistin actions is still unknown.
pids. Independently of the classical 1 and 2 receptors, adiponec-
Interestingly, RSTN is expressed in the mouse brain, partic-
tin binds to T-cadherin, and this interaction mediates its
ularly in the hypothalamus and in the pituitary gland (Morash
cardioprotective effects (Denzel et al., 2010) and also activates
et al., 2002). In rats, i.c.v. injection of resistin transiently
the NFkB pathway in many cell types by activating a still
reduces food intake in fasted and fed animals (Tovar et al.,
unidentified receptor entity (Yamauchi et al., 2014). AdipoR1
2005; Vázquez et al., 2008; Cifani et al., 2009). Although it
is expressed ubiquitously, whereas AdipoR2 is mostly
has no long-term effect (Park et al., 2008), resistin abolishes
expressed in the liver (Yamauchi and Kadowaki, 2013).
the anorectic effect of leptin when they are administered
Regarding adiponectin effects on the central nervous
together, in both control and diabetic animals (Park et al.,
system, its receptors are present in the brain (Yamauchi
2008). In fasted rats, central RSTN administration prevents
et al., 2003; Kubota et al., 2007), and the peptide is present
the normal increase in mRNA expression of the orexigenic
in the cerebrospinal fluid of rodents and humans (Kusminski
peptides NPY and AgRP and the decrease in the anorexigenic
et al., 2007; Kos et al., 2007). In lean animals, systemic treat-
CART, without effects on POMC in the ARC (Vázquez et al.,
ment with adiponectin increases energy expenditure, reduces
2008). In fed rats, RSTN reduced FAS mRNA expression in
body weight and fat mass, but does not alter food intake
the VMH (Vázquez et al., 2008), a mechanism that contributes
(Pajvani et al., 2003; Yamauchi et al., 2001; Berg et al.,
to decreased feeding (Chakravarthy et al., 2007). Consistent
2001). In obese animals it also improves insulin sensitivity
with this, RSTN reduces the orexigenic effect of NPY when
and glucose and lipid metabolism. These results are recapitu-
both are administered centrally (Cifani et al., 2009). Some
lated upon i.c.v. adiponectin injection (Qi et al., 2004). Intra-
contradictory results exist, such as the fact that in mice, central
venous injection of adiponectin increases cerebrospinal fluid
resistin increased the levels of NPY in the ARC nucleus, suggest-
levels of the hormone, suggesting serum to brain transport
ing possible species differences in the RSTN effects.
(Qi et al., 2004). Conversely, i.c.v. adiponectin injection
Additionally, central RSTN treatment promotes liver insulin
does not alter its circulating levels, suggesting adiponectin
resistance (Muse et al., 2007; Singhal et al., 2007), stimulates
receptor-mediated actions on the brain (Qi et al., 2004).
fatty acid synthesis, and pro-inflammatory cytokine expression
In contrast, others have shown that adiponectin stimulates
in liver and in adipose tissue (Vázquez et al., 2008).
food intake and decreases energy expenditure through activa-
tion of AdipoR1 and AMPK in the ARC of the hypothalamus
(ARH), without changes in body weight (Kubota et al.,
1.06.8.3 Nesfatin
2007). In agreement, adiponectin-deficient mice fed a high-
fat diet exhibited reduced food intake (Kubota et al., 2007). Nesfatin or nonesterified fatty acid/nucleobindin2 (NEFA/
Levels of adiponectin in serum and cerebrospinal fluid (CSF) NUCB2) was identified in 2006 as a satiety factor produced
and of its receptor in the ARH increase during fasting condi- throughout the hypothalamus (Oh et al., 2006) and later found
tions and decrease upon refeeding (Kubota et al., 2007). Adi- to be expressed in many different areas of the brain such as the
ponectin is proposed as a satiety signal, promoting food intake insular cortex, amygdala, periventricular nucleus, raphei nuclei,
and fat storage during fasting. These contradictory results may ventrolateral medulla, LC, cerebellum, DMV, NTS, etc. (Stengel
reflect different adiponectin preparations or molecular forms, et al., 2013b). Despite its recent discovery, there is a vast litera-
as it exists in trimers (main form in human CSF) (Kusminski ture describing the effects of NUCB2 in reducing food intake
et al., 2007), hexamers, and high molecular weight adiponec- (recently reviewed by Stengel et al., 2013b). NUCB2 is cleaved
tin containing 18–36 subunits. Thus, further studies are by prohormone convertases into an N-terminal fragment
needed to clarify the effects of the various adiponectin multi- named Nesfatin-1 that exerts potent anorexigenic activity (Oh
mers on food intake in different physiological and patholog- et al., 2006); however, it is controversial whether Nesfatin-1 is
ical conditions. produced in vivo (Stengel and Tache, 2013). Nevertheless, the
198 Food Intake and Its Control by Signaling Molecules

full-length NUCB2 molecule (50 KDa, 396 aa) exerts the same endogenous ligand for the orphan G protein-coupled receptor
anorexigenic activity as the cleaved product (9.7 kDa, aa APJ (Tatemoto et al., 1998). Apelin is expressed by many cell types
1–82), so it may well be the endogenous active molecule including the hypothalamus (Brailoiu et al., 2002; Reaux et al.,
(Stengel and Tache, 2013). Immunoneutralization or 2002), and it was described as an adipokine in 2005 because it
knockdown of NUCB2/Nesfatin-1 increases food intake (Oh is expressed by mature adipocytes in both rodents and humans
et al., 2006; Sedbazar et al., 2013) and conversely, i.c.v. or i.p. (Boucher et al., 2005). Apelin expression in adipocytes is
administration of this molecule reduces feeding (Oh et al., inhibited by fasting and increased by refeeding conditions, and
2006; Shimizu et al., 2009). The anorexigenic effects of central its circulating levels are increased in obese rodents and humans
nesfatin treatment lead to decreased body weight and adiposity (Boucher et al., 2005). Apelin is synthesized as a propeptide
in rats. Nesfatin-1 expression in the PVN and SON is regulated and processed to different peptides, from which apelin-13 is the
by the nutritional status: it is reduced upon fasting and main isoform found in the circulation and has the greatest activity
increases after re-feeding in rats (Oh et al., 2006; Kohno et al., upon receptor binding (Hosoya et al., 2000). Apelin receptors are
2008). Also, supporting its physiological role in food-intake present in the hypothalamus and the molecule regulates fluid
regulation, NUCB2 expression has a circadian expression homeostasis (De Mota et al., 2004; Mitra et al., 2006), pituitary
pattern, decreasing during the light phase, when rats reduce hormone release (Taheri et al., 2002), cardiovascular function
their feeding behavior, and increasing in the dark phase (Lee et al., 2005; Tatemoto et al., 2001), neuroprotective effects
coincidentally with increased feeding (Sedbazar et al., 2013). (O’donnell et al., 2007; Zeng et al., 2010), gastrointestinal
The anorexigenic effects of NUB2/Nesfatin-1 are indepen- motility (Lv et al., 2011; Yang et al., 2010), etc.
dent of leptin, as Zucker rats are still responsive to this protein Regarding effects on food intake, i.c.v. injection of apelin-13
and in the same fashion; nesfatin-1 neutralization does not inhibits food intake in fed and fasted rats and mice during the
alter the anorexigenic effects of leptin (Oh et al., 2006; Maejima light and dark phase, respectively (Sunter et al., 2003; Lv et al.,
et al., 2009). Nesfatin actions involve oxytocin and fix a-MSH 2012). However, apelin-13 has no effect on food intake in
signaling, as intraventricular administration of oxytocin and of mice during the light period (Lv et al., 2012). Apelin-13 effects
a-MSH receptor antagonists blunts the anorexigenic effect of reducing food intake are dependent on the APJ receptor, as
Nesfatin-1 (Maejima et al., 2009; Yosten and Samson, 2010, demonstrated by the use of an APJ antagonist that blocks
2009; Oh et al., 2006). Nesfatin-1 also induces oxytocin secre- apelin-13 effects (Lv et al., 2012). Apelin-13 effects are mediated
tion from PVN neurons in vitro (Maejima et al., 2009). NUCB2 by corticotropin-releasing factor (CRF); in vitro, apelin-13 stimu-
is increased by central administration of a-MSH, but it fails to lated CRF and AVP release from hypothalamic explants (Taheri
alter the expression of either orexigenic or anorexigenic et al., 2002), and a CRF receptor antagonist blocks the apelin-
peptides in the ARC or PVN (Oh et al., 2006). However, 13 inhibitory effect on food intake (Lv et al., 2012). Apelin
nesfatin-1 treatment increases expression of the mRNAs for also increases a-MSH release from POMC neurons, and both
POMC and CART in the nucleus tractus solitarius (NTS) apelin and its receptors are expressed by these neurons, suggest-
(Shimizu et al., 2009). Other mediators of Nesfatin-1 effects ing autocrine effects (Reaux-Le Goazigo et al., 2011). a-MSH
reducing food intake are CRF, histamine, serotonin, TRH stimulates CRF release from PVN neurons resulting in inhibition
(Stengel et al., 2013b), and dopamine (Chen et al., 2015). of food intake. Thus, apelin inhibits food intake by stimulating
The molecular mechanisms that mediate NUNCB2/Nesfatin- both a-MSH and CRF release (Reaux-Le Goazigo et al., 2011).
1 effects await the discovery of its receptor. Interestingly, apelin receptors are reduced in the hypothalamus
Just 5 years ago, NUNCB2 was found to be produced by of obese rats in parallel with reduced apelin effects on food
adipose tissue from mice and humans, adding to the list of adi- intake (Clarke et al., 2009). However, elevated apelin levels in
pokines with effects on feeding behavior. NUCB2/Nesfatin-1 obese rats and humans are suggested to occur as an attempt to
protein levels are reduced by fasting and increase in mice given maintain metabolic homeostasis (Carpéné et al., 2007).
a high fat diet (Ramanjaneya et al., 2010). In adipose tissue and
adipocytes, Nesfatin-1 levels are induced by proinflammatory
cytokine treatment. Interestingly, Nesfatin-1 is expressed by 1.06.9 Pancreas and Feeding Control
subcutaneous adipose tissue more than by other fat depots,
and its levels are positively correlated with body mass index In vertebrates the pancreas is an exocrine/endocrine gland that
in humans (Ramanjaneya et al., 2010). plays a crucial role in the metabolic status of the organism as
Besides its expression throughout the brain, NUCB2/ well as in the digestive processing of complex nutrients. Of
nesfatin-1 is also expressed in the pituitary gland, pancreas, endodermal origin, the pancreas communicates to the
testis (González et al., 2009; Stengel et al., 2013b), and the duodenum through the ampulla of Vater to release a variety
gastric mucosa of rodents and humans, in the same cells that of zymogens and enzymes that allow the digestion of proteins,
produce ghrelin (Stengel et al., 2009, 2013a). Given the wide- polysaccharides, and fats. Within the pancreatic tissue several
spread tissue localization of NUNCB2/Nesfatin-1, further specialized endocrine cells form aggregates known as the islets
studies are needed to determine the role of adipose tissue- of Langerhans. The number of islets varies from hundreds to
derived NUNCB2 in the regulation of food intake. thousands depending on the species; the diameter of each islet
is about 200 mm and overall, the islets constitute nearly 1.5% of
the pancreatic mass (Bockman, 2008).
1.06.8.4 Apelin
The exocrine pancreas is formed by ducts that are arranged
Apelin was first described and isolated from bovine stomach in in clusters known as acini. There are two principal types of cells:
1998 by the group of K. Tatemoto and colleagues, as an centroacinar cells that secrete bicarbonate (HCO 3 ) ions that
Food Intake and Its Control by Signaling Molecules 199

serve to neutralize acid coming from the stomach to the signaling process with physiological and pathological conse-
duodenum, and basophilic cells that synthesize a set of diges- quences. Within the target cells two transcription factors are
tive enzymes (trypsinogen, chymotrypsinogen, and pancreatic activated by the presence of cortisol/corticosterone, gluco-
lipase and amylase). Both types of cells are regulated by endo- corticoid receptors (GR), and mineralocorticoid receptors
crine cues. HCO 3 secretion is mainly stimulated by the peptide (MR). Paradoxically, the MR has a higher affinity for gluco-
hormone secretin which is produced in the S cells of the corticoids than the GR. This observation has been explained
duodenal glands, whereas the basophilic cells are activated by by postulating that glucocorticoids attach to GR only in
CCK, another peptide hormone synthesized in the mucosal stress conditions, when they are present at high circulating
epithelium of the small intestine (Bockman, 2008). levels after activation of the HPA. Nongenomic effects eli-
As to the endocrine pancreas, most of the cells within the cited by membrane receptors are faster than transcriptional
islets of Langerhans are b, which synthesize and secrete insulin actions, and they modulate the activity of signaling kinases
and amylin, also known as islet amyloid PP (in ratio of approx- as well as the permeability of ion channels (Prager and
imately 100:1). Cells producing and secreting glucagon are Johnson, 2009).
named a, and they are less abundant. Other cellular types Overproduction of glucocorticoids due to adrenal tumors or
present, but in a much smaller proportion, are d, ε, and PP cells. to prolonged administration of high therapeutic doses can
They produce and secrete somatostatin, ghrelin, and PP, respec- cause Cushing’s syndrome. This disease shows metabolic
tively. Endocrine pancreatic signals are key elements in the disturbances similar to those observed in patients with meta-
digestive process, prior to (glucagon) and after mealtime bolic syndrome. Interestingly, these pathologies may have in
(insulin and amylin) (Adler and Beglinger, 1990). common the enzyme 11b-HSD1, which in diverse organs and
The pancreas is under the modulation of the autonomic tissues converts inactive keto-steroids to active glucocorticoids.
nervous system (ANS): the sympathetic branch is active during For example, experimental overexpression of 11b-HSD1 in
stressful situations and physical exercise. ANS neurons promote adipose tissue produces abdominal obesity, hyperglycemia,
the secretion of glucagon from the a cells of the islets of Lang- insulin resistance, dyslipidaemia, and hypertension; in
erhans; the parasympathetic neurons become active during the contrast, overexpression of 11b-HSD1 in the liver yields an
digestive process, stimulating the secretion of insulin from the attenuated metabolic syndrome with mild insulin resistance,
b cells of the islets of Langerhans, as well as the digestive zymo- dyslipidaemia, hypertension, and fatty liver, but not obesity
gens from the exocrine ducts (Cerf, 2011). or glucose intolerance. These data corroborate the notion that
the enzyme may be a good therapeutic target in the treatment
of metabolic syndrome (Morton and Seckel, 2008).
1.06.10 Glucocorticoids and the Control of Food The enzyme 11b-HSD is also interesting from a biochemical
Intake perspective, since it catalyzes a redox reaction that depends on
NADPH. This coenzyme is produced by the activity of hexose-
Glucocorticoids are hormones produced and secreted from the 6-phosphate dehydrogenase (H6PDH), a microsomal protein
zona fasciculata in the adrenal cortex. They were named for their that fulfills a role similar to that of glucose-6-phosphate dehy-
ability to increase serum glucose. In primates, the main glucocor- drogenase, the marker enzyme of the pentose phosphate
ticoid is cortisol, whereas in rodents it is corticosterone. These pathway. Figure 7 illustrates the coordinated activity of these
hormones modulate the metabolism of carbohydrates, proteins,
and fats, and they are also important for their role in the fasting
response as well as their action as immunosuppressors. Access of
the active ligand to the mineralocorticoid and glucocorticoid
receptors is controlled by the activity of an enzyme called 11-
b hydroxysteroid dehydrogenase (11-b HSD, types 1 and 2).
The enzymatic activity is a NADP(H) redox reaction that
converts the active ligand to its corresponding inactive 11-keto
metabolite (cortisol to cortisone in humans, and corticosterone
to 11-dehydrocorticosterone in rats) (Ricketts et al., 1998).
The glucocorticoids are produced by activation of the
hypothalamic–pituitary–adrenal axis (HPA) by some stressor
(internal or environmental); thus, the release of cortisol activates
the mobilization of energy reserves. In fasting or starvation,
glucocorticoids induce skeletal muscle activation and the release
into the circulation of amino acids that are substrates for hepatic
gluconeogenesis (Braun and Marks, 2015).
Alterations in glucocorticoid homeostasis are associated
Figure 7 Coordinated action of 11b-hydroxy-steroid dehydrogenase
with severe diseases such as osteoporosis, metabolic syndrome,
(11b-HSD) and hexose-6-phosphate dehydrogenase (H6PDH). G6P,
cardiovascular complications, immune deficiencies, and even glucose-6-phosphate; 6 PG, 6-phosphogluconate; NADPþ/NADPH, nico-
psychiatric disorders. Glucocorticoids are the most prescribed tinamide adenine dinucleotide phosphate, oxidized and reduced form.
drugs around the world due to their anti-inflammatory effects. Modified from Odermatt, A., Klusonova, P., 2015. 11b-Hydroxysteroid
Glucocorticoids are recognized by both cytoplasmic and dehydrogenase 1: regeneration of active glucocorticoids is only part of
membrane receptors, and they are part of a complex the story. J. Steroid Biochem. 151 (3), 85–92.
200 Food Intake and Its Control by Signaling Molecules

Figure 8 Glucocorticoid response to endogenous and exogenous stress regulates the HPA axis and influences the satiety-related hormones (leptin,
ghrelin, and insulin) to alter feeding behavior long term. Stress reduces glucocorticoids receptor (GR) expression in hypothalamus and hippocampus
and increases PVN arginine vasopressin (AVP) and corticotropin-releasing hormone (CRH) by epigenetic mechanisms. Circulating glucocorticoids
(GC) and 11-bHSD activity within the hippocampus promote synaptic pruning. The circuit is closed when PVN is affected by hippocampal regulation.
Stress can also induce increased release of satiety-related hormones influencing appetite, feeding behavior, and metabolism throughout life. Modified
from Sominsky, L., Spencer, S.J., 2014. Eating behavior and stress: a pathway to obesity. Front. Psychol. 5 (434), 1–8.

two enzymes, their dependence on the NADPþ/NADPH ratio, a sophisticated system that provides appropriate central periph-
as well as some alternative substrates besides glucocorticoids, eral neuroendocrine responses (Figure 8).
which can act as inhibitors of the enzyme (Odermatt and The system responsive to stress functions through the activa-
Klusonova, 2015). tion of the HPA axis and the locus caeruleus (LC)/norepineph-
rine (NE)-autonomic nervous system. The end result of
activating this axis is the secretion of circulating glucocorti-
1.06.10.1 Glucocorticoids and the Stress System
coids. These hormones regulate the basal activity of the HPA
It has been reported that glucocorticoids have orexigenic effects axis and terminate the stress response by acting mainly at the
and can cause obesity in dogs. Besides, in humans, the admin- hypothalamus and the pituitary gland, forming negative feed-
istration of glucocorticoids increases energy intake by 20–59% back loops on the secretion of CRH and adrenocorticotropic
and also increases body weight in patients to 59%. The reports hormone (ACTH), respectively. CRH is released in response
suggest that these effects relate to the ability of glucocorticoids to acute fasting from the hypothalamic PVN. Consequently,
to act on the CNS. ACTH release from the pituitary follows and promotes the
As described in earlier sections leptin regulates body weight cascade of events that allows the secretion of glucocorticoids
by controlling energy balance and food intake; glucocorticoid (Sominsky and Spencer, 2014).
administration in humans increases circulating leptin by acti-
vating insulin release (Nishii et al., 2006). In addition, some
reports indicate that orexins regulate the HPA axis in response 1.06.11 Concluding Remarks
to stressful signals, for example, orexin A directly promotes the
secretion of ACTH and, eventually, corticosterone. This obser- After decades of intensive research, our knowledge of the phys-
vation has been confirmed in in vitro experiments, using human iological events that underlie the regulation of food intake,
and rat adrenal gland slices (Srinivasan et al., 2013). nutrient assimilation, and metabolic performance has
All living organisms must respond to many extrinsic and advanced considerably. Nowadays it is common to read in
intrinsic stressful stimuli that constantly challenge the equilib- scientific journals reports about the control of food intake
rium of the internal homeostasis. The term stress is used to that involve the removal or insertion of diverse genes in diverse
define a state of being threatened or perceived as being threat- tissues and organs. Indeed, conclusions are reached from
ening. To give a response, organisms have developed studying, e.g., the type of receptors, the cellular signaling
Food Intake and Its Control by Signaling Molecules 201

systems involved, and the resultant transcriptional responses. Ángeles-Castellanos, M., Aguilar-Roblero, R., Escobar, C., 2004. c-Fos expression in
Yet, impressive as it is, we must not forget the original contribu- hypothalamic nuclei of food-entrained rats. Am. J. Physiol. Regul. Integr. Comp.
Physiol. 286 (1), R158–R165.
tions and the first concepts elicited by a set of renowned phys-
Aponte, Y., Atasoy, D., Sternson, S.M., 2011. AGRP neurons are sufficient to
iologists that set the basis for the contemporary advances in the orchestrate feeding behavior rapidly and without training. Nat. Neurosci. 14 (3),
physiology of food intake. Overall, the understanding of the 351–355.
basic mechanisms underlying the physiology of food intake Arellanes-Licea, E.C., Báez-Ruiz, A., Carranza, M.E., Arámburo, C., Luna, M., Díaz-
will help us to address how to treat the pathologies of eating Muñoz, M., 2014. Daily patterns and adaptation of the ghrelin, growth hormone
and insulin-like growth factor-1 system under daytime food synchronisation in rats.
disorders. Effective therapies for obesity and anorexia nervosa, J. Neuroendocrinol. 26 (5), 282–295.
just to mention two of the more prevalent diseases related to Arita, Y., Kihara, S., Ouchi, N., et al., 1999. Paradoxical decrease of an adipose-
feeding, will benefit from the integration of the findings specific protein, adiponectin, in obesity. Biochem. Biophys. Res. Commun. 257
emerging from many biomedical research laboratories with (1), 79–83.
Badman, M.K., Flier, J.S., 2005. The gut and energy balance: visceral allies in the
the experience of clinicians and nutriologists.
obesity wars. Science 307 (5717), 1909–1914.
Báez-Ruiz, A., Escobar, C., Aguilar-Roblero, R., Vazquez-Martinez, O., Díaz-Muñoz, M.,
2005. Metabolic adaptations of liver mitochondria during restricted feeding
Acknowledgments schedules. Am. J. Physiol. Gastrointest. Liver Physiol. 289 (6), G1015–G1023.
Bass, J., Takahashi, J.S., 2010. Circadian integration of metabolism and energetics.
Science 330 (9), 1349–1354.
M.D.-M. and F.V.-C. are researchers from the Cellular and Molecular
Bates, S.H., Stearns, W.H., Dundon, T.A., et al., 2003. STAT3 signalling is required for
Department of the Neurobiology Institute, UNAM. Their research is leptin regulation of energy balance but not reproduction. Nature 421 (6925),
funded by the following grants: M.D.-M., DGAPA, PAPIIT, project 856–859.
IN200815 and F.V.-C., DGAPA, PAPIIT, IN205114, and CONACyT, Beck, B., Richy, S., Dimitrov, T., Stricker-Krongrad, A., 2001. Opposite regulation of
166725. We thank Dr Dorothy Pless for her professional and diligent hypothalamic orexin and neuropeptide Y receptors and peptide expressions in
review of the English grammar. We also thank Nutriologist Fernando obese Zucker rats. Biochem. Biophys. Res. Commun. 286 (3), 518–523.
López-Barrera for his support in the elaboration of the figures. Berg, A.H., Combs, T.P., Du, X., Beownlee, M., Scherer, P.E., 2001. The adipocyte-
secreted protein Acrp30 enhances hepatic insulin action. Nat. Med. 7 (8),
947–953.
Bernardis, L.L., Bellinger, L.L., 1993. The lateral hypothalamic area revisited: neuro-
References anatomy, body weight regulation, neuroendocrinology and metabolism. Neurosci.
Biobehav. Rev. 17 (2), 141–193.
Abrahamson, E.E., Moore, R.Y., 2006. Lesions of suprachiasmatic nucleus effer- Bernardis, L.L., Bellinger, L.L., 1998. The dorsomedial hypothalamic nucleus revisited:
ents selectively affect rest-activity rhythm. Mol. Cell. Endocrinol. 252 (1–2), 1998 update. Exp. Biol. Med. 218 (4), 284–306.
46–56. Berthoud, H.R., 2004. Neural control of appetite: cross-talk between homeostatic and
Aceves, C., Escobar, C., Rojas-Huidobro, R., et al., 2003. Liver 50 -deiodinase activity is non-homeostatic systems. Appetite 43 (3), 315–317.
modified in rats under restricted feeding schedules: evidence for post-translational Berthoud, H.R., 2007. Interactions between the “cognitive” and “metabolic” brain in
regulation. J. Endocrinol. 179 (1), 91–96. the control of food intake. Physiol. Behav. 91 (4), 486–498.
Acuña-Goycolea, C., Tamamaki, N., Yanagawa, Y., et al., 2005. Mechanisms of Berthoud, H.R., 2011. Metabolic and hedonic drives in the neural control of appetite:
neuropeptide Y, peptide YY, and pancreatic polypeptide inhibition of identified green who is the boss? Curr. Opin. Neurobiol. 21 (11), 888–896.
fluorescent protein-expressing GABA neurons in the hypothalamic neuroendocrine Berthoud, H.R., Münzberg, H., Richards, B.K., Morrison, C.D., 2012. Neural and
arcuate nucleus. J. Neurosci. 25 (32), 7406–7419. metabolic regulation of macronutrient intake and selection. Proc. Nutr. Soc. 71 (3),
Adan, R.A., Vanderschuren, L.J., la Fleur, S.E., 2008. Anti-obesity drugs and neural 390–400.
circuits of feeding. Trends Pharmacol. Sci. 29 (4), 208–217. Bi, S., Ladenheim, E.E., Schwartz, G.J., Moran, T.H., 2001. A role for NPY over-
Adler, G., Beglinger, C., 1990. Hormones as regulators of pancreatic secretion in man. expression in the dorsomedial hypothalamus in hyperphagia and obesity of OLETF
Eur. J. Clin. Invest. 20 (Suppl. 1), S27–S32. rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 281 (1), R254–R260.
Aguilar-Roblero, R., 2015. Chronostasis: the timing of physiological systems. In: Blouet, C., Schwartz, G.J., 2010. Hypothalamic nutrient sensing in the control of
Aguilar-Roblero, R., Díaz-Muñoz, M., Fanjul-Moles, M.L. (Eds.), Mechanisms of energy homeostasis. Behav. Brain Res. 209 (1), 1–12.
Circadian Systems in Animals and Their Clinical Relevance. Springer, New York, Blum, I.D., Patterson, Z., Khazall, R., et al., 2009. Reduced anticipatory locomotor
pp. 221–236. responses to scheduled meals in ghrelin receptor deficient mice. Neuroscience
Aguilar-Roblero, R., Díaz-Muñoz, M., 2010. Chronostatic adaptations in the liver to 164 (2), 351–359.
restricted feeding: the FEO as an emergent oscillator. Sleep Biol. Rhythms 8 (1), Bockman, D.L., 2008. Anatomy and fine structure. In: Beger, H.G., Buchler, M.,
9–17. Kozarek, R., et al. (Eds.), The Pancreas: An Integrated Textbook of Basic Science,
Aguilar-Roblero, R., Díaz-Muñoz, M., Báez-Ruíz, A., Quinto-Muñoz, D., Lundkvist, G., Medicine, and Surgery, second ed. Wiley – Blackwell (Chapter 4).
Stephan, M., 2015. Intracellular calcium as a clock output from SCN neurons. In: Bodosi, B., Gardi, J., Hajdu, I., Szentirmai, E., Obal Jr., F., Krueger, J.M., 2004.
Aguilar-Roblero, R., Díaz-Muñoz, M., Fanjul-Moles, M.L. (Eds.), Mechanisms of Rhythms of ghrelin, leptin, and sleep in rats: effects of the normal diurnal cycle,
Circadian Systems in Animals and Their Clinical Relevance. Marcel Dekker, Inc, restricted feeding, and sleep deprivation. Am. J. Physiol. Regul. Integr. Comp.
New York, NY, pp. 115–132. Physiol. 287 (5), R1071–R1079.
Ahima, R.S., Prabakaran, D., Mantzoros, C., et al., 1996. Role of leptin in the Boucher, J., Masri, B., Daviaud, D., et al., 2005. Apelin, a newly identified adipokine
neuroendocrine response to fasting. Nature 382 (6588), 250–252. up-regulated by insulin and obesity. Endocrinology 146 (4), 1764–1771.
Ahima, R.S., Flier, J.S., 2000. Leptin. Annu. Rev. Physiol. 62, 413–437. Bouret, S.G., Draper, S.J., Simerly, R.B., 2004. Trophic action of leptin on hypotha-
Albrecht, U., 2012. Timing to perfection: the biology of central and peripheral circadian lamic neurons that regulate feeding. Science 304 (5667), 108–110.
clocks. Neuron 74 (3), 246–260. Bouret, S.G., Simerly, R.B., 2006. Developmental programming of hypothalamic
Alizadeh, A., Zendehdel, M., Babapour, V., Charkhkar, S., Hassanpour, S., 2015. Role feeding circuits. Clin. Genet. 70 (4), 295–301.
of cannabinoidergic system on food intake in neonatal layer-type chicken. Vet. Res. Bradley, R.L., Cleveland, K.A., Cheatham, B., 2001. The adipocyte as a secretory
Commun. 39 (2), 151–157. organ: mechanisms of vesicle transport and secretory pathways. Recent Prog.
Alonso-Alonso, M., Woods, S.C., Pelchat, M., et al., 2015. Food reward system: Horm. Res. 56, 329–358.
current perspectives and future research needs. Nutr. Rev. 73, 296–307. Brady, L.S., Smith, M.A., Gold, P.W., Herkenham, M., 1990. Altered expression of
Anand, B.K., Brobeck, J.R., 1951. Hypothalamic control of food intake in rats and cats. hypothalamic neuropeptide mRNAs in food-restricted and food-deprived rats.
Yale J. Biol. Med. 24 (2), 123–140. Neuroendocrinology 52 (5), 441–447.
Andrew, S.F., Dinh, T.T., Ritter, S., 2007. Localized glucoprivation of hindbrain sites Brailoiu, G.C., Dun, S.L., Yang, J., Ohsawa, M., Chang, J.K., Dun, N.J., 2002. Apelin-
elicits corticosterone and glucagon secretion. Am. J. Physiol. Regul. Integr. Comp. immunoreactivity in the rat hypothalamus and pituitary. Neurosci. Lett. 327 (3),
Physiol. 292 (5), R1792–R1798. 193–197.
202 Food Intake and Its Control by Signaling Molecules

Braun, T.P., Marks, D.L., 2015. The regulation of muscle mass by endogenous Cheung, C.C., Clifton, D.K., Steiner, R.A., 1997. Proopiomelanocortin neurons are
glucocorticoids. Front. Physiol. 6 (12), 1–12. direct targets for leptin in the hypothalamus. Endocrinology 138 (10), 4489–4492.
Brobeck, J.R., 1948. Food intake as a mechanism of temperature regulation. Yale J. Chou, T.C., Scammell, T.E., Gooley, J.J., Gaus, S.E., Saper, C.B., Lu, J., 2003. Critical
Biol. Med. 20, 545–552. role of dorsomedial hypothalamic nucleus in a wide range of behavioral circadian
Broberger, C., Johansen, J., Johansson, C., Schalling, M., Hökfelt, T., 1998. The rhythms. J. Neurosci. 23 (33), 10691–10702.
neuropeptide Y/agouti gene-related protein (AGRP) brain circuitry in normal, Christian, C.A., Huguenard, J.R., 2013. Astrocytes potentiate GABAergic transmission
anorectic, and monosodium glutamate- treated mice. Proc. Natl. Acad. Sci. U.S.A. in the thalamic reticular nucleus via endozepine signaling. Proc. Natl. Acad. Sci.
95 (25), 15043–15048. U.S.A. 110 (50), 20278–20283.
Broberger, C., 2005. Brain regulation of food intake and appetite: molecules and Chronwall, B.M., DiMaggio, D.A., Massari, V.J., Pickel, V.M., Ruggiero, D.A.,
networks. J. Intern. Med. 258 (4), 301–327. O’Donohue, T.L., 1985. The anatomy of neuropeptide-Y-containing neurons in rat
Brüning, J.C., Gautam, D., Burks, D.J., et al., 2000. Role of brain insulin receptor in brain. Neuroscience 15 (4), 1159–1181.
control of body weight and reproduction. Science 289 (5487), 2122–2125. Cifani, C., Durocher, Y., Pathak, A., et al., 2009. Possible common central pathway for
Buijs, R.M., Wortel, J., van Heerikhuize, J.J., et al., 1999. Anatomical and functional resistin and insulin in regulating food intake. Acta Physiol. (Oxf.) 196 (4), 395–400.
demonstration of a multisynaptic suprachiasmatic nucleus adrenal (cortex) Civelli, O., Birnberg, N., Herbert, E., 1982. Detection and quantitation of pro-
pathway. Eur. J. Neurosci. 11 (5), 1535–1544. opiomelanocortin mRNA in pituitary and brain tissues from different species.
Buijs, R.M., Scheer, F.A., Kreier, F., Yi, C., Bos, N., Goncharuk, V.D., Kalsbeek, A., J. Biol. Chem. 257 (12), 6783–6787.
2006. Organization of circadian functions: interaction with the body. Prog. Brain Clark, J.T., Kalra, P.S., Kalra, S.P., 1985. Neuropeptide Y stimulates feeding but
Res. 153, 343–360. inhibits sexual behavior in rats. Endocrinology 117 (6), 2435–2442.
Buckman, L.B., Ellacott, K.L., 2014. The contribution of hypothalamic macroglia to the Clarke, K.J., Whitaker, K.W., Reyes, T.M., 2009. Diminished metabolic responses to
regulation of energy homeostasis. Front. Syst. Neurosci. 8, 212. http://dx.doi.org/ centrally-administered apelin-13 in diet-induced obese rats fed a high-fat diet.
10.3389/fnsys.2014.00212, eCollection 2014. J. Neuroendocrinol. 21 (2), 83–89.
Buxton, O.M., Cain, S.W., O’Connor, S.P., et al., 2012. Adverse metabolic conse- Coleman, D.L., 1978. Obese and diabetes: two mutant genes causing diabetes-obesity
quences in humans of prolonged sleep restriction combined with circadian syndromes in mice. Diabetologia 14 (3), 141–148.
disruption. Sci. Transl. Med. 4 (129), 129–143. Coll, A.P., Tung, Y.C.L., 2009. Pro-opiomelanocortin (POMC)-derived peptides and the
Cabanac, M., 2001. Regulation and the ponderostat. Int. J. Obesity Relat. Metab. regulation of energy homeostasis. Mol. Cell. Endocrinol. 300, 147–151.
Disord. 25 (Suppl. 5), S7–S12. Cone, R.D., 2005. Anatomy and regulation of the central melanocortin system. Nat.
Cai, X.J., Widdowson, P.S., Harrold, J., et al., 1999. Hypothalamic orexin expression: Neurosci. 8 (5), 571–578.
modulation by blood glucose and feeding. Diabetes 48 (11), 2132–2137. Considine, R.V., Sinha, M.K., Heiman, M.L., et al., 1996. Serum immunoreactive-leptin
Cammisotto, P.G., Bukowiecki, L.J., Deshaies, Y., Bendayan, M., 2006. Leptin concentrations in normal-weight and obese humans. N. Engl. J. Med. 334 (5),
biosynthetic pathway in white adipocytes. Biochem. Cell Biol. 84 (2), 207–214. 292–295.
Campbell, C.C., 1991. Food insecurity: a nutritional outcome or a predictable variable? Cota, D., Marsicano, G., Tschöp, M., et al., 2003. The endogenous cannabinoid
J. Nutr. 121 (3), 408–415. system affects energy balance via central orexigenic drive and peripheral lipo-
Campfield, L.A., Smith, F.J., 1986. Functional coupling between transient declines in genesis. J. Clin. Invest. 112 (3), 423–431.
blood glucose and feeding behavior: temporal relationships. Brain Res. Bull. 17 (3), Cowley, M.A., Smart, J.L., Rubinstein, M., et al., 2001. Leptin activates anorexigenic
427–433. POMC neurons through a neural network in the arcuate nucleus. Nature 411
Campfield, L.A., Smith, F.J., 1990. Systemic factors in the control of food intake. (6836), 480–484.
Evidence for patterns as signals. In: Stricker, E.M. (Ed.), Handbook of Behavioral Cowley, M.A., Smith, R.G., Diano, S., et al., 2003. The distribution and mechanism of
Neurobiology, Neurobiology of Food and Fluid Intake, vol. 10. Plenum Press, action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating
New York. energy homeostasis. Neuron 37 (4), 649–661.
Cannon, W.B., Washburn, A.L., 1912. An explanation of hunger. Am. J. Physiol. 29, Crespo, I., Gómez de Heras, R., Rodríguez de Fonseca, F., Navarro, M., 2008.
441–454. Pretreatment with subeffective doses of Rimonabant attenuates orexigenic actions
Carlson, A.J., 1916. The Control of Hunger in Health and Disease. The University of of orexin A-hypocretin 1. Neuropharmacology 54 (1), 219–225.
Chicago Press, Chicago. Cristino, L., Busetto, G., Imperatore, R., et al., 2013. Obesity-driven synaptic
Carnell, S., Wardle, J., 2008. Appetite and adiposity in children: evidence for remodeling affects endocannabinoid control of orexinergic neurons. Proc. Natl.
a behavioral susceptibility theory of obesity. Am. J. Clin. Nutr. 88 (1), 22–29. Acad. Sci. U.S.A. 110 (24), E2229–E2238.
Carpéné, C., Dray, C., Attane, C., et al., 2007. Expanding role for the apelin/APJ Cummings, D.E., Purnell, J.Q., Frayo, R.S., Schmidova, K., Wisse, B.E., Weigle, D.S.,
system in physiopathology. J. Physiol. Biochem. 63 (4), 359–373. 2001. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation
Casanueva, F.F., Diéguez, C., 1999. Neuroendocrine regulation and actions of leptin. in humans. Diabetes 50 (8), 1714–1719.
Front. Neuroendocrinol. 20 (4), 317–363. D’Addario, C., Micioni Di Bonaventura, M.V., Pucci, M., et al., 2014. Endocannabinoid
Cerf, M.E., 2011. Islets organogenesis, angiogenesis and innervation. Cell Biol. Int. 35 signaling and food addiction. Neurosci. Biobehav. Rev. 47, 203–224.
(11), 1065–1078. Dalamaga, M., Diakopoulos, K.N., Mantzoros, C.S., 2012. The role of adiponectin in
Chakravarthy, M.V., Zhu, Y., Lopez, M., et al., 2007. Brain fatty acid synthase acti- cancer: a review of current evidence. Endocrinol. Rev. 33 (4), 547–594.
vates PPARalpha to maintain energy homeostasis. J. Clin. Invest. 117 (9), Darling, R.A., Ritter, S., 2009. 2-Deoxy-D-glucose, but not mercaptoacetate, increases
2352–2539. food intake in decerebrate rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 297
Challis, B.G., Coll, A.P., Yeo, G.S., et al., 2004. Mice lacking proopiomelanocortin are (2), R382–R386.
sensitive to high-fat feeding but respond normally to the acute anorectic effects of Davenport, A.P., Bonner, T.I., Foord, S.M., et al., 2005. International union of phar-
peptide-YY(3-36). Proc. Natl. Acad. Sci. U.S.A. 101 (13), 4695–4700. macology. LVI. Ghrelin receptor nomenclature, distribution, and function. Phar-
Chang, A.C., Cochet, M., Cohen, S.N., 1980. Structural organization of human macol. Rev. 57 (4), 541–546.
genomic DNA encoding the pro-opiomelanocortin peptide. Proc. Natl. Acad. Sci. Degawa-Yamauchi, M., Bovenkerk, J.E., Juliar, B.E., et al., 2003. Serum resistin
U.S.A. 77 (8), 4890–4894. (FIZZ3) protein is increased in obese humans. J. Clin. Endocrinol. Metab. 88 (11),
Chemelli, R.M., Willie, J.T., Sinton, C.M., et al., 1999. Narcolepsy in orexin knockout 5452–5455.
mice: molecular genetics of sleep regulation. Cell 98 (4), 437–451. De Lecea, L., Kilduff, T.S., Peyron, C., et al., 1998. The hypocretins: hypothalamus-
Chen, H., Charlat, O., Tartaglia, L.A., et al., 1996. Evidence that the diabetes gene specific peptides with neuroexcitatory activity. Proc. Natl. Acad. Sci. U.S.A. 95 (1),
encodes the leptin receptor: identification of a mutation in the leptin receptor gene 322–327.
in db/db mice. Cell 84 (3), 491–495. Delhanty, P.J., Sun, Y., Visser, J.A., et al., 2010. Unacylated ghrelin rapidly modulates
Chen, X., Shu, X., Cong, Z.K., Jiang, Z.Y., Jiang, H., 2015. Nesfatin-1 acts on the lipogenic and insulin signaling pathway gene expression in metabolically active
dopaminergic reward pathway to inhibit food intake. Neuropeptides 53, tissues of GHSR deleted mice. PLoS One 5 (7), e11749.
45–50. Delporte, C., 2013. Structure and physiological actions of ghrelin. Scientifica (Cairo)
Chen, X.P., Yang, W., Fan, Y., et al., 2010. Structural determinants in the second 2013. http://dx.doi.org/10.1155/2013/518909, 518909.
intracellular loop of the human cannabinoid CB1 receptor mediate selective De Mota, N., Reaux-Le Goazigo, A., El Messari, S., et al., 2004. Apelin, a potent
coupling to G(s) and G(i). Br. J. Pharmacol. 161 (8), 1817–1834. diuretic neuropeptide counteracting vasopressin actions through inhibition of
Chen, Y., Lin, Y.C., Kuo, T.W., Knight, Z.A., 2015. Sensory detection of food rapidly vasopressin neuron activity and vasopressin release. Proc. Natl. Acad. Sci. U.S.A.
modulates arcuate feeding circuits. Cell 160 (5), 829–841. 101 (28), 10464–10469.
Food Intake and Its Control by Signaling Molecules 203

Denver, R.J., Bonett, R.M., Boorse, G.C., 2011. Evolution of leptin structure and Fisler, J.S., Egawa, M., Bray, G.A., 1995. Peripheral 3-hydroxybutyrate and food intake
function. Neuroendocrinology 94 (1), 21–38. in a model of dietary-fat induced obesity: effect of vagotomy. Physiol. Behav. 58
Denzel, M.S., Scimia, M.C., Zumstein, P.M., Walsh, K., Ruiz-Lozano, P., Ranscht, B., (1), 1–7.
2010. T-cadherin is critical for adiponectin-mediated cardioprotection in mice. Fliers, E., Noppen, N.M., Wiersinga, W.M., Visser, T.J., Swaab, D.F., 1994. Distri-
J. Clin. Invest. 120 (12), 4342–4352. bution of thyrotropin-releasing hormone (TRH)-containing cells and fibers in the
De Ruiter, L., 1967. Feeding behavior of vertebrates in the natural environment. In: human hypothalamus. J. Comp. Neurol. 350 (2), 311–323.
Code, C.F. (Ed.), Handbook of Physiology, Section 6: Alimentary Canal. Williams & Flores, A., Maldonado, R., Berrendero, F., 2013. Cannabinoid-hypocretin cross-talk in
Wilkins, Baltimore, pp. 97–116. the central nervous system: what we know so far. Front. Neurosci. 7 (256) http://
Devane, W.A., Hanus, L., Breuer, A., et al., 1992. Isolation and structure of a brain dx.doi.org/10.3389/fnins.2013.00256.
constituent that binds to the cannabinoid receptor. Science 258 (5090), Flynn, F.W., Grill, H.J., 1983. Insulin elicits ingestion in decerebrate rats. Science 221
1946–1949. (4606), 188–190.
Díaz-Muñoz, M., Vázquez-Martínez, O., Aguilar-Roblero, R., Escobar, C., 2000. Franklin, K., Muir, P., Scott, T., Wilcocks, L., Yates, P., 2010. Introduction to Biological
Anticipatory changes in liver metabolism and entrainment of insulin, glucagon, and Physics for the Health and Life Sciences. Chapter 17: Temperature and the Zeroth
corticosterone in food-restricted rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. Law. Wiley, USA.
279 (6), R2048–R2056. Friedman, J.M., Halaas, J.L., 1998. Leptin and the regulation of body weight in
Di Bella, L., Tarozzi, G., Rossi, M.T., Scalera, G., 1981. Effect of liver temperature mammals. Nature 395 (6704), 763–770.
increase on food intake. Physiol. Behav. 26 (1), 45–51. Friedman, J.M., Mantzoros, C.S., 2015. 20 years of leptin: from the discovery of
Di Marzo, V., Goparaju, S.K., Wang, L., et al., 2001. Leptin-regulated endocannabi- the leptin gene to leptin in our therapeutic armamentarium. Metabolism 64
noids are involved in maintaining food intake. Nature 410 (6830), 822–825. (1), 1–4.
Di Marzo, V., Ligresti, A., Cristino, L., 2009. The endocannabinoid system as a link Friedman, M.I., 1980. Hepatic-cerebral interactions in insulin-induced eating and
between homoeostatic and hedonic pathways involved in energy balance regula- gastric acid secretion. Brain Res. Bull. 5 (Suppl. 4), 63–68.
tion. Int. J. Obesity (Lond.) 33 (Suppl. 2), S18–S24. Friedman, M.I., Harris, R.B., Ji, H., Ramírez, I., Tordoff, M.G., 1999. Fatty acid
Di Marzo, V., Matias, I., 2005. Endocannabinoid control of food intake and energy oxidation affects food intake by altering hepatic energy status. Am. J. Physiol. 276
balance. Nat. Neurosci. 8 (5), 585–589. (4 Pt 2), R1046–R1053.
Dodd, G.T., Decherf, S., Loh, K., et al., 2015. Leptin and insulin act on POMC neurons Friedman, M.I., Horn, C.C., Ji, H., 2005. Peripheral signals in the control of feeding
to promote the browning of white fat. Cell 160 (1–2), 88–104. behavior. Chem. Senses 30 (Suppl. 1), i182–i183.
Drazen, D.L., Woods, S.C., 2003. Peripheral signals in the control of satiety and Gantz, I., Konda, Y., Tashiro, T., et al., 1993. Molecular cloning of a novel melanocortin
hunger. Curr. Opin. Clin. Nutr. Metab. Care 6 (6), 621–629. receptor. J. Biol. Chem. 268 (11), 8246–8250.
Drouin, J., Goodman, H.M., 1980. Most of the coding region of rat ACTH beta-LPH Gao, H., Fall, T., Van Dam, R.M., et al., 2013. Evidence of a causal relationship
precursor gene lacks intervening sequences. Nature 11 (5791), 610–613. between adiponectin levels and insulin sensitivity: a Mendelian randomization
Dryden, S., Pickavance, L., Frankish, H.M., Williams, G., 1995. Increased neuro- study. Diabetes 62 (4), 1338–1344.
peptide Y secretion in the hypothalamic paraventricular nucleus of obese (fa/fa) Garaulet, M., Madrid, J.A., 2010. Chronobiological aspects of nutrition, metabolic
Zucker rats. Brain Res. 690 (2), 185–188. syndrome and obesity. Adv. Drug Deliv. Rev. 62 (9–10), 967–978.
Eckel-Mahan, K., Sassone-Corsi, P., 2013. Metabolism and the circadian clock Gauna, C., Kiewiet, R.M., Janssen, J.A., et al., 2007. Unacylated ghrelin acts as
converge. Physiol. Rev. 93 (1), 107–135. a potent insulin secretagogue in glucose stimulated conditions. Am. J. Physiol.
Egertová, M., Cravatt, B.F., Elphick, M.R., 2003. Comparative analysis of fatty acid Endocrinol. Metab. 293 (3), E697–E704.
amide hydrolase and cb(1) cannabinoid receptor expression in the mouse brain: Gerard, C.M., Mollereau, C., Vassart, G., Parmentier, M., 1991. Molecular cloning of
evidence of a widespread role of fatty acid amide hydrolase in regulation of a human cannabinoid receptor which is also expressed in testis. Biochem. J. 279
endocannabinoid signaling. Neuroscience 119 (2), 481–496. (Pt 1), 129–134.
El-Haschimi, K., Pierroz, D.D., Hileman, S.M., Bjørbaek, C., Flier, J.S., 2000. Two Germain, N., Galusca, B., Le Roux, C.W., et al., 2007. Constitutional thinness and lean
defects contribute to hypothalamic leptin resistance in mice with diet-induced anorexia nervosa display opposite concentrations of peptide YY, glucagon-like
obesity. J. Clin. Invest. 105 (12), 1827–1832. peptide 1, ghrelin, and leptin. Am. J. Clin. Nutr. 85 (4), 967–971.
Elias, C.F., Lee, C., Kelly, J., et al., 1998a. Leptin activates hypothalamic CART Ghelardoni, S., Carnicelli, V., Frascarelli, S., Ronca-Testoni, S., Zucchi, R., 2006.
neurons projecting to the spinal cord. Neuron 21 (6), 1375–1385. Ghrelin tissue distribution: comparison between gene and protein expression.
Elias, C.F., Saper, C.B., Maratos-Flier, E., et al., 1998b. Chemically defined projections J. Endocrinol. Invest. 29 (2), 115–121.
linking the mediobasal hypothalamus and the lateral hypothalamic area. J. Comp. Gnanapavan, S., Kola, B., Bustin, S.A., et al., 2002. The tissue distribution of the
Neurol. 402 (4), 442–459. mRNA of ghrelin and subtypes of its receptor, GHS-R in humans. J. Clin.
Elias, C.F., Aschkenasi, C., Lee, C., et al., 1999. Leptin differentially regulates NPY and Endocrinol. Metab. 87 (6), 2988–2991.
POMC neurons projecting to the lateral hypothalamic area. Neuron 23 (4), Gold, R.M., 1973. Hypothalamic obesity: the myth of the ventromedial nucleus.
775–786. Science 182 (4111), 488–490.
Erickson, J.C., Clegg, K.E., Palmiter, R.D., 1996. Sensitivity to leptin and susceptibility Gonçalves, G.H., Li, W., Garcia, A.V., Figueiredo, M.S., Bjørbæk, C., 2014.
to seizures of mice lacking neuropeptide Y. Nature 381 (6581), 415–421. Hypothalamic agouti-related peptide neurons and the central melanocortin
Escobar, C., Díaz-Muñoz, M., Encinas, F., Aguilar-Roblero, R., 1998. Persistence of system are crucial mediators of leptin’s antidiabetic actions. Cell Rep. 7 (4),
metabolic rhythmicity during fasting and its entrainment by restricted feeding 1093–1103.
schedules in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 274 (5 pt 2), González, R., Tiwari, A., Unniappan, S., 2009. Pancreatic beta cells colocalize insulin
R1309–R1316. and pronesfatin immunoreactivity in rodents. Biochem. Biophys. Res. Commun.
Escobar, C., Hudson, R., Martínez-Gómez, M., Aguilar-Roblero, R., 2000. Metabolic 381 (4), 643–648.
correlates of the circadian pattern of suckling-associated arousal in young rabbits. Granata, R., Baragli, A., Settanni, F., Scarlatti, F., Ghigo, E., 2010. Unraveling the role
J. Comp. Physiol. Sens. Neural Behav. Physiol. 186 (1), 33–38. of the ghrelin gene peptides in the endocrine pancreas. J. Mol. Endocrinol. 45 (3),
Eva, C., Serra, M., Mele, P., Panzica, G., Oberto, A., 2006. Physiology and gene 107–118.
regulation of the brain NPY Y1 receptor. Front. Neuroendocrinol. 27 (3), 308–339. Green, E.D., Maffei, M., Braden, V.V., et al., 1995. The human obese (OB) gene: RNA
Fain, J.N., Cheema, P.S., Bahouth, S.W., Lloyd-Hiler, M., 2003. Resistin release by expression pattern and mapping on the physical, cytogenetic, and genetic maps of
human adipose tissue explants in primary culture. Biochem. Biophys. Res. Com- chromosome 7. Genome Res. 5 (1), 5–12.
mun. 300 (3), 674–678. Green, C.B., Takahashi, J.S., Bass, J., 2008. The meter of metabolism. Cell 134 (5),
Faouzi, M., Leshan, R., Björnholm, M., Hennessey, T., Jones, J., Münzberg, H., 2007. 728–742.
Differential accessibility of circulating leptin to individual hypothalamic sites. Grimaldi, B., Nakahata, Y., Kaluzova, M., Masubuchi, S., Sassone-Corsi, P., 2009.
Endocrinology 148 (11), 5414–5423. Chromatin remodeling, metabolism and circadian clocks: the interplay of CLOCK
Feng, D.D., Yang, S.K., Loudes, C., et al., 2011. Ghrelin and obestatin modulate and SIRT1. Int. J. Biochem. Cell Biol. 41 (1), 81–86.
growth hormone-releasing hormone release and synaptic inputs onto growth Gropp, E., Shanabrough, M., Borok, E., et al., 2005. Agouti-related peptide-expressing
hormone-releasing hormone neurons. Eur. J. Neurosci. 34 (5), 732–744. neurons are mandatory for feeding. Nat. Neurosci. 8 (10), 1289–1291.
Feng, N., Young, S.F., Aguilera, G., Puricelli, E., et al., 2005. Co-occurrence of two Guan, J.L., Suzuki, R., Funahashi, H., et al., 2002. Ultrastructural localization of orexin-
partially inactivating polymorphisms of MC3R is associated with pediatric onset 1 receptor in pre- and post-synaptic neurons in the rat arcuate nucleus. Neurosci.
obesity. Diabetes 54 (9), 2663–2667. Lett. 329 (2), 209–212.
204 Food Intake and Its Control by Signaling Molecules

Guan, X.M., Yu, H., Palyha, O.C., et al., 1997. Distribution of mRNA encoding the Kalra, S.P., Dube, M.G., Pu, S., Xu, B., Horvath, T.L., Kalra, P.S., 1999. Interacting
growth hormone secretagogue receptor in brain and peripheral tissues. Brain Res. appetite-regulating pathways in the hypothalamic regulation of body weight.
Mol. Brain Res. 48 (1), 23–29. Endocr. Rev. 20 (1), 68–100.
Hahn, T.M., Breininger, J.F., Baskin, D.G., Schwartz, M.W., 1998. Coexpression of Kalsbeek, A., Ruiter, M., La Fleur, S.E., Cailotto, C., Kreier, F., Buijs, R.M., 2006. The
Agrp and NPY in fasting-activated hypothalamic neurons. Nat. Neurosci. 1 (4), hypothalamic clock and its control of glucose homeostasis. Prog. Brain Res. 153,
271–272. 283–307.
Håkansson, M., de Lecea, L., Sutcliffe, J.G., Yanagisawa, M., Meister, B., 1999. Kalsbeek, A., Yi, C.X., La Fleur, S.E., Fliers, E., 2010. The hypothalamic clock and its
Leptin receptor- and STAT3-immunoreactivities in hypocretin/orexin neurons of the control of glucose homeostasis. Trends Endocrinol. Metab. 21 (7), 402–410.
lateral hypothalamus. J. Neuroendocrinol. 11 (8), 653–663. Kalsbeek, A., Scheer, F.A., Perreau-Lenz, S., et al., 2011. Circadian disruption and
Halaas, J.L., Gajiwala, K.S., Maffei, M., et al., 1995. Weight-reducing effects of the SCN control of energy metabolism. FEBS Lett. 585 (10), 1412–1426.
plasma protein encoded by the obese gene. Science 269 (5223), 543–546. Kamegai, J., Tamura, H., Shimizu, T., Ishii, S., Sugihara, H., Wakabayashi, I., 2000.
Hara, J., Beuckmann, C.T., Nambu, T., et al., 2001. Genetic ablation of orexin neurons Central effect of ghrelin, an endogenous growth hormone secretagogue, on
in mice results in narcolepsy, hypophagia, and obesity. Neuron 30 (2), 345–354. hypothalamic peptide gene expression. Endocrinology 141 (12), 4797–4800.
Harris, G.C., Aston-Jones, G., 2006. Arousal and reward: a dichotomy in orexin Kanamoto, N., Akamizu, T., Tagami, T., et al., 2004. Genomic structure and char-
function. Trends Neurosci. 29 (10), 571–577. acterization of the 5’-flanking region of the human ghrelin gene. Endocrinology 145
Haynes, A.C., Jackson, B., Chapman, H., Tadayyon, M., Johns, A., Porter, R.A., (9), 4144–4153.
Arch, J.R., 2000. A selective orexin-1 receptor antagonist reduces food Karatsoreos, I.N., Thaler, J.P., Borgland, S.L., Champagne, F.A., Hurd, Y.L., Hill, M.N.,
consumption in male and female rats. Regul. Peptides 96 (1–2), 45–51. 2013. Food for thought: hormonal, experiential, and neural influences on feeding
Herkenham, M., Lynn, A.B., Little, M.D., et al., 1990. Cannabinoid receptor localization and obesity. J. Neurosci. 33 (45), 17610–17616.
in brain. Proc. Natl. Acad. Sci. U.S.A. 87 (5), 1932–1936. Kaser, S., Kaser, A., Sandhofer, A., Ebenbichler, C.F., Tilg, H., Patsch, J.R., 2003.
Hervieu, G.J., Cluderay, J.E., Harrison, D.C., Roberts, J.C., Leslie, R.A., 2001. Gene Resistin messenger-RNA expression is increased by proinflammatory cytokines
expression and protein distribution of the orexin-1 receptor in the rat brain and in vitro. Biochem. Biophys. Res. Commun. 309 (2), 286–290.
spinal cord. Neuroscience 103 (3), 777–797. Kennedy, G.C., 1953. The role of depot fat in the hypothalamic control of food intake in
Higuchi, H., Hasegawa, A., Yamaguchi, T., 2005. Transcriptional regulation of neuronal the rat. Proc. R. Soc. Lond. Ser. B Biol. Sci. 140 (901), 578–596.
genes and its effect on neural functions: transcriptional regulation of neuropeptide Kirchner, H., Gutiérrez, J.A., Solenberg, P.J., et al., 2009. GOAT links dietary lipids
Y gene by leptin and its effect on feeding. J. Pharmacol. Sci. 98 (3), 225–231. with the endocrine control of energy balance. Nat. Med. 15 (7), 741–745.
Higuchi, H., 2012. Molecular analysis of central feeding regulation by neuropeptide Y Kirouac, G.J., 2015. Placing the paraventricular nucleus of the thalamus within the
(NPY) neurons with NPY receptor small interfering RNAs (siRNAs). Neurochem. Int. brain circuits that control behavior. Neurosci. Biobehav. Rev. 56 (1), 315–329.
61 (6), 936–941. Kleinridders, A., Konner, A.C., Bruning, J.C., 2009. CNS-targets in control of energy
Hilairet, S., Bouaboula, M., Carrière, D., Le Fur, G., Casellas, P., 2003. Hypersensi- and glucose homeostasis. Curr. Opin. Pharmacol. 9 (11), 794–804.
tization of the Orexin 1 receptor by the CB1 receptor: evidence for cross-talk Koch, M., Varela, L., Kim, J.G., et al., 2015. Hypothalamic PONC neurons promote
blocked by the specific CB1 antagonist, SR141716. J. Biol. Chem. 278 (26), cannabinoid-induced feeding. Nature 519 (7541), 45–50.
23731–23737. Biochem. J. 279 (Pt 1), 129–134. Kohno, D., Gao, H.Z., Muroya, S., Kikuyama, S., Yada, T., 2003. Ghrelin directly
Hill, R.A., Margetic, S., Pegg, G.G., Gazzola, C., 1998. Leptin: its pharmacokinetics interacts with neuropeptide-Y-containing neurons in the rat arcuate nucleus: Ca2þ
and tissue distribution. Int. J. Obesity Relat. Metab. Disord. 22 (8), 765–770. signaling via protein kinase A and N-type channel-dependent mechanisms and
Hirayama, K., Gillette, R., 2012. A neuronal network switch for approach/avoiding cross-talk with leptin and orexin. Diabetes 52 (4), 948–956.
toggled by appetitive state. Curr. Biol. 22 (2), 118–123. Kohno, D., Nakata, M., Maejima, Y., et al., 2008. Nesfatin-1 neurons in paraventricular
Hohmann, A.G., Herkenham, M., 1999. Localization of central cannabinoid CB1 and supraoptic nuclei of the rat hypothalamus coexpress oxytocin and vasopressin
receptor messenger RNA in neuronal subpopulations of rat dorsal root ganglia: and are activated by refeeding. Endocrinology 149 (3), 1295–1301.
a double-label in situ hybridization study. Neuroscience 90 (3), 923–931. Kojima, M., Hosoda, H., Date, Y., Nakazato, M., Matsuo, H., Kangawa, K., 1999.
Holliday, N.D., Michel, M.C., Cox, H.M., 2004. NPY receptor subtypes and their signal Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402
transduction. In: Michel, M.C. (Ed.), Neuropeptide Y and Related Peptides. (6762), 656–660.
Springer, Berlin, pp. 45–73. Kojima, M., Kangawa, K., 2005. Ghrelin: structure and function. Physiol. Rev. 85 (2),
Horvath, T.L., Diano, S., van den Pol, A.N., 1999. Synaptic interaction between 495–522.
hypocretin (orexin) and neuropeptide Y cells in the rodent and primate hypothal- Könner, A.C., Janoschek, R., Plum, L., et al., 2007. Insulin action in AgRP-expressing
amus: a novel circuit implicated in metabolic and endocrine regulations. J. Neu- neurons is required for suppression of hepatic glucose production. Cell Metab. 5
rosci. 19 (3), 1072–1087. (6), 438–449.
Hosoda, H., Kangawa, K., 2004. Ghrelin measurement: present and perspectives. In: Korner, J., Wissig, S., Kim, A., Conwell, I.M., Wardlaw, S.L., 2003. Effects of agouti-
Ghrelin. Springer Link, pp. 225–236 (Chapter 23). related protein on metabolism and hypothalamic neuropeptide gene expression.
Hosoya, M., Kawamata, Y., Fukusumi, S., et al., 2000. Molecular and functional J. Neuroendocrinol. 15 (12), 1116–11121.
characteristics of APJ. Tissue distribution of mRNA and interaction with the Kos, K., Harte, A.L., Da Silva, N.F., et al., 2007. Adiponectin and resistin in human
endogenous ligand apelin. J. Biol. Chem. 275 (38), 21061–21067. cerebrospinal fluid and expression of adiponectin receptors in the human hypo-
Howard, A.D., Feighner, S.D., Cully, D.F., et al., 1996. A receptor in pituitary and thalamus. J. Clin. Endocrinol. Metab. 92 (3), 1129–1136.
hypothalamus that functions in growth hormone release. Science 273 (5277), Koylu, E.O., Couceyro, P.R., Lambert, P.D., Ling, N.C., DeSouza, E.B., Kuhar, M.J.,
974–977. 1997. Immunohistochemical localization of novel CART peptides in rat hypothal-
Hu, E., Liang, P., Spiegelman, B.M., 1996. AdipoQ is a novel adipose-specific gene amus, pituitary and adrenal gland. J. Neuroendocrinol. 9 (11), 823–833.
dysregulated in obesity. J. Biol. Chem. 271 (18), 10697–10703. Krashes, M.J., Koda, S., Ye, C., et al., 2011. Rapid, reversible activation of AgRP
Inutsuka, A., Inui, A., Tabuchi, S., Tsunematsu, T., Lazarus, M., Yamanaka, A., 2014. neurons drives feeding behavior in mice. J. Clin. Invest. 121 (4), 1424–1428.
Concurrent and robust regulation of feeding behaviors and metabolism by orexin Kristensen, P., Judge, M.E., Thim, L., et al., 1998. Hypothalamic CART is a new
neurons. Neuropharmacology 85, 451–460. anorectic peptide regulated by leptin. Nature 393 (6680), 72–76.
Jequier, E., Acheson, K., Schutz, Y., 1987. Assessment of energy expenditure and fuel Krude, H., Biebermann, H., Luck, W., Horn, R., Brabant, G., Grüters, A., 1998. Severe
utilization in man. Annu. Rev. Nutr. 7, 187–208. early-onset obesity, adrenal insufficiency and red hair pigmentation caused by
Ji, H., Graczyk-Milbrandt, G., Friedman, M.I., 2000. Metabolic inhibitors synergistically POMC mutations in humans. Nat. Genet. 19 (2), 155–157.
decrease hepatic energy status and increase food intake. Am. J. Physiol. Regul. Kubota, N., Terauchi, Y., Yamauchi, T., et al., 2002. Disruption of adiponectin causes
Integr. Comp. Physiol. 278 (6), R1579–R1582. insulin resistance and neointimal formation. J. Biol. Chem. 277 (29), 25863–25866.
Jobst, E.E., Enriori, P.J., Cowley, M.A., 2004. The electrophysiology of feeding circuits. Kubota, N., Yano, W., Kubota, T., et al., 2007. Adiponectin stimulates AMP-activated
Trends Endocrinol. Metab. 15 (10), 488–499. protein kinase in the hypothalamus and increases food intake. Cell Metab. 6 (1),
Juárez, R., Tongju-Koh, E., 1978. Anatomical evidence of direct projections from the 55–68.
nucleus of the solitary tract to the hypothalamus, amygdala, and other forebrain Kunii, K., Yamanaka, A., Nambu, T., Matsuzaki, I., Goto, K., Sakurai, T., 1999.
structures in the rat. Brain Res. 153 (1), 1–26. Orexins/hypocretins regulate drinking behaviour. Brain Res. 842 (1), 256–261.
Kalra, S.P., Dube, M.G., Sahu, A., Phelps, C.P., Kalra, P.S., 1991. Neuropeptide Y Kusminski, C.M., McTernan, P.G., Schraw, T., et al., 2007. Adiponectin complexes in
secretion increases in the paraventricular nucleus in association with increased human cerebrospinal fluid: distinct complex distribution from serum. Diabetologia
appetite for food. Proc. Natl. Acad. Sci. U.S.A. 88 (23), 10931–10935. 50 (3), 634–642.
Food Intake and Its Control by Signaling Molecules 205

La Cava, A., Matarese, G., 2004. The weight of leptin in immunity. Nat. Rev. Immunol. Louis-Sylvestre, J., Larue-Achagiotis, C., Michel, A., Houssin, D., 1990. Feeding
4 (5), 371–379. pattern of liver-transplanted rats. Physiol. Behav. 48 (2), 321–326.
Lamia, K.A., Sachdeva, U.M., DiTacchio, L., et al., 2009. AMPK regulates the circadian Luiten, P.G.M., ter Horst, G.J., Karst, H., Steffens, A.B., 1985. The course of para-
clock by cryptochrome phosphorylation and degradation. Science 326 (5951), ventricular hypothalamic efferents to autonomic structures in medulla and spinal
437–440. cord. Brain Res. 329 (1), 374–378.
Lamy, C.M., Sanno, H., Labouebe, G., et al., 2014. Hypoglycemia-activated GLUT2 Luquet, S., Perez, F.A., Hnasko, T.S., Palmiter, R.D., 2005. NPY/AgRP neurons are
neurons of the nucleus tractus solitarius stimulate vagal activity and glucagon essential for feeding in adult mice but can be ablated in neonates. Science 310
secretion. Cell Metab. 19 (3), 527–538. (5748), 683–685.
Lanfranco, F., Motta, G., Baldi, M., et al., 2010. Ghrelin and anterior pituitary function. Lv, S.Y., Yang, Y.J., Qin, Y.J., Xiong, W., Chen, Q., 2011. Effect of centrally
Front. Horm. Res. 38, 206–211. administered apelin-13 on gastric emptying and gastrointestinal transit in mice.
Lanfray, D., Arthaud, S., Ouellet, J., et al., 2013. Gliotransmission and brain glucose Peptides 32 (5), 978–982.
sensing: critical role of endozepines. Diabetes 62 (3), 801–810. Lv, S.Y., Yang, Y.J., Qin, Y.J., et al., 2012. Central apelin-13 inhibits food intake via
Langhans, W., 2003. Role of the liver in the control of glucose-lipid utilization and body the CRF receptor in mice. Peptides 33 (1), 132–138.
weight. Curr. Opin. Clin. Nutr. Metab. Care 6 (4), 449–455. Maccarrone, M., Guzmán, M., Mackie, K., Doherty, P., Harkany, T., 2014.
LeBlanc, J., Diamond, P., Griggio, M., Nadeau, A., Richard, D., 1991. Control of Programming of neural cells by (endo)cannabinoids: from physiological roles to
cephalic thermogenic phase of feeding. In: Ailhaud, G., Guy-Graad, B., emerging therapies. Nat. Rev. Neurosci. 15 (12), 786–801.
Lafontan, M., Ricquier, D. (Eds.), Obesity in Europe, vol. 91. John Libbey, London, Mackie, K., 2005. Distribution of cannabinoid receptors in the central and peripheral
pp. 241–247. nervous system. Handb. Exp. Pharmacol. 168, 299–325.
Lechan, R.M., Toni, R., 2013. Functional anatomy of the hypothalamus and pituitary. Maeda, K., Okubo, K., Shimomura, I., Funahashi, T., Matsuzawa, Y., Matsubara, K.,
Endotext [Internet] MDtext.com, Inc., South Dartmouth (MA). 1996. cDNA cloning and expression of a novel adipose specific collagen-like factor,
Lee, D.K., Saldivia, V.R., Nguyen, T., Cheng, R., George, S.R., O’Dowd, B.F., 2005. apM1 (AdiPose most abundant gene transcript 1). Biochem. Biophys. Res. Com-
Modification of the terminal residue of apelin-13 antagonizes its hypotensive action. mun. 221 (2), 286–289.
Endocrinology 146 (1), 231–236. Maejima, Y., Sedbazar, U., Suyama, S., et al., 2009. Nesfatin-1-regulated oxy-
Lee, J.S., Lee, E.Y., Lee, H.S., 2015. Hypothalamic, feeding/arousal-related pepti- tocinergic signaling in the paraventricular nucleus causes anorexia through a lep-
dergic projections to the paraventricular thalamic nucleus in the rat. Brain Res. tin-independent melanocortin pathway. Cell Metab. 10 (5), 355–365.
1598, 97–113. Maffei, M., Halaas, J., Ravussin, E., et al., 1995. Leptin levels in human and rodent:
Lehrke, M., Reilly, M.P., Millington, S.C., Iqbal, N., Rader, D.J., Lazar, M.A., measurement of plasma leptin and ob RNA in obese and weight-reduced subjects.
2004. An inflammatory cascade leading to hyperresistinemia in humans. PLoS Nat. Med. 1 (11), 1155–1161.
Med. 1 (2), e45. Marchant, E.G., Mistlberger, R.E., 1997. Anticipation and entrainment to feeding
Leibel, R.L., 2008. Molecular physiology of weight regulation in mice and humans. Int. time in intact and SCN-ablated C57BL/6j mice. Brain Res. 765 (2),
J. Obesity 32 (Suppl. 7), S98–S108. 273–282.
Leibowitz, S.F., 1978. Adrenergic stimulation of the paraventricular nucleus and its Marsh, D.J., Hollopeter, G., Kafer, K.E., Palmiter, R.D., 1998. Role of the Y5 neuro-
effects on ingestive behavior as a function of drug dose and time of injection in the peptide Y receptor in feeding and obesity. Nat. Med. 4 (6), 718–721.
light-dark cycle. Brain Res. Bull. 3 (4), 357–363. Martínez-Merlos, M.T., Ángeles-Castellanos, M., Díaz-Muñoz, M., Aguilar-Roblero, R.,
Leibowitz, S.F., Wortley, K.E., 2004. Hypothalamic control of energy balance: different Mendoza, J., Escobar, C., 2004. Dissociation between adipose tissue signals,
peptides, different functions. Peptides 25 (3), 473–504. behavior and the food-entrained oscillator. J. Endocrinol. 181 (1), 53–63.
Lenard, N.R., Berthoud, H.R., 2008. Central and peripheral regulation of food intake Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., Bonner, T.T., 1990.
and physical activity: pathways and genes. Obesity (Silver Spring MD) 16 (Suppl. Structure of a cannabinoid receptor and functional expression of the cloned cDNA.
3), S11–S22. Nature 346 (6284), 561–564.
LeRoith, D., Bondy, C., Yakar, S., Liu, J.L., Butler, A., 2001. The somatomedin Mayer, J., 1953. Glucostatic mechanism of regulation of food intake. N. Engl. J. Med.
hypothesis: 2001. Endocr. Rev. 22 (1), 53–74. 249 (1), 13–16.
LeSauter, J., Hoque, N., Weintraub, M., Pfaff, D.W., Silver, R., 2009. Stomach ghrelin- McCabe, J.T., DeBellis, M., Leibowitz, S.F., 1984. Clonidine-induced feeding: analysis
secreting cells as food-entrainable circadian clocks. Proc. Natl. Acad. Sci. U.S.A. of central sites of action and fiber projections mediating this response. Brain Res.
106 (32), 13582–13587. 309 (1), 85–104.
Li, J., Yi, S.Q., Wang, H.X., Yi, N., Ogawa, Y., Ozaki, N., Itoh, M., 2011. Immuno- Mechoulam, R., Ben-Shabat, S., Hanus, L., et al., 1995. Identification of an endog-
localization of ghrelin in the stomach of Sprague-Dawley rat. Anat. Histol. Embryol. enous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors.
40 (2), 95–99. Biochem. Pharmacol. 50 (1), 83–90.
Li, L., Zhang, L.K., Pang, Y.Z., et al., 2006. Cardioprotective effects of ghrelin and Meister, B., 2007. Neurotransmitters in key neurons of the hypothalamus that regulate
des-octanoyl ghrelin on myocardial injury induced by isoproterenol in rats. Acta feeding behavior and body weight. Physiol. Behav. 92 (1–2), 263–271.
Pharmacol. Sin. 27 (5), 527–535. Mellinkoff, S.M., Frankland, M., Boyle, D., Greipel, M., 1956. Relationship between
Li, S., Kirouac, G.J., 2008. Projections from the paraventricular nucleus of the thal- serum amino acid concentration and fluctuations in appetite. J. Appl. Physiol. 8 (5),
amus to the forebrain, with special emphasis on the extended amygdala. J. Comp. 535–538.
Neurol. 506 (2), 263–287. Menaker, M., Takahashi, J.S., Eskin, A., 1978. The physiology of circadian pace-
Lin, L., Faraco, J., Li, R., et al., 1999. The sleep disorder canine narcolepsy makers. Annu. Rev. Physiol. 40, 501–526.
is caused by a mutation in the hypocretin (orexin) receptor 2 gene. Cell 98 (3), Mercer, J.G., Hoggard, N., Williams, L.M., et al., 1996. Coexpression of leptin receptor
365–376. and preproneuropeptide Y mRNA in arcuate nucleus of mouse hypothalamus.
Lin, S., Boey, D., Couzens, M., Lee, N., Sainsbury, A., Herzog, H., 2005. Compen- J. Neuroendocrinol. 8 (10), 733–735.
satory changes in [125I]-PYY binding in Y receptor knockout mice suggest the Mercer, R.E., Chee, M.J., Colmers, W.F., 2011. The role of NPY in hypothalamic
potential existence of further Y receptor(s). Neuropeptides 39 (1), 21–28. mediated food intake. Front. Neuroendocrinol. 32 (4), 398–415.
Liu, C., Li, S., Liu, T., Borjigin, J., Lin, J.D., 2007. Transcriptional coactivator PGC- Mieda, M., Williams, S.C., Sinton, C.M., Richardson, J.A., Sakurai, T., Yanagisawa, M.,
1alpha integrates the mammalian clock and energy metabolism. Nature 447 2004. Orexin neurons function in an efferent pathway of a food-entrainable
(7143), 477–481. circadian oscillator in eliciting food-anticipatory activity and wakefulness.
Liu, S., Borgland, S.L., 2015. Regulation of the mesolimbic dopamine circuit by J. Neurosci. 24 (46), 10493–10501.
feeding peptides. Neuroscience 289, 19–42. Millington, G.W., 2007. The role of proopiomelanocortin (POMC) neurones in feeding
Loos, R.J., 2011. The genetic epidemiology of melanocortin 4 receptor variants. Eur. J. behavior. Nutr. Metab. (Lond.) 4, 18.
Pharmacol. 660 (1), 156–164. Mitra, A., Katovich, M.J., Mecca, A., Rowland, N.E., 2006. Effects of central and
López, M., Seoane, L., García, M.C., Lago, F., Casanueva, F.F., Senaris, R., peripheral injections of apelin on fluid intake and cardiovascular parameters in rats.
Diéguez, C., 2000. Leptin regulation of prepro-orexin and orexin receptor mRNA Physiol. Behav. 89 (2), 221–225.
levels in the hypothalamus. Biochem. Biophys. Res. Commun. 269 (1), 41–45. Mizuno, T.M., Kleopoulos, S.P., Bergen, H.T., Roberts, J.L., Priest, C.A., Mobbs, C.V.,
López, M., Lage, R., Saha, A.K., et al., 2008. Hypothalamic fatty acid metabolism 1998. Hypothalamic pro-opiomelanocortin mRNA is reduced by fasting in ob/ob
mediates the orexigenic action of ghrelin. Cell Metab. 7 (5), 389–399. and db/db mice, but is stimulated by leptin. Diabetes 47 (2), 294–297.
Louis-Sylvestre, J., Le Magnen, J., 1980. Fall in blood glucose level precedes meal Morash, B.A., Willkinson, D., Ur, E., Wilkinson, M., 2002. Resistin expression and
onset in free-feeding rats. Neurosci. Biobehav. Rev. 4 (Suppl. 1), 13–15. regulation in mouse pituitary. FEBS Lett. 526 (1–3), 26–30.
206 Food Intake and Its Control by Signaling Molecules

Morgado, E., Juárez-Portilla, C., Silverman, A.J., Silver, R., 2015. Relevance of Nishii, N., Takasu, M., Ohba, Y., et al., 2006. Effects of administration of glucocor-
network organization in SCN clock function. In: Aguilar-Roblero, R., Díaz- ticoids and feeding status on plasma leptin concentrations in dogs. Am. J. Vet.
Muñoz, M., Fanjul-Moles, M.L. (Eds.), Mechanisms of Circadian Systems in Res. 67 (2), 266–270.
Animals and Their Clinical Relevance. New York, NY, pp. 149–175. Norgren, R., Grill, H., 1982. Brain-stem control of ingestive behavior. In: Pfaff, D. (Ed.), The
Morton, G.J., Cummings, D.E., Baskin, D.G., Barsh, G.S., Schwartz, M.W., 2006. Physiological Mechanisms of Motivation. Springer-Verlag, New York, pp. 99–131.
Central nervous system control of food intake and body weight. Nature 443 (7109), Odermatt, A., Klusonova, P., 2015. 11b-Hydroxysteroid dehydrogenase 1: regeneration of
289–295. active glucocorticoids is only part of the story. J. Steroid Biochem. 151 (3), 85–92.
Morton, N.M., Seckel, J.R., 2008. 11-beta-hydroxysteroid dehydrogenase type 1 and O’donnell, L.A., Agrawal, A., Sabnekar, P., Dichter, M.A., Lynch, D.R., Kolson, D.L.,
obesity. Front. Horm. Res. 36, 146–164. 2007. Apelin, an endogenous neuronal peptide, protects hippocampal neurons
Mountjoy, K.G., Robbins, L.S., Mortrud, M.T., Cone, R.D., 1992. The cloning of against excitotoxic injury. J. Neurochem. 102 (6), 1905–1917.
a family of genes that encode the melanocortin receptors. Science 257 (5074), Oh, I.S., Shimizu, H., Satoh, T., et al., 2006. Identification of nesfatin-1 as a satiety
1248–1251. molecule in the hypothalamus. Nature 443 (7112), 709–712.
Mountjoy, K.G., 2010. Distribution and function of melanocortin receptors within the Ollmann, M.M., Wilson, B.D., Yang, Y.K., et al., 1997. Antagonism of central mela-
brain. Adv. Exp. Med. Biol. 681, 29–48. nocortin receptors in vitro and in vivo by agouti-related protein. Science 278
Mrosovsky, N., 1990. Rheostasis: The Physiology of Change. Chapter 3: Terminology (5335), 135–138.
and Definitions. Oxford University Press, USA. Oomura, Y., Ono, T., Ooyama, H., Wayner, M.J., 1969. Glucose and osmosensitive
Mu, J., Zhuang, S.-Y., Kirby, M.T., Hampson, R.E., Deadwyler, S.A., 1999. Canna- neurones of the rat hypothalamus. Nature 222 (5190), 282–284.
binoid receptors differentially modulate potassium A and D currents in hippocampal Ovesjo, M.L., Gamstedt, M., Collin, M., Meister, B., 2001. GABAergic nature of
neurons in culture. J. Pharmacol. Exp. Ther. 291 (2), 893–902. hypothalamic leptin target neurones in the ventromedial arcuate nucleus.
Müller, T.D., Nogueiras, R., Andermann, M.L., et al., 2015. Ghrelin. Mol. Metab. 4 (6), J. Neuroendocrinol. 13 (6), 505–516.
437–460. Pajvani, U.B., Du, X., Combs, T.P., et al., 2003. Structure-function studies of the
Munro, S., Thomas, K.L., Abu-Shaar, M., 1993. Molecular characterization of adipocyte-secreted hormone Acrp30/adiponectin. Implications for metabolic
a peripheral receptor for cannabinoids. Nature 365 (6441), 61–65. regulation and bioactivity. J. Biol. Chem. 278 (11), 9073–9085.
Münzberg, H., Morrison, C.D., 2015. Structure, production and signaling of leptin. Palkovits, M., 1987. Anatomy of neural pathways affecting CRH secretion. Ann. N.Y.
Metabolism 64 (1), 13–23. Acad. Sci. 512 (1), 139–148.
Muroya, S., Yada, T., Shioda, S., Takigawa, M., 1999. Glucose-sensitive neurons in Parent, M.B., Darling, J.N., Henderson, Y.O., 2014. Remembering to eat: hippocampal
the rat arcuate nucleus contain neuropeptide Y. Neurosci. Lett. 264 (1–3), regulation of meal onset. Am. J. Physiol. Regul. Integr. Comp. Physiol. 306 (10),
113–116. R701–R713.
Muroya, S., Funahashi, H., Yamanaka, A., et al., 2004. Orexins (hypocretins) directly Park, C.R., Seeley, R.J., Benthem, L., Friedman, M.I., Woods, S.C., 1995. Whole body
interact with neuropeptide Y, POMC and glucose-responsive neurons to regulate energy expenditure and fuel oxidation after 2,5-anhydro-D-mannitol administration.
Ca2þ signaling in a reciprocal manner to leptin: orexigenic neuronal pathways in Am. J. Physiol. 268 (1 Pt 2), R299–R302.
the mediobasal hypothalamus. Eur. J. Neurosci. 19 (6), 1524–1534. Park, H.K., Ahima, R.S., 2013. Resistin in rodents and humans. Diabetes Metab. J. 37
Murphy, K.G., Bloom, S.R., 2006. Gut hormones and the regulation of energy (6), 404–414.
homeostasis. Nature 444 (7121), 854–859. Park, S., Hong, S.M., Sung, S.R., Jung, H.K., 2008. Long-term effects of central leptin
Muse, E.D., Lam, T.K., Scherer, P.E., Rossetti, L., 2007. Hypothalamic resistin induces and resistin on body weight, insulin resistance, and beta-cell function and mass by
hepatic insulin resistance. J. Clin. Invest. 117 (6), 1670–1678. the modulation of hypothalamic leptin and insulin signaling. Endocrinology 149 (2),
Myers Jr., M.G., 2010. Outstanding scientific achievement award lecture 2010: 445–454.
deconstructing leptin: from signals to circuits. Diabetes 59 (11), 2708–2714. Parker, R.M., Herzog, H., 1999. Regional distribution of Y-receptor subtype mRNAs in
Myers Jr., M.G., Cowley, M.A., Munzberg, H., 2008. Mechanisms of leptin action and rat brain. Eur. J. Neurosci. 11 (4), 1431–1448.
leptin resistance. Annu. Rev. Physiol. 70, 537–556. Patel, L., Buckels, A.C., Kinghorn, I.J., et al., 2003. Resistin is expressed in human
Nagai, K., Nagai, N., Shimizu, K., Chun, S., Nakagawa, H., Niijima, A., 1996. SCN macrophages and directly regulated by PPAR gamma activators. Biochem. Bio-
output drives the autonomic nervous system: with special reference to the auto- phys. Res. Commun. 300 (2), 472–476.
nomic function related to the regulation of glucose metabolism. Prog. Brain Res. Pedragosa-Badia, X., Stichel, J., Beck-Sickinger, A.G., February 4, 2013. Neuro-
111, 253–272. peptide Y receptors: how to get subtype selectivity. Front. Endocrinol. (Lausanne) 4
Nakahata, Y., Kaluzova, M., Grimaldi, B., et al., 2008. The NADþ-dependent (5). http://dx.doi.org/10.3389/fendo.2013.00005.
deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and Pertwee, R.G., 2005. Pharmacological actions of cannabinoids. Handbook of Experi-
circadian control. Cell 134 (2), 329–340. mental Pharmacology, vol. 168. Springer-Verlag, Heidelberg, pp. 1–51.
Nakano, Y., Tobe, T., Choi-Miura, N.H., Mazda, T., Tomita, M., 1996. Isolation and Power, M.L., Schulkin, J., 2008. Anticipatory physiological regulation in feeding
characterization of GBP28, a novel gelatin-binding protein purified from human biology: cephalic phase responses. Appetite 50 (2–3), 194–206.
plasma. J. Biochem. 120 (4), 803–812. Prager, E.M., Johnson, L.R., 2009. Stress at the synapse: signal transduction
Nakazato, M., Murakami, N., Date, Y., et al., 2001. A role for ghrelin in the central mechanisms of adrenal steroids at neuronal membranes. Sci. Signal. 2 (86), 1–10.
regulation of feeding. Nature 409 (6817), 194–198. Pritchard, L.E., Turnbull, A.V., White, A., 2002. Pro-opiomelanocortin processing in the
Nambu, T., Sakurai, T., Mizukami, K., Hosoya, Y., Yanagisawa, M., Goto, K., 1999. hypothalamus: impact on melanocortin signalling and obesity. J. Endocrinol. 172
Distribution of orexin neurons in the adult rat brain. Brain Res. 827 (1–2), (3), 411–421.
243–260. Qi, Y., Takahashi, N., Hileman, S.M., et al., 2004. Adiponectin acts in the brain to
Naveilhan, P., Neveu, I., Arenas, E., Ernfors, P., 1998. Complementary and overlapping decrease body weight. Nat. Med. 10 (5), 524–529.
expression of Y1, Y2 and Y5 receptors in the developing and adult mouse nervous Qu, D., Ludwig, D.S., Gammeltoft, S., et al., 1996. A role for melanin-concentrating
system. Neuroscience 87 (1), 289–302. hormone in the central regulation of feeding behaviour. Nature 380 (6571), 243–247.
Nawrocki, A.R., Rajala, M.W., Tomas, E., et al., 2006. Mice lacking adiponectin show Racotta, R., Islas-Chaires, M., Vega, C., Soto-Mora, M., Russek, M., 1984. Glyco-
decreased hepatic insulin sensitivity and reduced responsiveness to peroxisome genolytic substances, hepatic and systemic lactate, and food intake in rats. Am. J.
proliferator-activated receptor gamma agonists. J. Biol. Chem. 281 (5), Physiol. 246 (2 Pt 2), R247–R250.
2654–2660. Raffin-Sanson, M.L., de Keyzer, Y., Bertagna, X., 2003. Eur. J. Endocrinol. 149 (2),
Newman, E.A., Chai, B.X., Zhang, W., Li, J.Y., Ammori, J.B., Mulholland, M.W., 2006. 79–90.
Activation of the melanocortin-4 receptor mobilizes intracellular free calcium in Ramanjaneya, M., Chen, J., Brown, J.E., et al., 2010. Identification of nesfatin-1 in
immortalized hypothalamic neurons. J. Surg. Res. 132 (2), 201–207. human and murine adipose tissue: a novel depot-specific adipokine with increased
Nguyen, A.D., Mitchell, N.F., Lin, S., et al., 2012. Y1 and Y5 receptors are both levels in obesity. Endocrinology 151 (7), 3169–3180.
required for the regulation of food intake and energy homeostasis in mice. PLoS Ravussin, E., Swinburn, B.A., 1992. Pathophysiology of obesity. Lancet 340 (8816),
One 7 (6), e40191. 404–408.
Nicolaides, N.C., Kyratzi, E., Lamprokostopoulou, A., Chrousos, G.P., Charmandari, E., Reaux, A., Gallatz, K., Palkovits, M., Llorens-Cortes, C., 2002. Distribution of apelin-
2014. Stress, the stress system and the role of glucocorticoids. Neuro- synthesizing neurons in the adult rat brain. Neuroscience 113 (3), 653–662.
immunomodulation 22 (5), 6–19. Reaux-Le Goazigo, A., Bodineau, L., De Mota, N., et al., 2011. Apelin and the
Nicolaidis, S., Even, P., 1985. Physiological determinant of hunger, satiation, and proopiomelanocortin system: a new regulatory pathway of hypothalamic alpha-MSH
satiety. Am. J. Clin. Nutr. 42 (Suppl. 5), 1083–1092. release. Am. J. Physiol. (Endocrinol. Metab.) 301 (5), E955–E966.
Food Intake and Its Control by Signaling Molecules 207

Ricketts, M.M., Shoesmith, K.J., Hewison, M., Strain, A., Eggo, M.C., Stewart, P.M., Schoeller, D.A., Cella, L.K., Sinha, M.K., Caro, J.F., 1997. Entrainment of the diurnal
1998. Regulation of 11-beta-hydroxysteroid dehydrogenase type 1 in primary rhythm of plasma leptin to meal timing. J. Clin. Invest. 100 (7), 1882–1887.
culture of rat and human hepatocytes. J. Endocrinol. 156 (1), 159–168. Scholzen, T.E., Kalden, D.H., Brzoska, T., et al., 2000. Expression of proopiomela-
Rinaldi-Carmona, M., Barth, F., Heaulme, M., et al., 1994. SR141716A, a potent and nocortin peptides in human dermal microvascular endothelial cells: evidence for
selective antagonist of the brain cannabinoid receptor. FEBS Lett. 350 (2–3), 240–244. a regulation by ultraviolet light and interleukin-1. J. Invest. Dermatol. 115 (6),
Ritter, R.C., Slusser, P.G., Stone, S., 1981. Glucoreceptors controlling feeding and 1021–1028.
blood glucose: location in the hindbrain. Science 213 (4506), 451–452. Schwartz, M.W., Seeley, R.J., Campfield, L.A., Burn, P., Baskin, D.G., 1996a. Iden-
Ritter, S., Taylor, J.S., 1990. Vagal sensory neurons are required for lipoprivic but not tification of targets of leptin action in rat hypothalamus. J. Clin. Invest. 98 (5),
glucoprivic feeding in rats. Am. J. Physiol. 258 (6 Pt 2), R1395–R1401. 1101–1106.
Ritter, S., Dinh, T.T., Friedman, M.I., 1994. Induction of Fos-like immunoreactivity and Schwartz, M.W., Baskin, D.G., Bukowski, T.R., et al., 1996b. Specificity of leptin
stimulation of feeding by 2,5-anhydro-D-mannitol (2,5-AM) require the vagus action on elevated blood glucose levels and hypothalamic neuropeptide Y gene
nerve. Brain Res. 646 (1), 53–64. expression in ob/ob mouse. Diabetes 45 (4), 531–535.
Ritter, S., Llewellyn-Smith, I., Dinh, T.T., 1998. Subgroups of hindbrain catecholamine Schwartz, G.J., Moran, T.H., 1996. Sub-diaphragmatic vagal afferent integration of
neurons are selectively activated by 2-deoxy-D-glucose induced metabolic meal-related gastrointestinal signals. Neurosci. Biobehav. Rev. 20 (1), 47–56.
challenge. Brain Res. 805 (1–2), 41–54. Schwartz, M.W., Sipols, A.J., Marks, J.L., et al., 1992. Inhibition of hypothalamic
Rodrigues, A.R., Almeida, H., Gouveia, A.M., 2015. Intracelular signaling mechanisms neuropeptide Y gene expression by insulin. Endocrinology 130 (6),
of the melanocortin receptors: current state of the art. Cell. Mol. Life Sci. 72 (7), 3608–3616.
1331–1345. Schwartz, M.W., Seeley, R.J., Woods, S.C., et al., 1997. Leptin increases hypotha-
Ronveaux, C.C., Tomé, D., Raybould, H.E., 2015. Glucagon-like peptide 1 interacts lamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus.
with ghrelin and leptin to regulate glucose metabolism and food intake through Diabetes 46 (12), 2119–2123.
vagal afferent neuron signaling. J. Nutr. 145 (4), 672–680. Schwartz, M.W., Woods, S.C., Porte, D., Seeley, R.J., Baskin, D.G., 2000. Central
Roseberry, A.G., Liu, H., Jackson, A.C., Cai, X., Friedman, J.M., 2004. Neuropeptide nervous system control of food intake. Nature 404 (6778), 661–671.
Y-mediated inhibition of proopiomelanocortin neurons in the arcuate nucleus shows Sedbazar, U., Maejima, Y., Nakata, M., Mori, M., Yada, T., 2013. Paraventricular
enhanced desensitization in ob/ob mice. Neuron 41 (5), 711–722. NUCB2/nesfatin-1 rises in synchrony with feeding suppression during early light
Rosenbaum, M., Leibel, R.L., 1999. Clinical review 107: role of gonadal steroids in the phase in rats. Biochem. Biophys. Res. Commun. 434 (3), 434–438.
sexual dimorphisms in body composition and circulating concentrations of leptin. Seidah, N.G., Chretien, M., 1992. Proprotein and prohormone convertases of the
J. Clin. Endocrinol. Metab. 84 (6), 1784–1789. subtilisin family: recent developments and future perspectives. Trends Endocrinol.
Rosenwasser, A.M., Adler, N.T., 1986. Structure and function in circadian timing Metab. 3 (4), 133–140.
systems: evidence for multiple coupled circadian oscillators. Neurosci. Biobehav. Seidah, N.G., Marcinkiewicz, M., Benjannet, S., et al., 1991. Cloning and primary
Rev. 10 (4), 431–448. sequence of a mouse candidate prohormone convertase PC1 homologous to
Routh, V.H., 2002. Glucose-sensing neurons: are they physiologically relevant? PC2, Furin, and Kex2: distinct chromosomal localization and messenger RNA
Physiol. Behav. 76 (3), 403–413. distribution in brain and pituitary compared to PC2. Mol. Endocrinol. 5 (1),
Russell, C.D., Petersen, R.N., Rao, S.P., et al., 1998. Leptin expression in adipose 111–122.
tissue from obese humans: depot-specific regulation by insulin and dexametha- Seim, I., Amorim, L., Walpole, C., Carter, S., Chopin, L.K., Herington, A.C., 2010.
sone. Am. J. Physiol. 275 (3 Pt 1), E507–E515. Ghrelin gene-related peptides: multifunctional endocrine/autocrine modulators in
Russek, M., 1963. Participation of hepatic glucoreceptors in the control of intake of health and disease. Clin. Exp. Pharmacol. Physiol. 37 (1), 125–131.
food. Nature 197, 79–80. Shapiro, L., Scherer, P.E., 1998. The crystal structure of a complement-1q family
Russek, M., Cabanac, M., 1982. Régulation et contrôle en biologie. Le Presses De protein suggests an evolutionary link to tumor necrosis factor. Curr. Biol. 8 (6),
L’Unverstite Laval, Quebec. 335–338.
Sainsbury, A., Schwarzer, C., Couzens, M., et al., 2002. Y4 receptor knockout rescues Sharma, K.N., 1967. Receptor mechanisms in the alimentary tract: their excitation and
fertility in ob/ob mice. Gene Dev. 16 (9), 1077–1088. functions. In: Code, C.F. (Ed.), Handbook of Physiology, Section 6: Alimentary
Sakata, I., Park, W.M., Walker, A.K., et al., 2012. Glucose-mediated control of ghrelin Canal. Williams & Wilkins, Baltimore, pp. 225–237.
release from primary cultures of gastric mucosal cells. Am. J. Physiol. Endocrinol. Shi, S-q., Ansari, T., McGuinness, O.P., Wasserman, D.H., Johnson, C.H., 2013.
Metab. 302 (10), E1300–E1310. Circadian disruption leads to insulin resistance and obesity. Curr. Biol. 23 (5),
Sakurai, T., Amemiya, A., Ishii, M., et al., 1998. Orexins and orexin receptors: a family 372–381.
of hypothalamic neuropeptides and G protein-coupled receptors that regulate Shimizu, H., Oh-, I.S., Hashimoto, K., et al., 2009. Peripheral administration of
feeding behaviour. Cell 92 (4), 573–585. nesfatin-1 reduces food intake in mice: the leptin-independent mechanism.
Saladin, R., De Vos, P., Guerre-Millo, M., et al., 1995. Transient increase in obese Endocrinology 150 (2), 662–671.
gene expression after food intake or insulin administration. Nature 377 (6549), Shimizu, S., Nakamachi, T., Konno, N., Matsuda, K., 2014. Orexin A enhances food
527–529. intake in bullfrog larvae. Peptides 59, 79–82.
Salehi, A., de La Cour, C.D., Hakanson, R., Lundquist, I., 2004. Effects of ghrelin on Shin, A.C., Zheng, H., Berthoud, H.R., 2009. An expanded view of energy homeo-
insulin and glucagon secretion: a study of isolated pancreatic islets and intact stasis: neural integration of metabolic, cognitive, and emotional drives to eat.
mice. Regul. Peptides 118 (3), 143–150. Physiol. Behav. 97 (5), 572–580.
Sánchez-Lasheras, C., Konner, A.C., Bruning, J.C., 2010. Integrative neurobiology of Shrestha, Y.B., Wickwire, K., Giraudo, S.Q., 2009. Direct effects of nutrients,
energy homeostasis-neurocircuits, signals and mediators. Front. Neuroendocrinol. acetylcholine, CCK, and insulin on ghrelin release from the isolated stomachs of
31 (1), 4–15. rats. Peptides 30 (6), 1187–1191.
Saper, C.B., Loewy, A.D., Swanson, L.W., Cowan, W.M., 1976. Direct hypothalamo- Shutter, J.R., Graham, M., Kinsey, A.C., Scully, S., Lüthy, R., Stark, K.L., 1997.
autonomic connections. Brain Res. 117 (2), 305–312. Hypothalamic expression of ART, a novel gene related to agouti, is up-regulated in
Saper, C.B., Loewy, A.D., 1980. Efferent connections of the parabrachial nucleus in obese and diabetic mutant mice. Genes Dev. 11 (5), 593–602.
the rat. Brain Res. 197 (2), 291–317. Siegel, J.M., 2004. Hypocretin (OREXIN): role in normal behavior and neuropathology.
Saper, C.B., Scammell, T.E., Lu, J., 2005. Hypothalamic regulation of sleep and Annu. Rev. Psychol. 55 (1), 125–148.
circadian rhythms. Nature 437 (7063), 1257–1263. Singhal, N.S., Lazar, M.A., Ahima, R.S., 2007. Central resistin induces hepatic insulin
Savage, D.B., Sewter, C.P., Klenk, E.S., et al., 2001. Resistin/Fizz3 expression in resistance via neuropeptide Y. J. Neurosci. 27 (47), 12924–12932.
relation to obesity and peroxisome proliferator-activated receptor-gamma action in Sinha, M.K., Opentanova, I., Ohannesian, J.P., et al., 1996. Evidence of free and
humans. Diabetes 50 (10), 2199–2202. bound leptin in human circulation. Studies in lean and obese subjects and during
Scharrer, E., 1999. Control of food intake by fatty acid oxidation and ketogenesis. short-term fasting. J. Clin. Invest. 98 (6), 1277–1282.
Nutrition 15 (9), 704–714. Smith, P.M., Ferguson, A.V., 2008. Neurophysiology of hunger and satiety. Dev.
Schauer, E., Trautinger, F., Kock, A., et al., 1994. Proopiomelanocortin-derived Disabil. Res. Rev. 14 (2), 96–104.
peptides are synthesized and released by human keratinocytes. J. Clin. Invest. Sominsky, L., Spencer, S.J., 2014. Eating behavior and stress: a pathway to obesity.
93 (5), 2258–2262. Front. Psychol. 5 (434), 1–8.
Scherer, P.E., Williams, S., Fogliano, M., Baldina, G., Lodish, H.F., 1995. A novel Soria-Gómez, E., Massa, F., Bellocchio, L., et al., 2014. Cannabinoid type-1 receptors
serum protein similar to C1q, produced exclusively in adipocytes. J. Biol. Chem. in the paraventricular nucleus of the hypothalamus inhibit stimulated food intake.
270 (45), 26746–26749. Neuroscience 263, 46–53.
208 Food Intake and Its Control by Signaling Molecules

Srinivasan, S., Shariff, M., Bartlett, S.E., 2013. The role of the glucocorticoids in Tung, Y.C., Piper, S.J., Yeung, D., O’Rahilly, S., Coll, A.,P., 2006. A comparative study
developing resilience to stress and addiction. Front. Psychiatry 4 (68), 1–11. of the central effects of specific proopiomelancortin (POMC)-derived melanocortin
Stanley, B.G., Leibowitz, S.F., 1985. Neuropeptide Y injected in the paraventricular peptides on food intake and body weight in pomc null mice. Endocrinology 147
hypothalamus: a powerful stimulant of feeding behavior. Proc. Natl. Acad. Sci. (12), 5940–5947.
U.S.A. 82 (11), 3940–3943. Turek, F.W., Joshu, C., Kohsaka, A., et al., 2005. Obesity and metabolic syndrome in
Stellar, E., 1954. The physiology of motivation. Psychol. Rev. 61 (1), 5–22. circadian clock mutant mice. Science 308 (5724), 1043–1045.
Stengel, A., Goebel, M., Yakubov, I., et al., 2009. Identification and characterization of Turer, A.T., Khera, A., Ayers, C.R., et al., 2011. Adipose tissue mass and location
nesfatin-1 immunoreactivity in endocrine cell types of the rat gastric oxyntic affect circulating adiponectin levels. Diabetologia 54 (10), 2515–2524.
mucosa. Endocrinology 150 (11), 232–238. Turer, A.T., Scherer, P.E., 2012. Adiponectin: mechanistic insights and clinical
Stengel, A., Hofmann, T., Goebel-Stengel, M., et al., 2013a. Ghrelin and NUCB2/ implications. Diabetologia 55 (9), 2319–2326.
nesfatin-1 are expressed in the same gastric cell and differentially correlated van den Top, M., Lee, K., Whyment, A.D., Blanks, A.M., Spanswick, D., 2004.
with body mass index in obese subjects. Histochem. Cell. Biol. 139 (6), Orexigen-sensitive NPY/AgRP pacemaker neurons in the hypothalamic arcuate
909–918. nucleus. Nat. Neurosci. 7 (5), 493–494.
Stengel, A., Mori, M., Tache, Y., 2013b. The role of nesfatin-1 in the regulation of food Valassi, E., Scacchi, M., Cavagnini, F., 2008. Neuroendocrine control of food intake.
intake and body weight: recent developments and future endeavors. Obesity Rev. Nutr. Metab. Cardiovasc. Dis. 18 (2), 158–168.
14 (11), 859–870. Vázquez, M.J., González, C.R., Varela, L., et al., 2008. Central resistin regulates
Stengel, A., Tache, Y., 2013. Role of NUCB2/Nesfatin-1 in the hypothalamic control of hypothalamic and peripheral lipid metabolism in a nutritional-dependent fashion.
energy homeostasis. Horm. Metab. Res. 45 (13), 975–979. Endocrinology 149 (9), 4534–4543.
Stephan, F.K., 1983. Circadian rhythm dissociation induced by periodic feeding in rats Veldhuis, J.D., Bowers, C.Y., 2010. Integrating GHS into the ghrelin system, 879503
with suprachiasmatic lesions. Behav. Brain Res. 7 (1), 81–98. Int. J. Peptides. http://dx.doi.org/10.1155/2010/879503. Epub Mar 18, 2010.
Stephan, F.K., 1989. Entrainment of activity to multiple feeding times in rats with Verty, A.N., Allen, A.M., Oldfield, B.J., 2009. The effects of rimonabant on brown
suprachiasmatic lesions. Physiol. Behav. 46 (3), 489–497. adipose tissue in rat: implications for energy expenditure. Obesity (Silver Spring) 17
Steppan, C.M., Bailey, S.T., Bhat, S., et al., 2001. The hormone resistin links obesity (2), 254–261.
to diabetes. Nature 409 (6818), 307–312. Vindrola, O., Lindberg, I., 1992. Biosynthesis of the prohormone convertase mPC1 in
Stieg, M.R., Sievers, C., Farr, O., Stalla, G.K., Mantzoros, C.S., 2015. Leptin: AtT-20 cells. Mol. Endocrinol. 6 (7), 1088–1094.
a hormone linking activation of neuroendocrine axes with neuropathology. Psy- Voisin, T., Rouet-Benzineb, P., Reuter, N., Laburthe, M., 2003. Orexins and their
choneuroendocrinology 51, 47–57. receptors: structural aspects and role in peripheral tissues. Cell. Mol. Life Sci. 60
Sun, Y., Ahmed, S., Smith, R.G., 2003. Deletion of ghrelin impairs neither growth nor (1), 72–87.
appetite. Mol. Cell Biol. 23 (22), 7973–7981. Wajnrajch, M.I., Gertner, J., Leibel, R., 2000. Genomic organization of the ghrelin
Sunter, D., Hewson, A.K., Dickson, S.L., 2003. Intracerebroventricular injection of gene. J. Endocr. Genet. 1 (2), 231–233.
apelin-13 reduces food intake in the rat. Neurosc. Lett. 353 (1), 1–4. Wang, Q., Liu, C., Uchida, A., et al., 2013. Arcuate AgRP neurons mediate orexigenic
Suzuki, R., Shimojima, H., Funahashi, H., et al., 2002. Orexin-1 receptor immuno- and glucoregulatory actions of ghrelin. Mol. Metab. 3 (1), 64–72.
reactivity in chemically identified target neurons in the rat hypothalamus. Neurosc. Watts, A.G., Swanson, L.W., 1987. Efferent projections of the suprachiasmatic
Lett. 324 (1), 5–8. nucleus: II. Studies using retrograde transport of fluorescent dyes and simul-
Swaab, D.F., Hofman, M.A., Lucassen, P.J., Purba, J.S., Raadsheer, F.C., Van de taneous peptide immunohistochemistry in the rat. J. Comp. Neurol. 258 (2),
Nes, J.A., 1993. Functional neuroatonomy and neuropathology of the human 230–252.
hypothalamus. Anat. Embryol. (Berl.) 187 (4), 317–330. Watts, A.G., Swanson, L.W., Sanchez-Watts, G., 1987. Efferent projections of the
Switonski, M., Mankowska, M., Salamon, S., 2013. Family of melanocortin receptor suprachiasmatic nucleus: I. Studies using anterograde transport of Phaseolus
(MCR) genes in mammals-mutations, polymorphisms and phenotypic effects. J. vulgaris leucoagglutinin in the rat. J. Comp. Neurol. 258 (2), 204–229.
Appl. Genet. 54 (4), 461–472. Weiss, G.F., Leibowitz, S.F., 1985. Efferent projections from the paraventricular
Tabuchi, S., Tsunematsu, T., Black, S.W., et al., 2014. Conditional ablation of orexin/ nucleus mediating a2-noradrenergic feeding. Brain Res. 347 (2), 225–238.
hypocretin neurons: a new mouse model for the study of narcolepsy and orexin Wells, T., 2009. Ghrelin-defender of fat. Prog. Lipid Res. 48 (5), 257–274.
system function. J. Neurosci. 34 (19), 6495–6509. Willesen, M.G., Kristensen, P., Romer, J., 1999. Co-localization of growth hormone
Taheri, S., Murphy, K., Cohen, M., et al., 2002. The effects of centrally administered secretagogue receptor and NPY mRNA in the arcuate nucleus of the rat. Neuro-
apelin-13 on food intake, water intake and pituitary hormone release in rats. endocrinology 70 (5), 306–316.
Biochem. Biophys. Res. Commun. 291 (5), 1208–1212. Williams, C.M., Rogers, P.J., Kirkham, T.C., 1998. Hyperphagia in pre-fed rats
Tartaglia, L.A., Dembski, M., Weng, X., et al., 1995. Identification and expression following oral delta9-THC. Physiol. Behav. 65 (2), 343–346.
cloning of a leptin receptor, OB-R. Cell 83 (7), 1263–1271. Williams, C.M., Kirkham, T.C., 1999. Anandamide induces overeating: mediation by
Tatemoto, K., Carlquist, M., Mutt, V., 1982. Neuropeptide Y a novel brain peptide with central cannabinoid (CB1) receptors. Psychopharmacol. (Berl.) 143 (3), 315–317.
structural similarities to peptide YY and pancreatic polypeptide. Nature 296 (5858), Williams, G., Harrold, J.A., Cutler, D.J., 2000. The hypothalamus and the regulation of
659–660. energy homeostasis: lifting the lid on a black box. Proc. Nutr. Soc. 59 (3),
Tatemoto, K., Hosoya, M., Habata, Y., et al., 1998. Isolation and characterization of 385–396.
a novel endogenous peptide ligand for the human APJ receptor. Biochem. Biophys. Wilson, B.D., Bagnol, D., Kaelin, C.B., et al., 1999. Physiological and anatomical
Res. Commun. 251 (2), 471–476. circuitry between agouti-related protein and leptin signaling. Endocrinology 140 (5),
Tatemoto, K., Takayama, K., Zou, M.X., et al., 2001. The novel peptide apelin lowers 2387–2397.
blood pressure via a nitric oxide-dependent mechanism. Regul. Peptides 99 (2–3), Wintzen, M., Yaar, M., Burbach, J.P., Gilchrest, B.A., 1996. Proopiomelanocortin gene
87–92. product and regulation in keratinocytes. J. Invest. Dermatol. 106 (4), 673–678.
Thorens, B., 2015. GLUT2, glucose sensing and glucose homeostasis. Diabetologia 58 Wirth, M.M., Giraudo, S.Q., 2000. Agouti-related protein in the hypothalamic para-
(2), 221–232. ventricular nucleus: effect on feeding. Peptides 21 (9), 1369–1375.
Thornton, J.E., Cheung, C.C., Clifton, D.K., Steiner, R.A., 1997. Regulation of hypo- Woods, S.C., 1991. The eating paradox: how we tolerate food. Psychol. Rev. 98 (1),
thalamic proopiomelanocortin mRNA by leptin in ob/ob mice. Endocrinology 138 488–505.
(11), 5063–5066. Woods, S.C., Seeley, R.J., Porte, D., Schwartz, M.W., 1998. Signals that regulate food
Tomaru, T., Steger, D.J., Lefterova, M.I., Schupp, M., Lazar, M.A., 2009. Adipocyte- intake and energy homeostasis. Science 280 (5368), 1378–1383.
specific expression of murine resistin is mediated by synergism between peroxi- Woods, S.C., Lutz, T.A., Geary, N., Langhans, W., 2006. Pancreatic signals controlling
some proliferator-activated receptor gamma and CCAAT/enhancer-binding food intake; insulin, glucagon and amylin. Philos. Trans. R. Soc. Lond. Ser. B Biol.
proteins. J. Biol. Chem. 284 (10), 6116–6125. Sci. 361 (1471), 1219–1235.
Tovar, S., Nogueiras, R., Tung, L.Y., et al., 2005. Central administration of resistin Wortley, K.E., Anderson, K.D., García, K., et al., 2004. Genetic deletion of ghrelin does
promotes short-term satiety in rats. Eur. J. Endocrinol. 153 (5), R1–R5. not decrease food intake but influences metabolic fuel preference. Proc. Natl.
Trivedi, P., Yu, H., MacNeil, D.J., Van der Ploeg, L.H., Guan, X.M., 1998. Distribution Acad. Sci. U.S.A. 101 (21), 8227–8232.
of orexin receptor mRNA in the rat brain. FEBS Lett. 438 (1–2), 71–75. Wortley, K.E., del Rincón, J.P., Murray, J.D., et al., 2005. Absence of ghrelin protects
Tschöp, M., Devanarayan, V., Weyer, C., Tataranni, P.A., Ravussin, E., Heiman, M.L., against early-onset obesity. J. Clin. Invest. 115 (12), 3573–3578.
2001. Circulating ghrelin levels are decreased in human obesity. Diabetes 50 (4), Wren, A.M., Small, C.J., Abbott, C.R., et al., 2001. Ghrelin causes hyperphagia and
707–709. obesity in rats. Diabetes 50 (11), 2540–2547.
Food Intake and Its Control by Signaling Molecules 209

Wynne, K., Stanley, S., McGowan, B., Bloom, S., 2005. Appetite control. J. Endocrinol. Yildiz, B.O., Suchard, M.A., Wong, M.L., McCann, S.M., Licinio, J., 2004. Alterations
184 (2), 291–318. in the dynamics of circulating ghrelin, adiponectin, and leptin in human obesity.
Yamanaka, A., Beuckmann, C.T., Willie, J.T., et al., 2003. Hypothalamic orexin Proc. Natl. Acad. Sci. U.S.A. 101 (28), 10434–10439.
neurons regulate arousal according to energy balance in mice. Neuron 38 (5), Yin, Y., Li, Y., Zhang, W., 2014. The growth hormone secretagogue receptor: its
701–713. intracellular signaling and regulation. Int. J. Mol. Sci. 15 (3), 4837–4855.
Yamauchi, T., Kamon, J., Waki, H., et al., 2001. The fat-derived hormone adiponectin Yokobori, E., Kojima, K., Azuma, M., Kang, K.S., Maejima, S., Uchiyama, M.,
reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. Matsuda, K., 2011. Stimulatory effect of intracerebroventricular administration of
7 (8), 941–946. orexin A on food intake in the zebrafish, Danio rerio. Peptides 32 (7), 1357–1362.
Yamauchi, T., Kamon, J., Ito, Y., et al., 2003. Cloning of adiponectin receptors that Yosten, G.L., Samson, W.K., 2009. Nesfatin-1 exerts cardiovascular actions in brain:
mediate antidiabetic metabolic effects. Nature 423 (6941), 762–769. possible interaction with the central melanocortin system. Am. J. Physiol. (Regul.
Yamauchi, T., Kadowaki, T., 2013. Adiponectin receptor as a key player in healthy Integr. Comp. Physiol.) 297 (2), R330–R336.
longevity and obesity-related diseases. Cell Metab. 17 (2), 185–196. Yosten, G.L., Samson, W.K., 2010. The anorexigenic and hypertensive effects of
Yamauchi, T., Iwabu, M., Okada-Iwabu, M., Kadowaki, T., 2014. Adiponectin recep- nesfatin-1 are reversed by pretreatment with an oxytocin receptor antagonist. Am.
tors: a review of their structure, function and how they work. Best Pract. Res. Clin. J. Physiol. (Regul. Integr. Comp. Physiol.) 298 (6), R1642–R1647.
Endocrinol. Metab. 28 (1), 15–23. Zarjevski, N., Cusin, I., Vettor, R., Rohner-Jeanrenaud, F., Jeanrenaud, B., 1993. Chronic
Yamanaka, A., Kunii, K., Nambu, T., et al., 2000. Orexin-induced food intake involves intracerebroventricular neuropeptide-Y administration to normal rats mimics
neuropeptide Y pathway. Brain Res. 859 (2), 404–409. hormonal and metabolic changes of obesity. Endocrinology 133 (4), 1753–1758.
Yang, J., Brown, M.S., Liang, G., Grishin, N.V., Goldstein, J.L., 2008. Identification of Zeng, X.J., Yu, S.P., Zhang, L., Wei, L., 2010. Neuroprotective effect of the endog-
the acyltransferase that octanoylates ghrelin, an appetite-stimulating peptide enous neural peptide apelin in cultured mouse cortical neurons. Exp. Cell Res. 316
hormone. Cell 132 (3), 387–396. (11), 1773–1783.
Yang, Y., Atasoy, D., Su, H.H., Sternson, S.M., 2011. Hunger states switch a flip-flop Zhang, F., Basinski, M.B., Beals, J.M., et al., 1997. Crystal structure of the obese
memory circuit via a synaptic AMPK-dependent positive feedback loop. Cell 146 protein leptin-E100. Nature 387 (6629), 206–209.
(6), 992–1003. Zhang, Y., Proenca, R., Maffei, M., Barone, M., Leopold, L., Friedman, J.M., 1994.
Yang, Y.J., Lv, S.Y., Xiu, M.H., Xu, N., Chen, Q., 2010. Intracerebroventricular Positional cloning of the mouse obese gene and its human homologue. Nature 372
administration of apelin-13 inhibits distal colonic transit in mice. Peptides 31 (12), (6505), 425–432.
2241–2246. Zheng, H., Berthoud, H.R., 2007. Eating for pleasure or calories. Curr. Opin.
Yang, Y.-K., Thompson, D.A., Dickinson, C.J., et al., 1999. Characterization of agouti- Pharmacol. 7 (5), 607–612.
related protein binding to melanocortin receptors. Mol. Endocrinol. 13 (1), 148–155. Zheng, H., Lenard, N.R., Shin, A.C., Berthoud, H.R., 2009. Appetite control and energy
Yaswen, L., Diehl, N., Brennan, M.B., Hochgeschwender, U., 1999. Obesity in the balance regulation in the modern world: reward-driven brain overrides repletion
mouse model of pro-opiomelanocortin deficiency responds to peripheral melano- signals. Int. J. Obesity (Lond.) 33 (Suppl. 2), S8–S13.
cortin. Nat. Med. 5 (9), 1066–1070. Zhou, A., Bloomquist, B.T., Mains, R.E., 1993. The prohormone convertases PC1 and
Ye, R., Scherer, P.E., 2013. Adiponectin, driver or passenger on the road to insulin PC2 mediate distinct endoproteolytic cleavages in a strict temporal order during
sensitivity? Mol. Metab. 2 (3), 133–141. proopiomelanocortin biosynthetic processing. J. Biol. Chem. 268 (3), 1763–1769.
Yi, C.X., Serlie, M.J., Ackermans, M.T., et al., 2009. A major role for perifornical orexin Zigman, J.M., Nakano, Y., Coppari, R., et al., 2005. Mice lacking ghrelin receptors resist
neurons in the control of glucose metabolism in rats. Diabetes 58 (9), 1998–2005. the development of diet-induced obesity. J. Clin. Invest. 115 (12), 3564–3572.
This page intentionally left blank
1.07 Body Fluid Homeostasis
Eric G Krause, College of Pharmacy, University of Florida, Gainesville, FL, USA
Annette D de Kloet, College of Medicine, University of Florida, Gainesville, FL, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by D. Daniels, S.J. Fluharty, volume 1, pp. 259–289, Ó 2009, Elsevier Inc.

1.07.1 Introduction 211


1.07.2 In Defense of Osmolality 211
1.07.2.1 Vasopressin and OT 211
1.07.2.2 Thirst 212
1.07.2.3 Peripheral Osmoreceptors 213
1.07.2.4 Central Osmoreceptors 213
1.07.2.5 Sodium Deficit/Reduced Osmolality 213
1.07.3 In Defense of Volume 214
1.07.3.1 The Renin–Angiotensin–Aldosterone System 214
1.07.3.2 Hypovolemic Thirst 215
1.07.3.3 Salt Appetite 216
1.07.3.3.1 Aldosterone and Salt Appetite 217
1.07.3.3.2 Angiotensin-II and Salt Appetite 217
1.07.3.3.3 Angiotensin/Aldosterone Interactions 218
1.07.3.3.4 Baroreceptors, Osmoreceptors, and Salt Appetite 219
1.07.3.3.5 Taste and Salt Appetite 219
1.07.3.4 Blood Pressure 219
1.07.3.5 Hypervolemia 220
1.07.4 Body Fluid Homeostasis and Psychological Stress 220
1.07.5 Summary and Conclusion 221
References 221

1.07.1 Introduction 1.07.2 In Defense of Osmolality

Maintaining body fluid homeostasis requires neural and endo- In most mammals, extracellular fluid osmolality is tightly regu-
crine control of behavioral, cardiovascular, and renal responses lated near 290 mOsm, and this maintains optimal water distri-
that adjust the osmolality and volume of body fluid to levels bution between vascular, interstitial, and intracellular fluid
that are optimal for physiological function. Perturbations to compartments. Solutes that elicit the movement of water across
osmolality and volume are detected by sensors that relay the phospholipid bilayer that comprises the semipermeable
imbalance to the appropriate effector organs, which in turn, membrane separating the intracellular and extracellular fluid
initiate compensatory responses that maintain or restore blood are capable of generating ‘osmotic pressure’ that activates
pressure and hydromineral balance. Ultimately, these compen- osmoreceptors located within the brain and periphery. Osmor-
satory responses are orchestrated by the brain, which utilizes eceptors are thought to be mechanoreceptors that detect the
endocrine signals that exert effects on target organs expressing swelling or shrinkage of specialized neurons during osmotic
receptors that promote precise imposition of appropriate challenges. These ‘osmosensing neurons’ detect changes in
responding. osmolality and, subsequently, transduce this information
Challenges to body fluid homeostasis can result in concom- into action potentials relayed to effector brain nuclei that
itant changes in plasma osmolality and effective circulating initiate responses alleviating such perturbations and restore
volume; however, plasma osmolality and volume are regulated osmolality to homeostatic levels (Figure 1).
independently by distinct behavioral and endocrine responses.
Plasma osmolality is maintained by changes in water balance,
1.07.2.1 Vasopressin and OT
accomplished by thirst, water intake, and activation of the argi-
nine vasopressin (AVP)/oxytocin (OT) renal axes. Conversely, In humans and rodents, a 1–2% increase in systemic plasma
blood volume and pressure is largely maintained by activation osmolality causes the release of AVP and OT into the systemic
of the renin–angiotensin–aldosterone system (RAAS) which circulation and also causes the sensation of thirst (Rennke and
promotes vasoconstriction as well as sodium conservation Denker, 2010). Magnocellular neurons within the supraoptic
and consumption. Here, we review the neurohumoral systems (SON) and paraventricular (PVN) nuclei of the hypothalamus
that control behavioral, cardiovascular, and renal compensa- send their axons to the median eminence and posterior pitui-
tory mechanisms that maintain body fluid homeostasis. tary and release AVP and OT into the systemic circulation.

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00007-9 211


212 Body Fluid Homeostasis

Figure 1 Response to hyperosmotic (intracellular) dehydration. ACC, anterior cingulate cortex; MnPO, median preoptic nucleus; PVN, paraventricular
nucleus of the hypothalamus; SON, supraoptic nucleus.

Elevated circulating AVP binds to V2 receptors in the kidney increases in plasma osmolality elicit activation of OVLT
that initiate a Gs-mediated intracellular signaling cascade that neurons that likely synapse onto PVN neurons to evoke their
results in the insertion of aquaporin 2 channels into the apical excitation and the release of AVP and OT into the systemic
membrane of cells within the collecting tubule. These aqua- circulation (Shi et al., 2008). Consistent with this idea, lesion
porin channels allow the reabsorption of water, which of the OVLT attenuates the AVP and OT secretion that occurs
decreases urine output but increases urine osmolality. The with increased plasma osmolality in dogs, rats, or sheep
release of OT into the systemic circulation elicits activation of (Thrasher and Keil, 1987; Negoro et al., 1988; McKinley
OT receptors that promote natriuresis or renal sodium excre- et al., 2003). In addition to the SON and PVN, the OVLT is con-
tion (Verbalis et al., 1991; Sjoquist et al., 1999). In the end, nected to the MnPO, which has robust efferent projections to
the renal actions of AVP and OT allow the conservation of the SON that synapse onto AVP neurons (Oldfield et al.,
body water and the excretion of solutes in urine, which buffer 1991). Interestingly, brain lesions that encompass both the
against further elevations in plasma osmolality. OVLT and MnPO result in greater impairments to AVP secre-
The AVP and OT secretion that follows increased plasma tion relative to lesions confined to the OVLT (Johnson et al.,
osmolality is believed to be controlled by osmosensing 1978; Thrasher and Keil, 1987), suggesting that the MnPO
neurons within the organum vasculosum of the lamina termi- also contributes to the defense of body fluid homeostasis
nalis (OVLT). The OVLT is a circumventricular organ residing that occurs subsequent to osmotic challenges.
near the ventral surface of the forebrain. Circumventricular
organs lack a blood–brain barrier, and this allows the move-
1.07.2.2 Thirst
ment of water in or out of OVLT neurons during decreases or
increases in plasma osmolality, respectively (McKinley et al., While activation of neuroendocrine axes buffer against further
2003; Ciura et al., 2011). The activity of OVLT neurons is elevations in plasma osmolality, it is the sensation of thirst and
tightly coupled to the osmolality of the extracellular fluid, the resulting water consumption that ultimately dilutes the
and increasing concentrations elicit depolarization and plasma osmolality and restores its concentration to homeo-
increased action potential frequency (Ciura and Bourque, static levels. In this regard, the OVLT also plays a prominent
2006). Neurons in the OVLT have direct axonal projections role in the thirst and water intake that results from elevated
to the SON, PVN, and median preoptic nucleus (MnPO) plasma osmolality. Lesion of the OVLT decreases water
(Johnson and Gross, 1993; Bourque, 2008), and these connec- consumption subsequent to a hyperosmotic challenge
tions are believed to constitute a neurosecretory circuit that (Thrasher and Keil, 1987; McKinley et al., 2003). Similar to
controls the systemic release of AVP and OT. Evidence support- neuroendocrine secretion, lesions including the MnPO reduce
ing this idea comes from neuroanatomical and lesion studies osmotically induced water intake to a greater extent than those
demonstrating functional connections between these brain specifically targeting the OVLT (Johnson et al., 1978), indi-
nuclei that are sensitive to changes in osmolality. For example, cating that the MnPO may serve as a relay center within brain
hyperosmotic stimulation elicits activation of OVLT neurons circuits controlling drinking behavior. However, the OVLT
with monosynaptic connections to the PVN, suggesting that also has projections to thalamic and cortical nuclei, and these
Body Fluid Homeostasis 213

brain regions likely control the perception of thirst (Bourque, to changes in osmolality (Adachi et al., 1976; Adachi, 1984),
2008; Hollis et al., 2008). In addition to the SON, PVN, and and transection of the hepatic vagus nerve suppresses AVP
MnPO, the OVLT sends direct efferent projections to various release when osmolality is elevated via infusion of hypertonic
thalamic nuclei, the anterior cingulate cortex (ACC), and saline into the hepatic portal vein (Chwalbinska-Moneta,
insular cortex (Hollis et al., 2008). In particular, the ACC has 1979). Similar to AVP secretion, water intake stimulated by
been implicated in the generation of thirst because electrical intragastric infusion of hypertonic saline is reduced by
stimulation of the ACC in conscious monkeys elicits drinking vagotomy, indicating that the vagal afferent signals are capable
behavior within seconds of onset (Robinson and Mishkin, of influencing both behavioral and endocrine responses to
1968) though ablation of the ACC does not completely abolish increased osmolality (Starbuck et al., 2002). Thus, peripheral
water intake (Hollis et al., 2008). Functional magnetic reso- osmoreceptors monitor the concentration of fluid within
nance imaging studies show that the lamina terminalis and discrete vascular beds and use ascending vagal signals to prime
ACC are robustly activated in humans rendered thirsty via the brain for impending alterations in systemic osmolality.
delivery of intravenous hypertonic saline (Egan et al., 2003).
Remarkably, when these subjects drank water to satiation, the
1.07.2.4 Central Osmoreceptors
perception of thirst quickly subsided as did the intensity of
ACC activation; however, the plasma osmolality and activation The molecular identity of the peripheral osmoreceptor is
of the lamina terminalis persisted (Egan et al., 2003). Taken largely unexplored. In contrast, the molecular mechanism
together, these results implicate the OVLT and MnPO as sensors underlying central osmosensing is an active area of research,
that detect osmotic stimuli that initiate thirst, the perception of and investigations have centered on ion channels gated by
which arises within cortical brain nuclei, like the ACC. changes in osmolality. Specifically, the transient receptor
potential vanilloid 1 receptors (TRPV1) are cation channels
that have been implicated in osmosensing because TRPV1
1.07.2.3 Peripheral Osmoreceptors
knockout mice have impaired thirst and AVP responses subse-
It is interesting that drinking water relieves the sensation of quent to elevated plasma osmolality (Ciura and Bourque,
thirst and eliminates the activation of the ACC but not the 2006; Sharif Naeini et al., 2006). The increased action poten-
OVLT, and this occurs prior to resolving the elevated plasma tial discharge that occurs in OVLT neurons subjected to hyper-
osmolality (Egan et al., 2003). In other words, drinking water osmolality is absent in TRPV1 knockout mice, further
causes the perception of thirst to subside before plasma osmo- substantiating that TRPV1 is the molecular mechanism under-
lality is lowered and brain osmoreceptors are deactivated. This lying central osmosensing (Ciura and Bourque, 2006).
phenomenon suggests the existence of peripheral osmorecep- However, subsequent studies reported that TRPV1 knockout
tors that anticipate the effects of fluid consumption and, subse- mice and wild-type mice have similar increases in plasma
quently, elicit compensatory changes that prepare the organism osmolality, water intake, and activation of neurons residing
for subsequent increases or decreases in plasma osmolality. in the OVLT and MnPO subsequent to cellular dehydration
Indeed, in the periphery, osmoreceptors are expressed in the elicited by administration of hypertonic saline or mannitol
oropharyngeal cavity (Kuramochi and Kobayashi, 2000), (Taylor et al., 2008; Kinsman et al., 2014). Moreover, TRPV1
gastrointestinal tract (Dooley and Valenzuela, 1984; Carlson reporter mice that have had the TRPV1 genetic locus modified
et al., 1997; Andersen et al., 2000), splanchnic mesentery to accurately report the functional expression of TRPV1 gene
(Choi-Kwon and Baertschi, 1991), and hepatic portal vein products do not demonstrate the presence of these channels
(Baertschi and Vallet, 1981). In regard to the hepatic portal in brain nuclei implicated in osmosensing (Cavanaugh
vein, delivering intragastric concentrated saline to rats signifi- et al., 2011). Collectively, these studies indicate that the iden-
cantly increases portal venous osmolality several minutes tity of the central osmosensor is controversial and additional
before circulating plasma osmolality is elevated, and this effect research is required to resolve underlying mechanisms.
is associated with increased AVP secretion and neuronal activa-
tion of brain networks implicated in the regulation of body
1.07.2.5 Sodium Deficit/Reduced Osmolality
fluid osmolality (Carlson et al., 1997). Thus, peripheral osmor-
eceptors can prime the brain to respond to an osmotic chal- As may be expected, decreases in plasma osmolality also stim-
lenge prior to actual elevations in circulating plasma ulate osmoreceptors residing in the brain and periphery, but
osmolality. Inversely, in both osmotically dehydrated humans such stimulation inhibits rather than activates renal and
and rats, consuming water or gastric infusion of water rapidly behavioral mechanisms that promote thirst and water reten-
lowers plasma levels of AVP and OT prior to restoring plasma tion. It is important to note that decreased fluid osmolality
osmolality (Geelen et al., 1984; Baertschi and Pence, 1995; is often the result of lowering the plasma sodium concentra-
Huang et al., 2000). Taken together, these studies indicate the tion, which is a strong determinant of blood volume.
existence of peripheral osmoreceptors that detect the concen- Lowering of the plasma sodium concentration is often
tration of fluids within specific viscera and, subsequently, coupled to a reduction in blood volume and decreased perfu-
convey this information to brain circuits controlling body fluid sion pressure, which increases the activity of the RAAS, which
homeostasis through ascending afferent signals. In this regard, in turn, initiates physiological and behavioral compensatory
several lines of evidence suggest the vagus, cranial nerve X, is responses that restore blood volume and perfusion pressure
the afferent connection that relays signals from peripheral to homeostatic levels. These compensatory responses and
osmoreceptors to the CNS. For example, the hepatic branch their underlying neural and humoral mechanisms are dis-
of the vagus nerve increases or decreases firing rate in response cussed in the subsequent sections.
214 Body Fluid Homeostasis

1.07.3 In Defense of Volume promote vasoconstriction and water reabsorption (Aoyagi


et al., 2009; Sampey et al., 1999). Furthermore, this reduced
Much like the maintenance of plasma osmolality, complex pressure is sensed by baroreceptors in the renal vasculature to
humoral, neural, and behavioral mechanisms exist that main- initiate activation of the RAAS (Hainsworth, 2014; Schweda
tain blood volume (Figure 2). In contrast to increases in and Kurtz, 2011).
plasma osmolality, however, decreased blood volume requires
the conservation and replacement of both water and solute
1.07.3.1 The Renin–Angiotensin–Aldosterone System
(i.e., sodium) to correct the extracellular volume loss that
occurs. Therefore, hypovolemia triggers physiological changes The RAAS is a critical regulator of hydromineral balance, that
that involve renal and other peripheral mechanisms that mini- impacts both peripheral mechanisms (e.g., systemic blood
mize further loss of sodium and water and behavioral changes pressure, urinary sodium excretion, and renal hemodynamics)
that maximize their intake. For example, aldosterone and AVP and central mechanisms (e.g., sympathetic outflow, thirst, and
act at the level of the kidney to maximize the reabsorption of sodium appetite) that aim to restore blood volume and pres-
sodium and water, thereby minimizing their loss in urine. sure during hypovolemic threats. Stimuli such as hemorrhage
Furthermore, angiotensin-II (Ang-II) and aldosterone are (Yamaguchi and Hama, 2011), pharmacologically induced
largely responsible for the consumption of water and sodium vasodilation (Stocker et al., 2000), sodium loss (Badauê-Passos
that occur as a result of volume loss, and collectively, these et al., 2007), or hypovolemia induced by extravascular admin-
physiological and behavioral changes act to restore volume to istration of polyethylene glycol (Stricker and Verbalis, 1986)
both the extra- and intracellular compartments. potently trigger the activation of the RAAS, the consequent
Another important point is that, at least in a clinical setting, synthesis of the effector peptide, Ang-II, and its stimulation
a loss in blood volume is most often also associated with of aldosterone secretion. While there is some evidence for
a decrease in blood pressure, and this decrease in pressure is tissue-specific formation and actions of Ang-II, as discussed
responsible for the initiation of many of the physiological in Chapter 3.20, The Brain Renin–Angiotensin System, it has
mechanisms that allow one to overcome hypovolemia. In long-been established that the formation of this octapeptide
this regard, the carotid sinus and aortic baroreceptors respond occurs predominantly within the systemic circulation and
to this decrease in pressure by reducing their tonic firing requires the coordination of many organ systems (Figure 3).
frequency, thereby activating neurons within the hindbrain Initially, liver-generated angiotensinogen is cleaved into
nucleus of the solitary tract (NTS) that elevate sympathetic angiotensin-I by renin that arises predominantly from the
and decrease parasympathetic tone (Fisher and Paton, 2012). juxtaglomerular apparatus of the kidney. Renin is the rate-
If the magnitude of the reduced pressure is considerable, this limiting enzyme for Ang-II synthesis, and consequently, hypo-
then prompts the release of AVP that acts systemically to volemia leads to elevations in Ang-II by increasing the synthesis

Figure 2 Response to hypovolemic (extracellular) dehydration. IML, intermediolateral cell column of the spinal cord; NTS, nucleus of the solitary
tract; PVN, paraventricular nucleus of the hypothalamus; RVLM, rostral ventrolateral medulla.
Body Fluid Homeostasis 215

(Sasamura et al., 1992; MacTaggart et al., 1997). Within the


brain of rodents, AT1R is primarily present in its AT1aR form;
however, in some cases, AT1bR has been detected in the
brain, and it is also abundant in the anterior pituitary and
adrenal cortex (Kakar et al., 1992; Burson et al., 1994; Lenkei
et al., 1997). For the purposes of this chapter, however, it is
only important to distinguish between the AT1R (which in
rodents refers to both AT1aR and AT1bR) and the AT2R.
At the level of the brain, blood-borne Ang-II acts at CVOs
which, as discussed above, are brain regions with a ‘leaky’
blood–brain barrier that serve as an interface between signaling
factors within the systemic circulation and the brain. As a result
of this positioning, CVOs are ideally suited to relay hydromin-
eral imbalance and cardiovascular function to the brain, and
one way that they do this is by communicating the level of
Ang-II in circulation. In particular, the subfornical organ
(SFO) and the OVLT are known to express AT1R and to initiate
central responses to hypotension and hypovolemia. Ang-II
Figure 3 The classical endocrine renin–angiotensin system. Hypo- receptors are also expressed on neurons residing on the
volemia leads to the synthesis of Ang-II by stimulating the secretion of protected side of the blood–brain barrier and are heavily
renin from the kidney. Reduced renal arterial perfusion, reduced sodium implicated in the brain circuits that control responses to hypo-
delivery to the macula densa, increased sympathetic nerve activity to volemia and hypotension. The modulation of Ang-II levels
the b1 receptors of the juxtaglomerular apparatus, and increased circu-
and/or actions at its receptors within these blood–brain
lating catecholamines all occur in response to hypovolemic threats and
barrier-protected areas is known to impact physiology and
lead to increased secretion of renin.
behavior, and it is hypothesized that a brain-specific renin–
angiotensin system generates Ang-II that acts on these recep-
and secretion of renin from the kidney. Reduced renal arterial tors. This notion is discussed in detail in Chapter 3.20,
perfusion, reduced sodium delivery to the macula densa, The Brain Renin–Angiotensin System.
increased sympathetic nerve activity to the b1 receptors of the
juxtaglomerular apparatus, and increased circulating catechol-
1.07.3.2 Hypovolemic Thirst
amines all occur in response to hypovolemic threats and lead
to increased secretion of renin. Angiotensin-converting enzyme Much like an increase in plasma osmolality, a reduction in
(ACE; found in the lungs) then removes two amino acids from blood volume is also a potent stimulus of thirst and water
angiotensin-I to form Ang-II, which acts at its receptors in consumption. In contrast to osmotically driven thirst, however,
various areas of the body to maintain blood pressure and which is stimulated by a minimal, 1–2 % increase in plasma
volume via effects on vasoconstriction, renal sodium reabsorp- osmolality, a decrease in plasma volume by 10% is necessary
tion, sympathetic nervous system activity, aldosterone secre- to stimulate hypovolemic thirst. Clinically, such pronounced
tion, and water and sodium consumption. reductions in plasma volume occur with hemorrhage or with
The receptors to which Ang-II binds are G-protein-coupled excessive vomiting, diarrhea, or sweating. Some experimental
receptors that, in humans, can be subdivided into two types: methods by which hypovolemia is provoked include bleeding
the angiotensin type-1 receptor (AT1R) and the angiotensin (Fitzsimons, 1961; Oatley, 1964), surgical methods to reduce
type-2 receptor (AT2R) (Mendelsohn et al., 1984; Tsutsumi venous return to the heart (Fitzsimons, 1969; Fitzsimons and
and Saavedra, 1991; Johren et al., 1995, 1996; Johren and Moore-Gillon, 1980), induced sodium deficiency (Cizek
Saavedra, 1996; Lenkei et al., 1997; Hauser et al., 1998; et al., 1951), and the extravascular administration of colloidal
Gonzalez et al., 2012; de Kloet et al., 2016). Activation of solutions (Fitzsimons, 1961; Stricker, 1966). The latter leads to
these receptors triggers a number of intracellular signal the sequestration, in particular, of extracellular fluid allowing
transduction pathways, such as G-protein-mediated (Gq and for a reduction in plasma volume without impacting plasma
Gi), JAK/STAT, and MAPK or ERK intracellular signaling osmolality and pressure (Fitzsimons, 1961; Stricker, 1966),
cascades which have been implicated in Ang-II’s impact on thereby allowing for the determination of physiological and
physiology and behavior (Sadoshima et al., 1995; Velloso behavioral responses to volume loss, per se.
et al., 1996; Daniels et al., 2005, 2009; Daniels, 2014). Among the hormones involved in the physiological
Importantly, AT1Rs are the target of most of the known responses to volume loss is the above-described AVP, which
physiologic actions of Ang-II, such as vasoconstriction and causes vasoconstriction and water retention not only during
water consumption. Conversely, AT2R activation is thought increased plasma osmolality, but also in response to severe
to antagonize AT1R activation in many regards (Yayama and hypovolemia (Fitzsimons, 1961; Stricker, 1966; Dunn et al.,
Okamoto, 2008; Sumners et al., 2015). It is also relevant to 1973). That is, AVP secretion is stimulated in the absence of
note here that in rodents, the AT1R subtype is further any changes in plasma osmolality, if the volume loss is
subdivided into the angiotensin type-1a receptor (AT1aR) substantial enough (Fitzsimons, 1961; Stricker, 1966; Dunn
and type-1b receptor (AT1bR) whose genes are present et al., 1973). Again, this response is advantageous because
on separate chromosomes, but are highly homologous AVP acts at the vasculature to promote vasoconstriction as
216 Body Fluid Homeostasis

well as at the kidney to conserve urinary water, and these among fluid compartments. Perhaps consequently, several
combined effects allow maintenance of perfusion pressure RAAS mechanisms are centered on promoting salt appetite or
and fluid conservation subsequent to severe volume depletion. the ingestion of sodium that follows volume depletion.
Similar to AVP, Ang-II promotes vasoconstriction and renal
fluid reabsorption; however, it is also a potently acting dipso-
1.07.3.3 Salt Appetite
gen. As discussed, hypovolemia stimulates the secretion of
renin resulting in elevated circulating Ang-II that acts centrally From a historical perspective, much of our understanding of
to restore blood volume and pressure. In support of a role for salt appetite stems from the pioneering work of Curt Richter,
Ang-II in thirst and drinking behavior, administration of which focused largely on how changes in behavior compensate
this peptide systemically or intracranially causes prompt for perturbations in the internal environment (Krause and
and intense drinking responses in water-replete animals Sakai, 2007). In his experiments, Richter manipulated diet,
(Fitzsimons, 1969; Fitzsimons and Simons, 1969; Simpson interfered with the nervous system, and removed or replaced
and Routtenberg, 1978; Vanhouten et al., 1980; Mendelsohn glandular secretions in attempt to understand the neural and
et al., 1984). This phenomenon is true for many species, endocrine signals that contribute to behavior. One of the
including mammals (Epstein et al., 1970; Lotter et al., 1980; most striking behaviors that Richter observed was that of rats
Weisinger et al., 1997), birds (Schwob and Johnson, 1977), voluntarily ingesting salt when faced with sodium deficit.
reptiles (Fitzsimons and Kaufman, 1977), and fish (Beasley Sodium chloride was removed from the rats’ food and they
et al., 1986). The most sensitive region in the generation of were given access to water and 3% NaCl solution. The intake
Ang-II-induced drinking responses is the SFO (Simpson and of the NaCl solution was 1% of the total diet, which is the
Routtenberg, 1973, 1978; McKinley et al., 1983), which is a cir- amount of salt that was calculated as necessary for healthy
cumventricular organ that binds circulating factors and development of the rat (Richter, 1956). These results suggested
communicates their levels to the brain. Specific inhibition of that the rats were consuming NaCl, which effectively compen-
AT1R in the SFO decreases water intake elicited by increasing sated for the loss of salt in their food.
the circulating concentration of Ang-II but has no effect on Subsequent studies examined the behavioral responses to
drinking induced by increasing the plasma sodium concentra- sodium deficiency when physiological mechanisms to conserve
tion (Krause et al., 2008). This study suggests that the water sodium were surgically eliminated. While it was previously
intake that occurs as a consequence of increased circulating known that adrenalectomy produced sodium insufficiency
Ang-II is dependent, in part, on activation of AT1R residing (Rubin and Krick, 1933), Richter found that immediately after
in the SFO. That being said, the ability of Ang-II to stimulate adrenalectomy the consumption of NaCl solution was
water intake requires the coordination of numerous brain augmented while water intake was reduced (Richter, 1936).
structures, including not only CVOs but also areas that are pro- The increased consumption of NaCl was robust and occurred
tected by the blood–brain barrier (e.g., MnPO, BNST, lateral the first time the rats were given access to the solutions, suggest-
hypothalamus, and portions of the amygdala). The brain ing that the behavior was innate because there was little time to
nuclei and neural circuits that are involved in the central learn of the postingestive consequences. Importantly, adrenal-
actions of Ang-II are discussed in detail in Chapter 3.20, ectomized rats did not increase intake of chloride supplied as
The Brain Renin–Angiotensin System. In regard to Ang-II’s magnesium chloride or aluminum chloride, but did readily
role in the brain’s control of water balance, circulating Ang-II ingest different sodium compounds indicating that the appetite
stimulates water consumption by activating neurons in SFO was specific for sodium (Richter, 1956). As a result of having
that express AT1R, and the efferent projections of these neurons access to saline, the survival rate of the adrenalectomized rats
terminate within brain nuclei (e.g., preoptic, anterior hypotha- was greatly increased, and except for the propensity to consume
lamic, and limbic regions) that coordinate the perception of large volumes of salt solutions, there were no indications of
thirst with drinking behavior (Fitzsimons, 1998; McKinley deficiency. However, when the salt solutions were removed,
et al., 2003). Furthermore, there is evidence for Ang-II actions almost 90% of the rats died within 5 days, indicating that the
within blood–brain barrier-protected regions in the stimula- increased appetite for salt was keeping these animals alive.
tion of thirst and water consumption. For example, injection This is also of clinical relevance, as described in a case study
of Ang-II into the preoptic area also induces drinking (Richard- by Wilkins and Richter (1940) that concerned a 3 1/2-year-old
son and Mogenson, 1981). boy who was admitted to the hospital with symptoms of early-
It is important to note that, in spite of the hypovolemic drive onset puberty. Shortly after admittance the child died, and it
to consume water, the powerful inhibitory effect of osmotic dilu- was only after his death that it was determined that he was
tion on water intake is able to override hypovolemic thirst. That unable to retain sodium due to adrenal pathology. Interviews
is, a 2–3 % decrease in plasma osmolality will abolish water with the child’s parents revealed that at an early age the boy
intake and AVP secretion in volume-depleted rats (Stricker, had an insatiable appetite for salt, which likely kept him alive
1969). This phenomenon may be due to the fact that although until the relatively low-sodium diet at the hospital resulted in
increased water intake is a compensatory response to hypovole- his death.
mia, consuming only water will not alleviate deficits in vascular While Richter’s initial studies demonstrated that sodium
volume because water is stored mostly within intracellular deficiency was accompanied by an increased salt appetite that
compartments. A more advantageous response would be to promotes vitality, the signals relating to the arousal of the appe-
ingest sodium and water to produce a mixture near the concen- tite were poorly understood. Richter found that grafting adrenal
tration of isotonic saline, which would more effectively replace tissue into the eye of adrenalectomized rats decreased sodium
the volume lost, thereby allowing normal water distribution intake to that of intact rats within 10 days, suggesting that
Body Fluid Homeostasis 217

the increased intake is due to the loss of some adrenal secretion Behavior). First of all, aldosterone is known to bind to cytosolic
(Richter, 1956). Consequently, it was hypothesized that salt receptors that act as transcription factors and initiate changes in
appetite was dependent on the presence of the salt-retaining gene expression. This mode of action takes hours to days and is
hormone, aldosterone, which is produced in the adrenal glo- likely a factor in the long-term regulation of sodium intake.
merulosa. Richter treated adrenalectomized rats with the aldo- Second, aldosterone can also induce rapid changes in sodium
sterone precursor, deoxycorticosterone (DOC), and NaCl consumption through actions at the cell surface (Fluharty
intake returned to normal, presumably, because excessive et al., 1995; Sakai et al., 2000). Specifically, when converted
amounts of sodium were no longer lost in urine (Richter and into its tetrahydro metabolites, aldosterone interacts with
Eckert, 1938). However, subsequent studies revealed that treat- ligand-gated ion channels to influence neuronal firing. This
ing intact rats with DOC elicited a salt appetite similar to that of nongenomic mechanism was thought to contribute to the
adrenalectomized rats, which at the time seemed paradoxical rapid ingestion of sodium that accompanies DOC administra-
(Rice and Richter, 1943). Adrenalectomized rats lacking endog- tion (Fluharty et al., 1995; Sakai et al., 2000). Thus, it was
enous mineralocorticoids had a robust salt appetite that could hypothesized that aldosterone regulates short- and long-term
be attenuated by DOC, but administration of this agonist in consumption of sodium through genomic as well as nonge-
normal rats increased NaCl intake with increasing doses. Subse- nomic actions within the brain.
quent research would attempt to resolve this conundrum by Sakai et al. (2000) provided evidence supporting this
investigating the mechanisms underlying DOC-induced NaCl hypothesis by using antisense oligodeoxynucleotides (ASDNs),
intake and evaluating other factors contributing to salt appetite. single-stranded DNA molecules that inhibit the expression of
targeted genes, to evaluate the role of MR in DOC-induced
1.07.3.3.1 Aldosterone and Salt Appetite salt intake. Antisense oligodeoxynucleotides directed against
Wolf (1965) expanded on Richter’s initial observations by the GR or MR were injected into the amygdala, thereby
examining the NaCl intake of intact and adrenalectomized inhibiting the expression of these receptors locally within this
rats after administration of various doses of DOC. As expected, brain region. Subsequently, DOC was administered and salt
intact rats increased NaCl intake with elevating doses of DOC. intake was measured. Administration of ASDNs directed
In contrast, the dose–response curve of the adrenalectomized against MR significantly reduced DOC-induced sodium
rats resembled an inverted U-shape, with DOC diminishing intake, whereas ASDNs directed against the GR had no effect.
intakes at low doses, but increasing ingestion at higher Interestingly, the same study reported that salt intake could
doses. Intriguingly, pretreating adrenalectomized rats with be elicited 15 min after direct injection of tetrahydro-
corticosterone, an endogenous adrenal steroid, decreased aldosterone into the amygdala, and this effect was not
the doses of DOC required to produce NaCl intake. In blocked by administration of MR antagonists that inhibit the
addition, Wolf and others found that aldosterone, another genomic actions of this receptor. Collectively, these results
endogenous adrenal steroid, specifically augmented the suggested that salt appetite could be aroused through both
intake of sodium salts (Wolf and Handal, 1966; Weisinger genomic and nongenomic effects of mineralocorticoids.
and Woods, 1971). Taken together, these studies suggested However, the fact that adrenalectomized rats lacking
that endogenous adrenal steroids contributed to the arousal endogenous adrenal steroids demonstrated a robust salt
of salt appetite; however, the mechanism underlying the appetite made it unlikely that aldosterone was the only signal
natrorexigenic effects of these hormones remained unknown. capable of eliciting sodium intake.
Early investigation of adrenal steroid regulation of salt appe-
tite focused on the central actions of corticosterone and aldoste- 1.07.3.3.2 Angiotensin-II and Salt Appetite
rone because certain brain lesions inhibited the salt intake that It has long-been appreciated that Ang-II contributes to the elic-
follows DOC administration (Wolf, 1964; Nitabach et al., itation of salt appetite indirectly by stimulating the release of
1989). Brain regions that readily took up these hormones were aldosterone (Davis and Spielman, 1974), while the direct
identified with the use of receptor autoradiography. Specifically, involvement of Ang-II as a stimulator of sodium intake was
corticosterone and aldosterone were found to compete for often disregarded (Stricker, 1971). However, Buggy and Fisher
binding sites in the hippocampus, septum, and amygdala (1974) found that intracranial administration of Ang-II
(Gerlach and McEwen, 1972; Birmingham et al., 1979; McEwen increased water and sodium intake. Importantly, the elevated
et al., 1986). However, the relative contribution of adrenal consumption of NaCl was specific to Ang-II because adminis-
steroids to salt appetite in specific brain areas was uncertain until tration of carbachol, another centrally acting dipsogen, elicited
specific antagonists for glucocorticoid (GR) and mineralocorti- only water intake. Subsequent studies further implicated the
coid (MR) receptors were developed. In this regard, McEwen peptide in the arousal of salt appetite and provided potential
et al. (1986) administered RU-28318, an MR-specific sites of actions for this effect. For example, salt appetite result-
antagonist, to adrenalectomized rats and observed that the ing from the natriuretic–diuretic furosemide was attenuated by
same dose of RU-28318 that inhibited aldosterone binding in pharmacological reduction of circulating Ang-II and was
the hippocampus, septum, and amygdala also blunted the restored by intravenous administration of this peptide
hormone’s effects on salt intake. These results provided (Weisinger et al., 1987a, 1987b, 1997).
indirect evidence that aldosterone regulation of salt appetite As with Ang-II-induced water intake, investigators considered
involves, in part, the actions of aldosterone in the brain. the SFO and OVLT potential sites of action for Ang-II arousal of
Aldosterone activates central pathways mediating salt appe- salt appetite. In support of a role for these CVOs in this process,
tite via two mechanisms (Fluharty et al., 1995; Karst et al., ablation of the SFO decreased sodium intake elicited by
2005; see Chapter 3.09, Aldosterone Action on Brain and administration of the furosemide (Weisinger et al., 1990;
218 Body Fluid Homeostasis

Thunhorst et al., 1999) and lesion of the OVLT attenuated Because DOC is a precursor for both aldosterone and corti-
sodium-depletion-induced intake (Chiaraviglio and Perez costerone and its administration leads to elevations in both of
Guaita, 1984), while the site-specific injection of Ang-II into these adrenal steroids, it was not clear whether the observed
the OVLT stimulated NaCl consumption (Fitts and Masson, upregulation of Ang-II receptors was due to the actions of
1990). Although these studies suggested that Ang-II acted cen- MRs, GRs, or a combination of both. Nonetheless, a role for
trally to elicit salt appetite, the biological relevance of this GRs was proposed because DOC increased Ang-II receptor
phenomenon was called into question because supraphysiolog- binding to a greater degree than aldosterone (Wilson et al.,
ical doses of Ang-II were required to produce the behavior. Large 1986) and because pretreatment with dexamethasone, a GR
doses of Ang-II administered centrally or intravenously elevate agonist, also potentiated drinking stimulated by Ang-II
blood pressure and can cause natriuresis (Findlay and Epstein, (Ganesan and Sumners, 1989). Moreover, intravenous infu-
1980; Fluharty and Manaker, 1983). This complicated interpre- sion of dexamethasone, corticosterone, or the GR receptor
tation of studies examining Ang-II-induced salt appetite because agonist RU 28362 increased aldosterone-induced sodium
it was possible that the NaCl intake caused by Ang-II treatment consumption, further suggesting that GRs contribute to the
was the consequence of renal sodium excretion. However, the arousal of salt appetite (Ma et al., 1993).
recognition that circulating levels of Ang-II and aldosterone The strongest support for the regulation of salt appetite by
increased under normal conditions of sodium depletion Stricker glucocorticoids comes from studies examining NaCl intake
et al. (1979) led to a new understanding of hormonal regulation and AT1R expression subsequent to DOC and dexamethasone
of salt appetite. administration (Shelat et al., 1999b). Rats treated with both
DOC and dexamethasone ingested more NaCl than DOC given
1.07.3.3.3 Angiotensin/Aldosterone Interactions by itself. Additionally, coadministration of DOC and dexa-
Because sodium depletion leads to the concurrent elevations in methasone augmented Ang-II receptor binding in the PVN,
aldosterone and Ang-II, it has been hypothesized that these two SFO and area postrema; DOC alone evoked no such effect
hormones work in concert, or synergistically, to arouse sodium (Shelat et al., 1999a). Consequently, some models of the
appetite (Epstein, 1982). Consistent with this, Fluharty and hormonal regulation of salt appetite combine the contribu-
Epstein (1983) determined that simultaneous AT1R and MR tions of MRs as well as GRs.
activation via the coadministration of Ang-II and DOC It is worth noting that more recent studies have implicated
produced a salt intake that was greater than the sum of the hindbrain neurons that are uniquely sensitive to aldosterone in
amounts consumed when the same doses of Ang-II or DOC the control of sodium appetite. The MR has affinity for both
were separately administered. Furthermore, low doses of Ang- aldosterone and corticosterone with the latter circulating at
II and DOC that were ineffective at evoking salt appetite concentration orders of magnitude greater than the former
when given separately became potent at inducing sodium (Funder et al., 1988). Consequently, MR-expressing cells
intake when given concurrently. This enhanced salt intake that are uniquely responsive to aldosterone also express
was not accompanied by pressure-induced natriuresis, obvi- 11-b-hydroxysteriod dehydrogenase type 2 (11-bHSD2) which
ating the possibility that renal sodium excretion was respon- inactivates glucocorticoids that under normal conditions
sible for the augmented ingestion of NaCl. Additionally, occupy MRs, which in turn, permits their binding of aldoste-
central antagonism of MR using RU-28318 reduced, but did rone (Funder et al., 1988). Anatomical studies conducted by
not eliminate, the salt appetite elicited by sodium depletion; Geerling and Loewy (2006) identified neurons residing in the
however, when both Ang-II and MR actions were pharmacolog- NTS that expressed both MRs and 11-bHSD2. These ‘HSD2’
ically inhibited, depletion-induced sodium intake was neurons were found to send direct axonal projections to the
completely abolished (Sakai et al., 1986). Again, these effects ventral portion of the bed nucleus of the stria terminalis, the
were likely not due to altered renal sodium handling or general pre-locus coeruleus, the external lateral parabrachial nucleus,
inhibition of ingestive behaviors, as they occurred independent the ventral tegmental area, the lateral and paraventricular
of alterations in urinary sodium excretion or food intake (Sakai nuclei of the hypothalamus, and the central nucleus of the
et al., 1986). Thus, it was concluded that AT1R and MRs did amygdala (Geerling and Loewy, 2006). Given the hypothesized
indeed work synergistically to arouse salt appetite. role of brain MRs in salt appetite, the authors proposed that the
The synergistic interactions between Ang-II were also connectivity of HSD2 neurons comprised a neuronal circuit
quickly appreciated to be brain-mediated, as blocking the controlling the drive to consume sodium when challenged
effects of Ang-II and aldosterone in the brain, but not in the with such deficit. Indeed, their subsequent study determined
periphery, eliminates the salt appetite of furosemide-treated that HSD2 neurons in the NTS were mostly sensitive to
rats (Sakai et al., 1986, 1990). Furthermore, chronic adminis- aldosterone and were activated by DOC or sodium depletion,
tration of DOC increases Ang-II receptor binding in brain tissue and this activation was inhibited by voluntary consumption
homogenates that included the hypothalamus, thalamus, and of NaCl (Geerling et al., 2006). Consistent with these results,
septum (Wilson et al., 1986), and drinking stimulated by studies conducted by the Mifflin laboratory found that salt
central injection of Ang-II is augmented after DOC administra- appetite elicited by exogenous administration of aldosterone
tion, demonstrating the behavioral consequences of these is greatly reduced in rats delivered viral constructs that knock-
increased Ang-II binding sites (Wilson et al., 1986). Subsequent down MR expression in the NTS (Koneru et al., 2014).
studies identified increased Ang-II receptor binding in specific However, it is interesting to note that adrenalectomized rats
brain regions following DOC administration (Gutkind et al., that cannot synthesize aldosterone or corticosterone have
1988; De Nicola et al., 1993) as well as within cultured neurons robust activation of HSD2 neurons that is abrogated by NaCl
(Sumners and Fregly, 1989). consumption, indicating that adrenal steroids are not necessary
Body Fluid Homeostasis 219

for the activation of HSD2 neurons or the emergence of salt investigate whether sodium deficiency enhanced the detection
appetite (Geerling et al., 2006). These results suggest that threshold for NaCl solutions. Using a two-bottle preference
HSD2 neurons in the NTS are activated by aldosterone to drive test, Richter determined that intact rats preferred a NaCl solu-
salt appetite, but in the absence of adrenal steroids sodium tion over water when the concentration was about 0.055%;
deficiency still engages these HSD2 neurons to drive sodium however, immediately after adrenalectomy rats drank more of
consumption. the NaCl solution when it was at a nearly 15-fold lesser concen-
tration of 0.0037% (Richter, 1939). That is, these results indi-
1.07.3.3.4 Baroreceptors, Osmoreceptors, and Salt Appetite cated that sodium insufficiency decreases the threshold at
Baroreceptors are mechanoreceptors located in blood vessels which rats prefer NaCl solutions over water, possibly by
near the heart that provide the brain with information pertain- increasing the sensitivity for the taste of sodium. Also along
ing to blood volume and pressure, by detecting the level of these lines, examination of the ingestion of saline solutions
stretch on vascular walls. As blood volume increases, vessels at various concentrations revealed that while intact rats have
are stretched and the firing rate of baroreceptors increases. In a preference curve that resembles an inverted U-shape with
contrast, as vessels contract in response to reduced blood maximum consumption occurring near the concentration of
volume, baroreceptors decrease firing. Since sodium insuffi- isotonic saline (Richter, 1956), sodium deficiency dramatically
ciency is often accompanied by changes in blood volume, it changes this preference curve by elevating intake of NaCl at low
was reasoned that baroreceptors were involved in the regula- and high concentrations (Epstein and Stellar, 1955; Nachman,
tion of sodium appetite. Consistent with this, surgical manipu- 1962; Curtis et al., 2001).
lation of baroreceptors (Thunhorst et al., 1994) or the Of particular relevance, sodium-deficient rats develop this
sinoaortic nerves that communicate their status to the brain preference for sodium within 15 s after they are given a choice
(Thunhorst et al., 1994) alters sodium consumption. For between sodium and nonsodium solutions (Nachman, 1962).
example, Toth et al. (1987) implanted small balloons into This short latency to develop a preference is also consistent
the superior vena caval–right atrial junction of rats in order with the effects being taste-mediated as there is little time for
to model volume expansion and determine its impact on the animal to learn of the consequences of the ingestion.
sodium appetite. Inflation of these balloons led to stretching Furthermore, sodium depletion alters orofacial patterns of
of the baroreceptors and signaled volume expansion to the rats in response to intraoral infusions of high concentrations
brain. This manipulation also attenuated NaCl intake elicited of NaCl in a manner suggestive of the depletion rendering
either by peritoneal dialysis or by administration of DOC, the taste of sodium more palatable (Flynn and Grill, 1988).
and intakes returned to normal when the balloons were High concentrations of NaCl that elicit aversive orofacial
deflated. Similarly, NaCl intake elicited by furosemide is signif- behaviors in normal rats elicit ingestive behaviors when rats
icantly reduced when baroreceptor input is eliminated by are salt deficient (Berridge et al., 1984). In sum, sodium deficit
sinoaortic denervation (Thunhorst et al., 1994). Collectively, enhances the gustatory processing of sodium, thereby facili-
these studies suggest that baroreceptors provide the brain tating the likelihood that sodium will be detected and ingested.
with neural signals pertaining to blood volume that are critical
for the normal expression of sodium appetite.
1.07.3.4 Blood Pressure
While cardiac baroreceptors regulate salt appetite by detect-
ing changes in blood volume, the above-described cerebral Other important aspects of overcoming hypovolemia and the
osmoreceptors influence sodium consumption by monitoring concomitant hypotension include factors that are involved in
the concentration of sodium within cerebrospinal fluid elevating blood pressure. To this end, hypotension is remedied,
(CSF). Along these lines, increasing the sodium concentration at least in part, by the above-described mechanisms to mini-
within the brain via the intraventricular injection of hyperos- mize sodium and water excretion and maximize their intake.
motic CSF [Naþ, 500 mM] leads to an attenuation of That is, an important outcome of increasing blood volume is
deprivation-induced sodium intake (Weisinger et al., 1979). to also increase blood pressure. Also important, however, are
On the other hand, when osmolality of the CSF is increased Ang-II’s (and AVP’s) more direct roles in elevating blood pres-
by intraventricular injection of mannitol, which decreases the sure by activating receptors in the peripheral vasculature and
sodium concentration, sodium intake is augmented (Weisinger within certain brain nuclei. In regard to central effects of Ang-
et al., 1979). These results suggest that in addition to regulating II, the sensing of elevated Ang-II levels by CVOs, such as the
thirst, AVP, and OT secretion (as described above) by detecting SFO also communicates to the brain the need to elevate blood
central changes in osmolality cerebral osmoreceptors may also pressure; the goal being to restore blood pressure to normal
regulate the ingestion of sodium. levels (Ferguson, 1988, 2009; Ferguson and Bains, 1997;
Krause et al., 2011b; Osborn et al., 2012). This ultimately leads
1.07.3.3.5 Taste and Salt Appetite to an increase in blood pressure that, again, is partly an indirect
The sensory modality that is most important for the detection consequence of the behavioral effects discussed above.
of sodium is taste, and early studies conducted by Richter However, it is also due to its ability to impact blood pressure
and others revealed that adrenalectomy and the resulting by other means, such as activation of the sympathetic nervous
sodium insufficiency creates a clear preference for sodium solu- system (SNS).
tions that were previously indistinguishable from water Much like with Ang-II’s behavioral actions, in order to
(Richter, 1939). Therefore, it was hypothesized that sodium exert these influences over the SNS, CVO neurons send projec-
depletion led to changes in the oral cavity that contributed to tions to and influence the activity of numerous cardiovascular
the increased salt intake, and experiments were conducted to control centers that are generally protected by the BBB,
220 Body Fluid Homeostasis

including the PVN (Ferguson, 1988; Krause et al., 2011b). to body fluid homeostasis can have a profound impact on
Upon activation of these CVO projections, the release of tradi- the responses to psychological stress.
tional neurotransmitters (e.g., glutamate) from their nerve For example, sodium depletion is anxiogenic in rats
terminals and onto neurons situated in downstream cardio- (Leshem, 2011), and several factors that are traditionally
vascular control nuclei is modulated, and sympathetic known to regulate hydromineral balance have also been impli-
outflow and blood pressure become elevated (Tanaka et al., cated in the responses to psychological stress. The RAAS, which
2001; Saxena et al., 2014). That being said, much like the again, is activated during hypovolemic threats, is thought to
positioning and influence of the AT1R within the BBB- facilitate physiological and behavioral responses to psycholog-
protected nuclei involved in the behavioral responses to ical stress (Saavedra et al., 2006; Krause et al., 2011b; Saavedra,
Ang-II, many of the downstream brain nuclei involved in 2011). Psychological stress activates the RAAS and upregulates
sympathetic outflow also densely express AT1R and Ang-II’s certain populations of AT1R in the absence of threats to hydro-
actions at its AT1R within these nuclei impact cardiovascular mineral balance (Yang et al., 1993; Aguilera et al., 1995; Krause
function (see Chapter 3.20, The Brain Renin–Angiotensin et al., 2011b) and inhibition of Ang-II’s actions at the SFO also
System). inhibits anxiety-like behavior and stress-induced activation of
the HPA axis (Krause et al., 2008, 2011b). Along these lines,
the RAAS has gained considerable attention over the past
1.07.3.5 Hypervolemia
decade as a potential therapeutic target for treating stress-
An increase in blood volume, hypervolemia, is also sensed by related disorders (Baghai et al., 2002, 2006; Saavedra et al.,
the stretch-sensitive atrial baroreceptors to trigger the secretion 2006; Krause et al., 2011b; Khoury et al., 2012; Hurt et al.,
of atrial natriuretic peptide (ANP), from the cardiac atria (Chiu 2015). Angiotensin receptor blockers administered either
et al., 1987; Palkovits et al., 1995). This volume-regulatory peripherally or centrally improve mood and cognition as well
hormone inhibits the secretion of renin and aldosterone as attenuate HPA axis and sympathetic responses to stressors
(Atlas and Maack, 1987), promotes natriuresis (Soares et al., having strong psychogenic components (Barnes et al., 1990;
1999), and opposes the stimulatory actions of Ang-II on water Kaiser et al., 1992; Jezova et al., 1998; Kubo et al., 2001;
intake and vasoconstriction (Antunes-Rodrigues et al., 1985; Saavedra et al., 2006).
Saavedra, 1990; McCann et al., 2003). There is also evidence On the other hand, challenges to body fluid homeostasis
that ANP reduces sodium appetite and consumption (Ehrlich that manifest as increases in osmolality oppose or dampen
and Fitts, 1990; Stellar and Epstein, 1991; McCann et al., the physiological and behavioral responses to psychological
1994). Collectively, ANP acts at its receptors throughout the stress (Krause et al., 2011a; Frazier et al., 2013; Smith et al.,
body to facilitate restoration of blood volume and pressure 2014). As discussed above, excess body sodium or hypernatre-
to normal levels subsequent to volume expansion. mia creates a distinct neural and humoral milieu that alleviates
elevations in body sodium levels. One important aspect of this
milieu is the inhibition of the RAAS, and it is possible that this
1.07.4 Body Fluid Homeostasis and Psychological reduction contributes to the stress-dampening effects of hyper-
Stress osmotic conditions by removing or reducing the facilitating
influence of Ang-II on stress responses. That being said, there
Stressors can be classified as either real (i.e., physiological) or is also evidence for a prominent role of OT in these stress-
perceived (i.e., psychological) in nature and lead to behavioral, dampening actions. This is supported by rodent studies
endocrine, and autonomic adjustments that allow one to demonstrating that increased plasma osmolality induced by
attempt to cope with the stressor. Some examples of physiolog- the systemic administration of hypertonic (2.0 M) saline fol-
ical stressors include the threats to hydromineral balance dis- lowed by 60 min water deprivation significantly increases
cussed above, such as hypovolemia, while examples of plasma OT concentrations that correspond to a reduced HPA
psychogenic stressors may include the loss of a loved one or axis response to an acute psychological stressor (Krause et al.,
social isolation (Jankord and Herman, 2008). The origin and 2011a). In addition to blunting the HPA axis response to the
initiation of the responses to these types of stressors are distinct psychological stressor, this paradigm also results in a blunted
from one another; however, they are similar in that they cardiovascular response to stress and decreased anxiety-like
both activate the hypothalamic–pituitary–adrenal (HPA) axis behavior in the elevated plus maze and social interaction para-
(see Chapter 4.07, Hypothalamic–Pituitary–Thyroid Axis). digm (Krause et al., 2011a). Subsequent anatomical studies
Activation of the HPA axis is initiated by stimulation of revealed that acute hypernatremia elicits robust activation of
corticotrophin-releasing hormone (CRH) neurons within the OT neurons in the PVN but attenuates stress-induced activation
PVN that trigger the release of adrenocorticotrophin hormone of CRH neurons (Smith et al., 2014). Furthermore, whole-cell
(ACTH) from the anterior pituitary and glucocorticoid patch-clamp recordings found that acute hypernatremia
synthesis and release from the adrenal cortex into the systemic causes an inhibition of CRH neurons in the PVN that is
circulation. In addition to the fact that physiological stress dependent on activation of OT receptors; in other words, the
(such as threats to body fluid homeostasis) and psychological increased plasma osmolality is associated with an elevated
stress both activate the HPA axis, it is also evident that many inhibitory oxytocinergic tone that is exerted onto CRH neurons
brain regions and neural circuits that are involved in the regu- (Frazier et al., 2013). The implication is that this oxytocinergic
lation of body fluid homeostasis are also essential for the tone on CRH neurons observed in electrophysiological
neural, humoral, and behavioral responses to psychological experiments suppresses stress-induced activation of CRH
stress. It is perhaps not surprising, therefore, that challenges neurons and consequent elevations in ACTH and
Body Fluid Homeostasis 221

glucocorticoids in vivo. Therefore, while disturbances to body that also supports neuronal networks implicated in a variety
fluid homeostasis that manifest in reduced blood volume of behaviors ranging from stress and anxiety to sociality and
may heighten responses to psychological stress, those that affiliation.
manifest in increased plasma osmolality may act to dampen
responses to psychological stress.

References
1.07.5 Summary and Conclusion Adachi, A., 1984. Thermosensitive and osmoreceptive afferent fibers in the hepatic
branch of the vagus nerve. J. Aut. Nerv. Syst. 10 (3–4), 269–273.
As discussed throughout this chapter, alterations in body fluid Adachi, A., Niijima, A., Jacobs, H.L., 1976. An hepatic osmoreceptor mechanism in
homeostasis resulting from changes in the osmolality, the rat: electrophysiological and behavioral studies. Am. J. Physiol. 231 (4),
volume, and/or pressure of the blood are detected by sensors 1043–1049.
Aguilera, G., Young, W.S., Kiss, A., Bathia, A., 1995. Direct regulation of hypothalamic
in the periphery and brain that relay the imbalance to effector corticotropin-releasing-hormone neurons by angiotensin-II. Neuroendocrinology 61
organs. Consequently, neural, humoral, and behavioral (4), 437–444.
compensatory responses are initiated and maintain or restore Andersen, L.J., Jensen, T.U., Bestle, M.H., Bie, P., 2000. Gastrointestinal osmor-
body fluid homeostasis. Although changes in plasma osmo- eceptors and renal sodium excretion in humans. Am. J. Physiol. Regul. Integr.
Comp. Physiol. 278 (2), R287–R294.
lality and changes in effective circulating volume and blood
Antunes-Rodrigues, J., McCann, S.M., Rogers, L.C., Samson, W.K., 1985. Atrial
pressure are similarly considered threats to body fluid homeo- natriuretic factor inhibits dehydration- and angiotensin II-induced water intake in
stasis, the behavioral and endocrine mechanisms by which the conscious, unrestrained rat. Proc. Natl. Acad. Sci. U.S.A. 82 (24),
osmolality, volume, and pressure are regulated are distinct 8720–8723.
and these mechanisms are summarized in Figures 1 and 2. Aoyagi, T., Koshimizu, T.A., Tanoue, A., 2009. Vasopressin regulation of blood
pressure and volume: findings from V1a receptor-deficient mice. Kidney Int. 76
That is, plasma osmolality is maintained largely by changes (10), 1035–1039.
in water balance and activation of the AVP/OT renal axes. Atlas, S.A., Maack, T., 1987. Effects of atrial natriuretic factor on the kidney and the
On the other hand, the RAAS is largely responsible for the renin-angiotensin-aldosterone system. Endocrinol. Metabol. Clin. N. Am. 16 (1),
mechanisms by which blood volume and pressure are main- 107–143.
Badauê-Passos Jr., D., Godino, A., Johnson, A.K., Vivas, L., Antunes-Rodrigues, J.,
tained. That being said, AVP is involved both in the restora-
2007. Dorsal raphe nuclei integrate allostatic information evoked by depletion-
tion of osmolality and in the restoration of blood volume induced sodium ingestion. Exp. Neurol. 206 (1), 86–94.
and pressure subsequent to hyperosmotic or hypovolemic Baertschi, A.J., Pence, R.A., 1995. Gut-brain signaling of water absorption inhibits
threats, respectively. That is, AVP secretion is stimulated in vasopressin in rats. Am. J. Physiol. 268 (1 Pt 2), R236–R247.
response to increased plasma osmolality and also in response Baertschi, A.J., Vallet, P.G., 1981. Osmosensitivity of the hepatic portal vein area and
vasopressin release in rats. J. Physiol. 315, 217–230.
to profound hypovolemia. In both conditions, it acts to regu- Baghai, T.C., Binder, E.B., Schule, C., et al., 2006. Polymorphisms in the angiotensin-
late body fluid homeostasis by activating its receptors in the converting enzyme gene are associated with unipolar depression, ACE activity and
kidney and vasculature, to promote water reabsorption and hypercortisolism. Mol. Psychiatry 11 (11), 1003–1015.
vasoconstriction. Baghai, T.C., Schule, C., Zwanzger, P., et al., 2002. Hypothalamic-pituitary-adreno-
cortical axis dysregulation in patients with major depression is influenced by the
In regard to the behavioral responses that act to preserve
insertion/deletion polymorphism in the angiotensin I-converting enzyme gene.
body fluid homeostasis, it is particularly important to note Neurosci. Lett. 328 (3), 299–303.
that although both hypovolemic and hyperosmotic stimuli Barnes, N.M., Costall, B., Kelly, M.E., Murphy, D.A., Naylor, R.J., 1990. Anxiolytic-like
both induce the sensation of thirst and the subsequent act action of DuP753, a non-peptide angiotensin II receptor antagonist. Neuroreport 1
of drinking, hypovolemic thirst differs in that it is also accom- (1), 20–21.
Beasley, D., Shier, D.N., Malvin, R.L., Smith, G., 1986. Angiotensin-stimulated
panied by an increase in sodium conservation, appetite, and drinking in marine fish. Am. J. Physiol. 250 (6 Pt 2), R1034–R1038.
consumption; the goal being to restore the extracellular deficit Berridge, K.C., Flynn, F.W., Schulkin, J., Grill, H.J., 1984. Sodium depletion enhances
in fluid which requires the restoration of both solute salt palatability in rats. Behav. Neurosci. 98 (4), 652–660.
and water. Birmingham, M.K., Stumpf, W.E., Sar, M., 1979. Nuclear localization of aldosterone in
rat brain cells assessed by autoradiography. Experientia 35 (9), 1240–1241.
The regulation of body fluid homeostasis is critical for all
Bourque, C.W., 2008. Central mechanisms of osmosensation and systemic osmo-
living organisms and requires coordinated responses with regulation. Nat. Rev. Neurosci. 9 (7), 519–531.
cardiovascular, renal, neuroendocrine, and behavioral nodes. Buggy, J., Fisher, A.E., 1974. Evidence for a dual central role for angiotensin in water
The humoral and neural mechanisms that orchestrate these and sodium intake. Nature 250 (5469), 733–735.
responses are highly conserved and in some instances are Burson, J.M., Aguilera, G., Gross, K.W., Sigmund, C.D., 1994. Differential expression
of angiotensin receptor 1A and 1B in mouse. Am. J. Physiol. 267 (2 Pt 1),
hardwired. Consequently, investigation of such mechanisms E260–E267.
reveals fundamental brain circuits controlling the compensa- Carlson, S.H., Beitz, A., Osborn, J.W., 1997. Intragastric hypertonic saline increases
tory responses and motivated behaviors that promote the vasopressin and central Fos immunoreactivity in conscious rats. Am. J. Physiol.
homeostatic concentrations of water and solute that are 272 (3 Pt 2), R750–R758.
Cavanaugh, D.J., Chesler, A.T., Jackson, A.C., et al., 2011. Trpv1 reporter mice reveal
essential for normal physiological function. Given their
highly restricted brain distribution and functional expression in arteriolar smooth
importance to the vitality of an organism, it is likely that muscle cells. J. Neurosci. 31 (13), 5067–5077.
such mechanisms were first established to maintain hydro- Chiaraviglio, E., Perez Guaita, M.F., 1984. Anterior third ventricle (A3V) lesions and
mineral balance, but subsequently, evolved to promote other homeostasis regulation. J. Physiol. (Paris) 79 (6), 446–452.
types of adaptive physiological and behavioral responses. In Chiu, P.J., Vemulapalli, S., Policelli, M., Kifor, I., Sybertz, E.J., Dzau, V.J., 1987. The
responses of atrial natriuretic factor concentrations to acute volume changes in
this way, studying the neuroendocrine control of thirst and conscious rats. Life Sci. 41 (21), 2339–2347.
salt appetite, as well as those mediating blood pressure and Choi-Kwon, S., Baertschi, A.J., 1991. Splanchnic osmosensation and vasopressin:
renal function, provides insight toward an infrastructure mechanisms and neural pathways. Am. J. Physiol. 261 (1 Pt 1), E18–E25.
222 Body Fluid Homeostasis

Chwalbinska-Moneta, J., 1979. Role of hepatic portal osmoreception in the control of Fitzsimons, J.T., Moore-Gillon, M.J., 1980. Drinking and antidiuresis in response to
ADH release. Am. J. Physiol. 236 (6), E603–E609. reductions in venous return in the dog: neural and endocrine mechanisms.
Ciura, S., Bourque, C.W., 2006. Transient receptor potential vanilloid 1 is required for J. Physiol. 308, 403–416.
intrinsic osmoreception in organum vasculosum lamina terminalis neurons and for Fitzsimons, J.T., Simons, B.J., 1969. The effect on drinking in the rat of intravenous
normal thirst responses to systemic hyperosmolality. J. Neurosci. 26 (35), infusion of angiotensin, given alone or in combination with other stimuli of thirst.
9069–9075. J. Physiol. 203 (1), 45–57.
Ciura, S., Liedtke, W., Bourque, C.W., 2011. Hypertonicity sensing in organum vas- Fluharty, S.J., Epstein, A.N., 1983. Sodium appetite elicited by intracerebroventricular
culosum lamina terminalis neurons: a mechanical process involving TRPV1 but not infusion of angiotensin II in the rat: II. Synergistic interaction with systemic
TRPV4. J. Neurosci. 31 (41), 14669–14676. mineralocorticoids. Behav. Neurosci. 97 (5), 746–758.
Cizek, L.J., Semple, R.E., Huang, K.C., Gregersen, M.I., 1951. Effect of extracellular Fluharty, S.J., Flanagancato, L.M., Sakai, R.R., 1995. Adrenal-steroid regulation of
electrolyte depletion on water intake in dogs. Am. J. Physiol. 164 (2), 415–422. neuropeptide receptor expression and function. Appetite 24 (1), 81.
Curtis, K.S., Krause, E.G., Contreras, R.J., 2001. Altered NaCl taste responses precede Fluharty, S.J., Manaker, S., 1983. Sodium appetite elicited by intracerebroventricular
increased NaCl ingestion during Na(þ) deprivation. Physiol. Behav. 72 (5), 743–749. infusion of angiotensin II in the rat: I. Relation to urinary sodium excretion. Behav.
Daniels, D., 2014. Diverse roles of angiotensin receptor intracellular signaling path- Neurosci. 97 (5), 738–745.
ways in the control of water and salt intake. In: De Luca Jr., L.A., Menani, J.V., Flynn, F.W., Grill, H.J., 1988. Intraoral intake and taste reactivity responses elicited by
Johnson, A.K. (Eds.), Neurobiology of Body Fluid Homeostasis: Transduction and sucrose and sodium chloride in chronic decerebrate rats. Behav. Neurosci. 102 (6),
Integration. Boca Raton (FL). 934–941.
Daniels, D., Mietlicki, E.G., Nowak, E.L., Fluharty, S.J., 2009. Angiotensin II stimulates Frazier, C.J., Pati, D., Hiller, H., et al., 2013. Acute hypernatremia exerts an inhibitory
water and NaCl intake through separate cell signalling pathways in rats. Exp. oxytocinergic tone that is associated with anxiolytic mood in male rats. Endocri-
Physiol. 94 (1), 130–137. nology 154 (7), 2457–2467.
Daniels, D., Yee, D.K., Faulconbridge, L.F., Fluharty, S.J., 2005. Divergent behavioral Funder, J.W., Pearce, P.T., Smith, R., Smith, A.I., 1988. Mineralocorticoid action:
roles of angiotensin receptor intracellular signaling cascades. Endocrinology 146 target tissue specificity is enzyme, not receptor, mediated. Science 242 (4878),
(12), 5552–5560. 583–585.
Davis, J.O., Spielman, W.S., 1974. The renin-angiotensin system in the control of Ganesan, R., Sumners, C., 1989. Glucocorticoids potentiate the dipsogenic action of
aldosterone secretion in the rat. Acta Physiol. Lat. Am. 24 (5), 399–404. angiotensin II. Brain Res. 499 (1), 121–130.
de Kloet, A.D., Wang, L., Ludin, J.A., et al., 2016. Reporter mouse strain provides Geelen, G., Keil, L.C., Kravik, S.E., et al., 1984. Inhibition of plasma vasopressin after
a novel look at angiotensin type-2 receptor distribution in the central nervous drinking in dehydrated humans. Am. J. Physiol. 247 (6 Pt 2), R968–R971.
system. Brain Struct. Funct. 221 (2), 891–912. Geerling, J.C., Engeland, W.C., Kawata, M., Loewy, A.D., 2006. Aldosterone target
De Nicola, A.F., Seltzer, A., Tsutsumi, K., Saavedra, J.M., 1993. Effects of deoxy- neurons in the nucleus tractus solitarius drive sodium appetite. J. Neurosci. 26 (2),
corticosterone acetate (DOCA) and aldosterone on Sar1-angiotensin II binding and 411–417.
angiotensin-converting enzyme binding sites in brain. Cell. Mol. Neurobiol. 13 (5), Geerling, J.C., Loewy, A.D., 2006. Aldosterone-sensitive neurons in the nucleus of the
529–539. solitary tract: efferent projections. J. Comp. Neurol. 497 (2), 223–250.
Dooley, C.P., Valenzuela, J.E., 1984. Duodenal volume and osmoreceptors in the Gerlach, J.L., McEwen, B.S., 1972. Rat brain binds adrenal steroid hormone:
stimulation of human pancreatic secretion. Gastroenterology 86 (1), 23–27. radioautography of hippocampus with corticosterone. Science 175 (4026),
Dunn, F.L., Brennan, T.J., Nelson, A.E., Robertson, G.L., 1973. The role of blood 1133–1136.
osmolality and volume in regulating vasopressin secretion in the rat. J. Clin. Invest. Gonzalez, A.D., Wang, G., Waters, E.M., et al., 2012. Distribution of angiotensin type
52 (12), 3212–3219. 1a receptor-containing cells in the brains of bacterial artificial chromosome
Egan, G., Silk, T., Zamarripa, F., et al., 2003. Neural correlates of the emergence of transgenic mice. Neuroscience 226, 489–509.
consciousness of thirst. Proc. Natl. Acad. Sci. U.S.A. 100 (25), 15241–15246. Gutkind, J.S., Kurihara, M., Saavedra, J.M., 1988. Increased angiotensin II receptors
Ehrlich, K.J., Fitts, D.A., 1990. Atrial natriuretic peptide in the subfornical organ in brain nuclei of DOCA-salt hypertensive rats. Am. J. Physiol. 255 (3 Pt 2),
reduces drinking induced by angiotensin or in response to water deprivation. H646–H650.
Behav. Neurosci. 104 (2), 365–372. Hainsworth, R., 2014. Cardiovascular control from cardiac and pulmonary vascular
Epstein, A.N., 1982. Mineralocorticoids and cerebral angiotensin may act together to receptors. Exp. Physiol. 99 (2), 312–319.
produce sodium appetite. Peptides 3 (3), 493–494. Hauser, W., Johren, O., Saavedra, J.M., 1998. Characterization and distribution of
Epstein, A.N., Fitzsimons, J.T., Rolls, B.J., 1970. Drinking induced by injection of angiotensin II receptor subtypes in the mouse brain. Eur. J. Pharmacol. 348 (1),
angiotensin into the rain of the rat. J. Physiol. 210 (2), 457–474. 101–114.
Epstein, A.N., Stellar, E., 1955. The control of salt preference in the adrenalectomized Hollis, J.H., McKinley, M.J., D’Souza, M., Kampe, J., Oldfield, B.J., 2008. The
rat. J. Comp. Physiol. Psychol. 48 (3), 167–172. trajectory of sensory pathways from the lamina terminalis to the insular and
Ferguson, A.V., 1988. Systemic angiotensin acts at the subfornical organ to control the cingulate cortex: a neuroanatomical framework for the generation of thirst. Am. J.
activity of paraventricular nucleus neurons with identified projections to the median Physiol. Regul. Integr. Comp. Physiol. 294 (4), R1390–R1401.
eminence. Neuroendocrinology 47 (6), 489–497. Huang, W., Sved, A.F., Stricker, E.M., 2000. Water ingestion provides an early signal
Ferguson, A.V., 2009. Angiotensinergic regulation of autonomic and neuroendocrine inhibiting osmotically stimulated vasopressin secretion in rats. Am. J. Physiol.
outputs: critical roles for the subfornical organ and paraventricular nucleus. Regul. Integr. Comp. Physiol. 279 (3), R756–R760.
Neuroendocrinology 89 (4), 370–376. Hurt, R.C., Garrett, J.C., Keifer Jr., O.P., et al., 2015. Angiotensin type 1a receptors on
Ferguson, A.V., Bains, J.S., 1997. Actions of angiotensin in the subfornical organ and corticotropin-releasing factor neurons contribute to the expression of conditioned
area postrema: implications for long term control of autonomic output. Clin. Exp. fear. Genes Brain Behav. 14 (7), 526–533.
Pharmacol. Physiol. 24 (1), 96–101. Jankord, R., Herman, J.P., 2008. Limbic regulation of hypothalamo-pituitary-adreno-
Findlay, A.L., Epstein, A.N., 1980. Increased sodium intake is somehow induced in rats cortical function during acute and chronic stress. Ann. N.Y. Acad. Sci. 1148,
by intravenous angiotensin II. Horm. Behav. 14 (1), 86–92. 64–73.
Fisher, J.P., Paton, J.F., 2012. The sympathetic nervous system and blood pressure Jezova, D., Ochedalski, T., Kiss, A., Aguilera, G., 1998. Brain angiotensin II modulates
in humans: implications for hypertension. J. Hum. Hypertens. 26 (8), 463–475. sympathoadrenal and hypothalamic pituitary adrenocortical activation during stress.
Fitts, D.A., Masson, D.B., 1990. Preoptic angiotensin and salt appetite. Behav. J. Neuroendocrinol. 10 (1), 67–72.
Neurosci. 104 (4), 643–650. Johnson, A.K., Gross, P.M., 1993. Sensory circumventricular organs and brain
Fitzsimons, J.T., 1961. Drinking by rats depleted of body fluid without increase in homeostatic pathways. FASEB J. 7 (8), 678–686.
osmotic pressure. J. Physiol. 159, 297–309. Johnson, A.K., Hoffman, W.E., Buggy, J., 1978. Attenuated pressor responses to
Fitzsimons, J.T., 1969. The role of a renal thirst factor in drinking induced by intracranially injected stimuli and altered antidiuretic activity following preoptic-
extracellular stimuli. J. Physiol. 201 (2), 349–368. hypothalamic periventricular ablation. Brain Res. 157 (1), 161–166.
Fitzsimons, J.T., 1998. Angiotensin, thirst, and sodium appetite. Physiol. Rev. 78 (3), Johren, O., Inagami, T., Saavedra, J.M., 1995. AT1A, AT1B, and AT2 angiotensin II
583–686. receptor subtype gene expression in rat brain. Neuroreport 6 (18), 2549–2552.
Fitzsimons, J.T., Kaufman, S., 1977. Cellular and extracellular dehydration, and Johren, O., Inagami, T., Saavedra, J.M., 1996. Localization of AT2 angiotensin II
angiotensin as stimuli to drinking in the common iguana Iguana iguana. J. Physiol. receptor gene expression in rat brain by in situ hybridization histochemistry. Mol.
265 (2), 443–463. Brain Res. 37 (1–2), 192–200.
Body Fluid Homeostasis 223

Johren, O., Saavedra, J.M., 1996. Expression of AT1A and AT1B angiotensin II receptor Nitabach, M.N., Schulkin, J., Epstein, A.N., 1989. The medial amygdala is part of
messenger RNA in forebrain of 2-wk-old rats. Am. J. Physiol. 271 (1 Pt 1), a mineralocorticoid-sensitive circuit controlling NaCl intake in the rat. Behav. Brain
E104–E112. Res. 35 (2), 127–134.
Kaiser, F.C., Palmer, G.C., Wallace, A.V., Carr, R.D., Fraser-Rae, L., Hallam, C., 1992. Oatley, K., 1964. Changes of blood volume and osmotic pressure in the production of
Antianxiety properties of the angiotensin II antagonist, DUP 753, in the rat using the thirst. Nature 202, 1341–1342.
elevated plus-maze. Neuroreport 3 (10), 922–924. Oldfield, B.J., Miselis, R.R., McKinley, M.J., 1991. Median preoptic nucleus projections
Kakar, S.S., Riel, K.K., Neill, J.D., 1992. Differential expression of angiotensin II to vasopressin-containing neurones of the supraoptic nucleus in sheep. A light and
receptor subtype mRNAs (AT-1A and AT-1B) in the brain. Biochem. Biophys. Res. electron microscopic study. Brain Res. 542 (2), 193–200.
Commun. 185 (2), 688–692. Osborn, J.W., Hendel, M.D., Collister, J.P., Ariza-Guzman, P.A., Fink, G.D., 2012. The
Karst, H., Berger, S., Turiault, M., Tronche, F., Schutz, G., Joels, M., 2005. Miner- role of the subfornical organ in angiotensin II-salt hypertension in the rat. Exp.
alocorticoid receptors are indispensable for nongenomic modulation of hippo- Physiol. 97 (1), 80–88.
campal glutamate transmission by corticosterone. Proc. Natl. Acad. Sci. U.S.A. Palkovits, M., Bahner, U., Geiger, H., 1995. Preoptic neuronal circuit: atrial natriuretic
102 (52), 19204–19207. peptide-containing neurons are sensitive to acute and chronic alterations in body
Khoury, N.M., Marvar, P.J., Gillespie, C.F., et al., 2012. The renin-angiotensin pathway fluid volume. Min. Electrolyte Metabol. 21 (6), 423–427.
in posttraumatic stress disorder: angiotensin-converting enzyme inhibitors and Rennke, H.G., Denker, B.M., 2010. Renal Pathophysiology: The Essentials, third ed.
angiotensin receptor blockers are associated with fewer traumatic stress symp- Wolters Kluwer/Lippincott Williams & Wilkins, Baltimore, MD, USA.
toms. J. Clin. Psychiatry 73 (6), 849–855. Rice, K.K., Richter, C.P., 1943. Increased sodium chloride and water intake of
Kinsman, B., Cowles, J., Lay, J., Simmonds, S.S., Browning, K.N., Stocker, S.D., normal rats treated with desoxycorticosterone acetate. Endocrinology 33 (2),
2014. Osmoregulatory thirst in mice lacking the transient receptor potential 106–115.
vanilloid type 1 (TRPV1) and/or type 4 (TRPV4) receptor. Am. J. Physiol. Regul. Richardson, D.B., Mogenson, G.J., 1981. Water intake elicited by injections of
Integr. Comp. Physiol. 307 (9), R1092–R1100. angiotensin II into preoptic area of rats. Am. J. Physiol. 240 (1), R70–R74.
Koneru, B., Bathina, C.S., Cherry, B.H., Mifflin, S.W., 2014. Mineralocorticoid receptor Richter, C.P., 1936. Increased salt appetite in adrenalectomized rats. Am. J. Physiol.
in the NTS stimulates saline intake during fourth ventricular infusions of aldoste- 115 (1), 155–161.
rone. Am. J. Physiol. Regul. Integr. Comp. Physiol. 306 (1), R61–R66. Richter, C.P., 1939. Salt taste thresholds of normal and adrenalectomized rats.
Krause, E.G., de Kloet, A.D., Flak, J.N., et al., 2011a. Hydration state controls stress Endocrinology 24 (3), 367–371.
responsiveness and social behavior. J. Neurosci. 31 (14), 5470–5476. Richter, C.P., 1956. Salt appetite of mammals: its dependence on instinct and
Krause, E.G., de Kloet, A.D., Scott, K.A., et al., 2011b. Blood-borne angiotensin II acts metabolism. In: L’instinct dans le comportement des animaux et de l’homme,
in the brain to influence behavioral and endocrine responses to psychogenic stress. pp. 577–632. Paris, France.
J. Neurosci. 31 (42), 15009–15015. Richter, C.P., Eckert, J.F., 1938. Mineral metabolism of adrenalectomized rats studied
Krause, E.G., Melhorn, S.J., Davis, J.F., et al., 2008. Angiotensin type 1 receptors in by the appetite method. Endocrinology 22 (2), 214–224.
the subfornical organ mediate the drinking and hypothalamic-pituitary-adrenal Robinson, B.W., Mishkin, M., 1968. Alimentary responses to forebrain stimulation in
response to systemic isoproterenol. Endocrinology 149 (12), 6416–6424. monkeys. Exp. Brain Res. 4 (4), 330–366.
Krause, E.G., Sakai, R.R., 2007. Richter and sodium appetite: from adrenalectomy to Rubin, M.I., Krick, E.T., 1933. Effect of adrenalectomy on salt metabolism in rats.
molecular biology. Appetite 49 (2), 353–367. Proc. Soc. Exp. Biol. Med. 31 (2), 228–229.
Kubo, T., Numakura, H., Endo, S., Hagiwara, Y., Fukumori, R., 2001. Angiotensin Saavedra, J.M., 1990. Interactions between the circulating hormones angiotensin and
receptor blockade in the anterior hypothalamic area inhibits stress-induced pressor atrial natriuretic peptide and their receptors in brain. Adv. Exp. Med. Biol. 274,
responses in rats. Brain Res. Bull. 56 (6), 569–574. 191–210.
Kuramochi, G., Kobayashi, I., 2000. Regulation of the urine concentration mechanism Saavedra, J.M., 2011. Angiotensin II AT(1) receptor blockers ameliorate inflammatory
by the oropharyngeal afferent pathway in man. Am. J. Nephrol. 20 (1), 42–47. stress: a beneficial effect for the treatment of brain disorders. Cell. Mol. Neurobiol.
Lenkei, Z., Palkovits, M., Corvol, P., Llorens-Cortès, C., 1997. Expression of angio- Saavedra, J.M., Armando, I., Bregonzio, C., et al., 2006. A centrally acting, anxiolytic
tensin Type-1 (AT1) and Type-2 (AT2) receptor mRNAs in the adult rat brain: angiotensin II AT1 receptor antagonist prevents the isolation stress-induced
a functional neuroanatomical review. Front. Neuroendocrinol. 18 (4), 383. decrease in cortical CRF1 receptor and benzodiazepine binding. Neuro-
Leshem, M., 2011. Low dietary sodium is anxiogenic in rats. Physiol. Behav. 103 (5), psychopharmacology 31 (6), 1123–1134.
453–458. Sadoshima, J., Qiu, Z., Morgan, J.P., Izumo, S., 1995. Angiotensin II and other
Lotter, E.C., McKay, L.D., Mangiapane, M.L., et al., 1980. Intraventricular angiotensin hypertrophic stimuli mediated by G protein-coupled receptors activate tyrosine
elicits drinking in the baboon. Proc. Soc. Exp. Biol. Med. 163 (1), 48–51. kinase, mitogen-activated protein kinase, and 90-kD S6 kinase in cardiac myo-
Ma, L.Y., McEwen, B.S., Sakai, R.R., Schulkin, J., 1993. Glucocorticoids facilitate cytes. The critical role of Ca(2þ)-dependent signaling. Circ. Res. 76 (1), 1–15.
mineralocorticoid-induced sodium intake in the rat. Horm. Behav. 27 (2), Sakai, R.R., Chow, S.Y., Epstein, A.N., 1990. Peripheral angiotensin II is not the cause
240–250. of sodium appetite in the rat. Appetite 15 (3), 161–170.
MacTaggart, T.E., Ito, M., Smithies, O., John, S.W., 1997. Mouse angiotensin receptor Sakai, R.R., McEwen, B.S., Fluharty, S.J., Ma, L.Y., 2000. The amygdala: site of
genes Agtr1a and Agtr1b map to chromosomes 13 and 3. Mamm. Genome 8 (4), genomic and nongenomic arousal of aldosterone-induced sodium intake. Kidney
294–295. Int. 57 (4), 1337–1345.
McCann, S.M., Gutkowska, J., Antunes-Rodrigues, J., 2003. Neuroendocrine control Sakai, R.R., Nicolaidis, S., Epstein, A.N., 1986. Salt appetite is suppressed by
of body fluid homeostasis. Braz. J. Med. Biol. Res. 36 (2), 165–181. interference with angiotensin II and aldosterone. Am. J. Physiol. 251 (4 Pt 2),
McCann, S.M., Gutkowska, J., Franci, C.R., Favaretto, A.L., Antunes-Rodrigues, J., R762–R768.
1994. Hypothalamic control of water and salt intake and excretion. Braz. J. Med. Sampey, D.B., Burrell, L.M., Widdop, R.E., 1999. Vasopressin V2 receptor enhances
Biol. Res. 27 (4), 865–884. gain of baroreflex in conscious spontaneously hypertensive rats. Am. J. Physiol.
McEwen, B.S., Lambdin, L.T., Rainbow, T.C., De Nicola, A.F., 1986. Aldosterone effects 276 (3 Pt 2), R872–R879.
on salt appetite in adrenalectomized rats. Neuroendocrinology 43 (1), 38–43. Sasamura, H., Hein, L., Krieger, J.E., Pratt, R.E., Kobilka, B.K., Dzau, V.J., 1992. Cloning,
McKinley, M.J., Denton, D.A., Leventer, M., Penschow, J., Weisinger, R.S., characterization, and expression of two angiotensin receptor (AT-1) isoforms from the
Wright, R.D., 1983. Morphology of the organum vasculosum of the lamina ter- mouse genome. Biochem. Biophys. Res. Commun. 185 (1), 253–259.
minalis (OVLT) of the sheep. Brain Res. Bull. 11 (6), 649–657. Saxena, A., Little, J.T., Nedungadi, T.P., Cunningham, J.T., 2014. Angiotensin II type
McKinley, M.J., McAllen, R.M., Davern, P., et al., 2003. The sensory circumventricular 1a receptors in subfornical organ contributes towards chronic intermittent hypoxia
organs of the mammalian brain. Adv. Anat. Embryol. Cell Biol. 172, III–XII, 1-122, associated sustained increase in mean arterial pressure. Am. J. Physiol. 308 (5),
back cover. H435–H446.
Mendelsohn, F.A., Quirion, R., Saavedra, J.M., Aguilera, G., Catt, K.J., 1984. Auto- Schwob, J.E., Johnson, A.K., 1977. Angiotensin-induced dipsogenesis in domestic
radiographic localization of angiotensin II receptors in rat brain. Proc. Natl. Acad. fowl (Gallus gallus). J. Comp. Physiol. Psychol. 91 (1), 182–188.
Sci. U.S.A. 81 (5), 1575–1579. Schweda, F., Kurtz, A., 2011. Regulation of renin release by local and systemic
Nachman, M., 1962. Taste preferences for sodium salts by adrenalectomized rats. factors. Rev. Physiol. Biochem. Pharmacol. 161, 1–44.
J. Comp. Physiol. Psychol. 55, 1124–1129. Sharif Naeini, R., Witty, M.F., Seguela, P., Bourque, C.W., 2006. An N-terminal variant
Negoro, H., Higuchi, T., Tadokoro, Y., Honda, K., 1988. Osmoreceptor mechanism for of Trpv1 channel is required for osmosensory transduction. Nat. Neurosci. 9 (1),
oxytocin release in the rat. Jpn. J. Physiol. 38 (1), 19–31. 93–98.
224 Body Fluid Homeostasis

Shelat, S.G., Flanagan-Cato, L.M., Fluharty, S.J., 1999a. Glucocorticoid and miner- Thrasher, T.N., Keil, L.C., 1987. Regulation of drinking and vasopressin secretion:
alocorticoid regulation of angiotensin II type 1 receptor binding and inositol role of organum vasculosum laminae terminalis. Am. J. Physiol. 253 (1 Pt 2),
triphosphate formation in WB cells. J. Endocrinol. 162 (3), 381–391. R108–R120.
Shelat, S.G., King, J.L., Flanagan-Cato, L.M., Fluharty, S.J., 1999b. Mineralocorticoids Thunhorst, R.L., Beltz, T.G., Johnson, A.K., 1999. Effects of subfornical organ lesions
and glucocorticoids cooperatively increase salt intake and angiotensin II receptor on acutely induced thirst and salt appetite. Am. J. Physiol. Regul. Integr. Comp.
binding in rat brain. Neuroendocrinology 69 (5), 339–351. Physiol. 277 (1), R56–R65.
Shi, P., Martinez, M.A., Calderon, A.S., Chen, Q., Cunningham, J.T., Toney, G.M., Thunhorst, R.L., Lewis, S.J., Johnson, A.K., 1994. Effects of sinoaortic baroreceptor
2008. Intra-carotid hyperosmotic stimulation increases Fos staining in forebrain denervation on depletion-induced salt appetite. Am. J. Physiol. 267 (4 Pt 2),
organum vasculosum laminae terminalis neurones that project to the hypothalamic R1043–R1049.
paraventricular nucleus. J. Physiol. 586 (21), 5231–5245. Toth, E., Stelfox, J., Kaufman, S., 1987. Cardiac control of salt appetite. Am. J.
Simpson, J.B., Routtenberg, A., 1973. Subfornical organ: site of drinking elicitation by Physiol. 252 (5 Pt 2), R925–R929.
angiotensin II. Science 181 (4105), 1172–1175. Tsutsumi, K., Saavedra, J.M., 1991. Characterization and development of angiotensin
Simpson, J.B., Routtenberg, A., 1978. Subfornical organ – dipsogenic site of action of II receptor subtypes (AT1 and AT2) in rat brain. Am. J. Physiol. 261 (1 Pt 2),
angiotensin-Ii. Science 201 (4353), 379–380. R209–R216.
Sjoquist, M., Huang, W., Jacobsson, E., Skott, O., Stricker, E.M., Sved, A.F., 1999. Vanhouten, M., Schiffrin, E.L., Mann, J.F.E., Posner, B.I., Boucher, R., 1980. Auto-
Sodium excretion and renin secretion after continuous versus pulsatile infusion of radiographic localization of specific binding-sites for blood-borne angiotensin-Ii in
oxytocin in rats. Endocrinology 140 (6), 2814–2818. the rat-brain. Brain Res. 186 (2), 480–485.
Smith, J.A., Wang, L., Hiller, H., Taylor, C.T., de Kloet, A.D., Krause, E.G., 2014. Acute Velloso, L.A., Folli, F., Sun, X.J., White, M.F., Saad, M.J., Kahn, C.R., 1996. Cross-talk
hypernatremia promotes anxiolysis and attenuates stress-induced activation of the between the insulin and angiotensin signaling systems. Proc. Natl. Acad. Sci.
hypothalamic-pituitary-adrenal axis in male mice. Physiol. Behav. 136, 91–96. U.S.A. 93 (22), 12490–12495.
Soares, T.J., Coimbra, T.M., Martins, A.R., et al., 1999. Atrial natriuretic peptide and Verbalis, J.G., Mangione, M.P., Stricker, E.M., 1991. Oxytocin produces natriuresis in
oxytocin induce natriuresis by release of cGMP. Proc. Natl. Acad. Sci. U.S.A. 96 (1), rats at physiological plasma concentrations. Endocrinology 128 (3), 1317–1322.
278–283. Weisinger, R.S., BlairWest, J.R., Burns, P., Denton, D.A., Tarjan, E., 1997. Role of
Starbuck, E.M., Wilson, W.L., Fitts, D.A., 2002. Fos-like immunoreactivity and thirst brain angiotensin in thirst and sodium appetite of rats. Peptides 18 (7), 977–984.
following hyperosmotic loading in rats with subdiaphragmatic vagotomy. Brain Res. Weisinger, R.S., Considine, P., Denton, D.A., McKinley, M.J., 1979. Rapid effect of
931 (2), 159–167. change in cerebrospinal fluid sodium concentration on salt appetite. Nature 280
Stellar, E., Epstein, A.N., 1991. Neuroendocrine factors in salt appetite. J. Physiol. (5722), 490–491.
Pharmacol. 42 (4), 345–355. Weisinger, R.S., Denton, D.A., Dinicolantonio, R., et al., 1990. Subfornical organ lesion
Stocker, S.D., Sved, A.F., Stricker, E.M., 2000. Role of renin-angiotensin system in decreases sodium appetite in the sodium-depleted rat. Brain Res. 526 (1), 23–30.
hypotension-evoked thirst: studies with hydralazine. Am. J. Physiol. Regul. Integr. Weisinger, R.S., Denton, D.A., Dinicolantonio, R., Mckinley, M.J., 1987a. Effect of
Comp. Physiol. 279 (2), R576–R585. captopril on the Na appetite of Na-deplete sheep. Aust. N.Z. J. Med. 17 (1), 170.
Stricker, E.M., 1966. Extracellular fluid volume and thirst. Am. J. Physiol. 211 (1), Weisinger, R.S., Denton, D.A., Dinicolantonio, R., Mckinley, M.J., Muller, A.F.,
232–238. Tarjan, E., 1987b. Role of angiotensin in sodium appetite of sodium-deplete sheep.
Stricker, E.M., 1969. Osmoregulation and volume regulation in rats: inhibition of Am. J. Physiol. 253 (3), R482–R488.
hypovolemic thirst by water. Am. J. Physiol. 217 (1), 98–105. Weisinger, R.S., Woods, S.C., 1971. Aldosterone-elicited sodium appetite. Endocri-
Stricker, E.M., 1971. Effects of hypovolemia and-or caval ligation on water and NaCl nology 89 (2), 538–544.
solution drinking by rats. Physiol. Behav. 6 (4), 299–305. Wilkins, L., Richter, C.P., 1940. A great craving for salt by a child with corticoadrenal
Stricker, E.M., Verbalis, J.G., 1986. Interaction of osmotic and volume stimuli in insufficiency. J. Am. Med. Assoc. 114, 866–868.
regulation of neurohypophyseal secretion in rats. Am. J. Physiol. 250 (2 Pt 2), Wilson, K.M., Sumners, C., Hathaway, S., Fregly, M.J., 1986. Mineralocorticoids
R267–R275. modulate central angiotensin-Ii receptors in rats. Brain Res. 382 (1), 87–96.
Stricker, E.M., Vagnucci, A.H., Mcdonald, R.H., Leenen, F.H., 1979. Renin and Wolf, G., 1964. Effect of dorsolateral hypothalamic lesions on sodium appetite elicited
aldosterone secretions during hypovolemia in rats – relation to Nacl intake. Am. J. by desoxycorticosterone and by acute hyponatremia. J. Comp. Physiol. Psychol.
Physiol. 237 (1), R45–R51. 58, 396–402.
Sumners, C., de Kloet, A.D., Krause, E.G., Unger, T., Steckelings, U.M., 2015. Wolf, G., 1965. Effect of deoxycorticosterone on sodium appetite of intact and
Angiotensin type 2 receptors: blood pressure regulation and end organ damage. adrenalectomized rats. Am. J. Physiol. 208, 1281–1285.
Curr. Opin. Pharmacol. 21, 115–121. Wolf, G., Handal, P.J., 1966. Aldosterone-induced sodium appetite: dose-response
Sumners, C., Fregly, M.J., 1989. Modulation of angiotensin II binding sites in neuronal and specificity. Endocrinology 78 (6), 1120–1124.
cultures by mineralocorticoids. Am. J. Physiol. 256 (1 Pt 1), C121–C129. Yamaguchi, K., Hama, H., 2011. Changes in vasopressin release and autonomic
Tanaka, J., Hayashi, Y., Sakamaki, K., Okumura, T., Nomura, M., 2001. Activation of function induced by manipulating forebrain GABAergic signaling under euvolemia
the subfornical organ enhances extracellular noradrenaline concentrations in the and hypovolemia in conscious rats. Endocr. J. 58 (7), 559–573.
hypothalamic paraventricular nucleus in the rat. Brain Res. Bull. 54 (4), 421–425. Yang, G., Xi, Z.X., Wan, Y., Wang, H., Bi, G., 1993. Changes in circulating and tissue
Taylor, A.C., McCarthy, J.J., Stocker, S.D., 2008. Mice lacking the transient receptor angiotensin II during acute and chronic stress. Biol. Signals 2 (3), 166–172.
vanilloid potential 1 channel display normal thirst responses and central Fos Yayama, K., Okamoto, H., 2008. Angiotensin II-induced vasodilation via type 2
activation to hypernatremia. Am. J. Physiol. Regul. Integr. Comp. Physiol. 294 (4), receptor: role of bradykinin and nitric oxide. Int. Immunopharmacol. 8 (2),
R1285–R1293. 312–318.
1.08 Stress and Opioid Systems
Ryszard Przewlocki, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
Osborne FX Almeida, Max Planck Institute of Psychiatry, Munich, Germany
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by R. Przewlocki, volume 1, pp. 289–332, Ó 2009, Elsevier Inc.

1.08.1 Introduction: The Concept of Stress 226


1.08.2 Opioid Systems 227
1.08.2.1 Multiple Opioid Peptides 227
1.08.2.2 Opioid Receptor Subtypes and Their Ligands 227
1.08.3 Distribution of Opioid Peptides and Receptors in the Stress Network 232
1.08.3.1 Proopiomelanocortin System 232
1.08.3.2 Proenkephalin System 232
1.08.3.3 Prodynorphin System 232
1.08.3.4 Endomorphins 232
1.08.3.5 Opioid Receptors 232
1.08.4 Stress-Induced Alterations in Endogenous Opioidergic Systems 233
1.08.4.1 Proopiomelanocortin System 233
1.08.4.2 Proenkephalin System 234
1.08.4.3 Prodynorphin System 234
1.08.4.4 Opioid Receptors 235
1.08.5 Multiple Neurochemical Interactions with Opioid System as a Consequence of Stress 236
1.08.5.1 Dopaminergic System 236
1.08.5.2 Adrenergic System 236
1.08.6 Involvement of Opioids in Modulation of the HPA Axis 237
1.08.6.1 Corticotropin Releasing Factor 237
1.08.6.2 Vasopressin System 238
1.08.7 Involvement of Opioids in the Behavioral and Physiological Response to Stress 238
1.08.7.1 Locomotor Activity 238
1.08.7.2 Reward 239
1.08.7.3 Nociception: SIA 240
1.08.7.4 Thermoregulation: Stress-Induced Hyperthermia 243
1.08.7.5 Feeding Behavior 243
1.08.7.6 Cardiovascular Effects 244
1.08.7.7 Respiration 245
1.08.7.8 Reproduction 245
1.08.7.9 Autonomic Nervous System 245
1.08.7.10 Immune System 246
1.08.8 Therapeutic Potential of Opioid Agonists and Antagonists in Stress-Related Psychiatric Disorders 247
1.08.9 Opioidergic Control of Stress Responses – Conclusions 248
Acknowledgments 248
References 249

Nomenclature
ACTH adrenocorticotropic hormone GAS general adaptation syndrome
CRF corticotrophin-releasing factor HPA hypothalamo–pituitary–adrenal axis
CTOP D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH 2 (MOP ICI 174,864 [allyl]2-Tyr-alpha-amino-isobutyric acid
receptor selective agonist) (Aib)-Aib-Phe-Leu-OH (DOP receptor antagonist)
DAMGO [D-Ala2, MePhe4, Gly-ol]-enkephalin (selective m- ICSS intracranial self-stimulation
receptor agonist) IFN-a interferon-a
DAT dopamine transporter KOP k-opioid peptide
DOP d-opioid peptide LH lateral hypothalamus
EOPs endogenous opioid peptides LPS lipopolysaccharide
b-FNA beta-funaltrexamine (MOP receptor antagonist) MOP m-opioid peptide

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00008-0 225


226 Stress and Opioid Systems

Mr2266 (1R,5R,9R)-5,9-diethyl-2-(3-furylmethyl)-20 - POMC proopiomelanocortin


hydroxy-6,7-benzomorphan (opioid antagonist) RU (11b,17b)-11-[4-(dimethylamino)phenyl]-17-
MSH melanocyte-stimulating hormone hydroxy-17-(1-propynyl)estra-4,9-dien-3-one
NTS nucleus tractus solitarii SIA stress-induced analgesia
PDYN prodynorphin U50,488H trans-()-3,4-dichloro-N-methyl-N-[7-(1-
PENK proenkephalin pyrrolidinyl)cyclohexyl] (selective k-receptor agonist)

1.08.1 Introduction: The Concept of Stress reserves (e.g., energy) to help it cope. Selye referred to the third
phase of the stress response as the exhaustion stage when the
The term ‘stress’ refers to the mental or bodily tension that organism has failed to overcome or adapt successfully to the
occurs upon experiencing a stressful stimulus (‘stressor’) that environmental challenge, thereby increasing its chances of death.
challenges the organism’s homeostatic mechanisms and poten- While earlier theories of stress tended to focus on physiolog-
tially threatening its health and well-being. The stress response ical responses only, it is now common to also consider the
is the compensatory reaction of the body to the disturbance emotional and psychological aspects of stress. Importantly,
imposed by the stressor. Generally, the stress response evolves stressors that are not accompanied by an emotional component
as an adaptive mechanism, but in extreme cases, depending on elicit only minor physiological responses (Mason, 1971). Munck
the quality, intensity, or duration of the noxious stimulus, the et al. (1984) suggested that the initial response was specific to
response can produce maladaptive responses (McEwen and a particular stressor and targeted toward maintaining homeo-
Wingfield, 2003; de Kloet et al., 2005). Importantly, the indi- stasis. Accordingly, the subsequent and slower hormonal
vidual’s state at the moment the stressor arrives, is also an response of the HPA axis is a response to the initial behavioral
important, albeit often neglected, consideration; it is important response rather than to the stressor per se. In an important
because the way the individual perceives the stressor will deter- conceptual advance, Kopin et al. (1988) defined stress as a state
mine its subsequent psychological as well as physiological reac- in which expectations did not match current or anticipated
tion (Lovallo, 1997; Koolhaas et al., 2011). It is worthwhile to perceptions of the internal or external environment and that
briefly mention some historical landmarks that helped make the maintenance of homeostasis involves (usually) unconscious
stress the intensely studied subject it is today. adjustments in multiple systems; failure in any of these would
The idea of the critical need to maintain constancy of the lead to a state of distress associated with emotional components
milieu de l’intérieur (internal environment) was introduced by such as fear or anxiety. All physical stressors have a psychological
Claude Bernard in the mid-nineteenth century and later extended component (Lovallo, 1997), and as alluded to above, physiolog-
by Walter Cannon (1939) who introduced the term homeostasis to ical and psychological factors are equally important determi-
describe physiological reactions which maintain the steady state nants of the stress response.
of the organism in the face of external stimuli. Cannon noted An activated HPA axis is the most overt and easily measure-
that the brain plays an important role in maintaining homeo- able physiological response to stress. Stress stimulates the secre-
stasis as it communicates with the rest of the body via specialized tion of proopiomelanocortin (POMC)-derived peptides such
sensory nerves to recognize the external threats and to detect the as adrenocorticotropic hormone (ACTH) and opioid peptide
internal state of the body. He also suggested that the brain can b-endorphin as well as corticotropin releasing factor (CRF)
activate multiple mechanisms to compensate for any disequilib- and glucocorticoids (GCs). This reaction is accompanied by
rium caused, and importantly, he observed that psychological autonomic responses, namely, activation of the sympathetic
as well as physiological disturbances could elicit responses nervous system and the release of noradrenaline from sympa-
from the sympathetic nervous system and adrenals. thetic nerve endings and adrenaline from the adrenal medulla.
Today’s concept of stress was framed first by Selye (1936). He These changes trigger rapid alterations in cardiovascular and
observed that, irrespective of the nature (or identity) of the immune system functioning.
stressor(s), the physiological changes were relatively similar The discovery of the endogenous opioid peptides (EOPs),
and were primarily mediated by the hypothalamo–pituitary– and research into their physiology, pharmacology, and molec-
adrenal (HPA) axis. He used the term ‘stress’ to refer to the phys- ular biology, provided new insights into our understanding of
iological response to ‘stressor’ such as heat, cold, toxic, and infec- stress. The EOPs modulate many behaviors and affect physio-
tious agents. He discovered that exposure to stressors caused logical functions in both the brain and periphery, ranging
enlargement of the adrenal cortex, reduction of thymus and from motor activity and nociceptive threshold to feeding and
lymph gland weights, and appearance of stomach ulcers. Selye sexual behavior; all of these functions are sensitive to the dis-
described the process in terms of a three-stage model called the rupting effects of stress and interestingly encompass interac-
General Adaptation Syndrome (GAS). In the first, alarm stage, tions between central opioidergic pathways, neuroendocrine
there is an adaptive reaction to the external challenge that is likely systems, the sympathetic nervous system, and the immune
to be flight or fight; this involves increased heart rate, pupillary system. The many nodes at which EOP can act to influence
dilatation, a stop to digestion, muscle tension, and activation physiology and behavior suggest a potentially important role
of the adrenals. If this reaction is unsuccessful, the organism for them in facilitating the organism’s interaction with, and
enters the state of arousal or resistance for which it uses existing response to, the ever-changing external environment.
Stress and Opioid Systems 227

In this chapter, we will discuss the possible involvement of (Evans et al., 1992; Kieffer et al., 1992), then the MOP receptor
opioids in mediating physiological as well as behavioral reac- (Chen et al., 1993a; Fukuda et al., 1993; Thompson et al.,
tions to stress. Further, we will focus on the neurobiological 1993), and lastly, the k-opioid peptide (KOP) receptor (Chen
mechanisms that underlie interactions between EOP systems et al., 1993b; Li et al., 1993; Meng et al., 1993; Minami et al.,
and other stress-processing pathways. We will also discuss alter- 1993; Nishi et al., 1993). All three receptors share extensive
ations in EOP systems and multiple opioid receptors which structural homologies and belong to the family of seven trans-
occur as a result of the organism’s reaction and adaptation to membrane G-protein-coupled receptors (GPCR). In some
stressful stimuli. Lastly, we will consider current research on cases, certain EOPs bind to more than a single opioid
the use of opioid receptor agonists and antagonists for the treat- receptor, albeit with differing affinities; the binding properties
ment of stress-related disorders in psychiatry. of EOPs and of related opioidomimetics are reviewed
elsewhere (Okada et al., 2002, 2003). Generally, the
enkephalins and dynorphins are considered to be high-
1.08.2 Opioid Systems affinity agonists of the DOP and KOP receptors, respectively.
However, b-endorphin binds similarly to both MOP and
1.08.2.1 Multiple Opioid Peptides
DOP receptors.
Over the last two decades, our understanding of the biogenesis Functional studies led to postulation that different subtypes
of various EOPs, their anatomical distribution, and characteris- of main opioid receptors exist (Cox et al., 2015). That postulate
tics of the multiple receptors with which they interact, has was confirmed by molecular analysis which also showed the
advanced considerably. The EOPs derive from three different existence of several variants of opioid receptor subtypes (Pas-
precursor proteins: POMC, prodynorphin (PDYN), and proen- ternak and Pan, 2013; Dietis et al., 2011). In addition, oligo-
kephalin (PENK), which were cloned in the late 1970s and early merization of various opioid receptors has been shown to
1980s (Nakanishi et al., 1979; Kakidani et al., 1982; Noda et al., generate receptors with unique functional properties; for
1982). Details concerning EOP discoveries, their body and brain example, an association between DOP receptors and MOP
distribution, and properties have been reviewed elsewhere receptors was recently demonstrated, with the occupancy of
(Höllt, 1990). The main groups derived from PENK, PDYN, DOP receptors (by DOP receptor antagonists) enhancing
and POMC are enkephalins, dynorphins, and b-endorphin, MOP receptor binding and signaling activity (Alfaras-Melainis
respectively. PENK is the source of Met- and Leu-enkephalins et al., 2009).
and several longer peptides. EOPs such as dynorphin A, dynor- Pharmacological studies have led to the description of
phin B and a- and b-neoendorphin and several larger molecules several opioid receptor subtypes (MOP1, MOP2; DOP1,
are generated from PDYN. POMC is the precursor of b-endor- DOP2; KOP1, KOP2, KOP3) (Dietis et al., 2011). While
phin, a-endorphin, and several nonopioid peptides. Recently, such categorization has not been confirmed by molecular
a novel group of brain peptides, the endomorphins, biology to date, the existence of different variants of the
endomorphin-1 (Tyr-Pro-Trp-Phe-NH2) and endomorphin-2 known opioid receptors has been suggested (Koch et al.,
(Tyr-Pro-Phe-Phe-NH2), have been discovered. These unique 1998; Uhl et al., 1999; Abbadie et al., 2000; Pasternak and
EOPs have a characteristic structure and anatomical distribution Pan, 2000). Alternatively spliced MOP receptor subtypes
and show high selectivity for the m-opioid peptide (MOP) have in fact been described in mice (Pan et al., 1999, 2000,
receptor (Zadina et al., 1997). 2001, 2003), rats (Zimprich et al., 1995), and humans (Bare
EOP-containing neurons are commonly found in tissues that et al., 1994). The particular MOP receptor domains encoded
make up the HPA axis, namely the hypothalamus, pituitary, and by various exons of MOP-1 mediate different mechanisms
adrenal glands and all of which are involved in mounting the of MOP receptor activation (Schuller et al., 1999), and various
neuroendocrine response to stress. In addition, central and MOP receptor ligands, acting at different MOP-1 splice vari-
peripheral opioidergic neurons innervate the autonomic ants, can be ranked differentially according to efficacy
nervous system centers. Further, EOPs are widely distributed in (Pasternak, 2004). Although it is not known how the alterna-
brain areas (Figure 1(a) and 1(b)), also in structures associated tive variants correspond to the pharmacologically defined
with emotional components of stress. It is important to note receptors types, their existence may help explain some of
that, most EOPs are usually not tonically active, and hence, various phenomena observed in clinical settings, e.g., incom-
opioid antagonists have little or no effect under homeostatic plete cross-tolerance to different MOP receptor agonists
states. However, they are activated by stressful stimuli and act (Pasternak, 2004). Interestingly, genetic studies suggest that
to influence the physiological responses to stress. Interestingly, humans carrying one or two copies of the 118G MOP receptor
b-endorphin derives from the same precursor (POMC) as that allele may be at increased risk for opiate and alcohol addic-
from which adrenocorticotropin (ACTH) is cleaved, and both tion (Kreek and LaForge, 2007), and of pertinence to the
molecules are cosecreted by hypothalamic neurons and present chapter, Lovallo (2006) suggested individuals with
pituitary corticotropes. Colocalization and corelease also an altered MOP receptor allele coding for the high-affinity
occurs in the case of adrenaline and the enkephalins in the MOP receptor may be more vulnerable to stress and more
adrenals, and the dynorphins and CRF in hypothalamic neurons. prone to develop addiction as a consequence of stressful situ-
ation. The pharmacologically defined subtypes of KOP
receptor have also been proposed to have functional signifi-
1.08.2.2 Opioid Receptor Subtypes and Their Ligands
cance; for example, dopamine and acetylcholine release in
Three members of opioid receptor family were cloned in the rat neostriatum was shown to be modulated by KOP1 and
early 1990s, first, the mouse d-opioid peptide (DOP) receptor KOP receptors, respectively (Schoffelmeer et al., 1997).
228 Stress and Opioid Systems

Figure 1 Anatomical distribution of opioid peptides (a) their place of gene expression (b) and opioid receptors and (c) and sites of their expression
(d) in the rat brain. Colors correspond to each of the three opioid receptor or peptide precursor. Densities are represented by symbols from low
(L) medium (M) to high (D). AD, anterodorsal thalamus; AL, anterior lobe, pituitary; AMB, nucleus ambiguus; AON, anterior olfactory nucleus; ARC,
arcuate nucleus, hypothalamus; BLA, basolateral nucleus, amygdala; BNST, bed nucleus of the stria terminalis; CEA, central nucleus, amygdala; Cl,
claustrum; CL, centrolateral thalamus; CM, centromedial thalamus; COA, cortical nucleus, amygdala; CPU, caudate putamen; C, cerebellum; DMH, dor-
somedial hypothalamus; DMR, dorsal and medial raphe; DR, dorsal raphe; DTN, dorsal tegmental nucleus; EN, endopiriform cortex; ENT, entorhinal
cortex; FRCX, frontal cortex; G, nucleus gelatinosus, thalamus; GP, globus pallidus; HbL, lateral habenula; HbM, medial habenula; HPC, hippocampus;
IL, intermediate lobe, pituitary; IP, interpeduncular nucleus; LC, locus coeruleus; LD, laterodorsal thalamus; LG, lateral geniculate, thalamus; LH, lateral
hypothalamus; LRN, lateral reticular nucleus; MD, mediodorsal thalamus; ME, median eminence; MEA, median nucleus, amygdala; MG, medial genicu-
late; MM, medial mammillary nucleus; NAc, nucleus accumbens; NL, neuronal lobe, pituitary; NRGC, nucleus reticularis gigantocellularis; NST, nucleus
tractus solitarius; OB, olfactory bulb; OCX, occipital cortex; PAG, periaqueductal gray; PCX, parietal cortex; PIR, piriform cortex; PN, pontine nucleus;
PNR, pontine reticular; PO, posterior thalamus; POA, preoptic area; PV, paraventricular thalamus; PVN, paraventricular hypothalamus; RE, reuniens
Stress and Opioid Systems 229

Figure 1 (continued).

=thalamus; RM, raphe magnus; S, septum; SON, supraoptic nucleus; SN, substantia nigra; STN, spinal trigeminal nucleus; TCx, temporal cortex; Tu,
olfactory tubercle; VL, ventrolateral thalamus; VM, ventromedial thalamus; VMH, ventromedial hypothalamus; VP, ventral pallidum; VPL, ventropostero-
lateral thalamus; VR, ventral raphe; VTA, ventral tegmental area; ZI, zona incerta. Adapted from Le Merrer, J., Becker, J.A., Befort, K., Kieffer, B.L.,
2009. Reward processing by the opioid system in the brain. Physiol. Rev. 89 (4), 1379–1412; Erbs, E., Faget, L., Veinante, P., Kieffer, B.L., Massotte,
D., 2014. In vivo neuronal co-expression of mu and delta opioid receptors uncovers new therapeutic perspectives. Recept. Clin. Investig. 1 (5), 210.
230 Stress and Opioid Systems

Figure 1 (continued).
Stress and Opioid Systems 231

Figure 1 (continued).
232 Stress and Opioid Systems

1.08.3 Distribution of Opioid Peptides and posterior pituitary pituicytes, the adrenal glands; in the latter
Receptors in the Stress Network PDYN mRNA is strongly expressed in the adrenal cortex (Day
et al., 1991).
1.08.3.1 Proopiomelanocortin System
b-Endorphin and related peptides are present in the nucleus
1.08.3.4 Endomorphins
arcuatus of the mediobasal hypothalamus (Sofroniew, 1979;
Bugnon et al., 1979; Khachaturian et al., 1985). An extensive Endomorphins are endogenous peptides that were relatively
nerve fiber system originating in the arcuate nucleus terminates recently isolated from bovine and human brain (Zadina
in many areas of the brain which have been implicated in the et al., 1997; Hackler et al., 1997). These peptides differ in their
stress response, e.g., various hypothalamic nuclei, limbic and amino acid sequences from other known EOPs; rather than
raphe nuclei, and some pontine nuclei. Moreover, POMC having a Tyr residue followed by Gly, endomorphins resemble
mRNA is also found in the reward system, specifically in the members of another related family of peptides (e.g., morphi-
medial prefrontal cortex, nucleus accumbens, and ventral ceptin, hemorphin, and casomorphins) that instead contain
tegmental area (Leriche et al., 2007). Some of these structures Tyr–Pro residues. Endomorphins are localized in neuronal
may also be innervated by laterally projecting POMC neurons circuits involved in processing nociceptive information and
located in the nucleus tractus solitarii (NTS) of the caudal also in many stress-responsive areas of the brain and auto-
medulla; POMC projections from the NTS are also found in nomic nervous system where MOP receptors are present
the spinal cord (Bronstein et al., 1992; Maley, 1996). Endocrine (Zadina et al., 1999). Endomorphins act as endogenous ligands
cells of the intermediate lobe of the pituitary, cells of some of MOP receptors and thus are important in modulating
peripheral tissues, and immunocytes also synthesize and responses to pain and stress by acting on autonomic and neuro-
release POMC peptides (Bloom et al., 1978; Blalock et al., endocrine systems (Zadina, 2002; Wang et al., 2003; Glatzer
1985) and may contribute to the peripheral responses to stress; and Smith, 2005; Silverman et al., 2005; Greenwell et al.,
further, POMC peptides secreted from mast cells in the skin 2007). In addition, the common distribution of endomorphins
have been implicated in the regulation of stress response to and other neurotransmitters suggests their modulatory role in
exogenous stressful stimuli (Slugg et al., 2000; Arck et al., neurotransmission (Greenwell et al., 2007).
2006; Slominski et al., 2006).
1.08.3.5 Opioid Receptors
1.08.3.2 Proenkephalin System
Opioid receptors are differentially distributed in brain areas
PENK neurons are widely distributed throughout the central (Figure 1( c) and 1(d)), also in structures involved in the regu-
and peripheral nervous systems, primarily within interneu- lation of the stress response. Dense MOP and KOP receptor
rons, some of which form longer local tract projections. binding and high expression is present in many hypothalamic
They are especially abundant in the paraventricular and arcua- nuclei in the monkey (Mansour et al., 1988) and human
tus nuclei of the hypothalamus and are relatively well repre- (Peckys and Landwehrmeyer, 1999). However, equivalent areas
sented in limbic structures such as the hippocampus, of the rat brain display few or no MOP-binding sites (Mansour
septum, and bed nucleus of the stria terminalis. Septal et al., 1987). DOP receptors are also very poorly distributed in
PENK neurons project directly to the amygdala, extend hypothalamic nuclei, e.g., weakly detectable in the
throughout the bed nucleus of the stria terminalis, and from ventromedial nucleus in the rat. All three opioid receptors
there, project to the hypothalamic paraventricular nucleus (MOP, DOP, and KOP) are present in the monkey median
and median eminence. PENK neurons have also been mapped eminence, with KOP receptors being most densely expressed
in the spinal cord, cranial sensory systems, and in the major in this area; KOP receptors are also richly expressed in the
pain-signaling network. Further a variety of PENK- monkey and rat posterior pituitary lobe (Mansour et al.,
containing cells are present in the adrenal medulla (Viveros 1988). This pattern of opioid receptor distribution is
et al., 1979). consistent with the view that opioids are important regulators
of neuroendocrine secretion. In addition, a moderate density
of MOP and KOP receptor binding has been found in the
1.08.3.3 Prodynorphin System
periaqueductal gray, locus coeruleus, substania nigra, ventral
PDYN neurons are widely distributed in brain areas associated tegmental area, raphe nuclei, and NTS, while low DOP
with stress (Watson et al., 1981; Khachaturian et al., 1985). receptor binding has been reported in the substantia nigra
Dynorphin and related peptides are coexpressed with arginine and NTS. Further, opioid receptor-containing neurons have
vasopressin in the magnocellular neurons of the paraventricu- also been described in the limbic system where they may
lar nucleus of the hypothalamus and also present in the NTS, modulate the emotional component of stress and in areas of
an area closely associated with the regulation of vagal and other the spinal cord involved in the transmission of nociceptive
autonomic functions. Further, PDYN neurons occur in the stimuli. Opioid receptors have also been found in the
limbic system and in areas of the spinal cord involved in the peripheral nervous system (Bechara and van der Kooy, 1985;
transmission of nociceptive stimuli. Cells expressing PDYN Hedner and Cassuto, 1987; Wittert et al., 1996) as well as in
mRNA are also present in a subpopulation of anterior pituitary various immune cells (Wybran et al., 1979; Blalock et al.,
lobe gonadotrophs (Khachaturian et al., 1985), in POMC- 1985; Sibinga and Goldstein, 1988; Carr et al., 1988, 1989;
expressing melanotrophs of the intermediate pituitary lobe Stein et al., 1990; Gaveriaux et al., 1995; Chuang, 1995;
(Day et al., 1993) as well as in neuronal terminals of Peterson et al., 1998).
Stress and Opioid Systems 233

1.08.4 Stress-Induced Alterations in Endogenous decreased following repeated immobilization (Makino et al.,
Opioidergic Systems 1999) and food deprivation (Gayle et al., 1999). In contrast,
water restriction had no effect on hypothalamic levels of
Most studies on the interrelationship between stress and the POMC mRNA (Zhou et al., 1999).
endogenous opioid system have been on male laboratory Stress leads to a substantial release of b-endorphin into the
rodents. The main reason for this is that male physiology is blood of animals and humans (Molina, 2006), most likely
not subject to cyclical changes in the secretion of gonadal originating from corticotrophs in the anterior pituitary lobe.
hormones; the short duration of the rodent ovarian cycle in Increased blood levels of b-endorphin are seen following
commonly used experimental species (4–5 days) makes study different acute stressors, such as electrical footshock (Guille-
design and results interpretation in rodents particularly diffi- min et al., 1977; Rossier et al., 1977; Millan et al., 1981a;
cult. Greater effort should be placed on overcoming these diffi- Przewlocki et al., 1982), swim stress (Przewlocka et al.,
culties, however, because of well-known sex differences in the 1988; Vaswani et al., 1988; Young et al., 1993a), burn injury
stress response (Bale and Epperson, 2015; Ferin, 1993; Kokras (Osgood et al., 1987), immobilization (Forman and Estilow,
and Dalla, 2014; Patchev and Almeida, 1999). Readers inter- 1988; Kjaer et al., 1993b; Larsen and Mau, 1994), cold stress
ested in the modulation of opioidergic activity by sex (Forman et al., 1988), hemorrhagic shock, muscle crush
hormones are referred to studies by Almeida and Pfeiffer injury, administration of LPS (Molina, 2002), food (Majeed
(1991), Chartoff and Mavrikaki (2015), Ferin et al. (1984), et al., 1986) and water deprivation (Zhou et al., 1999), labor
and Piva et al. (1995). (Pancheri et al., 1985; McLean et al., 1994), electroacupunc-
ture (Pan et al., 1996), surgical stress (Roth-Isigkeit et al.,
2000), septic shock and sepsis (Legakis et al., 1998), and
1.08.4.1 Proopiomelanocortin System
simulated combat military flight (Leino et al., 1998). These
Acute and chronic stress appear to influence the activity of changes in peptide release are paralleled by increases in
EOP systems in the brain. Evidence indicates that releasable the rate of POMC synthesis and processing in the anterior
pools of b-endorphin exist within the hypothalamus pituitary (Akil et al., 1982; Höllt et al., 1986; Harbuz and
(Osborne et al., 1979), and early in vivo studies showed Lightman, 1992; Zhou et al., 1999).
that short-term footshock stress depletes b-endorphin in The intermediate lobe of pituitary responds to stress in
the hypothalamus, septum, and periaqueductal gray of rats a similar way to the anterior lobe (Przewlocki et al., 1982;
(Rossier et al., 1977; Millan et al., 1981a; Przewlocki et al., Berkenbosch et al., 1983; Akil et al., 1985; Young et al.,
1982). Interestingly, conditioned fear-induced stress mark- 1993b). Acute exposure to 30-min swim stress at room temper-
edly decreased hypothalamic content of b-endorphin (Przew- ature increased plasma b-endorphin levels (derived from the
locka et al., 1990), a finding in line with an earlier intermediate lobe) several fold (Young, 1990; Young et al.,
observation that there is a measurable decrease of b-endor- 1993b); the effect was abolished by apomorphine, indicating
phin levels in the midbrain when chronically stressed rats the involvement of dopaminergic receptors. Acute ether stress
are acutely restressed (Akil et al., 1986). On the other followed by 1-h restraint stress also increased POMC mRNA
hand, cold swim stress was found to increase hypothalamic in the intermediate lobe cells, which appeared to be mediated
b-endorphin levels (Vaswani et al., 1988), although a more by tonic activity of the serotoninergic system (Garcia-Garcia
severe stress (acute prolonged intermittent footshock) did et al., 1998).
not alter hypothalamic and midbrain levels of the peptide POMC-containing cell types in the anterior and
(Akil et al., 1986; Przewlocki et al., 1987). Thus, the stress- intermediate pituitary lobes are derived from a single
induced decrease in hypothalamic b-endorphin levels might embryonic rudiment and synthesize the same hormone
be detectable only after mild, short-term stress. Various kinds precursor, POMC, but differ in the pattern of precursor
of chronic stress (repeated footshock, repeated electroconvul- processing and regulation of peptide secretion. Whereas
sive shock) reportedly do not induce substantial alterations b-endorphin is the predominant EOP produced and secreted
in brain levels of b-endorphin (Lason et al., 1987; Przewlocki by anterior pituitary (Young and Akil, 1985), the
et al., 1987). b-Endorphin levels were found to increase in intermediate lobe shows a slight increase in its content of
the nucleus accumbens septi following acute intermittent N-acetyl-b-endorphin following stress; this peptide is devoid
footshock and after extinction of opiate self-administration of opioidergic activity but contributes to the changes in the
(Zangen and Shalev, 2003). On the other hand, hypotha- circulating b-endorphin levels (Akil et al., 1985; Young,
lamic and amygda levels of b-endorphin were found to be 1990). The secretion of POMC-derived peptides from the
increased by social isolation of adult gerbils (Raab et al., anterior lobe and intermediate lobe is differentially
1985), probably pointing to the intricate control of tissue regulated, and the relative contributions of each lobe may
neuropeptide levels that reflect peptide release, biosynthesis, vary with the stimulus. For example, in response to restraint
and axonal transport. It is also worth noting that the POMC stress, b-endorphin and ACTH are secreted in equal
gene is under the negative control of adrenal steroids amounts from the anterior lobe and intermediate lobe,
(Beaulieu et al., 1988). whereas the intermediate lobe is the most important source
Studies examining how POMC mRNA levels are modulated of b-endorphin and a-MSH when ether stress is used (Kjaer
by various stressors have produced divergent results. An acute et al., 1995).
(1 h) restraint stress resulted in increased levels of POMC Chronic stress leads to a substantial increase in b-endorphin
mRNA in the arcuate nucleus of the hypothalamus (Larsen levels in both, the anterior and intermediate lobes of the pitui-
and Mau, 1994), whereas arcuate POMC mRNA levels were tary (Akil et al., 1985; Höllt et al., 1986; Przewlocki et al., 1987).
234 Stress and Opioid Systems

Interestingly, prolonged stress decreases the proportion of levels returned to control levels after 28 days of isolation
b-endorphin in the releasable pool of anterior lobe cortico- (Angulo et al., 1991).
tropes. Chronic footshock releases proportionally more A few data are available on the effects of PENK in the pitu-
b-lipotropin than b-endorphin in the rat (Young et al., itary. Zhu and Desiderio (1994) observed that the space flight
1993b). Repeated immobilization (Lopez-Calderon et al., stress diminished the Met-enkephalin level in the posterior
1991; Marti et al., 1999; Slominski, 2006), water restriction pituitary of rats and suggested that posterior pituitary enkepha-
(Zhou et al., 1999), repeated injection of hypertonic saline linergic system may respond to this type of unique stress.
(Kiss and Aguilera, 1993), and adjuvant-induced inflammation Further the same authors demonstrated that the level of Met-
(Harbuz and Lightman, 1992; Aguilera et al., 1997) increase enkephalin significantly increased 10 days after head injury in
POMC mRNA in the anterior pituitary. Interestingly, the level rats (Grigoriants et al., 1995).
of POMC mRNA increases selectively in the anterior, but not Lewis et al. (1982) demonstrated the role of adrenal medul-
in the intermediate, lobe after repeated footshock (Höllt et al., lary PENK opioids in some behavioral responses to stress. The
1986). This finding suggests selective activation of POMC exposure of rats to short, intermittent footshock caused
synthesis in the anterior lobe only in response to chronic stress. a decrease in the adrenal medullary content of PENK-derived
In contrast, other studies have indicated that rats subjected to peptides (Lewis et al., 1982), pointing to an enhanced release
chronic footshock or forced swim stress display an increase in of these opioids. Levels of PENK peptides in the adrenal
POMC mRNA levels in the intermediate pituitary (Kelsey medulla were decreased after acute stress, but did not differ
et al., 1984). Therefore, it is likely that POMC biosynthesis in from control levels in chronically stressed rats. The PENK
the intermediate lobe is regulated differentially, depending on mRNA levels increased several fold over control after
the type of stressor. hypoglycemic stress (Kanamatsu et al., 1986).
One study in humans demonstrated biphasic changes in the
plasma levels of peptide F, the peptide derived from PENK, in
1.08.4.2 Proenkephalin System
response to heavy exercise. Initially the level of the peptide F
Stressful stimuli strongly activate the PENK system within the decreased but significantly increased during recovery period
hypothalamus. Early studies demonstrated that footshock (Bush et al., 1999). This result suggests that the biosynthetic
stress (Rossier et al., 1978; McGivern et al., 1983) and social activity of adrenal PENK cells increases upon prolonged stress,
isolation (Raab et al., 1985) induced a decrease in the hypo- thus compensating for the enhanced peptide release into the
thalamic content of PENK-derived peptides. In contrast, blood. However, Van Loon et al. (1990) suggested that plasma
other studies reported no obvious effect of immobilization, Met-enkephalin in rats derived from sympathetic peripheral
footshock, or cold swim stress upon PENK peptide levels and neurons and not only from the adrenals. Repeated daily expo-
its mRNA in the rat hypothalamus (Millan et al., 1981a; sure to restraint stress resulted in an adaptive loss of the plasma
Takayama et al., 1986; Przewlocki et al., 1987; Vaswani et al., Met-enkephalin response. Repeated immobilization stress,
1988; Vellucci and Parrott, 1997). However, a plethora of however, increased PENK mRNA in sympathetic neurons of
other studies clearly indicated that stressful stimuli enhance the rat cervical and stellate ganglia (Nankova et al., 1996).
PENK gene expression in cells localized in the parvocellular Thus, it seems that, in the periphery, adrenal opioidergic cells
part of the hypothalamic paraventricular nucleus (Harbuz and sympathetic neurons contribute to the stress response.
and Lightman, 1989; Lightman and Young, 1989; Larsen and Additionally, it is likely that different types of stress selectively
Mau, 1994; Priest et al., 1997; Garcia-Garcia et al., 1998). activate different pools of PENK peptides.
Stress induced by the injection of hypertonic saline increased
PENK mRNA levels in the paraventricular nucleus, an effect
1.08.4.3 Prodynorphin System
blocked by the administration of the GC receptor antagonist,
RU-38486 (Garcia-Garcia et al., 1998). Levels of PENK Hypothalamic levels of PDYN-derived peptides were
mRNA were upregulated in the medial parvocellular significantly increased after acute swim stress and starvation
subdivision of the hypothalamic paraventricular nucleus (Przewlocki et al., 1983a, 1988a), probably indicating an
following 1 h restraint stress (Young and Lightman, 1992; inhibition of their release. In contrast, a pronounced fall in
Larsen and Mau, 1994); novelty and hypertonic saline hypothalamic dynorphin levels was observed in rats subjected
injections induced a similar response (Yukhananov and to electroconvulsive shocks, cold swim stress, or 2-h exposure
Handa, 1997). Social deprivation (14–20 days) in rats was to 4  C (Morley et al., 1982; Lason et al., 1987; Vaswani et al.,
also associated with an activation of the central PENK opioid 1988). This finding may indicate the enhancement of PDYN-
system. This paradigm increased the levels of PENK mRNA derived peptide release during seizures and extreme stress. In
and Met-enkephalin immunoreactivity in the hypothalamus. contrast, repeated electroconvulsive shock markedly increased
Interesting studies were performed in transgenic mice with dynorphin content and PDYN mRNA levels in the
human PENK-b-galactosidase fusion transgene; the transgene hypothalamus (Hong et al., 1985; Lason et al., 1987). Thus,
resulted in correct phenotypic expression and was regulated hypothalamic PDYN neurons appear to be particularly
by osmotic and hypovolemia stress in the hypothalamic sensitive to seizures. There is no evidence that chronic stress,
paraventricular and supraoptic nuclei (Borsook et al., e.g., recurrent footshock (Przewlocki et al., 1987), or
1994a,b,c). In the forebrain, proenkephalin mRNA levels conditioned fear-induced stress (Przewlocka et al., 1990)
were found to be transiently decreased in the anterior and influences the hypothalamic PDYN system. Also no evidence
medial aspects of the caudate–putamen and the nucleus of the changes in PDYN biosynthesis has been found in
accumbens after 7 or 14 days of isolation stress, but the hippocampal dentate neurons after chronic restraint stress
Stress and Opioid Systems 235

(Watanabe et al., 1995). However, isolation induced (Dantas et al., 2005). Chronic, recurrent stress induced by
a significant accumulation of PDYN mRNA selectively in the repetitive electroconvulsive shocks was shown to downregu-
paraventricular nucleus, but no changes were noted in mRNA late both DOP and MOP receptors in the rat brain (Nakata
content within the supraoptic nucleus (Matthews et al., 1993). et al., 1985). Sleep deprivation-induced stress decreased the
No significant differences were observed in the level of PDYN Bmax of DOP and MOP receptors in the limbic system (Fadda
mRNA in the nucleus accumbens between low and high et al., 1991, 1992). It is reasonable to assume that these effects
responders to novelty stress in rats (Hooks et al., 1994). On reflect an adaptive response to the enhanced release of EOPs
the contrary, a profound increase in PDYN mRNA levels was during stress. Rank-related stress has been shown to change
found in the brain, in particular in the nucleus accumbens, responsiveness of the DOP system, and social dominance
during intake and withdrawal from various drugs of abuse was linked to increased excitatory effects of DOP receptor
(Trujillo et al., 1995; Przewlocka et al., 1996; Wang et al., 1999). agonists, indicating enhanced sensitivity of this receptor in
The hypothalamic PDYN system clearly responded to dehy- dominant versus nondominant rats (Pohorecky et al.,
dration and water deprivation, evidenced by an increase in 1999). In line with these findings, Holaday et al. (1982) re-
hypothalamic PDYN-derived peptides (Przewlocki et al., ported an upregulation of DOP receptors in response to
1983b; Majeed et al., 1986; Höllt, 1990) and PDYN mRNA repeated electroconvulsive shock treatments. Further, an
levels (Sherman et al., 1986). Dehydration significantly increase in the number of DOP in the corpus striatum was re-
increased PDYN mRNA in the magnocellular neurons of the ported after repeated immobilization stress (Zeman et al.,
paraventricular and supraoptic nucleus (Matthews et al., 1993). 1988). Moreover, another study showed that stressors such
Intermittent footshock and swim stress did not alter PDYN- as intermittent footshock and 4 days of water deprivation
derived peptide content in the anterior pituitary (Przewlocki induced an increase in DOP- and MOP-binding sites in the
et al., 1987; Vaswani et al., 1988). Another study showed that rat limbic system. A 90-h water deprivation induced
dynorphin levels in the posterior pituitary (containing increases in [3H]DAMGO binding in the septum as well as
dynorphin neuronal terminals) were unchanged after acute increases in[3H]DSTLE binding in the caudate and
footshock (Przewlocki et al., 1987). Thus, the accumbens nuclei (Stein et al., 1992). Offspring from
abovementioned observations do not provide a clear prenatally stressed female rats showed increases in MOP
indication as to whether stress activates PDYN peptide receptors in brain areas such as the striatum, lateral
biosynthesis and release in the pituitary. amygdala, and pyriform nucleus (Insel et al., 1990). Further,
mice selectively bred for high swim stress-induced analgesia
(SIA) were found to have significantly higher m receptor
1.08.4.4 Opioid Receptors
density in the whole brain than those with low analgesic
Several studies have assessed stress-induced alterations in response to stress (Mogil et al., 1994). In contrast, defeat
opioid receptors. The presumptive mild stress of handling stress was without effect on the expression of MOP receptor-
was sufficient to decrease MOP or DOP receptor binding in encoding mRNA in the substantia nigra (Nikulina et al.,
several investigated brain regions, including the frontal cortex 1999). However, Lewis et al. (1987) were unsuccessful in
and olfactory tubercle when compared to unhandled control demonstrating any changes in the number or affinity of
animals (Stein et al., 1992). Fear-induced stress decreased DOP, KOP and MOP receptor sites in various brain and
[3H]Leu-enkephalin binding to the rat brain (Chance et al., spinal cord regions of rats exposed to chronic stress.
1978; Sumova and Jakoubek, 1989). Similar results were ob- A number of studies have analyzed the changes in the
tained in rats after exposure to forced swimming (Christie biosynthesis of opioid receptors as a result of stress. Novelty
et al., 1981). Low-affinity [3H]Leu-enkephalin binding to brain stress increased the levels of KOP receptor mRNA in the
homogenates at low temperature was significantly reduced in ventral zone of the medial parvocellular part but not in the
mice exposed to acute and chronic forced swimming. It is likely lateral parvocellular part of the paraventricular nucleus, claus-
that the reduced binding reflects increased in vivo occupation of trum, nucleus accumbens, or the nucleus of the lateral olfac-
opioid binding sites by EOPs (Christie and Chesher, 1983). tory tract (Yukhananov and Handa, 1996). Within 30 min
Seeger et al. (1984) reported that either prolonged intermittent (and for up to 6 h) after social defeat stress, the levels of
footshock or forced swimming caused a significant reduction in MOP-encoding mRNA increased in the lateral ventral
[3H]diprenorphine binding in the hypothalamus and other tegmental area, as detected and quantified by in situ
brain structures, as measured by autoradiography. A single hybridization histochemistry. These data suggest that stress-
20-min footshock resulted in diminished [3H]DAMGO induced alteration of MOP receptor-encoding mRNA
binding in the septum (Stein et al., 1992). A decrease in expression in the ventral tegmental area may be involved in
high-affinity [3H]etorphine binding after restraint stress in rat the consequences of social defeat stress (Nikulina et al.,
brain membranes (Hnatowich et al., 1986) and a drop in the 1999, 2005). On the other hand, KOP receptor mRNA but
number of MOP receptors in the midbrain after inescapable not MOP receptor mRNA levels were decreased in the ventral
shock (Stuckey et al., 1989) and acute noise stress (Vitale tegmental area of mice that were repeatedly winners in
et al., 2005) have also been documented. Thus, acute stress aggressive episodes (Goloshchapov et al., 2005). Evidently,
appears to decrease the binding of opioid receptor ligands, sug- there are discrepancies between the results reported by
gesting a persistent activation of opioid receptors due to an different labs, and some results are contradictory. Clearly,
enhanced release of EOPs. more studies are needed before a conclusive picture
Repeated restraint stress for 40 days reduced opioid regarding the influence of stress on the regulation of specific
receptor binding in several CNS structures in the rat opioid receptors can be obtained.
236 Stress and Opioid Systems

1.08.5 Multiple Neurochemical Interactions with of KOP receptor agonists observed following their peripheral or
Opioid System as a Consequence of Stress intraventricular administration may be mediated at least partly
by the modulation of dopaminergic cell activity. The effect of
1.08.5.1 Dopaminergic System
exogenous or endogenous opioids appears to be mediated
Stress has been shown to selectively activate the mesolimbic through local GABA interneurons, e.g., via disinhibition of
dopaminergic system located in the nucleus accumbens, the GABA neurons, thereby releasing dopamine neurons from
prefrontal cortex, and the ventral tegmental area (VTA) (Deutch tonic GABA inhibition.
et al., 1985; Scatton et al., 1988; Fadda et al., 1993; Cuadra Under basal (stress-free) conditions, EOP tonic activity is
et al., 1999; George et al., 2000), while the nigrostriatal dopa- minimal or absent, allowing a dominant role for GABAergic
minergic neurons seem not to be affected by stressful stimuli inhibition; however, during stress, EOPs inhibit GABA interneu-
(Kalivas and Abhold, 1987). Further, dopamine turnover in rons, resulting in an activation of dopaminergic transmission.
the mesolimbic system is accelerated by cues associated with On the other hand, EOP-containing terminals provide direct
a previously applied stressor (Deutch et al., 1985). Thus, fear synaptic input to dopaminergic neurons in the VTA and may
and anxiety that are evoked by stress appear to be related to directly modulate the activity of dopaminergic neurons during
the changes in dopaminergic transmission. stress. EOPs may also influence the activity of the mesolimbic
Stress-induced alterations in the activity of the central dopa- dopaminergic system in the nucleus accumbens by altering
minergic system can be modulated by opioids. Opioid admin- activity at dopaminergic terminals. Interestingly, a single
istration into the VTA has been shown to enhance dopamine immobilization stress produced an increase in striatal PENK
turnover in both the nucleus accumbens septi and the mRNA levels, while repeated immobilization stress resulted in
prefrontal cortex (Leone et al., 1991; Spanagel et al., 1992; reduction of PENK mRNA (Lucas et al., 2007). It is known
Noel and Gratton, 1995) and to influence the motor stimulant that the release of dopamine from the nucleus accumbens is
effect of footschock stress (Kalivas and Abhold, 1987). On the enhanced by some opioids (Di Chiara and Imperato, 1988;
other hand, the administration of opioid antagonists prevented Spanagel et al., 1990). This effect appears to be mediated by
stress-induced variations in dopamine turnover (Kalivas MOP as well as by DOP receptors (Spanagel et al., 1990). In
and Abhold, 1987). Further, repeated immobilization stress contrast, KOP receptor agonists inhibit dopamine release from
enhanced sensitivity to dopamine agonists as a result of hyper- this structure (Di Chiara and Imperato, 1988; Spanagel et al.,
activity of opioidergic systems. In fact, Kalivas et al. (1988) 1990).
showed that daily exposure to mild footshock stress enhanced Restraint stress triggers dopamine release in the frontal
the motor stimulatory effect of DAMGO, a DOP receptor cortex, an effect that becomes further sensitized when the stress
agonist, injections into the VTA, whereas daily intra-VTA is applied chronically. The sensitization is preventable by
administration of this enkephalin analog potentiated the naloxone pretreatment, implicating an endogenous opioider-
capacity of acute footshock to elevate dopamine metabolism gic mechanism in the frontal cortex in the development of
in the nucleus accumbens. Furthermore, it was shown that stress-induced sensitization (Cuadra et al., 1999).
daily pretreatment with naltrexone prior to stress prevented The interaction of hypothalamic tuberoinfundibular dopa-
augmentation of the effects of DAMGO when both substances minergic neurons with EOPs may be of some relevance in
were administered into the VTA, indicating the involvement of stress. The hypothalamic tuberoinfundibular dopaminergic
EOPs released locally into the structure during stress. Microin- neurons localized in the nucleus arcuatus are inhibited by
jection of enkephalin analogs into the VTA produced an opiates (Haskins et al., 1981). The inhibition of dopamine
increase in spontaneous motor activity, and this effect was release into the portal vessels during stress, as a consequence
antagonized by intra-accumbens or peripheral administration of enhanced release of EOPs (most likely b-endorphin), may
of dopamine receptor antagonists (Broekkamp et al., 1979; promote the release of POMC peptides and vasopressin from
Kelley et al., 1980). Systemic administration or microinjection the intermediate lobe and posterior lobe of pituitary, respec-
of morphine into dopamine cells into the VTA excited dopami- tively. On the other hand, an intrinsic dopaminergic system
nergic neurons in the rat (Gysling and Wang, 1983; Matthews exerts an inhibitory control on POMC peptide release via D2
and German, 1984). The enkephalin-evoked increase in motor dopaminergic receptors (Tiligada and Wilson, 1990; Tong
activity was associated with an increase in dopamine metabo- and Pelletier, 1992; Yamaguchi et al., 1996). It is apparent
lism in the mesolimbic system and appeared to be mediated from the preceding paragraph that the opioidergic network in
by MOP receptor (Latimer et al., 1987) most likely localized both the VTA and nucleus accumbens is critically involved in
on local GABAergic interneurons in the VTA since the the regulation of dopaminergic neuron activity during stress.
DAMGO-induced effects could be antagonized by GABA Opioids modulate activity of the mesocorticolimbic dopa-
antagonists (Kalivas et al., 1990). minergic system, and this interaction appears to underlie major
Both, DOP and KOP receptors appear to be involved in the aspects of stress-coping behaviors, motor response to stress,
modulation of dopaminergic neuron activity in the VTA. Acti- and reward and drug-seeking.
vation of DOP (but not KOP) receptors in the VTA were found
to facilitate the brain reward system (Jenck et al., 1987) and
1.08.5.2 Adrenergic System
dopamine release therein (Devine et al., 1993). On the other
hand, administration of the KOP receptor agonist U50,488H It is well established that stress activates the adrenergic system
and the stable dynorphin analog E 2078 into the VTA induced in several brain structures (Tsuda et al., 1986; Tanaka, 1999).
dose-dependent aversion in the place-conditioning procedure Psychological stress and conditioned fear cause increases in
(Bals-Kubik et al., 1993). Thus interestingly, the aversive effects noradrenaline release in the hypothalamus, amygdala, and
Stress and Opioid Systems 237

locus coeruleus (Tanaka, 1999). Noradrenergic neurons in the chronic opiates may alter the homeostatic balance and lead
locus coeruleus neurons have been suggested to regulate states to hyperactivity of the adrenergic system (Valentino and Van
of attention and vigilance as well as activity of the sympathetic Bockstaele, 2001).
nervous system. These neurons have also been implicated not Together, these findings indicate that coactivation of the
only in the actions of stress but also as a critical target for EOP system, which inhibits the activity of locus coeruleus
opioid action. Some data indicate that CRF release upon stress and lateral tegmental noradrenergic neurons, would favor
stimulates the brain systems by triggering the activity of cate- adaptive behavioral coping in response to the emotional
cholaminergic neurons (Emoto et al., 1993; Otagiri et al., elements of stress.
2000) and opioidergic systems. On the other hand, opioids
influence catecholaminergic neurons. Several in vitro studies
have documented that the activation of MOP (but not KOP 1.08.6 Involvement of Opioids in Modulation of the
and DOP) receptors inhibits [3H]noradrenaline release from HPA Axis
rat brain cortical slices (Illes, 1989). In hypothalamic and neo-
1.08.6.1 Corticotropin Releasing Factor
striatal slices of rats, morphine also depresses the evoked secre-
tion of [3H]noradrenaline, again suggesting the involvement of Stress of various origins induces the secretion of CRF and
presynaptic inhibitory MOP receptors (Diez-Guerra et al., enhances its synthesis in neurons of the paraventricular nucleus
1986; Schoffelmeer et al., 1988), although other studies suggest of the hypothalamus (Haas and George, 1988; de Goeij et al.,
that the activation of MOP and KOP receptors does not modu- 1991; Bartanusz et al., 1993; Imaki et al., 1996). CRF is a potent
late noradrenaline release from the basal hypothalamus secretagogue of the three major endogenous opioid peptides
(Heijna et al., 1990). MOP receptors also seem to activate (b-endorphin, Met-enkephalin, and dynorphin) acting via
noradrenaline release from the rat hippocampus (Jackisch specific CRF receptors (Nikolarakis et al., 1986) and stimu-
et al., 1988). On the contrary, presynaptic KOP receptors lating opioidergic neurons in the hypothalamus (Almeida
appear to inhibit noradrenergic transmission in the rabbit et al., 1993). Interestingly, CRF neurons appear to control
hippocampus (Jackisch et al., 1988). EOPs release tonically, since the application of the CRF
Microinjections of morphine suppress the activity of norad- receptor antagonist, a-helical CRF9-41, lowered the rate of the
renergic neurons in the locus coeruleus of freely moving cats basal release of both b-endorphin and Met-enkephalin from
(Abercombie et al., 1988), and iontophoretic application of the hypothalamus. On the other hand, EOPs may act on CRF
MOP (but not KOP and DOP) receptor agonists cause a marked neurons to modulate CRF secretion, although there is no clear
and naloxone-reversible inhibition of locus coeruleus cell evidence for this. Enkephalins as well as low doses of b-endor-
activity in vitro due to an increase in Kþ conductance and subse- phin were shown to stimulate CRF release from the hypothal-
quent hyperpolarization. This suggests that MOP receptors amus in vitro in a naloxone-reversible fashion (Buckingham,
influence excitatory transmission to the locus coeruleus. Thus, 1986). An increase in CRF mRNA levels in the paraventricular
the vast majority of current data support the view that the nucleus and increased plasma ACTH concentration were
noradrenergic system is under tonic inhibitory control of an observed in vivo after the intraventricular injection of moderate
opioidergic network in several structures of the central nervous doses of b-endorphin in rats (Wang et al., 1996). Both effects
system. The enhanced opioidergic activity occurring during were mediated via opioid receptors. The effect of b-endorphin
stress seems to modulate the function of the noradrenergic on ACTH release was inhibited by intravenous injection of anti-
system. CRF antiserum. These results suggest that the injection of
How do EOPs communicate with catecholaminergic b-endorphin increases the neuronal activity and the biosyn-
neurons upon stress, and what is the evidence for such interac- thesis of CRF in the paraventricular nucleus, and a rise in
tions? An early study reported that naloxone administration CRF secretion stimulates the secretion of ACTH. On the other
enhanced stress-induced increases in noradrenaline turnover hand, higher doses of b-endorphin administered intraventricu-
in the rat brain (Tanaka et al., 1983), and it was later demon- larly inhibited both basal and acetylcholine- and serotonin-
strated that stressor-induced conditioning of the locus coeru- stimulated CRF release (Buckingham, 1986). Furthermore,
leus unit activity was profoundly potentiated by systemic intraventricular administration of both b-endorphin and
naloxone administration in freely moving cats (Abercombie dynorphin induced a dose-related inhibition of CRF secretion
et al., 1988). Consistent with these reports is the observation into the hypophysial portal circulation of rats (Plotsky,
that immobilization stress increased noradrenaline release 1986), and the effect was antagonized by naltrexone, suggest-
from the hypothalamus, amygdala, and thalamus, an effect ing the involvement of opioid receptors. Centrally adminis-
attenuated by morphine and enhanced by naloxone (Tanaka tered EOPs inhibited hypoglycemia-induced CRF gene
et al., 1983, 1988, 1991). The intraventricular administration expression in the hypothalamus and suppressed CRF release
of Met-enkephalin during the early phase of stress attenuated which, in turn, led to a decrease in ACTH secretion and
the stress-induced increase in noradrenaline turnover in several POMC mRNA levels in the anterior pituitary (Suda et al.,
brain structures (Tanaka et al., 1985, 1989). 1992). Further, both KOP and MOP (but not DOP) agonists
These studies suggest that various kinds of stress activate inhibited the stimulated release of CRF from the rat hypothal-
noradrenergic neurons in the brain, and EOPs counteract this amus in vitro (Tsagarakis et al., 1990), the effects being specifi-
enhanced activity. CRF neurons and enkephalin neuronal fibers cally reversed by opioid antagonists. Moreover, it was shown
impinge on locus coerelus cell dendrites and may have oppo- that chronic morphine administration changes the anxiogenic
site actions in controlling their activity and homeostasis response to CRF intracerebroventricular administration, which
(Valentino and Van Bockstaele, 2015). Prolonged stress and suggests that alteration in central CRF transmission may evoke
238 Stress and Opioid Systems

the psychological disturbances after chronic morphine admin- inhibition of vasopressin release by EOPs. EOPs appear to
istration (Blatchford et al., 2006). Interestingly, naltrindole, inhibit excitatory input to vasopressinergic and oxytocinergic
a selective antagonist of DOP receptors, clearly enhanced basal neurons via the MOP receptors (Liu et al., 1999a).
and stimulated CRF release. Thus, a complex relationship Knepel and Reimann (1982) showed that morphine and
between the EOP and CRF systems appears to exist. It is b-endorphin inhibited the electrical field-stimulated release
possible that depending on circumstances, EOP systems act of vasopressin from the mediobasal hypothalamus in vitro.
directly on CRF neurons, or indirectly via other neuronal The inhibition of secretion can occur both at the level of the
systems. Several pieces of evidence indicate that the EOP and terminals and cell bodies of magnocellular cells. In fact, their
CRF systems interact in opposing manners to regulate the adap- activity is modulated by presynaptic inhibition of afferent
tive response to stress (Valentino and Van Bockstaele, 2015). inputs to magnocellular cells by opioids as well as direct effects
of afferent input cells on the cell bodies (Brown et al., 2000).
Systemic administration of morphine to rats reduced vaso-
1.08.6.2 Vasopressin System
pressin release into the pituitary stalk blood. This effect was
Stress activates the neurohormone vasopressin in animals antagonized by naloxone, which itself was without any effect.
(Knepel et al., 1985; Vellucci and Parrott, 1997). In humans, On the other hand, the inhibition of vasopressin release from
anticipation of novelty seems to be a human-specific stress posterior lobe terminals appears to be mediated rather through
stimulus for sustained elevation of plasma vasopressin in KOP than through MOP or DOP receptors. KOP selective
men (Ehrenreich et al., 1996). Vasopressin, released upon agonists as well as dynorphin A1-13 inhibited secretion of vaso-
stress, appears to affect EOP systems. Subcutaneous administra- pressin, and this effect was antagonized by naloxone and selec-
tion of arginine vasopressin to conscious rats induces a dose- tive KOP receptor antagonist nor-binaltorphimine (Bondy
dependent increase in plasma b-endorphin levels (Mormede et al., 1988; Zhao et al., 1988). KOP receptors may also directly
et al., 1986). This effect seems to be mediated via both arginine modulate the activity and release of vasopressin (but not
vasopressin V1- and V2-receptors (Kjaer et al., 1993a). Further, oxytocin) in the hypothalamic magnocellular neurosecretory
passive immunization with antivasopressin antibodies neurons (Brown and Leng, 2000). Furthermore, the KOP
inhibited stress-induced POMC-derived peptides secretion receptor might be an autoreceptor in this system because of
(Linton et al., 1985). Vasopressin system may directly a high degree of colocalization of KOP receptor, dynorphin,
contribute to the regulation of POMC-derived peptide release and vasopressin in magnocellular nerve terminals (Shuster
from the pituitary. In vitro, vasopressin was shown to release et al., 2000).
both b-endorphin and ACTH from the anterior pituitary
(Arimura et al., 1969; Vale et al., 1978; Przewlocki et al.,
1979). In fact, vasopressin acts synergistically with CRF both 1.08.7 Involvement of Opioids in the Behavioral and
in vitro and in vivo (Gillies et al., 1982; Rivier et al., 1984). Physiological Response to Stress
Studies have shown that vasopressin is a potent secretogogue
of hypothalamic b-endorphin in vitro and in vivo (Burns et al., The behavioral and physiological responses to stress are modu-
1989; Barna et al., 1990). Interestingly, vasopressin receptor lated by EOPs. The responses are varied, and they include
blockade reduced CRF-induced b-endorphin release in vitro. motor activity, nociception, reward, learning, memory, regula-
Therefore, vasopressin appears to participate in the effects of tion of emotion, and mood. The EOPs are also involved in the
CRF. While the significance of this control of the mechanism modulation of endocrine, cardiovascular, respiratory, gastroin-
of b-endorphin release remains to be established, an testinal, autonomic, and immune functions.
interesting possibility is that CRF tonically activates some
central b-endorphin neurons (as well as PENK and, to some
1.08.7.1 Locomotor Activity
extent, PDYN cells) and controls basal release of these
peptides. On the other hand, in stress, vasopressin may Stressors in the form of restraint (Zurita and Molina, 1999), tail-
participate in the mediation of opioid release. Hypothalamic pinch pressure (Amir, 1986), footshock (Van den Berg et al.,
vasopressinergic neurons appear to be an essential mediator 1998), and forced swimming (Walker et al., 1981; Overstreet
of CRF effect, suggesting the occurrence of CRF synapses on et al., 1986; Abel, 1993) affect motor and investigatory behavior.
or in the vicinity of vasopressinergic neurons (Almeida et al., The motor responses are modified by opioids, as demonstrated
1993). Thus, it appears that vasopressin might be an by experiments in which opioid receptor antagonists decreased
important modulator of POMC-derived peptide secretion, motor activity (Arnsten and Segal, 1979; Katz, 1979; Walker
making it a potentially important factor in the mediation of et al., 1981; Roth et al., 1981; Kavaliers and Innes, 1987). The
stress reactions. opioid receptor antagonist naloxone was found to decrease loco-
On the other hand, EOPs appear to participate in the motor activity and rearing in rats exposed to an open field arena
control of vasopressinergic neurons in the hypothalamus (Rodgers and Deacon, 1979; Walker et al., 1981) and potenti-
and posterior pituitary. In rats, EOPs were shown to inhibit ated the effect of immobilization stress (acute and chronic) on
vasopressin release in response to stressors such as footshock amphetamine-induced hyperlocomotion (Holtzman, 1974;
and immobilization (Knepel et al., 1985). Electrophysiolog- Diez-Otanez et al., 1997). Likewise, naloxone reverses the
ical evidence obtained in hypothalamic slices in vitro enhanced immobility time in the forced swimming test
confirmed a role for EOP in the regulation of magnocellular following restraint stress (Zurita and Molina, 1999). Emotional
vasopressinergic (and oxytocinergic) neuronal activity (Kne- stimulus (forced perception of another rat receiving footshocks)
pel and Reimann, 1982). These results suggest lack of tonic induces a transient decrease in ambulation and rearing
Stress and Opioid Systems 239

immediately after the last session, but an increase in ambulation, by administration of inhibitors (thiorphan and bestatin) of
rearing, and sniffing for at least 15 days; this emotion-driven Met-enkephalin degradation as well as by naloxone. The results
motor effect is blocked by naloxone (Van den Berg et al., from the experiments with thiorphan and bestatin suggest that
1998). The results suggest that activation of EOP systems under- the evoked attenuation of the conditioned suppression of
lies some of the changes in motor behavior evoked by exposure mobility may be directly proportional to increased striatal levels
to stress. In contrast, however, the decrease in ambulation, rear- of Met-enkephalin (Nabeshima et al., 1988). Confirming a role
ing, and sniffing and an increase in immobility in open field for Met-enkephalin is the observation that mice lacking the pro-
observed after footshock stress are not antagonizable by enkephalin gene display increased freezing (immobility) in
naloxone (Van den Berg et al., 1998). Thus, EOPs appear to be response to fear (Ragnauth et al., 2001). On the other hand,
involved in the changes in motor activity that are induced by overexpression of proenkephalin in the olfactory tubercle
very selective stressors. Interestingly, naloxone potentiated decreased freezing behavior in response to mild footshock and
stress-evoked freezing in male, but not female, rats (Klein produced behavioral activation in the open field arena
et al., 1998), pointing to sex-specific regulation of EOPs during (Primeaux et al., 2003). Administration of enkephalin analog
the emotional response to stress. Notably, stress-induced D-Ala2-Met5-enkephalinamide into the ventral tegmental area
increases in locomotor activity in the mainland deer mice can of rats increased spontaneous motor activity (Kalivas and
be blocked by the DOP receptor antagonist, ICI 154,129, Abhold, 1987). Furthermore, rats receiving daily footshock
whereas naloxone had the opposite effect in a related, island- stress for few days exhibited a significantly greater motor
residing, species. These findings suggest the involvement of stimulant response to intraventral tegmental area injection
different opioid receptor types (DOP and possibly MOP) in with the peptide than control rats. These results indicate that
the behavioral responses to stress (Kavaliers and Innes, 1987); the endogenous enkephalin system in the ventral tegmental
they also suggest that EOPs and their receptors differentially area may participate in stress-induced motor sensitization.
modulate the stress response in different species. The KOP receptor agonists tifluadom, bremazocine, and
Exposure to stressful situations has been also shown to alter U50,488H reduced locomotor activity in C57BL/6 and DB/2
the effects of opiates. Both exogenous opioids and EOPs have mice, and the effect of the agonists was enhanced by immobi-
been observed to exert bidirectional (depending on the dose) lization stress (Castellano et al., 1988). This finding is consis-
effects on motor activity. Moreover, there are contradictory tent with suggestions that PDYN and KOP receptor may
reports regarding the influence of opioids and EOPs on loco- contribute to stress-induced behavioral immobilization
motor activity, with increases and decreases, being reported. (Pliakas et al., 2001; Newton et al., 2002; Mague et al., 2003;
Given this, one may assume that stress elicits various biochem- McLaughlin et al., 2003; Shirayama et al., 2004). Indeed,
ical changes that then differentially intervene and modulate the immobilization stress in rats increases dynorphin A and dynor-
effects of opiate administration (Stohr et al., 1999). In general, phin B immunoreactivity in the hippocampus and nucleus
low opiate doses produce arousal and increase activity, while accumbens, although it should also be noted that forced
higher doses decrease motor activity, leading to sedation, swim stress transiently increases dynorphin A levels in the
stupor, or catalepsy. Further, stress may alter the locomotor hippocampus (Shirayama et al., 2004).
stimulatory effects of morphine in rats, a phenomenon called In summary, various opioid receptors appear to be differen-
stress-induced behavioral sensitization to morphine (Frances tially involved in the modulation of stress-induced alterations
et al., 2000). Thus, the locomotor effects of morphine admin- of motor activity. Stress-induced motor suppression may be
istration are augmented by low, but not high, doses of mediated or potentiated by PDYN and KOP receptors, while
morphine administered after repeated stressors (restraint, PENK and DOP receptors (Hebb et al., 2005) may act to atten-
handling, and social defeat) (Stohr et al., 1999). Similar effects uate the locomotor response to stress.
were observed after food deprivation, a condition which poten-
tiates sensitivity to opioids. Interestingly, the latter effects were
1.08.7.2 Reward
inhibited by blockade of corticosterone secretion (Deroche
et al., 1993). It would therefore appear that stress-evoked corti- Morphine (Katz and Gormezano, 1979; Van der Kooy et al.,
costerone release underlies stress-induced behavioral sensitiza- 1982; Mucha and Iversen, 1984; Mucha and Herz, 1986),
tion to morphine (Stohr et al., 1999; Deroche et al., 1992). EOPs (Bals-Kubik et al., 1993; Zangen et al., 2002; Terashvili
Interestingly, morphine elicits hyperactivity in hamsters et al., 2004; Dong et al., 2006; Der-Avakian et al., 2007), as
habituated to handling, whereas it produces hypoactivity in well as various MOP and DOP receptor agonists (Spanagel
nonhabituated animals. The effects of handling diminished et al., 1992) elicit conditioned place preference; for example,
across test days, and morphine eventually elicited hyperactivity animals receiving an opioid are inclined to choose an initially
in both habituated and nonhabituated animals (Schnur et al., nonpreferred environment. In contrast, the opioid antagonist
1988). These results are in line with the suggestion that stress naloxone (Mucha and Iversen, 1984; Mucha and Herz, 1986)
induces the release of EOPs which summate with exogenous and KOP receptor agonists (Iwamoto, 1985; Mucha and
opiates to exert the final effect. Herz, 1985) evoke place aversion. EOP systems play a key
Several authors have sought to identify the specific EOP role in modulating reward and mood and in regulating neural
mechanisms activated by stress. For example, the immobility hedonic homeostasis. Opioid-induced place preference is abol-
induced in mice subjected to a conditioned footshock (fear- ished in m receptor KO mice (Marquez et al., 2007) but is unaf-
conditioning) paradigm was associated with decreased Met- fected when the genes encoding the KOP receptor or
enkephalinergic activity in the striatum (Nabeshima et al., prodynorphin are deleted (Simonin et al., 1998; Mizoguchi
1986). The conditioned suppression of mobility was prevented et al., 2010; Zimmer et al., 2001). On the other hand, higher
240 Stress and Opioid Systems

level of prodynorphin expression in the nucleus accumbens of 1.08.7.3 Nociception: SIA


DBA/2J and SWR/J inbred mice correlated with lower
A year after the discovery of enkephalins in 1975, Akil et al.
morphine place preference, and pretreatment of these mice
(1976) made the pioneering observation that footshock stress
with KOP receptor antagonist nor-BNI enhanced the place pref-
produces potent antinociception in rats. These authors also
erence (Gieryk et al., 2010).
found that naloxone partially reversed this analgesia, suggest-
Other work suggests that enkephalins, but not b-endor-
ing an involvement of EOPs. A number of later studies showed
phin, may be responsible for opioid-mediated reward since
that various kinds of stress such as exposure to novel stimuli
PENK knockout mice failed to show aversion to naloxone,
(Kavaliers and Innes, 1988), inescapable footshocks (DeVries
while b-endorphin-deficient mice displayed conditioned
et al., 1979; Lewis et al., 1980; Terman et al., 1984; Rosecrans
place aversion to the opioid antagonist (Skultetyova and
et al., 1986; Hemingway and Reigle, 1987), restraint (Kelly
Jezova, 1999). Briefly, the current consensus is that both
and Franklin, 1987; Kurumaji et al., 1987), food deprivation
PENK- and POMC-containing neurons and MOP and DOP
(Konecka et al., 1985; Wideman et al., 1996), acute noise stress
opioid receptors mediate reward mechanisms while the
(Vitale et al., 2005), forced swimming (Cooper and Carmody,
PDYN/KOP receptor system is involved in dysphoria
1982; Terman et al., 1986a; Suaudeau and Costentin, 2000),
(Chavkin, 2013); importantly, it is thought that PDYN may
cold water stress (Bodnar et al., 1979; Girardot and Holloway,
limit drug-produced reward (Shippenberg et al., 2007;
1985b), burn injury (Osgood et al., 1987), learned helplessness
Gieryk et al., 2010). Stress appears to influence opioid-
(Maier et al., 1983; Hemingway and Reigle, 1987), social isola-
conditioned place preference, albeit through poorly
tion (Konecka and Sroczynska, 1990), food deprivation
understood mechanisms; however, GC receptors in the
(Konecka et al., 1985; Hodgson and Bond, 1996), pregnancy
hippocampus and nucleus accumbens are apparently
(Baron and Gintzler, 1984, 1987; Iwasaki and Namiki,
essential for place preference for morphine (Dong et al.,
1997), conditioned fear in animals and man (Levine et al.,
2006). There are now numerous examples of how stress
1984; Fanselow, 1986; Przewlocka et al., 1990; Pitman et al.,
influences the rewarding effects of opiates and opioids.
1990), handling stress (Fanselow and Sigmundi, 1986),
Inescapable shock enhances morphine-conditioned place
mild social stress (Kulling et al., 1988; Pohorecky et al.,
preference for several days thereafter (Will et al., 1998), and
1999), defeat stress (Miczek et al., 1982), stressful odors
an acute emotional stress or mild intermittent stress,
(Fanselow and Sigmundi, 1986), and even exposure of
respectively, resulted in an increase in the sensitivity to the
domestic and wild animals to fight-inflicted wounds (Colwell
rewarding effects of morphine (Kuzmin et al., 1996) and
and Kavaliers, 1992) elicit antinociception, in a naloxone-
enhanced the reinforcing efficacy of heroin in rats (Shaham
sensitive manner (Akil et al., 1984, 1986; Girardot and
and Stewart, 1994; Spanagel et al., 1998; Shaham et al.,
Holloway, 1985a; Terman et al., 1985). The analgesic effects
2000). Interestingly, footshock reliably reinstated
of opioids were comparable to the stress-like responses of
extinguished heroin-taking behavior, suggesting that stress
rodents given analgesic doses of morphine (5–10 mg kg1),
may be a critical factor leading to relapse (Spanagel et al.,
albeit with a faster onset and of shorter duration.
1998). Remarkably, the controllability of the stress is
Interestingly, experiments demonstrated cross-tolerance
important since escapable tail shock does not enhance
between morphine and swim SIA (Sadowski and Panocka,
morphine CPP in the rat (Der-Avakian et al., 2007).
1993; Sadowski and Konarzewski, 1999). In rodents, mild
Recent findings suggest that chronic social defeat stress acti-
stressors usually produce opioid-dependent analgesia while
vates the KOP system resulting in potentiation of the acute
severe stressors induced analgesia independent of opioids
rewarding properties of cocaine (McLaughlin et al., 2003).
(Izumi et al., 1983; Hamm and Knisely, 1987; Hawranko
On the other hand Bruchas et al. (2007) showed that repeated
et al., 1994; Mogil et al., 1996; Sadowski and Konarzewski,
swim stress caused activation of both KOP receptors and p38
1999). Stress severity plays an important role in determining
mitogen-activated protein kinase coexpressed in GABAergic
the neurochemical basis of SIA; increasing severity (duration
neurons in the nucleus accumbens, cortex, and hippocampus,
or intensity) of stress causes a shift from opioidergic to nonop-
indicating that the endogenous PDYN-KOP receptor system
ioidergic mechanisms. For example, the severe conditions of
constitutes a key component of the molecular mechanisms
swimming (longer duration or lower water temperature)
mediating the aversive properties of stress (Bilkei-Gorzo
produce SIA that is insensitive to the opiate antagonist,
et al., 2012). Furthermore, administration of ethanol under
naltrexone, whereas naloxone significantly attenuates analgesia
conditional fear stress leads to development of place
induced by less severe swimming conditions (Terman et al.,
preference, and KOP receptors modulate the development of
1986b). Similar findings have been reported with other
the rewarding effects of ethanol under psychological stress
stressors such as footshock (Rodgers and Deacon, 1981;
(Matsuzawa et al., 1999). Stress also appears to influence
Cannon et al., 1984). Interestingly, SIA is followed by a period
opioid self-administration (Piazza and Le Moal, 1998). Thus,
of hyperalgesia with a series of symptoms characteristic of the
mild footshock applied before each self-administration
exogenous opioid abstinence syndrome (Cristea et al., 1993).
session was seen to evoke higher rates of lever pressing for
Some studies have indicated that there are substantial sex and
heroin in rats (Shaham et al., 1997). However, a previous
population/genetic differences in the novelty-induced anal-
study reported that acute footshock stress did not
gesia displayed by various populations of deer mice (Kavaliers
significantly affect opioid self-administration (Kuzmin et al.,
and Innes, 1987, 1988; Marek et al., 1989; Mogil et al., 1997).
1996). Stress has been shown to reduce self-stimulation from
Some findings suggest an early evolutionary development
ventral tegmental area that was attenuated by intracerebral
and phylogenetic continuity of stress responses to aversive
administration of opioid peptides (Zacharko et al., 1998).
Stress and Opioid Systems 241

stimuli mediated by opioidergic and nonopioidergic mecha- morphine. Chronically stressed mice were hyposensitive to
nisms (Kavaliers, 1987). Exposure to either cold or warm stress MOP-mediated antinociception (Omiya et al., 2000).
increased the thermal nociceptive thresholds of the terrestrial Interestingly, rats exposed to the stress of repeated exposure to
snail, Cepaea nemoralis. Warm SIA was blocked by opiate antag- noxious heat exhibited SIA, but antinociception was reduced
onists while cold SIA was unaffected. Exposure to tail-pinch (detected using the tail-flick test) after the administration of
stress increased the thermal nociceptive thresholds (which b-endorphin into the periaqueductal gray region of the brain
was antagonized by naloxone) of the slug, Limax maximus. (Hawranko et al., 1999).
These results indicate that this mollusk displays both opioider- Several studies demonstrated the potential involvement of
gic and nonopioidergic forms of SIA in a manner analogous to DOP in SIA (Hart et al., 1983, 1985; Kitchen and Pinker,
that reported in mammals (Kavaliers, 1987). Tolerance to the 1990; Killian et al., 1995). Those studies showed that naltrindole
analgesia induced by repeated forced intermittent cold water and ICI 174874 inhibited analgesia induced by warm water
swim has been reported (Girardot and Holloway, 1985b). (20  C) swim stress in adult and young rats (Kitchen and Pinker,
Naltrexone antagonized the adaptive aspect of all those forms 1990). Also cold water swim stress (5  C) was antagonized by
of analgesia. Thus, EOPs may play a functional role in the the DOP antagonist ICI 174,864. This antinociceptive response
behavioral adaptation to aversive stressful environmental was antagonized by prior administration of the DOP2-opioid
situations. receptor antagonist, naltriben, but not by the DOP1 receptor
A series of studies in humans have shown that physical exer- antagonist, 7-benzylidenenaltrexone in mice (Killian et al.,
cise leads to temporary hypoalgesia. Reduced sensitivity to pain 1995). Another study showed that the cold water swim stress
is demonstrable after long-distance exercise (such as marathon response was selectively antagonized by the naltrindole, but
run) but also after intensive physical exercise. Pain threshold not by the DOP1 antagonist, [D-Ala2-Leu5- Cys6]enkephalin,
elevation is most pronounced during maximal exertion, but the MOP antagonist, b-funaltrexamine, or the KOP antagonist,
hypoalgesia persists after exercise has been stopped, demon- nor-binaltorphimine (Vanderah et al., 1993). These studies
strating that a systemic analgesic effect is induced by the exer- suggest that cold water swim stress analgesia is mediated
cise process (Droste, 1992). by DOP2-opioid receptors. On the other hand, the selective
Exposure to acute stress potentiates the magnitude and dura- DOP receptor antagonist naltrindole had no influence on
tion of analgesia following both the peripheral and intracerebro- psychological SIA (Takahashi et al., 1990), indicating the
ventricular administration of several opioid agonists as specific involvement of DOP in certain kinds of stress, such as
compared to nonstressed controls (Hassen et al., 1982). Both swim stress.
supraspinal and spinal analgesic opioidergic mechanisms signif- The involvement of KOP receptor in SIA has also been
icantly contribute to the enhanced analgesic potency of opioids demonstrated. An early study suggested that KOP receptor
in subjects exposed to various types of stress. Opioid-treated rats antagonists reversed nonopioid SIA while MOP receptor might
exposed to restraint stress showed potentiation of the magnitude mediate opioid analgesia (Panerai et al., 1984, 1987). It was
and duration of opioid analgesia (Calcagnetti et al., 1990; subsequently demonstrated that nor-binaltorphimine, a selec-
Calcagnetti and Holtzman, 1992; Woolfolk and Holtzman, tive KOP receptor antagonist, blocked footshock SIA when
1995). However, rats given agonists with high intrinsic activity administered systemically or intrathecally in rats (Menendez
at the MOP displayed the most potent and consistent potentia- et al., 1993). Further, the stress analgesia induced by exposure
tion of analgesia compared to unrestrained controls. Thus, the to psychological stress was also antagonized by pretreatment
results suggest that the activation of MOP is of primary impor- with nor-binaltorphimine and Mr2266 (Takahashi et al.,
tance for restraint to potentiate analgesia (Calcagnetti et al., 1990). Furthermore, chronic cold swim stress increased the
1990, 1992). Naloxone or naltrexone at low doses antagonized antinociceptive response via KOP receptor (Omiya et al.,
certain kinds of SIA. However, neither antagonist is selective for 2000); thus, the antinociceptive activity of U50,488H was
the MOP, and this experiment demonstrates only the involve- demonstrated in mice subjected to the repeated cold stress.
ment of an opioid receptor in the phenomenon. Few studies Interestingly, the antinociceptive activity of the MOP receptor
authenticated the above conclusion using more specific MOP agonist DAMGO was attenuated by stress. These results suggest
receptor antagonist b-FNA and CTOP (Fanselow et al., 1989). that spinal KOP receptors may be responsible for endogenous
The octapeptide CTOP dose-dependently reversed fear-induced analgesia induced by some types of stress.
conditional analgesia in rats when administered intraventricu- The potential involvement of the brain POMC and b-endor-
larly in rats (Fanselow et al., 1989). More recently, other authors phin systems in SIA was indicated by early evidence that
have demonstrated that CTOP suppressed SIA, when injected showed that hypothalamic and midbrain b-endorphin levels
into the periaqueductal gray (Wiedenmayer and Barr, 2000). were changed upon footshock stress, possibly due to the
Thus, MOP localized in the midbrain periaqueductal gray appear enhanced release of this peptide (Przewlocki et al., 1987,
to be involved in opioid SIA. Fields (2000) has described the 1991). Conditioned fear stress evoked naloxone-sensitive anal-
neuronal pain-modulating circuit, which includes the periaque- gesia and a marked decrease in b-endorphin levels in the hypo-
ductal gray, amygdala, and raphe nuclei in the brain stem. thalamus and the anterior and intermediate lobes of the
Through descending projections, this circuit controls both the pituitary, together with an increase in the peptide level in
spinal and trigeminal dorsal horn pain-transmitting neurons. plasma (Przewlocka et al., 1990). On the other hand, b-endor-
This system may be activated by acute stress and mediates stress phin levels were increased in the periaqueductal gray, where
analgesia in animals and humans. Interestingly, prolonged terminals of b-endorphinergic neurons are located (Kulling
chronic stress such as repeated cold swim, in contrast to acute et al., 1989; Nakagawasai et al., 1999), as well as in the arcuate
stress, decreases sensitivity to the antinociceptive effects of nucleus of the medial basal hypothalamus, after exposure of
242 Stress and Opioid Systems

rats to mild social stress (aggressive confrontation) and forced mice to forced swim stress in cold water produced opioid
walking stress (Nakagawasai et al., 1999), respectively. Lesions analgesia that was blocked by intrathecal pretreatment with
of the arcuate nucleus reduced opioid-mediated SIA, but inter- antiserum to Met-enkephalin, but not to Leu-enkephalin,
estingly, they enhanced a form of footshock SIA that was not b-endorphin, or dynorphin (Mizoguchi et al., 1997). The
blocked by injections of the opiate receptor blocker, naltrexone results suggest that swim SIA is mediated by spinal Met-
(Kelsey et al., 1986). Thus, the arcuate lesions led to compen- enkephalin. Inhibition of PENK peptide degradation has
satory changes in the nonopioid analgesic system, resulting been reported to potentiate this phenomena, when applied
in the enhanced nonopiate-mediated stress-induced analgesia. intraventricularly in mice (Chipkin et al., 1982; Christie and
Further, naloxone injections into the periaqueductal gray and Chesher, 1983). The enkephalinase inhibitor thiorphan
arcuate nucleus blocked analgesia in defeated mice (Miczek evoked a dose-related potentiation of both the peak effect
et al., 1985). In addition, microinjections of antibodies against and the duration of the SIA after exposure of rats to
b-endorphin into the midbrain periaqueductal gray attenuated inescapable footshock (Chipkin et al., 1982). Moreover, it
antinociception elicited by electroacupuncture (Xie et al., was shown that adrenal demedullation abolished the
1983). Another study suggested the potential role of b-endor- analgesic response (Lewis et al., 1982), suggesting the
phinergic cells within the NTS in SIA. Electrical stimulation participation of circulating PENK peptides. Unexpectedly,
of this structure evoked opioid-mediated analgesia in the rat however, enkephalin-deficient knockout mice exhibited
(Lewis et al., 1987). Such results suggest that b-endorphin- normal SIA (Konig et al., 1990; Bilkei-Gorzo et al., 2004).
containing neuronal cells may, at least partly, be involved in The involvement of the PDYN system in the mediation of SIA
this phenomenon. remains less clear. A recent study in mice demonstrated that
There are some suggestions that SIA may also depend on psychological SIA was fully antagonized by the selective KOP
peripheral pools of b-endorphin. It is not clear whether pitui- antagonist nor-binaltorphimine, while this compound was
tary pools of b-endorphin play a role in the phenomenon since without any effect on footshock- and swim-induced antinocicep-
some kinds of stress induce the b-endorphin release into the tion (Takahashi et al., 1990). Further, Starec et al. (1997)
blood without affecting nociception. Other studies have sug- demonstrated that dynorphin exerted an analgesic effect in
gested that although the integrity of the adenohypophysis is mice when combined with a stressor. On the other hand, an
essential for the manifestation of SIA, probably involving intermittent fasting diet in mice produced analgesia that was
neither ACTH nor b-endorphin is essential for the development blocked by nor-binaltorphimine, accompanied by increased
of the analgesia which accompanies stress (Millan et al., 1980, spinal PDYN mRNA expression (de los Santos-Arteaga et al.,
1981a). Lim et al. (1982) have suggested that plasma levels of 2003). Furthermore, the KOP receptor antagonist blocks SIA eli-
b-endorphin do not reflect changes in pain threshold, and cited by the repeated forced swim test in mice, and PDYN-
b-endorphin levels in the anterior pituitary, neurointermediate knockout mice fail to display SIA following the forced swim
lobe, and plasma are probably not causally related to stress test (McLaughlin et al., 2003).
effects such as footshock-induced analgesia. Some results Such results indicate that the PDYN system, as well as KOP
suggest that the stress of birth causes an increase in maternal receptors, may be involved in the mechanisms underlying the
secretion of b-endorphin which is not related to the degree of analgesia effects of certain kinds of stress. Notably, however,
pain itself (Jouppila et al., 1983). Mean levels of b-endorphin certain stressors may inhibit, rather than activate, the brain
in cerebrospinal fluid was significantly greater in the patients PDYN system. For example, it has been shown that the
with PTSD compared with normal subjects. Thus, the increased in vitro hypothalamic release of a-neoendorphin, a peptide
endorphinergic activity in the central nervous system may exist derived from PDYN, is lowered in rats subjected to conditioned
in PTSD patients, and that hypersecretion of opioids might fear-induced stress, which itself is accompanied by naloxone-
constitute an adaptive response to traumatic experience (Baker reversible analgesia (Przewlocka et al., 1990). Further, a rise
et al., 1997). However, there is poor correlation between cere- in the hypothalamic content of dynorphin after footshock
brospinal fluid and plasma b-endorphin levels in combat stress has been reported, suggesting a suppression of dynorphin
veterans with PTSD (Baker et al., 1997). The study on mice release (Millan et al., 1981b).
with a selective deficiency of b-endorphin clearly demonstrated It has been demonstrated that SIA is abolished in prodynor-
the critical role of b-endorphin in SIA. In these mice, the lack of phin knockout mice (McLaughlin et al., 2003). Furthermore,
opioid-dependent naloxone-reversible analgesia was observed MOP receptor-deficient CXBK mice displayed no opioid SIA
in response to mild swim stress (Rubinstein et al., 1996). The (Moskowitz and Goodman, 1985), and the late component
mice exhibited normal analgesia in response to morphine, of opioid SIA was reduced in MOP knockout mice (LaBuda
indicating the presence of functional MOP receptors. Mutant et al., 2000). A recent study demonstrated that mild SIA (3-
mice also displayed significantly greater nonopioid analgesia min swim at 32  C) was reversed by the triple mutation of
in response to cold water swim stress compared with the the MOP, KOP, and DOP receptors and decreased in MOP
controls and displayed paradoxical naloxone-induced anal- and DOP receptor knockout female mice (Contet et al.,
gesia. These changes may reflect compensatory upregulation 2006). Interestingly, the stress-induced opioid-mediated
of alternative opioid-independent pain inhibitory mecha- responses were modified in CB1 cannabinoid receptor knock-
nisms. More recently, SIA was further studied in b-endorphin outs. Indeed, these mutants did not exhibit antinociception
knockout mice; results demonstrated attenuated opioid- following a forced swim in warm water. However, absence of
mediated SIA (Skoubis et al., 2005). the CB1 cannabinoid receptor did not modify the antinocicep-
There is some evidence suggesting a role of the brain PENK- tive effects induced by different opioid agonists. These results
derived peptides in SIA (Kurumaji et al., 1987). Exposure of indicate that a physiological interaction between the opioid
Stress and Opioid Systems 243

and cannabinoid systems is necessary to allow the develop- repeated swim stress developed thermal hyperalgesia that was
ment of opioid-mediated responses to stress (Valverde et al., blocked by naloxone and naloxonazine, but not by naltrindole,
2000). DOP receptor antagonist, and nor-binaltophimine, a KOP
Experimental data clearly demonstrate that opioids are able receptor antagonist (Suarez-Roca et al., 2006) suggesting
to inhibit nociception arising in inflamed tissue by a local involvement of MOP receptors in development of SIA.
peripheral action, presumably via the terminal region of the Restraint stress affects the changes in body temperature
sensory nerves. Similar effects are observed when endogenous induced by morphine and opioid peptides administered
opioid peptides are released under stress conditions from either systemically or intracerebroventricularly (ICV) in rats.
immune cells present in the inflamed tissue (Stein et al., The unstressed group of rats responded to all doses of
1990; Przewlocki et al., 1992; Herz, 1995, 1996). Immunoreac- morphine, DAMGO, DADLE, and D-Met2-Pro5-enkephalin,
tive b-endorphin and enkephalins, processed in these cells, with an increase in core temperature. In contrast, restrained
seem to be the relevant peptides in this respect. Although the rats showed a decrease in core temperature following the
mechanism of stress-induced release of opioid peptides from injection of opioids (Szikszay et al., 1983; Spencer et al.,
the immunocytes is presently not clear, there is an indication 1985; Appelbaum and Holtzman, 1986). Interestingly, the
that this process involves cytokines and CRF. administration of cholecystokinin reduced the hypothermic
EOPs appear to be involved in the mechanisms of placebo- response to systemic morphine in restrained rats, while
analgesic effects in human (Bendetti and Amanzio, 1997; Sher, hyperthermia elicited by an administration of morphine to
2004). In fact, positron-emission tomography with [(11)C]car- freely moving rats was not diminished by cholecystokinin
fentanil has revealed that placebo effects were accompanied by pretreatment. These results support the hypothesis that
release of EOPs, as measured by enhanced MOP receptor- cholecystokinin may contribute to the regulation of the
dependent neurotransmission in the rostral anterior cingulate, endogenous opioid system (Kapas et al., 1989).
dorsolateral prefrontal cortex, insular cortex, nucleus accum- There is indirect evidence for involvement of the opioid
bens, amygdala, and ventral pallidum (Zubieta et al., 2005). peptide b-endorphin in the hypothalamic regulation of the
These studies suggest that one of the possible mechanisms of development of fever and stress-induced hyperthermia. In
placebo analgesia results from an enhanced EOP response to the unanesthetized rabbits, microinjection of b-endorphin in
noxious stimuli, and the results help explain how expectancy the preoptic/anterior hypothalamus resulted in the elevation
of noxious stimuli can influence affective and nociceptive of body temperature. It has been suggested that b-endorphin
components of pain (Gracely et al., 1983; Wager et al., 2007). reduces sensitivity of hypothalamic neurons to high ambient
Thus, whether or not EOPs play a role in the antinocicep- temperature and that this reduction leads to the increased
tion depends on the kind of stress and variables studied. peripheral vasoconstriction, inhibition of evaporative heat
Some stressors activate the EOP systems, whereas others do loss, and modification of behavioral thermoregulation, result-
not. Moreover, when EOPs are involved, they frequently ing in the elevation of body temperature (Gordon et al., 1984).
interact with other neuronal systems. Therefore, the analgesic Plasma b-endorphin response under three exercise-
effects of stress are complex and difficult to characterize. Never- thermoregulatory stress conditions was measured in humans
theless, the reactions to certain kinds of stress have been shown during stationary upright cycling. The b-endorphin response
to be mediated by specific neuronal opioid systems, since they pattern closely paralleled rectal temperature changes in all
are known to cause release of these opioid peptides and may be conditions. These data suggest that conditions of increasing
modulated by opiates. thermoregulatory stress caused by exercise are associated with
rising peripheral b-endorphin concentration (Kelso et al.,
1984).
1.08.7.4 Thermoregulation: Stress-Induced Hyperthermia
It has been previously reported that sauna-induced fevers
Several forms of stress have been shown to cause hyperthermia result in a rise in b-endorphin level in normal human volun-
in the rat. Footshock stress produced hyperthermia, the degree teers (Jezova et al., 1985). Another report described similar
of which was found to be a function of current intensity changes in plasma b-endorphin in cancer patients undergoing
(Pechnick and Morgan, 1987). Restraint stress and novelty whole body hyperthermia (Robins et al., 1987). The presented
stress also produced hyperthermia (Vidal et al., 1984). results show that there is a relationship between thermal stress,
Naloxone (Vidal et al., 1984; Kapas et al., 1989) slightly defined in terms of core temperature and/or duration of
reduced restraint and ‘novelty’ hyperthermias (Vidal et al., hyperthermia, and the quantitative rise in plasma b-endorphin
1984) or emotional hyperthermia (Blasig et al., 1978). These levels.
effects suggest that endogenous opioids are not significantly
involved in the thermal effects of the stressors. They may play
1.08.7.5 Feeding Behavior
only a minor role in the regulation of basal temperature (Vidal
et al., 1984). Peak rise in temperature following footshock Stress is known to influence feeding. Some kinds of stress
stress was not affected by naltrexone or chronic morphine augment feeding in animals, and opioid antagonists appear
administration; however, the rate of return to baseline temper- to abolish it (Antelman and Rowland, 1981; Ferin and Vande
ature was slowed by these treatments. Thus, the endogenous Wiele, 1984; Teskey and Kavaliers, 1988). Stress produced by
opioidergic system appears to be involved in the return to pinching the tail has been shown to compel satiated animals
normal body temperature following footshock, but not in the to eat and display oral stereotypies. Eating induced by the tail
footshock-induced rise in temperature (Pechnick and Morgan, pinch was reduced by microinjections of naloxone and the
1987). On the other hand, a recent study demonstrated that MOP-selective antagonist CTOP into the substantia nigra,
244 Stress and Opioid Systems

indicating the selective involvement of MOP. In fact, stress- rats since naloxone has been demonstrated to attenuate or
induced eating did occur after treatment with KOP and DOP reverse the elevation in blood pressure in both renovascular
receptor antagonists (Hawkins et al., 1994). Interestingly, and spontaneous hypertension (Szilagyi, 1991).
opioids are thought to influence the palatability of food and During periods of severe stress, opioid blockade increased
its relative reward value (Taha et al., 2006; Olszewski and ambulatory blood pressure in humans. These observations
Levine, 2007). MOP in the nucleus accumbens shell are suggest that opioidergic mechanisms inhibit ambulatory blood
postulated to mediate this influence (Ward et al., 2006; pressure responses during naturally occurring stress (Nordin
Woolley et al., 2007a,b). Secretion of enkephalin and et al., 1987; McCubbin et al., 1998). The results of another
dynorphin in the hypothalamus is also modulated by the study indicated that relaxation training reduced the diastolic
nutritional value of food (Chang et al., 2007; Naleid et al., pressure response to mental arithmetic stress. Opioid receptor
2007). Additionally, injection of morphine into the rostral blockade with naltrexone antagonized the effects of relaxation
portion of lateral hypothalamic area increases food intake in training. The study suggests that some of the physiological
the rat (Li et al., 2006). effects of relaxation training are mediated by augmentation
Opioid peptides may partly determine the rewarding of inhibitory opioid mechanisms (McCubbin et al., 1998).
aspects of eating. Mercer and Holder (1997) argue that altered Thus, endogenous opioid mechanism appears to inhibit the
EOP activity may elicit food cravings which, in turn, may influ- cardiovascular response to stress.
ence food consumption. Support for this opioidergic theory of In the brain, the POMC neurons are involved in the
food cravings is provided by various clinical conditions control of the function of the NTS, the structure known to
(bulimia nervosa, anorexia nervosa, Prader–Willi syndrome, participate in the control of cardiac function. Intravenous or
and eating-induced obesity) which are associated with the intracerebroventricular administration, as well as the injection
altered EOP levels, intensified food cravings, and increased or into the NTS of b-endorphin, has been shown to decrease
decreased food intake (Johnson, 1995; Morley et al., 1982, blood pressure (Hassen et al., 1982; Sitsen et al., 1982). The
1983). Furthermore, food deprivation results in alterations of localization of enkephalins in the paraventricular nucleus of
EOP levels in the brain and pituitary of the rat (Majeed et al., the hypothalamus, a brain region important for the regulation
1986; Tsuji et al., 1987), similar to those induced by chronic of the stress response, further substantiates the involvement of
stress. The data support the notion that EOPs, in particular, the opioid system in the modulation of stress-induced hyper-
dynorphin and the MOP receptors, play a role in the modula- tension. Studies on the cardiovascular responses to centrally
tion of food intake by stress. Stress-induced alterations in EOP administered PENK products are contradictory so far and
activity may elicit food craving, which may then influence food provide little insight into the physiological role of central
consumption (Mercer and Holder, 1997). PENK in cardiovascular functions. However, adrenal PENK
peptides, released by the stimulation of the splanchnic nerve,
may induce bradycardia and hypotension as shown in reserpi-
1.08.7.6 Cardiovascular Effects
nized dogs (Hanbauer et al., 1982).
Cardiovascular responses to stress include increased catechol- Dynorphin decreases blood pressure and produces brady-
amine secretion, tachycardia due to elevated cardiac sympa- cardia when applied intravenously or into the cisterna magna
thetic and reduced vagal efferent activity, peripheral (Laurent and Schmitt, 1983). In contrast, application of dynor-
vasoconstriction in certain vascular beds, arrhythmia, and phin into the NTS or cerebral ventricles does not alter cardio-
hypertension. Some data suggest the involvement of partic- vascular function (Hassen et al., 1982; Glatt et al., 1987).
ular EOP systems in these processes. EOPs are present in the Prodynorphin peptides appear to modulate the release of
cerebrospinal fluid and the cerebrovascular bed, and opioid vasopressin from the posterior lobe and may regulate diuresis
receptors have been found in cerebral perivascular nerves. in this way. KOP receptor agonists are powerful diuretics
Their activation may modulate the function of vasoregulatory (Leander, 1982). Therefore, prodynorphin peptides and KOP
mechanisms that are involved in the control of the cerebrovas- receptors may influence the cardiovascular system. In humans,
cular tone. Furthermore, EOPs have been found in cardiac mental stress affects blood pressure and increases various
tissue and EOPs of myocardial origin have also recently opioid peptides in the plasma. Subjects responding to stress
been shown to play a role in regulation of the heart functions with a low increase in blood pressure had high levels of
(van den Brink et al., 2003). PENK synthesis have been b-endorphin, while those who reacted with a high stress-
demonstrated in isolated rat heart (Younes et al., 2000), while induced blood pressure had elevated levels of dynorphin and
PDYN expression have been found in cultured rat myocytes Met-enkephalin. Pretreatment with naloxone enhanced blood
(Ventura et al., 1994). pressure in low responders but not in high blood pressure
Under resting conditions, EOPs do not appear to play an responders (Fontana et al., 1997). Interestingly, naloxone
important role in the regulation of the cardiovascular system, decreased blood pressure response in hypertensive subjects
but they become important under stress (Benyo and Wahl, with acute stress-induced increase in blood pressure suggesting
1996). Restraint stress evoked an increase in heart rate, blood the pressor effects of some EOPs, possibly dynorphins or
pressure, and plasma catecholamine levels in rats. Pretreatment PENK-derived peptides, in hypertensive patients (Fontana
with b-funaltrexamine partially attenuated the increase in heart et al., 1997).
rate in response to stress (Houdi et al., 1996). During MOP In summary, it seems likely that certain EOPs may, under
receptor activation by DAMGO, restraint stress resulted in certain circumstances, counteract the cardiovascular effect of
bradycardia. Psychosocial stress appears to elevate blood pres- moderate stress, e.g., tachycardia and increased blood pressure,
sure via an opioid-dependent mechanism in normotensive and mediate cardioprotection (Tanaka et al., 2014). On the
Stress and Opioid Systems 245

other hand, some EOPs appear to also mediate cardiovascular stress. Restraint stress reduced plasma testosterone levels in
depression which occurs in response to severe stress. In fact, control rats in a naloxone-sensitive manner (Akibami and
a number of studies have demonstrated that naloxone reverses Mann, 1996). Testicular steroidogenesis may also be locally
the hypotension induced by most cardiovascular shock states inhibited by opioid peptides via peripheral opioid receptors
(Reynolds et al., 1980; Vargish et al., 1980; Feuerstein et al., (Kostic et al., 1998), in particular, b-endorphin secreted into
1981; Boeuf et al., 2003). plasma and secreted locally by the Leydig cells (Fabbri et al.,
1988; Eskeland et al., 1992, 1989). On the other hand,
estrogen treatment enhanced PENK gene expression in the
1.08.7.7 Respiration
ventromedial hypothalamus (Quinones-Jenab et al., 1996),
It is well documented that opioids and EOPs influence and PENK expression therein can be associated with estrogen
respiration (McQueen, 1983) and that stress modulates as well as progesterone concentration during the estrous cycle
respiration. Rats exposed to inescapable footshock displayed (Funabashi et al., 1995). Although stress and estrogen appear
an increase in respiratory rate and naloxone potentiated to have specific effects on PENK expression in the hypotha-
the footshock-induced increase in ventilation (Isom and lamic neurons (Priest et al., 1997), it has also been demon-
Elshowihy, 1982). Chronic footshock stress attenuated both strated that the administration of 17-b-estradiol to
the respiratory stimulation produced by acute footshock ovariectomized females upregulates MOP receptor mRNA
and the potentiation induced by naloxone. These results expression in the ventromedial nucleus and arcuate nucleus
strongly suggest that stress can influence respiratory function of the hypothalamus, indicating estrogenic regulation of
through the activation of endogenous opioid systems and MOP receptors in the hypothalamus (Quinones-Jenab et al.,
release of the endogenous opioids as a compensatory reac- 1997). The involvement of MOP receptors in sexual function
tion which prevents excessive stimulation of respiration was recently observed in MOP receptor knockout mice. Inter-
(Isom and Elshowihy, 1982). estingly, male homozygotes showed unexpected changes in
sexual function as shown by reduced mating activity,
a decrease in sperm count and motility, and smaller litter
1.08.7.8 Reproduction
size (Tian et al., 1997).
Stress is frequently accompanied by an impairment of reproduc-
tive functions (Calogero et al., 1998). Stressors generally induce
1.08.7.9 Autonomic Nervous System
a depression of the HPA axis, seen as a fall in plasma luteinizing
hormone (LH) and testosterone levels (Petralgia et al., 1986). Stress results in a broad spectrum of autonomic effects. They
There is accumulating evidence that CRF is a critical stress factor include (1) elevation of plasma catecholamine levels due to
which exerts inhibitory actions upon sexual behavior and secre- the enhanced adrenomedullary and sympathetic outflow; (2)
tion of gonadotropins. However, CRF may inhibit hypothalamic an increase in mean arterial pressure and tachycardia (Fisher,
neurons producing gonadotropin-releasing hormone most 1989); (3) stress-induced intestinal effects (Tache et al.,
likely via endogenous opioidergic pathways although direct 1990); and (4) immunosuppression (Blazar et al., 1986;
effects of CRF on gonadotrophin-releasing hormone neurons Glaser et al., 1987; Fujiwara et al., 1999). Further, stress mobi-
are also likely to occur (Almeida et al., 1989). Endogenous lizes the release of b-endorphin from the pituitary gland and
opioids, acting through CRF-independent mechanisms in the PENK peptides from the adrenomedullary cells and sympa-
brain or pituitary, may also influence gonadotropin thetic nerves (Farrell et al., 1983; Jarry et al., 1985; Matthews
production (Chatterton, 1990). Acute opioid administration and Challis, 1995; Nankova et al., 1996). Little is known
decreases plasma LH levels which is due to an inhibitory about the effect of stress on peripheral PDYN-derived
modulation of gonadotrophin-releasing hormone discharge peptides. A source of these peptides in the periphery could
from the hypothalamic neurons (Briski et al., 1984; Ferin and also be the gastrointestinal tract since it has been shown
Vande Wiele, 1984; Petralgia et al., 1986; Doma nski et al., that they are released from the duodenum in vitro (Majeed
1989; Genazzani et al., 1993). It is indicated that EOPs et al., 1986).
released from the hypothalamus inhibit LH secretion, which Circulating EOPs released from pituitary and adrenals, as
results in an inhibition of ovulation and termination of well as those released from the postganglionic nerves, appear
pregnancy. Data suggesting the involvement of EOPs in the to regulate functions of the autonomic nervous system and
direct control of the neuroendocrine mechanism modulating may modulate the effects of stress. These EOPs may inhibit
gonadotrophin secretion have been reported. sympathetic outflow via the action on peripheral sympa-
In stress, the presence of low plasma LH levels and thetic nerves. This suggestion is supported by several obser-
abnormal LH pulsatile secretion has been related to the vations. It has been shown that tachycardia induced by the
increased opioidergic activity thus supporting the role of stimulation of the accelerans nerve was reduced by ethylketo-
opioids in the integration of hormonal and neuronal systems cyclazocine, a preferential KOP receptor agonist. The chrono-
of the brain (Genazzani et al., 1993). Inhibitory effects of tropic effect of noradrenaline was not changed by this
footshock stress on LH release were found to be antagonized compound, and MOP and DOP receptor agonists were
by MOP and KOP (but not DOP) receptor antagonists as well without effect. Therefore, Starke et al. (1985) have concluded
as by antibodies against both b-endorphin and dynorphin that postganglionic sympathetic neurons innervating the
(but not Met-enkephalin) (Petralgia et al., 1986). Therefore, sinus node of the rabbit heart may express presynaptic
these results suggest the involvement of POMC and PDYN KOP receptors. Further, it has been shown that KOP and
(but not PENK) neurons in the control of LH release during DOP receptor types (but not MOP) are localized on
246 Stress and Opioid Systems

sympathetic nerves in the isolated guinea pig atria (Ledda mediate the peripheral effects of stress (Stein et al., 1990;
et al., 1985). In isolated arteries, KOP and DOP receptor Przewlocki et al., 1992; Herz, 1995).
agonists depressed the response to sympathetic stimulation.
Agonists of the KOP receptors decreased noradrenaline
1.08.7.10 Immune System
release from postganglionic neurons into the blood. On
the other hand, EOPs and opiates may stimulate sympathetic A variety of stressors have been found to alter immune func-
nervous system. ICV administration of b-endorphin to rats tions. EOPs released in response to stress from endocrine and
increased plasma noradrenaline and adrenaline (Yamauchi neuronal systems may interact with the immune system by
et al., 1997) via opioid receptors. On the other hand, the modulating immune responses to various factors (Shavit
increase in catecholamine levels by restraint stress was not et al., 1985; Jodar et al., 1994; Sacerdote et al., 1994, 2003;
inhibited by anti-b-endorphin antisera but was clearly Sacerdote, 2003; Moynihan et al., 2000). On the other hand,
diminished by naloxone. The results suggest that some EOPs produced and released by immune cells display paracrine
EOPs other than b-endorphin are involved in restrain and autocrine action and demonstrate functional activities
stress-induced activation of autonomic nervous system. In that are in part similar to cytokines.
fact, immobilization stress activated enkephalin neurons in Several in vitro studies suggested that EOPs enhanced
the ventral medulla paragigantocellularis and lateral reticular immune responses (Plotnikoff and Miller, 1983; Wybran,
nuclei which might be involved in autonomic response to 1985; Gatti et al., 1993), while others led to the opposite
stress (Mansi et al., 2000). Exposure to stress increased conclusion (Greenberg et al., 1984; Shavit et al., 1986;
sympathetic nervous system activity inducing an elevation Ben-Eliyahu et al., 1990). In fact, stress suppresses immune
of plasma noradrenaline and might disturb glucose homeo- function. Greenberg et al. (1984) have found that stress
stasis. Liu et al. (1999b) found that a hypoglycemic effect induced by tail shock suppressed natural killer cell cytotox-
was produced in the rats with streptozotocin-induced dia- icity and that this effect was blocked by naltrexone. Subse-
betes after cold-exposure, and the effect was reversed by quent studies showed that inescapable footshock stress
naloxone. It was suggested that the hypoglycemia was medi- (Shavit et al., 1986) and forced swimming (Ben-Eliyahu
ated by b-endorphin since the increase in the plasma concen- et al., 1990) decreased natural killer cell activity and finally
tration was observed upon stress. Moreover, intravenous reduced the resistance of rats to a mammary ascites tumor
injection of b-endorphin in the rats with streptozotocin- (Shavit et al., 1984). Other studies demonstrated that heat
induced diabetes produced a lowering of plasma glucose stress-induced immunosuppression during pregnancy was
level. Therefore, b-endorphin appears to be responsible for mediated by opioid system most likely by placental b-endor-
the induction of hypoglycemic effects in diabetic rats after phin release into blood (Nakamura et al., 1998). The effect
cold stress. In rats, postexercise peak of insulin in response was inhibited by naloxone indicating the involvement of
to glucose was markedly reduced when compared to resting opioid receptors. Overnight restraint stress of mice decreased
controls. Interestingly, the administration of naloxone concanavalin A-driven lymphocyte proliferation, plaque-
further decreased the insulin response. These results suggest forming cell response to sheep red blood cells, and natural
that EOPs may participate in the physiological adaptation killer cell activity in the spleen, but the phagocytic activity
to exercise stress, maintaining postexercise insulin response was enhanced (Marotti et al., 1996). Interestingly, injection
to glucose (Bouix et al., 1996). of Met-enkephalin before restraint stress abolished these
Studies in humans provided evidence that DOP receptors changes (except for the natural killer cell activity) and
and possibly enkephalins might influence autonomic sympa- attenuated stress-induced elevation of GCs, although Met-
thetic reactivity. The selective DOP receptor agonist deltorphin enkephalin itself affected the immune responses to stress: it
failed to modify basal plasma levels of noradrenaline in the decreased natural killer cell activity and the plaque-forming
control rats, but completely suppressed the insulin-evoked cell response and enhanced phagocytic activity (Brown and
elevation of noradrenaline and the release of both noradrena- Van Epps, 1985). Further studies revealed that the
line and adrenaline elicited by cold stress. These findings concentrations of b-endorphin in splenocytes, peripheral
provide evidence that DOP receptors and possibly enkephalins blood mononuclear cells, and lymph node cells were
may modify autonomic sympathetic output (degli Uberti et al., significantly increased after the exposure to inescapable
1993). intermittent footshock for 20 min (Sacerdote et al., 1994).
Lymphoid organs, like many others, are innervated by the In contrast, the exposure to a continuous footshock for
autonomic nervous system, and there is a growing body of 3 min did not affect the b-endorphin concentrations
evidence that this system can have immunomodulatory effects. (Sacerdote et al., 1994). Recent studies have indicated that
Noradrenergic postganglionic nerve fibers are found in the the brain produces interferon-a (IFN-a) in response to
thymus, spleen, lymph nodes, and gut-associated lymphoid inflammatory stress and that the effect of IFN-a was
tissue, where they can make direct contact with immunocytes inhibited by naloxone. Central administration of IFN-
(Ader et al., 1990). It would appear that opioids may influence a inhibited natural killer cytotoxicity. Further study revealed
peripheral noradrenergic nerves and noradrenergic innervation that IFN-a decreased the activity of hypothalamic neurons
of lymphoid organs. On the other hand, there is evidence that via opioid receptors which, in turn, resulted in the
cells of the immune system produce EOPs (Blalock et al., activation of CRF neurons, thereby suppressing natural killer
1985). Therefore, it is likely that in situations such as inflam- cytotoxicity through the activation of the splenic sympathetic
mation, immunocytes may release EOPs under stress and nerves in splenocytes (Hori et al., 1998). Interestingly, EOPs are
through paracrine or direct synaptic-like communication synthesized and released under stress conditions from immune
Stress and Opioid Systems 247

cells present in the inflamed tissue (Stein et al., 1990, 2003; supported by reports of upregulated MOP receptors in the
Przewlocki et al., 1992; Herz, 1995). The majority of studies brains of depressed suicide victims (Gabilondo et al., 1995;
indicated that the effects of stress on immune response were Gross-Isseroff et al., 1998), as well as pioneering studies on
blocked by opioid receptor antagonists, indicating that they the antidepressant-like actions of b-endorphin (Kline et al.,
were mediated by endogenous EOPs mobilized during stress. 1977; Gerner et al., 1980), the partial opiate agonist cyclazo-
cine (Fink et al., 1970), and buprenorphine (Emrich et al.,
1982). Additional indirect evidence that opioids may be
1.08.8 Therapeutic Potential of Opioid Agonists and involved in the regulation of mood comes from the observa-
Antagonists in Stress-Related Psychiatric Disorders tion that withdrawal from exogenous opiate usage causes
a decline in dysphoria and a PET-scan report of reduced
Chronic stress is associated with several mental disorders; it is MOP receptors in the neural circuitry regulating emotions
strongly implicated in anxiety and depression in humans (Kennedy et al., 2006). Buprenorphine is an approved
(Holsboer and Ising, 2010; Lucassen et al., 2014; Patchev medication for chronic pain and addiction; the compound
et al., 2014) and thought to play a role in the development of has far fewer side effects (respiratory, immune, and cognitive)
PTSD (Daskalakis et al., 2013), eating disorders (both anorexia and displays lower abuse potential (dependence) than most
and hyperphagia; Bazhan and Zelena, 2013), drug and other opiates (Shmygalev et al., 2011; Davis, 2012; Karp
substance (Koob and Kreek, 2007; Koob and Le Moal, 1997; et al., 2014). Buprenorphine acts rapidly and effectively to
Koob and Mason, 2015), stroke (Craft and Devries, 2009), ameliorate depressive symptoms in patients who were
and more recently, neurodegenerative disorders such as Alz- otherwise resistant to other antidepressant medications
heimer’s (Machado et al., 2014; Sotiropoulos et al., 2008) (Emrich et al., 1982; Bodkin et al., 1995); treatment-resistant
and Parkinson’s (Ros-Bernal et al., 2011) diseases. These effects depression, which affects about 50% of depressed patients,
may be attributed to the hypersecretion of GCs that accom- remains one of psychiatry’s greatest challenges (Rush et al.,
panies the stress response, which may continue for an extended 2006). Buprenorphine is a partial MOP agonist and KOP
period well after the stressful stimulus has faded due to failure antagonist (Cowan, 2007), a characteristic referred to again
in the mechanisms mediating GC negative feedback at the later in this section. Lastly, mention should be made of
central and pituitary levels (Holsboer and Ising, 2010; Lucassen a compound (ALKS 5461) that combined buprenorphine
et al., 2014). Chronically elevated GC levels result in altered with a m opioid receptor antagonist (samidorphan); this drug
neuronal structure, namely synaptic dysfunction and loss, entered Phase III clinical trials on the basis of evidence from
dendritic atrophy, and even death of neural precursor cells or a placebo-controlled study that it is efficacious and safe and
mature neurons. Such morphological mal(adaptations) – lacks abuse potential (Ehrich et al., 2015).
primarily observed in the hippocampus and prefrontal cortex Considerable attention has been paid to the possible anti-
– may extend to electrophysiological and functional disconnec- depressant actions of DOP receptors (Pradhan et al., 2012;
tion between brain nuclei; all of them are linked with Saitoh and Yamada, 2012). The rationale for pursuing DOP
emotional, affective, and cognitive impairments (Sousa and receptors as possible therapeutic targets included the demon-
Almeida, 2012; Lucassen et al., 2014). This, and the previously stration that mice with a deletion of the DOP receptor-coding
discussed (1) distribution of EOPs and EOP receptors in fronto- gene display increased immobility in the Porsolt forced swim
cortical, limbic, and hypothalamic areas of the brain and (2) test (Filliol et al., 2000) and that various DOP receptor agonists
the reciprocal regulatory interactions between EOPs and the have antidepressant properties (Tejedor-Real et al., 1998;
HPA axis, has led to interest in the potential of opioid-based Torregrossa et al., 2006; Vergura et al., 2008; Jutkiewicz and
treatments for stress-related disorders of the brain such as major Roques, 2012). The latter effects may be attributed to increased
depression. However, the idea that opioid receptor ligands may availability of the brain levels of serotonin and/or brain-
be useful to alleviate psychiatric symptoms has been around derived neurotrophic factor (BDNF) (Narita et al., 2006;
and practiced for centuries (Weber and Emrich, 1988); in the Torregrossa et al., 2005). As already mentioned, DOP receptor
modern era of medicine, opium was widely used to treat agonists would appear to be candidate treatments because they
mood disorders until the early 1950s when tricyclic antidepres- also have some euphorigenic properties, but possibly lower
sants were introduced (Tenore, 2008). In the following para- abuse liability than MOP receptor-activating drugs (Jutkiewicz,
graphs, we will only review studies in which opioidergic 2006). Convulsions are a major undesired effects of many DOP
ligands were tested for their antidepressant efficacy. We will receptor agonists, but since similar side effects of conventional
not discuss such investigations in patients with addictive disor- antidepressant drugs can be avoided by appropriate dosage,
ders although depression and drug and alcohol addiction this would not necessarily be an insurmountable barrier. A
often coexist; readers interested in those aspects should refer number of DOP agonists have entered clinical trials in recent
to Nunes et al. (2004) and Lalanne et al. (2014). years (www.clinicaltrials.gov); while some trials were termi-
Anhedonia, the inability to sense pleasure, is a cardinal nated due to inconclusive evidence, published reports on
feature of depression (Nestler and Carlezon, 2006). Since others are still unavailable.
MOP receptor-activating opioids as well as the prototypic Antagonism of KOP receptors as a means to prevent or treat
opiate morphine, and DOP receptor agonists, induce feelings depression appears to be the most promising ‘opium cure’
of well-being, decreased anxiety, and tranquility (Jaffe and (Harrison, 2013) as Emil Kraepelin would have put it (Weber,
Martin, 1990), it has been hypothesized that EOP deficiency 1987). Recent PET studies suggest disturbed KOP receptor
may contribute to depressive illness (Byck, 1976; Herz and activity in depression (Pietrzak et al., 2014). Moreover, whereas
Emrich, 1983; Lutz and Kieffer, 2013). This hypothesis is MOP receptor agonists increase reward and produce euphoria,
248 Stress and Opioid Systems

KOP receptor-activating drugs are aversive and dysphoric in other hand, EOPs derived from POMC and, possibly, from
humans and animals (Chefer et al., 2013; Pfeiffer et al., 1986; PENK may enhance the activity of the mesolimbic dopaminergic
Shippenberg and Herz, 1986; Roth et al., 2002; Chavkin, system during stress, resulting in the reinforcement of a positive
2013). These KOP-mediated actions result from their emotional state, a decrease in anxiety, and better adaptation. In
inhibitory effects on dopaminergic, serotoninergic, and contrast, PDYN peptides may have an opposite effect on the
glutamatergic innervation of the corticolimbic reward pathway dopaminergic system under stress. Stress-induced activation of
(reviewed in Lalanne et al., 2014) and possibly from their the PDYN system produces dysphoria and depression.
ability to modulate BDNF synthesis (Zhang et al., 2007) and It seems likely that the endogenous PDYN system may act
hippocampal neurogenesis (Drake et al., 2007). The latter upon motor and emotional aspects of the stress response; the
phenomena are implicated in the development of, and PDYN system appears to act in a manner opposite to that of
recovery from, depression (Krishnan and Nestler, 2010; the POMC and PENK systems.
Mateus-Pinheiro et al., 2013; Miller and Hen, 2015). Stress Repeated or chronic stress leads to alterations in EOPs that
results in dynorphin release and KOP receptor activation and may, in turn, contribute to the onset of stress-related patholo-
concomitantly induces KOP receptor antagonist-reversible gies. Several findings suggest that peripheral EOPs released
anxiolytic and depression-like behaviors in animals (del during stress may contribute to the production of gastric ulcers,
Rosario et al., 2002; Mague et al., 2003; Shirayama et al., 2004; while the central pool of EOPs appears to have an opposite func-
McLaughlin et al., 2006; Carr et al., 2010; Van’t Veer and tion (Hernandez et al., 1983). In addition, EOPs participate in
Carlezon, 2013; Falcon et al., 2015). If stress-induced the regulation of the circulatory response to stress and the path-
hypersecretion of GC is indeed a key culprit in generating ogenesis of hypertension. Activation of EOPs in response to
depressive behavior (Holsboer and Ising, 2010), it is important traumatic injury, ischemia, and infection has been observed in
to note that mice with deletion of either the prodynorphin both preclinical and clinical studies (Molina, 2006).
(Bilkei-Gorzo et al., 2008) or KOP receptor genes (Wittmann While the reactions of EOP systems to stress generally help
et al., 2009) fail to increase GC secretion upon exposure to restore altered homeostasis, EOPs may also be, at least partly,
certain stressors. For a comprehensive and critical review of the responsible for the pathological effects of some forms of shock
involvement of the prodynorphin and KOP receptor systems in (e.g., circulatory shock and hypoxia) and may facilitate or even
the relationship between stress and depression/depressive-like accelerate death.
behavior, readers are referred to a recent review by Van’t Veer While the organism’s response to stress serves to maintain
and Carlezon (2013). With respect to the potential of KOP or restore physiological and behavioral homeostasis in the
receptor-targeted drugs for the alleviation of depression, readers event of adverse challenges from the environment, the EOP
are reminded that the efficacy of buprenorphine appears to system is implicated in dissociative symptoms in patients
depend as much on its MOP receptor activity as on its KOP with personality disorder and PTSD. Specifically, hyperactivity
receptor antagonism. Reference was also previously made to of the EOP system appears to contribute to flashbacks (a key
ALKS 5461, a drug presently in Phase II clinical trials (Ehrich feature of PTSD) and borderline personality disorder (Bohus
et al., 2015). This compound combines buprenorphine with an et al., 1999).
MOP receptor antagonist (samidorphan); presumably, the The role of stress hormones in the development of drug
added benefit of samidorphan is that it helps titrate down the addiction and depression is significant but the underlying
euphoric actions of buprenorphine, thus reducing abuse mechanisms remain poorly understood. Interestingly, human
potential of the new compound. addicts and depressive patients display dysregulation of the
HPA axis and these subjects are hyporesponsive to stress
(Lovallo, 2006). This blunted response to stress, which is
1.08.9 Opioidergic Control of Stress Responses – considered an important risk factor for relapse into drug abuse
Conclusions and depression (Sinha, 2007; Valentino and Van Bockstaele,
2015), may be, at least partly, caused by changes in the func-
EOP systems appear to play an important role in the interaction tioning of EOP systems; a disturbed balance in the activity of
of the organism with different stressors by limiting the harmful the PENK and PDYN systems may be especially important in
effects of stress (Drolet et al., 2001; Molina, 2006). While rela- this respect.
tively quiescent in the resting state, EOPs are released during Many stressors encountered by humans are chronic in
intense stressful stimuli. Acute, mild, and short-lasting stressors nature; therefore, elucidation of exactly how EOPs influence
appear to mobilize EOPs, which may, in turn, act to oppose the adaptation to stress can have potentially important thera-
stress-precipitated reactions and, in concert with other factors, peutic implications for a variety of systems that become disrup-
counteract the initial response and help restore homeostasis. ted by stress. More research on the interactions between EOP
Systemic stressors act predominantly by interacting with systems and stress therefore remain a high research priority.
hypothalamic neuropeptides and catecholaminergic systems.
EOPs can limit the HPA axis response to stress by dampening
the adrenocortical system during stress and uncoupling the Acknowledgments
adrenal gland from hypothalamo–pituitary stimulation. Interac-
tions of EOPs with brain catecholaminergic systems appear to This work was supported by research grant 2013/08/A/NZ3/00848
influence the ability of the CNS to cope with stress. Coactivation MAESTRO from National Science Foundation (NCN, Poland). We
of EOPs (especially b-endorphin) inhibits activity of the locus express thanks to Lidia Radwan for invaluable help with the preparation
coeruleus to promote adaptive behavioral responses. On the of the manuscript and Mikolaj Przewlocki for figure preparation.
Stress and Opioid Systems 249

References Bare, L.A., Mansson, E., Yang, D., 1994. Expression of two variants of the human l
opioid receptor mRNA in SK-N-SH cells and human brain. FEBS Lett. 354,
Abbadie, C., Pan, Y.X., Pasternak, G.W., 2000. Differential distribution in rat brain of 213–216.
mu opioid receptor carboxy terminal splice variants MOR-1C-like and MOR-1-like Barna, I., Sweep, C.G., Veldhuis, H.D., Wiegant, V.M., De Wied, D., 1990. Effects of
immunoreactivity: evidence for region-specific processing. J. Comp. Neurol. 419 pituitary b-endorphin secretagogues on the concentration of b-endorphin in rat
(2), 244–256. cerebrospinal fluid: evidence for a role of vasopressin in the regulation of brain
Abel, E.L., 1993. Ontogeny of immobility and response to alarm substance in the b-endorphin release. Neuroendocrinology 51, 104–110.
forced swim test. Physiol. Behav. 54 (4), 713–716. Baron, S.A., Gintzler, A.R., 1984. Pregnancy-induced analgesia: effects of adrenal-
Abercombie, E.D., Levine, E.S., Jacobs, B.L., 1988. Microinjected morphine ectomy and glucocorticoid replacement. Brain Res. 321, 341–346.
suppresses the activity of locus coeruleus noradrenergic neurons in freely moving Baron, S.A., Gintzler, A.R., 1987. Effects of hypophysectomy and dexamethasone
cats. Neurosci. Lett. 86, 334–339. treatment on plasma beta-endorphin and pain threshold during pregnancy. Brain
Ader, R., Felten, D., Cohen, N., 1990. Interaction between the brain and the immune Res. 418, 138–145.
system. Annu. Rev. Pharmacol. Toxicol. 30, 561–602. Bartanusz, V., Jezova, D., Bertini, L.T., Tilders, F.J., Aubry, J.M., Kiss, J.Z., 1993.
Aguilera, G., Jessop, D.S., Harbuz, M.S., Kiss, A., Lightman, S.L., 1997. Differential Stress-induced increase in vasopressin and corticotropin-releasing factor expression
regulation of hypothalamic pituitary corticotropin releasing hormone receptors in hypophysiotrophic paraventricular neurons. Endocrinology 132 (2), 895–902.
during development of adjuvant-induced arthritis in the rat. J. Endocrinol. 153 (2), Bazhan, N., Zelena, D., 2013. Food-intake regulation during stress by the
185–191. hypothalamo-pituitary-adrenal axis. Brain Res. Bull. 95, 46–53.
Akibami, M.A., Mann, D.R., 1996. Mechanism of stress-induced attenuation of the Beaulieu, S., Gagne, B., Barden, N., 1988. Glucocorticoid regulation of proopiome-
testicular response to gonadotropin: possible involvement of testicular opioids, lanocortin messenger ribonucleic acid content of rat hypothalamus. Mol. Endo-
a pertussis toxin-sensitive G-protein, and phosphodiesterase. J. Androl. 17, crinol. 2, 727–731.
10–16. Bechara, A., van der Kooy, D., 1985. Opposite motivational effects of endogenous
Akil, H., Mayer, D.J., Liebeskind, J.C., 1976. Antagonism of stimulation produced opioids in brain and periphery. Nature 314 (6011), 533–534.
analgesia by naloxone, a narcotic antagonist. Science 191, 961–962. Bendetti, F., Amanzio, M., 1997. The neurobiology of placebo analgesia: from
Akil, H., Shiomi, H., Walker, J.M., Watson, S.J., 1982. Characterization of multiple endogenous opioids to cholecystokinin. Prog. Neurobiol. 51, 109–125.
forms of beta-E in pituitary and brain: effect of stress. Adv. Biochem. Psycho- Ben-Eliyahu, S., Yirmiya, R., Shavit, Y., Liebeskind, J.C., 1990. Stress-induced
pharmacol. 33, 61–67. suppression of natural killer cell cytotoxicity in the rat: a naltrexone-insensitive
Akil, H., Watson, S.J., Young, E., Lewis, M.E., Khachaturian, H., Walker, J.M., 1984. paradigm. Behav. Neurosci. 104, 235–238.
Endogenous opioids: biology and function. Annu. Rev. Neurosci. 7, 223–255. Benyo, Z., Wahl, M., 1996. Opiate receptor-mediated mechanisms in the regulation of
Akil, H., Shiomi, H., Matthews, J., 1985. Induction of the intermediate pituitary by cerebral blood flow. Cerebrovasc. Brain Metab. Rev. 8 (4), 326–357.
stress. Synthesis and release of a non-opioid form of b-endorphin. Science 227, Berkenbosch, F., Tilders, R.J.H., Vermes, I., 1983. 3-Adrenoceptor activation mediates
424–442. stress-induced secretion of b-endorphin-related peptides from the intermediate but
Akil, H., Young, E., Walker, J.M., Watson, S.J., 1986. The many possible roles of not anterior pituitary. Nature 305, 237–239.
opioids and related peptides in stress-induced analgesia. Ann. N.Y. Acad. Sci. 467, Bilkei-Gorzo, A., Erk, S., Schürmann, B., Mauer, D., Michel, K., Boecker, H.,
140–153. Scheef, L., Walter, H., Zimmer, A., 2012. Dynorphins regulate fear memory: from
Alfaras-Melainis, K., Gomes, I., Rozenfeld, R., Zachariou, V., Devi, L., 2009. Modu- mice to men. J. Neurosci. 32 (27), 9335–9343.
lation of opioid receptor function by protein-protein interactions. Front. Biosci. Bilkei-Gorzo, A., Racz, I., Michel, K., Zimmer, A., Klingmüller, D., Zimmer, A., 2004.
(Landmark Ed.) 14, 3594–3607. Behavioral phenotype of pre-proenkephalin-deficient mice on diverse congenic
Almeida, O.F.X., Pfeiffer, D.G., 1991. A neuroendocrine perspective of sex and drugs. backgrounds. Psychopharmacology (Berl.) 176 (3–4), 343–352.
In: Almeida, O.F.X., Shippenberg, T.S. (Eds.), Neurobiology of Opioids. Springer, Bilkei-Gorzo, A., Racz, I., Michel, K., Mauer, D., Zimmer, A., Klingmüller, D.,
Berlin, pp. 301–330. Zimmer, A., 2008. Control of hormonal stress reactivity by the endogenous opioid
Almeida, O.F.X., Nikolarakis, K.E., Sirinathsinghji, D.J.S., Herz, A., 1989. Opioid- system. Psychoneuroendocrinology 33, 425–436.
mediated inhibition of sexual behaviour and luteinizing hormone secretion by Blalock, J.E., Harbour-McMenamin, D., Smith, E.M., 1985. Peptide hormones shared
corticotrophin-releasing hormone. In: Dyer, R.G., Bicknell, R.J. (Eds.), Brain Opioid by the neuroendocrine and immunologic systems. J. Immunol. 135, 858–861.
Systems in Reproduction. Oxford University Press, Oxford, pp. 149–164. Blasig, J., Hollt, V., Bauerle, U., Herz, A., 1978. Involvement of endorphins in emotional
Almeida, O.F., Hassan, A.H., Holsboer, F., 1993. Intrahypothalamic neuroendocrine hyperthermia of rats. Life Sci. 23, 2525–2531.
actions of corticotropin-releasing factor. Ciba Found. Symp. 172, 151–169. Blatchford, K.E., Choi, E.A., McNally, G.P., 2006. Altered responsivity to central
Amir, S., 1986. Catalepsy induced by body pinch: relation to stress-induced analgesia. administrations of corticotropin-releasing factor in rats with a history of opiate
Ann. N.Y. Acad. Sci. 467, 226–237. exposures. Behav. Neurosci. 120 (5), 1169–1174.
Angulo, J.A., Printz, D., Ledoux, M., McEwen, B.S., 1991. Isolation stress increases Blazar, B.A., Rodrick, M.L., O’Mahony, J.B., Wood, J.J., Bessey, P.Q., Wilmore, D.W.,
tyrosine hydroxylase mRNA in the locus coeruleus and midbrain and decreases Mannick, J.A., 1986. Suppression of natural killer-cell function in humans
proenkephalin mRNA in the striatum and nucleus accumbens. Brain Res. Mol. following thermal and traumatic injury. J. Clin. Immunol. 6 (1), 26–36.
Brain Res. 11, 301–308. Bloom, F.E., Battenberg, E., Rossier, J., Ling, N., Guillemin, R., 1978. Neurons containing
Antelman, S.M., Rowland, N., 1981. Endogenous opiates and stress-induced eating. b-endorphin in rat brain exist separately from those containing enkephalin:
Science 214, 1149–1151. immunocytochemical studies. Proc. Natl. Acad. Sci. U.S.A. 75, 1591–1595.
Appelbaum, B.D., Holtzman, S.G., 1986. Stress-induced changes in the analgesic and Bodkin, J.A., Zornberg, G.L., Lukas, S.E., Cole, J.O., 1995. Buprenorphine treatment
thermic effects of opioid peptides in the rat. Brain Res. 377, 330–336. of refractory depression. J. Clin. Psychopharmacol. 15, 49–57.
Arck, P.C., Slominski, A., Theoharides, T.C., Peters, E.M., Paus, R., 2006. Neuro- Bodnar, R.J., Glusman, M., Brutus, M., Spiaggia, A., Kelly, D.D., 1979. Analgesia
immunology of stress: skin takes center stage. J. Investig. Dermatol. 126 (8), induced by cold-water stress: attenuation following hypophysectomy. Physiol.
1697–1704. Behav. 23, 53–62.
Arimura, A., Schally, A.V., Bowers, C.Y., 1969. Corticotropin-releasing activity of lysine Boeuf, B., Poirier, V., Gauvin, F., Guerguerian, A.M., Roy, C., Farrell, C.A., Lacroix, J.,
vasopressin analogues. Endocrinology 84, 579–583. 2003. Naloxone for shock. Cochrane Database Syst. Rev. (4), CD004443.
Arnsten, A.T., Segal, D.S., 1979. Naloxone alters locomotion and interaction with Bohus, M.J., Landwehrmeyer, G.B., Stiglmayr, C.E., Limberger, M.F., Bohme, R.,
environmental stimuli. Life Sci. 25 (12), 1035–1042. Schmahl, C.G., 1999. Naltrexone in the treatment of dissociative symptoms in
Baker, D.G., West, S.A., Orth, D.N., Hill, K.K., Nicholson, W.E., Ekhator, N.N., patients with borderline personality disorder: an open-label trial. J. Clin. Psychiatry
Bruce, A.B., Wortman, M.D., Keck Jr., P.E., Geracioti Jr., T.D., October 1997. 60 (9), 598–603.
Cerebrospinal fluid and plasma beta-endorphin in combat veterans with post- Bondy, C.A., Gainer, H., Russell, J.T., 1988. Dynorphin A inhibits and naloxone
traumatic stress disorder. Psychoneuroendocrinology 22 (7), 517–529. increases the electrically stimulated release of oxytocin but not vasopressin from
Bale, T.L., Epperson, C.N., 2015. Sex differences and stress across the lifespan. Nat. the terminals of the neural lobe. Endocrinology 122 (4), 1321–1327.
Neurosci. 18, 1413–1420. Borsook, D., Falkowski, O., Burstein, R., Strassman, A., Konradi, C., Dauber, A.,
Bals-Kubik, R., Ableitner, A., Herz, A., Shippenberg, T.S., 1993. Neuroanatomical sites Comb, M., Hyman, S.E., 1994a. Stress-induced regulation of a human
mediating the motivational effects of opioids as mapped by the conditioned place proenkephalin-beta-galactosidase fusion gene in the hypothalamus of transgenic
preference paradigm in rats. J. Pharmacol. Exp. Ther. 264 (1), 489–495. mice. Mol. Endocrinol. 8, 116–125.
250 Stress and Opioid Systems

Borsook, D., Falkowski, O., Rosen, H., Comb, M., Hyman, S.E., 1994b. Opioids Carr, G.V., Bangasser, D.A., Bethea, T., Young, M., Valentino, R.J., Lucki, I., 2010.
modulate stress-induced proenkephalin gene expression in the hypothalamus of Antidepressant-like effects of kappa-opioid receptor antagonists in Wistar Kyoto
transgenic mice: a model of endogenous opioid gene regulation by exogenous rats. Neuropsychopharmacology 35, 752–763.
opioids. J. Neurosci. 14, 7261–7271. Castellano, C., Ammassari-Teule, M., Libri, V., Pavone, F., 1988. Effects of kappa-
Borsook, D., Konradi, C., Falkowski, O., Comb, M., Hyman, S.E., 1994c. opioid receptor agonists on locomotor activity and memory processes in mice.
Molecular mechanisms of stress-induced proenkephalin gene regulation: Pol. J. Pharmacol. Pharm. 40, 507–513.
CREB interacts with the proenkephalin gene in the mouse hypothalamus and Chance, W.T., White, A.C., Krynock, G.M., Rosecrans, J.A., 1978. Morphine-induced
is phosphorylated in response to hyperosmolar stress. Mol. Endocrinol. 8, changes in the activity of proopiomelanocortin and prodynorphin systems in
240–248. zymosan-induced peritonitis in mice. Brain Res. 141, 371–374.
Bouix, O., Najimi, A., Lenoir, V., Kerdelhue, B., Orsetti, A., 1996. Endogenous oploid Chang, G.Q., Karatayev, O., Ahsan, R., Gaysinskaya, V., Marwil, Z., Leibowitz, S.F.,
peptides stimulate post-exercise insulin response to glucose in rats. Int. J. Sports 2007. Dietary fat stimulates endogenous enkephalin and dynorphin in the para-
Med. 17, 80–84. ventricular nucleus: role of circulating triglycerides. AJP Endocrinol. Metab. 292
van den Brink, O.W., Delbridge, L.M., Rosenfeldt, F.L., Penny, D., Esmore, D.S., (2), E561–E570.
Quick, D., Kaye, D.M., Pepe, S., 2003. Endogenous cardiac opioids: Chartoff, E.H., Mavrikaki, M., 2015. Sex differences in kappa opioid receptor function
enkephalins in adaptation and protection of the heart. Heart Lung Circ. 12 and their potential impact on addiction. Front. Neurosci. 9, 466. http://dx.doi.org/
(3), 178–187. 10.3389/fnins.2015.00466.
Briski, K.P., Quigley, K., Meites, J., 1984. Endogenous opiate involvement in acute and Chatterton, R.T., 1990. The role of stress in female reproduction: animal and human
chronic stress-induced changes in plasma LH concentrations in the male rat. Life considerations. Int. J. Fertil. 35, 8–13.
Sci. 34, 2485–2493. Chavkin, C., 2013. Dynorphin-still an extraordinarily potent opioid peptide. Mol.
Broekkamp, C.L., Phillips, A.G., Cools, A.R., 1979. Facilitation of self stimulation Pharmacol. 83, 729–736.
behavior following intracerebral microinjection of opioid into the ventral tegmental Chefer, V.I., Bäckman, C.M., Gigante, E.D., Shippenberg, T.S., 2013. Kappa opioid
area. Pharmacol. Biochem. Behav. 11, 289–295. receptors on dopaminergic neurons are necessary for kappa-mediated place
Bronstein, D.M., Schafer, M.K., Watson, S.J., Akil, H., 1992. Evidence that beta- aversion. Neuropsychopharmacology 38, 2623–2631.
endorphin is synthesized in cells in the nucleus tractus solitarius: detection of Chen, Y., Mestek, A., Liu, J., Hurley, J.A., Yu, L., 1993a. Molecular cloning and
POMC mRNA. Brain Res. 587 (2), 269–275. functional expression of a mu-opioid receptor from rat brain. Mol. Pharmacol. 44
Brown, C.H., Leng, G., 2000. In vivo modulation of post-spike excitability in vaso- (1), 8–12.
pressin cells by kappa-opioid receptor activation. J. Neuroendocrinol. 12 (8), Chen, Y., Mestek, A., Liu, J., Yu, L., 1993b. Molecular cloning of a rat kappa opioid
711–714. receptor reveals sequence similarities to the mu and delta opioid receptors. Bio-
Brown, S.L., Van Epps, D.E., 1985. Suppression of T lymphocyte chemotactic factor chem. J. 295 (Pt 3), 625–628.
production by the opioid peptides beta-endorphin and met-enkephalin. J. Immunol. Chipkin, R.E., Latranyi, M.B., Iorio, L.C., 1982. Potentiation of stress-induced anal-
134 (5), 3384–3390. gesia (SIA) by thiorphan and its block by naloxone. Life Sci. 31, 1189–1192.
Brown, C.H., Russell, J.A., Leng, O., 2000. Opioid modulation of magnocellular Christie, M.J., Chesher, G.B., 1983. [3H]leu-enkephalin binding following chronic
neurosecretory cell activity. Neurosci. Res. 36 (2), 97–120. swim-stress in mice. Neurosci. Lett. 36, 323–328.
Bruchas, M.R., Land, B.B., Aita, M., Xu, M., Barot, S.K., Li, S., Chavkin, C., 2007. Christie, M.J., Chesher, G.B., Bird, K.D., 1981. The correlation between swim-stress
Stress-induced p38 mitogen-activated protein kinase activation mediates kappa- induced antinociception and [3H] leu-enkephalin binding to brain homogenates in
opioid-dependent dysphoria. J. Neurosci. 27 (43), 11614–11623. mice. Pharmacol. Biochem. Behav. 15, 853–857.
Buckingham, J.C., 1986. Stimulation and inhibition of corticotropin releasing factor Chuang, R.Y., 1995. Mu opioid receptor gene expression in immune cells. Biochem.
secretion by b-endorphin. Neuroendocrinology 42, 148–152. Biophys. Res. Commun. 16 (3), 922–930.
Bugnon, C., Bloch, B., Lenys, D., Gouget, A., Fellmann, D., 1979. Comparative Colwell, D.D., Kavaliers, M., 1992. Evidence for activation of endogenous opioid
study of the neuronal populations containing beta-endorphin, corticotropin systems in mice following short exposure to. Med. Vet. Entomol. 6, 159–164.
and dopamine in the arcuate nucleus of the rat hypothalamus. Neurosci. Lett. 14 Contet, C., Gaveriaux-Ruff, C., Matifas, A., Caradec, C., Champy, M.F., Kieffer, B.L.,
(1), 43–48. 2006. Dissociation of analgesic and hormonal responses to forced swim stress
Burns, G., Almeida, O.F.X., Passarelli, F., Herz, A., 1989. A two-step mechanism by using opioid receptor knockout mice. Neuropsychopharmacology 31 (8),
which corticotropin-releasing hormone releases hypothalamic b-endorphin: the role 1733–1744.
of vasopressin and G-proteins. Endocrinology 125, 1365–1372. Cooper, K., Carmody, J., 1982. The characteristics of the opioid-related analgesia
Bush, J.A., Kraemer, W.J., Mastro, A.M., Triplett-McBride, N.T., Volek, J.S., induced by the stress of swimming in the mouse. Neurosci. Lett. 31, 165–170.
Putukian, M., Sebastianelli, W.J., Knuttgen, H.G., 1999. Exercise and recovery Cowan, A., 2007. Buprenorphine: the basic pharmacology revisited. J. Addict. Med. 1,
responses of adrenal medullary neurohormones to heavy resistance exercise. Med. 68–72.
Sci. Sports Exerc. 31, 554–559. Cox, B.M., Christie, M.J., Devi, L., Toll, L., Traynor, J.R., 2015. Challenges for
Byck, R., 1976. Peptide transmitters: a unifying hypothesis for euphoria, respiration, opioid receptor nomenclature: IUPHAR review 9. Br. J. Pharmacol. 172 (2),
sleep, and the action of lithium. Lancet 2 (7976), 72–73. 317–323.
Calcagnetti, D.J., Holtzman, S.G., 1992. Potentiation of morphine analgesia in rats Craft, T.K., Devries, A.C., 2009. Vulnerability to stroke: implications of perinatal
given a single exposure to restraint stress immobilization. Pharmacol. Biochem. programming of the hypothalamic-pituitary-adrenal axis. Front. Behav. Neurosci.
Behav. 41 (2), 449–453. 3, 54.
Calcagnetti, D.J., Fleetwood, S.W., Holtzman, S.G., 1990. Pharmacological profile of Cristea, A., Restian, A., Vaduva, G., 1993. Endogenous opioid abstinence syndrome.
the potentiation of opioid analgesia by restraint stress. Pharmacol. Biochem. Rom. J. Physiol. 30, 241–247.
Behav. 37, 193–199. Cuadra, G., Zurita, A., Lacerra, C., Molina, V., 1999. Chronic stress sensitizes frontal
Calcagnetti, D.J., Stafinsky, J.L., Crisp, T., 1992. A single restraint stress exposure cortex dopamine release in response to a subsequent novel stressor: reversal by
potentiates analgesia induced by intrathecally administered DAGO. Brain Res. naloxone. Brain Res. Bull. 48, 303–308.
592, 305–309. Dantas, G., Torres, I.L., Crema, L.M., Lara, D.R., Dalmaz, C., 2005. Repeated restraint
Calogero, A.E., Bagdy, G., D’Agata, R., 1998. Mechanisms of stress on reproduction. stress reduces opioid receptor binding in different rat CNS structures. Neurochem.
Evidence for a complex intra-hypothalamic circuit. Ann. N.Y. Acad. Sci. 851, Res. 30 (1), 1–7.
364–370. Daskalakis, N.P., Lehrner, A., Yehuda, R., 2013. Endocrine aspects of post-traumatic
Cannon, J.T., Terman, G.W., Lewis, J.W., Liebeskind, J.C., 1984. Body region stress disorder and implications for diagnosis and treatment. Endocrinol. Metab.
shocked need not critically define the neurochemical basis of stress analgesia. Clin. N. Am. 42, 503–513.
Brain Res. 323, 316–319. Davis, M.P., 2012. Twelve reasons for considering buprenorphine as a frontline
Cannon, W.B., 1939. The Wisdom of the Body. Norton, New York. analgesic in the management of pain. J. Support. Oncol. 10, 209–219.
Carr, D.J., Kim, C.H., deCosta, B., Jacobson, A.E., Rice, K.C., Blalock, J.E., 1988. Day, R., Schafer, M.K., Collard, M.W., Watson, S.J., Akil, H., 1991. Atypical prody-
Evidence for a delta-class opioid receptor on cells of the immune system. Cell. norphin gene expression in corticosteroid-producing cells of the rat adrenal gland.
Immunol. 116 (1), 44–51. Proc. Natl. Acad. Sci. U.S.A. 88, 1320–1324.
Carr, D.J., DeCosta, B.R., Kim, C.H., Jacobson, A.E., Guarcello, V., Rice, K.C., Day, R., Schafer, M.K., Collard, M.W., Weihe, E., Akil, H., 1993. Prodynorphin gene
Blalock, J.E., 1989. Opioid receptors on cells of the immune system: evidence for expression in the rat intermediate pituitary lobe: gender differences and postpartum
delta- and kappa-classes. J. Endocrinol. 122, 161–168. regulation. Endocrinology 133 (6), 2652–2659.
Stress and Opioid Systems 251

Der-Avakian, A., Bland, S.T., Rozeske, R.R., Tamblyn, J.P., Hutchinson, M.R., Fadda, P., Tortorella, A., Fratta, W., 1991. Sleep deprivation decreases mu and delta
Watkins, L.R., Maier, S.F., 2007. The effects of a single exposure to uncontrollable opioid receptor binding in the rat limbic system. Neurosci. Lett. 129, 315–317.
stress on the subsequent conditioned place preference responses to oxycodone, Fadda, P., Martellotta, M.C., De Montis, M.G., Gessa, G.L., Fratta, W., 1992.
cocaine, and ethanol in rats. Psychopharmacology (Berl.) 191 (4), 909–917. Dopamine D1 and opioid receptor binding changes in the limbic system of sleep
Deroche, V., Piazza, P.V., Casolini, P., Maccari, S., Le Moal, M., Simon, H., 1992. deprived rats. Neurochem. Int. 20, 153S–156S.
Stress-induced sensitization to amphetamine and morphine psychomotor effects Fadda, P., Martellotta, M.C., Gessa, G.L., Fratta, W., 1993. Dopamine and opioids
depend on stress-induced corticosterone secretion. Brain Res. 598 (1–2), interactions in sleep deprivation. Prog. Neuropsychopharmacol. Biol. Psychiatry 17,
343–348. 269–278.
Deroche, V., Piazza, P.V., Casolini, P., Le Moal, M., Simon, H., 1993. Sensitization to Faden, A.J., Molineauks, C.J., Rosenberg, J.G., Jacobs, T.P., Cox, B.M., 1985.
the psychomotor effects of amphetamine and morphine induced by food restriction Endogenous opioid immunoreactivity in rat spinal cord following traumatic injury.
depends on corticosterone secretion. Brain Res. 611 (2), 352–356. Ann. Neurol. 17, 368–390.
Deutch, A.Y., Tam, S.Y., Roth, R.H., 1985. Footshock and conditioned stress increase Falcon, E., Maier, K., Robinson, S.A., Hill-Smith, T.E., Lucki, I., 2015. Effects of
DOPAC in the ventral tegmental area but not substantia nigra. Brain Res. 333, buprenorphine on behavioral tests for antidepressant and anxiolytic drugs in mice.
143–146. Psychopharmacology (Berl.) 232, 907–915.
Devine, D.P., Leone, P., Pocock, D., Wise, R.A., 1993. Differential involvement of Fanselow, M.S., Sigmundi, R.A., 1986. Species-specific danger signals, endogenous
ventral tegmental mu, delta and kappa opioid receptors in modulation of basal opioid analgesia, and defensive behavior. J. Exp. Psychol. Anim. Behav. Process.
mesolimbic dopamine release: in vivo microdialysis studies. J. Pharmacol. Exp. 12, 301–309.
Ther. 266 (3), 1236–1246. Fanselow, M.S., Calcagnetti, D.J., Helmstetter, F.J., 1989. Role of mu and kappa
DeVries, G.H., Chance, W.T., Payne, W.R., Rosecrans, J.A., 1979. Effect of auto- opioid receptors in conditional fear-induced analgesia: the antagonistic actions of
analgesia on CNS enkephalin receptors. Pharmacol. Biochem. Behav. 11, 741–744. nor-binaltorphimine and the cyclic somatostatin octapeptide, Cys2Tyr3Orn5Pen7-
Di Chiara, G., Imperato, A., 1988. Opposite effects of p- and K-opiate agonists on amide. J. Pharmacol. Exp. Ther. 250 (3), 825–830.
dopamine release in the nucleus accumbens and in the dorsal caudate of freely Fanselow, M.S., 1986. Conditioned fear-induced opiate analgesia: a competing
moving rats. J. Pharmacol. Exp. Ther. 244, 1067–1081. motivational state theory of stress analgesia. Ann. N.Y. Acad. Sci. 467,
Dietis, N., Rowbotham, D.J., Lambert, D.G., 2011. Opioid receptor subtypes: fact or 40–54.
artifact? Br. J. Anaesth. 107 (1), 8–18. Farrell, L.D., Harrison, T.S., Demers, L.M., 1983. Immunoreactive met-enkephalin in
Diez-Guerra, F.J., Augood, S., Emson, P.C., Dyer, R.G., 1986. Morphine inhibits the canine adrenal; response to acute hypovolemic stress. Proc. Soc. Exp. Biol.
electrically stimulated noradrenaline release from slices of rat medial preoptic area. Med. 173 (4), 515.
Neuroendocrinology 43, 89–91. Ferin, M., Vande Wiele, R., 1984. Endogenous opioid peptides and the control of the
Diez-Otanez, C.S., Capriles, N.R., Cancela, L.M., 1997. D1 and D2 dopamine and opiate menstrual cycle. Eur. J. Obstet. Gynecol. Reprod. Biol. 18, 365–373.
receptors are involved in the restraint stress-induced sensitization to the psychosti- Ferin, M., van Vugt, D., Wardlaw, S., 1984. The hypothalamic control of the menstrual
mulant effects of amphetamine. Pharmacol. Biochem. Behav. 58 (1), 9–14. cycle and the role of endogenous opioid peptides. Recent Prog. Horm. Res. 40,
Domanski, E., Przekop, F., Chomicka, L., Ostrowska, A., 1989. Effect of stress on the 441–445.
course of oestrous cycle and the release of luteinizing hormone; the role of Ferin, M., 1993. Neuropeptides, the stress response, and the hypothalamo-pituitary-
endorphin in these processes. Acta Physiol. Pol. 40, 64–73. gonadal axis in the female rhesus monkey. Ann. N.Y. Acad. Sci. 697, 106–116.
Dong, Z., Han, H., Wang, M., Xu, L., Hao, W., Cao, J., 2006. Morphine conditioned Feuerstein, G., Chiueh, C.C., Kopin, I.J., 1981. Effect of naloxone on the cardiovas-
place preference depends on glucocorticoid receptors in both hippocampus and cular and sympathetic response to hypovolemic hypotension in the rat. Eur. J.
nucleus accumbens. Hippocampus 16 (10), 809–813. Pharmacol. 75 (1), 65–69.
Drake, C.T., Chavkin, C., Milner, T.A., 2007. Opioid systems in the dentate gyrus. Fields, H.L., 2000. Pain modulation: expectation, opioid analgesia and virtual pain.
Prog. Brain Res. 163, 245–263. Prog. Brain Res. 122, 245–253.
Drolet, G., Dumont, E.C., Gosselin, I., Kinkead, R., Laforest, S., Trottier, J.F., 2001. Filliol, D., Ghozland, S., Chluba, J., Martin, M., Matthes, H.W., Simonin, F., Befort, K.,
Role of endogenous opioid system in the regulation of the stress response. Biol. Gavériaux-Ruff, C., Dierich, A., LeMeur, M., Valverde, O., Maldonado, R.,
Psychiatry 25 (4), 729–741. Kieffer, B.L., 2000. Mice deficient for delta- and mu-opioid receptors exhibit
Droste, C., 1992. Transient hypoalgesia under physical exercise–relation to silent opposing alterations of emotional responses. Nat. Genet. 25, 195–200.
ischaemia and implications for cardiac rehabilitation. Ann. Acad. Med. Singap. 21, Fink, M., Simeon, J., Itil, T.M., Freedman, A.M., 1970. Clinical antidepressant activity
23–33. of cyclazocine–a narcotic antagonist. Clin. Pharmacol. Ther. 11, 41–48.
Ehrenreich, H., Stender, N., Gefeller, O., tom Dieck, K., Schilling, L., Kaw, S., 1996. Fisher, L.A., 1989. Corticotropin-releasing factor: endocrine and autonomic integration
A novelty-related sustained elevation of vasopressin plasma levels in young men is of responses to stress. Trends Pharmacol. Sci. 10, 189–193.
not associated with an enhanced response of adrenocorticotropic hormone (ACTH) Fontana, F., Bernardi, P., Pich, E.M., Boschi, S., De Iasio, R., Spampinato, S.,
to human corticotropin releasing factor (hCRF). Res. Exp. Med. (Berl.) 196 (5), Grossi, G., 1997. Opioid peptide modulation of circulatory and endocrine response
291–299. to mental stress in humans. Peptides 18, 169–175.
Ehrich, E., Turncliff, R., Du, Y., Leigh-Pemberton, R., Fernandez, E., Jones, R., Forman, L.J., Estilow, S., 1988. Estrogen influences the effect of immobilization stress
Fava, M., 2015. Evaluation of opioid modulation in major depressive disorder. on immunoreactive b-endorphin levels in the female rat pituitary. Proc. Soc. Exp.
Neuropsychopharmacology 40, 1448–1455. Biol. Med. 187, 190–196.
Emoto, H., Koga, C., Ishii, H., Yokoo, H., Yoshida, M., Tanaka, M.A., 1993. CRF Forman, L.J., Estilow, S., Mead, J., Vasilenko, P., 1988. Eight weeks of streptozotocin-
antagonist attenuates stress-induced increases in NA turnover in extended brain induced diabetes influences the effects of cold stress on immunoreactive beta-
regions in rats. Brain Res. 627 (1), 171–176. endorphin levels in female rats. Horm. Metab. Res. 10, 555–558.
Emrich, H.M., Vogt, P., Herz, A., Kissling, W., 1982. Antidepressant effects of Frances, H., Graulet, A., Debray, M., Coudereau, J.P., Gueris, J., Bourre, J.M.,
buprenorphine. Lancet 2 (8300), 709. 2000. Morphine-induced sensitization of locomotor activity in mice: effect of
Erbs, E., Faget, L., Veinante, P., Kieffer, B.L., Massotte, D., 2014. In vivo neuronal co- social isolation on plasma corticosterone levels. Brain Res. 860 (1–2),
expression of mu and delta opioid receptors uncovers new therapeutic perspec- 136–140.
tives. Recept. Clin. Investig. 1 (5), 210. Fujiwara, R., Shibata, H., Komori, T., Yokoyama, M.M., Okazaki, Y., Ohmori, M., 1999.
Eskeland, N.L., Lugo, D.I., Pintar, J.E., Schachter, B.S., 1989. Stimulation of beta- The mechanisms of immune suppression by high-pressure stress in mice.
endorphin secretion by corticotropin-releasing factor in primary rat Leydig cell J. Pharm. Pharmacol. 51 (12), 1397–1404.
cultures. Endocrinology 124 (6), 2914–2919. Fukuda, K., Kato, S., Mori, K., Nishi, M., Takeshima, H., 1993. Primary structures and
Eskeland, N.L., Molineaux, C.J., Schachter, B.S., 1992. Regulation of beta-endorphin expression from cDNAs of rat opioid receptor delta- and mu-subtypes. FEBS Lett.
secretion by corticotropin-releasing factor in the intact rat testis. Endocrinology 130 327 (3), 311–314.
(3), 1173–1179. Funabashi, T., Brooks, P.J., Kleopoulos, S.P., Grandison, L., Mobbs, C.V.,
Evans, C.J., Keith Jr., D.E., Morrison, H., Magendzo, K., Edwards, R.H., 1992. Cloning Pfaff, D.W., 1995. Changes in preproenkephalin messenger RNA level in the rat
of a delta opioid receptor by functional expression. Science 258 (5090), ventromedial hypothalamus during the estrous cycle. Brain Res. Mol. Brain Res.
1952–1955. 28 (1), 129–134.
Fabbri, A., Knox, G., Buczko, E., Dufau, M.L., 1988. Beta-endorphin production by the Gabilondo, A.M., Meana, J.J., García-Sevilla, J.A., 1995. Increased density of mu-
fetal Leydig cell: regulation and implications for paracrine control of Sertoli cell opioid receptors in the postmortem brain of suicide victims. Brain Res. 682,
function. Endocrinology 122 (2), 749–755. 245–250.
252 Stress and Opioid Systems

Garcia-Garcia, L., Harbuz, M.S., Manzanares, J., Lightman, S.L., Fuentes, J.A., 1998. Haas, D.A., George, S.R., 1988. Single or repeated mild stress increases synthesis
RU-486 blocks stress-induced enhancement of proenkephalin gene expression in and release of hypothalamic corticotropin-releasing factor. Brain Res. 461 (2),
the paraventricular nucleus of rat hypothalamus. Brain Res. 786, 215–218. 230–237.
Gatti, G., Masera, R.G., Pallavicini, L., Sartori, M.L., Staurenghi, A., Orlandi, F., Hackler, L., Zadina, J.E., Ge, L.J., Kastin, A.J., 1997. Isolation of relatively large
Angeli, A., 1993. Interplay in vitro between ACTH, beta-endorphin, and gluco- amounts of endomorphin-1 and endomorphin-2 from human brain cortex. Peptides
corticoids in the modulation of spontaneous and lymphokine-inducible human 18 (10), 1635–1639.
natural killer (NK) cell activity. Brain Behav. Immun. 7, 16–28. Hamm, R.J., Knisely, J.S., 1987. Ontogeny of an endogenous, nonopioid and
Gaveriaux, C., Peluso, J., Simonin, F., Laforet, J., Kieffer, B., 1995. Identification of hormonally mediated analgesic system. Dev. Psychobiol. 20, 539–548.
kappa- and delta-opioid receptor transcripts in immune cells. FEBS Lett. 369 (2– Hanbauer, I., Govoni, F., Majane E,Yang, H.T., Costa, E., 1982. In vivo regulation of the
3), 272–276. release of met enkephalin-like peptides from dog adrenal medulla. Adv. Biochem.
Gayle, D., Ilyin, S.E., Plata-Salaman, C.R., 1999. Feeding status and bacterial LPS- Psychopharmacol. 33, 63–69.
induced cytokine and neuropeptide gene expression in hypothalamus. Am. J. Harbuz, M.S., Lightman, S.L., 1989. Responses of hypothalamic and pituitary mRNA to
Physiol. 277 (4 Pt 2), R1188–R1195. physical and psychological sterss in rat. J. Endocrinol. 122, 705–711.
Genazzani, A.R., Genazzani, A.D., Volpogni, C., Pianazzi, F., Li, G.A., Surico, N., Harbuz, M.S., Lightman, S.L., 1992. Stress and the hypothalamo-pituitary-adrenal
Petraglia, F., 1993. Opioid control of gonadotrophin secretion in humans. Hum. axis: acute, chronic and immunological activation. J. Endocrinol. 134 (3),
Reprod. 8 (Suppl. 2), 151–153. 327–339.
George, T.P., Verrico, C.D., Xu, L., Roth, R.H., 2000. Effects of repeated nicotine Harrison, C., 2013. Trial watch: opioid receptor blocker shows promise in Phase II
administration and footshock stress on rat mesoprefrontal dopamine systems: depression trial. Nat. Rev. Drug Discov. 12, 415.
evidence for opioid mechanisms. Neuropsychopharmacology 23, 79–88. Hart, S.L., Slusarczyk, H., Smith, T.W., 1983. The involvement of opioid delta-
Gerner, R.H., Catlin, D.H., Gorelick, D.A., Hui, K.K., Li, C.H., 1980. Beta-Endorphin. receptors in stress induced antinociception in mice. Eur. J. Pharmacol. 95,
Intravenous infusion causes behavioral change in psychiatric inpatients. Arch. Gen. 283–285.
Psychiatry 37, 642–647. Hart, S.L., Slusarczyk, H., Smith, T.W., 1985. The effects of selective opioid delta-
Gieryk, A., Ziolkowska, B., Solecki, W., Kubik, J., Przewlocki, R., 2010. Forebrain receptor antagonists on stress-induced antinociception and plasma corticoste-
PENK and PDYN gene expression levels in three inbred strains of mice and their rone levels in mice. Neuropeptides 5, 303–306.
relationship to genotype-dependent morphine reward sensitivity. Psychopharma- Haskins, J.T., Gudelsky, G.A., Moss, R.L., Porter, J.C., 1981. Iontophoresis of
cology (Berl.) 208 (2), 291–300. morphine into the arcuate nucleus effects on dopamine concentrations in hypo-
Gillies, G.E., Linton, E.A., Lowry, P.J., 1982. Corticotropin-releasing activity of new physial portal plasma and serum prolactin concentrations. Endocrinology 08,
CRF is potentiated several times by vasopressin. Nature 299, 355–357. 767–771.
Girardot, M.N., Holloway, F.A., 1985a. Effect of age and long-term stress experience Hassen, A.K., Feuerstein, G.Z., Faden, A.I., 1982. Cardiovascular responses to opioid
on adaptation to stress analgesia in mature rats: role of opioids. Behav. Neurosci. agonists injected into the nucleus of the tractus solitarius of anaesthetized cats.
99, 411–422. Life Sci. 31, 2193–2196.
Girardot, M.N., Holloway, F.A., 1985b. Naltrexone antagonizes the biobehavioral Hawkins, M.F., Fuller, R.D., Baumeister, A.A., McCallum, M.D., 1994. Effects in the rat
adaptation to cold water stress in rats. Pharmacol. Biochem. Behav. 22, of intranigral morphine and DAGO on eating and gnawing induced by stress.
769–779. Pharmacol. Biochem. Behav. 49, 737–740.
Glaser, R., Rice, J., Sheridan, J., Fertel, R., Stout, J., Speicher, C., Pinsky, D., Hawranko, A.A., Monroe, P.J., Smith, D.J., 1994. Repetitive exposure to the hot-plate
Kotur, M., Post, A., Beck, M., 1987. Stress-related immune suppression: health test produces stress induced analgesia and alters beta-endorphin neuronal
implications. Brain Behav. Immun. 1 (1), 7–20. transmission within the periaqueductal gray of the rat. Brain Res. 667, 283–286.
Glatt, C.E., Kenner, J.R., Long, J.B., Holaday, J.W., 1987. Cardiovascular effects of Hawranko, A.A., Serafini, M., Smith, D.J., 1999. Anti-analgesia and reduced anti-
dynorphin A (1-13) in conscious rats and its modulation of morphine bradycardia nociception from supraspinally administered beta-endorphin in stressed rats:
over time. Peptides 8, 1089–1092. dependence on spinal cholecystokinin via cholecystokinin B receptors. Neurosci.
Glatzer, N.R., Smith, B.N., 2005. Modulation of synaptic transmission in the rat nucleus Lett. 267, 101–104.
of the solitary tract by endomorphin-1. J. Neurophysiol. 93 (5), 2530–2540. Hebb, A.L., Drolet, G., Mendella, P.D., Roach, S.P., Gauthier, M.S., Zacharko, R.M.,
de Goeij, D.C., Kvetnansky, R., Whitnall, M.H., Jezova, D., Berkenbosch, F., 2005. Intracerebroventricular D-Pen2, D-Pen5-enkephalin administration soon
Tilders, F.J., 1991. Repeated stress-induced activation of corticotropin-releasing after stressor imposition influences behavioral responsivity to a subsequent
factor neurons enhances vasopressin stores and colocalization with corticotropin- stressor encounter in CD-1 mice. Pharmacol. Biochem. Behav. 82 (3), 453–469.
releasing factor in the median eminence of rats. Neuroendocrinology 53 (2), Hedner, T., Cassuto, J., 1987. Opioids and opioid receptors in peripheral tissues.
150–159. Scand. J. Gastroenterol. 130 (Suppl.), 27–46.
Goloshchapov, A.V., Filipenko, M.L., Bondar, N.P., Kudryavtseva, N.N., Van Ree, J.M., Heijna, M.H., Hogenboom, F., Schoffelmee, A.N.M., Mulder, A.H., 1990. Opioid receptor
2005. Decrease of kappa-opioid receptor mRNA level in ventral tegmental area of mediated inhibition of dopamine release from rat basal hypothalamus slices;
male mice after repeated experience of aggression. Brain Res. Mol. Brain Res. 135 involvement of both p and K receptors. Eur. J. Pharmacol. 183, 2334–2335.
(1–2), 290–292. Hemingway, R.B., Reigle, T.G., 1987. The involvement of endogenous opiate systems
Gordon, C.J., Rezvani, A.H., Heath, J.E., 1984. Role of beta-endorphin in the control of in learned helplessness and stress-induced analgesia. Psychopharmacology (Berl.)
body temperature in the rabbit. Neurosci. Biobehav. Rev. 8, 73–82. 93, 353–357.
Gracely, R.H., Dubner, R., Wolskee, P.J., Deeter, W.R., 1983. Placebo and naloxone Hernandez, D.E., Nemeroff, C.B., Orlando, R.C., Prange, A.J., 1983. The effect of
can alter post-surgical pain by separate mechanisms. Nature 306, 264–265. centrally administered neuropeptides on the development of stress-induced gastric
Greenberg, A., Dyck, D., Sandler, L., 1984. Opponent processes, neurohormones and ulcers in rats. J. Neurosci. Res. 9, 145–157.
neural resistance. In: Fox, B., Newberry, B. (Eds.), Impact of Psychoendocrine in Herz, A., Emrich, H.M., 1983. Opioid systems and the regulation of mood: possible
Cancer and Immunity. Hogrefe, Toronto, p. 225. significance in depression? In: Angst, J. (Ed.), The Origins of Depression: Current
Greenwell, T.N., Martin-Schild, S., Inglis, F.M., Zadina, J.E., 2007. Colocalization and Concepts and Approaches. Springer, Berlin, pp. 221–234.
shared distribution of endomorphins with substance P, calcitonin gene-related Herz, A., 1995. Role of immune processes in peripheral opioid analgesia. Adv. Exp.
peptide, gamma-aminobutyric acid, and the mu opioid receptor. J. Comp. Neu- Med. Biol. 373, 193–199.
rol. 503 (2), 319–333. Herz, A., 1996. Peripheral opioid analgesia-facts and mechanisms. Prog. Brain Res.
Grigoriants, O.O., Pravdenkova, S.V., Andersen, B.J., Desiderio, D.M., 1995. Alteration 110, 95–104.
of opioid peptide concentrations in the rat pituitary following survivable closed head Hnatowich, M.R., Labella, F.S., Kiernan, K., Glavin, G.B., 1986. Cold-restraint stress
injury. Neurochem. Res. 20 (7), 827–831. reduces (3H) etorphine binding to rat brain membranes: influence of acute and
Gross-Isseroff, R., Biegon, A., Voet, H., Weizman, A., 1998. The suicide brain: chronic morphine and naloxone. Brain Res. 380, 107–113.
a review of postmortem receptor/transporter binding studies. Neurosci. Biobehav. Hodgson, D.M., Bond, N.W., 1996. The role of hypophyseal and adrenal mecha-
Rev. 22, 653–661. nisms in the hypoalgesic response to non-contingent food delivery in the rat.
Guillemin, R., Vargo, T., Rossier, J., Minick, S., Ling, N., Rivier, C., Vale, W., Bloom, F., Behav. Brain Res. 80, 27–32.
1977. b-Endorphin and adrenocorticotropin are secreted concomitantly by the Holaday, J.W., Hitzeman, R.J., Curell, I., Tortella, F.C., Belenky, G.I., 1982. Repeated
pituitary gland. Science 197, 1367–1369. electroconvulsive shock or chronic morphine treatment increases the number of
Gysling, K., Wang, R.Y., 1983. Morphine-induced activation of A10 dopamine neurons 3H-D-Ala2-D-Leu5-enkephalin binding sites in the rat rain membranes. Life Sci.
in the rat. Brain Res. 277, 119–127. 31, 2359–2362.
Stress and Opioid Systems 253

Holaday, J.W., 1983. Cardiovascular effects of endogenous opiate systems. Annu. Kalivas, P.W., Abhold, R., 1987. Enkephalin release into the ventral area in
Rev. Pharmacol. Toxicol. 23, 541–594. response to stress: modulation of mesocorticolimbic dopamine. Brain Res.
Höllt, V., Przewlocki, R., Haarmann, I., Almeida, O.F.X., Kley, N., Millan, M.J., Herz, A., 414, 339–348.
1986. Stress-induced alterations in the levels of messenger RNA coding for Kalivas, P.W., Duffy, P., Dilts, R., Abhold, R., 1988. Enkephalin modulation of A10
proopiomelanocortin and prolactin in rat pituitary. Neuroendocrinology 43, dopamine neurons: a role in dopamine sensitization. Ann. N.Y. Acad. Sci. 537,
277–282. 405–414.
Höllt, V., 1990. Regulation of opioid peptides gene expression. In: Herz, A. (Ed.), Kalivas, P.W., Duffy, P., Eberhardt, H., 1990. Modulation of A10 dopamine neurons
Opioids I, pp. 307–333. by gamma-aminobutyric acid antagonists. J. Pharmacol. Exp. Ther. 253,
Holsboer, F., Ising, M., 2010. Stress hormone regulation: biological role and translation 858–866.
into therapy. Annu. Rev. Psychol. 61, 81–109. Kanamatsu, T., Unsworth, C.D., Diliberto Jr., E.J., Viveros, O.H., Hong, J.S., 1986.
Holtzman, S.G., 1974. Behavioural effects of separate and combined administration Reflex splanchnic nerve stimulation increases levels of proenkephalin A mRNA and
of naloxone and amphetamine. J. Pharmacol. Exp. Ther. 189, 51–60. proenkephalin A-related peptides in the rat adrenal medulla. Proc. Natl. Acad. Sci.
Hong, J.S., Yoshikawa, K., Kanamatsu, T., McGinty, J.F., Sabol, S.L., 1985. Effects of U.S.A. 83, 9245–9249.
repeated electroconvulsive shock on the biosynthesis of enkephalin and concen- Kapas, L., Benedek, G., Penke, B., 1989. Cholecystokinin interferes with the ther-
tration of dynorphin in the rat brain. Neuropeptides 5, 557–560. moregulatory effect of exogenous and endogenous opioids. Neuropeptides 14,
Hooks, M.S., Sorg, B.A., Kalivas, P.W., 1994. The relationship between MRNA levels 85–92.
and the locomotor response to novelty. Brain Res. 663, 312–316. Karp, J.F., Butters, M.A., Begley, A.E., Miller, M.D., Lenze, E.J., Blumberger, D.M.,
Hori, T., Katafuchi, T., Take, S., Shimizu, N., 1998. Neuroimmunomodulatory Mulsant, B.H., Reynolds, C.F., 2014. Safety, tolerability, and clinical effect of low-
actions of hypothalamic interferon-alpha. Neuroimmunomodulation 5 (3–4), dose buprenorphine for treatment-resistant depression in midlife and older adults.
172–177. J. Clin. Psychiatry 75, e785–e793.
Houdi, A.A., Marson, L., Davenport, K.E., Van Loon, G.R., 1996. Effects of beta-FNA Katz, R.J., Gormezano, G.A., 1979. Rapid and inexpensive technique for assessing
on sympathoadrenal, cardiovascular, and analgesic responses to DAMPGO at rest the reinforcing effects of opiate drugs. Pharmacol. Biochem. Behav. 11 (2),
and during stress. Pharmacol. Biochem. Behav. 53, 927–933. 231–233.
Illes, P., 1989. Modulation of transmitter and hormone release by multiple neuronal Katz, R.J., 1979. Opiate stimulation increases exploration in the mouse. Int. J. Neu-
opioid receptors. Rev. Physiol. Biochem. Pharmacol. 112, 140–233. rosci. 9 (4), 213–215.
Imaki, T., Naruse, M., Harada, S., Chikada, N., Imaki, J., Onodera, H., Demura, H., Kavaliers, M., Innes, D., 1987. Stress-induced opioid analgesia and activity in deer
Vale, W., 1996. Corticotropin-releasing factor up-regulates its own receptor mRNA mice: sex and population differences. Brain Res. 425, 49–56.
in the paraventricular nucleus of the hypothalamus. Brain Res. Mol. Brain Res. 38 Kavaliers, M., Innes, D.G., 1988. Novelty-induced opioid analgesia in deer mice
(1), 166–170. (Peromyscus maniculatus): sex and population differences. Behav. Neural Biol. 49,
Insel, T.R., Kinsley, C.H., Mann, P.E., Bridges, R.S., 1990. Prenatal stress has long 54–60.
term effects on brain opiate receptors. Brain Res. 511, 93–97. Kavaliers, M., 1987. Evidence for opioid and non-opioid forms of stress-induced
Isom, G.E., Elshowihy, R.M., 1982. Interaction of acute and chronic stress with analgesia in the snail, Cepaea nemoralis. Brain Res. 410, 111–115.
respiration: modification by naloxone. Pharmacol. Biochem. Behav. 16, 599–603. Kelley, A.E., Stinus, L., Iversen, S.D., 1980. Interactions between D-Ala-Met-
Iwamoto, E.T., 1985. Place-conditioning properties of mu, kappa, and sigma opioid enkephalin, A10 dopaminergic neurons, and spontaneous behavior in the rat.
agonists. Alcohol Drug Res. 6 (5), 327–339. Behav. Brain Res. 1, 3–24.
Iwasaki, H., Namiki, A., 1997. A review of pregnancy-induced analgesia. Masui 46, Kelly, S.J., Franklin, K.B., 1987. Role of peripheral and central opioid activity in
598–606. analgesia induced by restraint stress. Life Sci. 41, 789–794.
Izumi, R., Takahashi, M., Kaneto, H., 1983. Involvement of different mechanisms, Kelsey, S.J., Watson, S.J., Akil, H., 1984. Changes in pituitary POMC mRNA levels.
opioid and non-opioid forms, in the analgesia induced by footshock (FS) and Soc. Neurosci. Abstr. 10, 359.
immobilized-water immersion (IW) stress. Jpn. J. Pharmacol. 33, 1104–1106. Kelsey, J.E., Hoerman, W.A., Kimball, L.D., Radack, L.S., Carter, M.V., 1986. Arcuate
Jackisch, R., Geppert, M., Lupp, A., Huang, H.Y., Illes, P., 1988. Types of opioid nucleus lesions reduce opioid stress-induced analgesia (SIA) and enhance non-
receptors modulating neurotransmitter release in discrete brain regions. In: Illes, P., opioid SIA in rats (published erratum appears in March 31, 1987. Brain Res.
Farsang, C. (Eds.), Regulatory Roles of Opioid Peptides. VCH, Weinheim, 407 (2), 412). Brain Res. 382, 278–290.
pp. 240–258. Kelso, T.B., Herbert, W.G., Gwazdauskas, F.C., Goss, F.L., Hess, J.L., 1984. Exercise-
Jaffe, J.H., Martin, W.R., 1990. Opioid analgesics and antagonists. In: Gilman, A.G., thermoregulatory stress and increased plasma beta-endorphin/beta-lipotropin in
et al. (Eds.), Goodman and Gilman’s the Pharmacological Basis of Therapeutics, humans. J. Appl. Physiol. 57, 444–449.
eighth ed. Pergamon, New York, pp. 485–521. Kennedy, S.E., Koeppe, R.A., Young, E.A., Zubieta, J.K., 2006. Dysregulation of
Jarry, H., Duker, E.M., Wuttke, W., 1985. Adrenal release of catecholamines and Met- endogenous opioid emotion regulation circuitry in major depression in women.
enkephalin before and after stress as measured by a novel in vivo dialysis method Arch. Gen. Psychiatry 63, 1199–1208.
in the rat. Neurosci. Lett. 60 (3), 273–278. Khachaturian, H., Lewis, M.E., Schafer, M.K.-H., Watson, S.J., 1985. Anatomy of CNS
Jenck, F., Gratton, A., Wise, R.A., 1987. Opioid receptor subtypes associated with opioid systems. Trends Neurosci. 8, 111–119.
ventral tegmental facilitation of lateral hypothalamic brain stimulation reward. Brain Kieffer, B.L., Befort, K., Gaveriaux-Ruff, C., Hirth, C.G., 1992. The delta-opioid
Res. 423, 34–38. receptor: isolation of a cDNA by expression cloning and pharmacological charac-
Jezova, D., Vigas, M., Tatar, P., 1985. Rise in plasma b-endorphin and ACTH in terization. Proc. Natl. Acad. Sci. U.S.A. 89 (24), 12048–12052.
response to hyperthermia sauna. Horm. Metab. Res. 17, 693–694. Killian, P., Holmes, B.B., Takemori, A.E., Portoghese, P.S., Fujimoto, J.M., 1995.
Jodar, L., Takahashi, M., Kaneto, H., 1994. Trends in physiological role of opioids Cold water swim stress- and delta-2 opioid-induced analgesia are modulated
in psychoneuroendocrine-immune network. Yakubutsu. Seishin. Kodo. 14, by spinal gamma-aminobutyric acidA receptors. J. Pharmacol. Exp. Ther. 274
195–214. (2), 730–734.
Johnson, R.D., 1995. Opioid involvement in feeding behaviour and the pathogenesis of Kiss, A., Aguilera, G., 1993. Regulation of the hypothalamic pituitary adrenal axis
certain eating disorders. Med. Hypotheses 45, 491–497. during chronic stress: responses to repeated intraperitoneal hypertonic saline
Jouppila, R., Jouppila, P., Karlqvist, K., Kaukoranta, P., Leppaluoto, J., injection. Brain Res. 630 (1–2), 262–270.
Vuolteenaho, O., 1983. Maternal and umbilical venous plasma immunoreactive Kitchen, I., Pinker, S.R., 1990. Antagonism of swim-stress-induced antinociception by
beta-endorphin levels during labor with and without epidural analgesia. Am. J. the delta-opioid receptor antagonist naltrindole in adult and young rats. Br. J.
Obstet. Gynecol. 147, 799–802. Pharmacol. 100 (4), 685–688.
Jutkiewicz, E.M., Roques, B.P., 2012. Endogenous opioids as physiological antide- Kjaer, A., Knigge, U., Vilhardt, H., Bach, F.W., Warberg, J., 1993a. Involvement of
pressants: complementary role of d receptors and dopamine. Neuro- vasopressin V1- and V2-receptors in histamine-and stress-induced secretion of
psychopharmacology 37, 303–304. ACTH and beta-endorphin. Neuroendocrinology 57, 503–509.
Jutkiewicz, E.M., 2006. The antidepressant -like effects of delta-opioid receptor Kjaer, A., Knigge, U., Bach, F.W., Warberg, J., 1993b. Permissive, mediating and
agonists. Mol. Interv. 6, 162–169. potentiating effects of vasopressin in the ACTH and beta-endorphin response to
Kakidani, H., Furutani, Y., Takahashi, H., Noda, M., Morimoto, Y., Hirose, T., Asai, M., histamine and restraint stress. Neuroendocrinology 58, 588–596.
Inayama, S., Nakanishi, S., Numa, S., 1982. Cloning and sequence analysis of Kjaer, A., Knigge, U., Bach, F.W., Warberg, J., 1995. Stress-induced secretion of pro-
cDNA for porcine beta-neo-endorphin/dynorphin precursor. Nature 298 (5871), opiomelanocortin-derived peptides in rats: relative importance of the anterior and
245–249. intermediate pituitary lobes. Neuroendocrinology 61, 167–172.
254 Stress and Opioid Systems

Klein, L.C., Popke, E.J., Grunberg, N.E., 1998. Sex differences in effects of opioid Lason, W., Przewlocka, B., Przewlocki, R., 1987. Single and repeated electrocon-
blockade on stress-induced freezing behavior. Pharmacol. Biochem. Behav. 61 (4), vulsive shock differentially affects the prodynorphin and proopiomelanocortin
413–417. system in the rat. Brain Res. 403, 301–307.
Kline, N.S., Li, C.H., Lehmann, H.E., Lajtha, A., Laski, E., Cooper, T., 1977. Beta- Latimer, L.G., Duffy, P., Kalivas, P.W., 1987. Mu opioid receptor involvement in
endorphin–induced changes in schizophrenic and depressed patients. Arch. enkephalin activation of dopamine neurons in the ventral tegmental area.
Gen. Psychiatry 34, 1111–1113. J. Pharmacol. Exp. Ther. 241, 328–337.
de Kloet, E.R., Joels, M., Holsboer, F., 2005. Stress and the brain: from adaptation to Laurent, S., Schmitt, H., 1983. Central cardiovascular effects of kappa agonists
disease. Nat. Rev. Neurosci. 6, 463–475. dynorphin-(1-13) and ethylketocyclazocine in the anaesthetized rat. Eur. J. Phar-
Knepel, W., Reimann, W., 1982. Inhibition by morphine and beta-endorphin of macol. 96, 165–169.
vasopressin release evoked by electrical stimulation of the rat medial basal Le Merrer, J., Becker, J.A., Befort, K., Kieffer, B.L., 2009. Reward processing by the
hypothalamus in vitro. Brain Res. 238, 484–488. opioid system in the brain. Physiol. Rev. 89 (4), 1379–1412.
Knepel, W., Przewlocki, R., Herz, A., 1985. Foot shock stress-induced release of Leander, J.D., 1982. A kappa opioid effects increased urination in the rat.
vasopressin in adenohypophysectomized and hypophysectomized rats. Endocri- J. Pharmacol. Exp. Ther. 224, 89–94.
nology 117, 292–299. Ledda, F., Mantelli, L., Corti, V., 1985. Sensitivity to dynorphin-(1-13) of the
Koch, T., Schulz, S., Schroder, H., Wolf, R., Raulf, E., Höllt, V., 1998. Carboxyl- presynaptic inhibitory opiate receptors of the guinea-pig heart. Eur. J. Pharmacol.
terminal splicing of the rat mu opioid receptor modulates agonist-mediated 117, 377–380.
internalization and receptor resensitization. J. Biol. Chem. 273 (22), Legakis, I., Saramantis, A., Voros, D., Chalevelakis, G., Tolis, G., 1998. Dissociation of
13652–13657. ACTH, beta-endorphin and cortisol in graded sepsis. Horm. Metab. Res. 30 (9),
Kokras, N., Dalla, C., 2014. Sex differences in animal models of psychiatric disorders. 570–574.
Br. J. Pharmacol. 171, 4595–4619. Leino, T.K., Leppaluoto, J., Ruokonen, A., Kurone, P., 1998. Pro-opiomelanocortin
Konecka, A.M., Sroczynska, I., 1990. Stressors and pain sensitivity in CFW mice. Role activation and simulated interceptor combat flight. Aviat. Space Environ. Med.
of opioid peptides. Arch. Int. Physiol. Biochim. 98, 245–252. 69, 486–490.
Konecka, A.M., Sroczynska, I., Przewlocki, R., 1985. The effect of food and water Leone, P., Pocock, D., Wise, R.A., 1991. Morphine-dopamine interaction: ventral
deprivation on post-stress analgesia in mice and levels of beta-endorphin and tegmental morphine increases nucleus accumbens dopamine release. Pharmacol.
dynorphin in blood plasma and hypothalamus. Arch. Int. Physiol. Biochim. 93 (4), Biochem. Behav. 39 (2), 469–472.
279–284. Leriche, M., Cote-Velez, A., Mendez, M., 2007. Presence of pro-opiomelanocortin
Konig, M., Zimmer, A.M., Steiner, H., Holmes, P.V., Crawley, J.N., 1990. Pain mRNA in the rat medial prefrontal cortex, nucleus accumbens and ventral
responses, anxiety and aggression in mice deficient in pre-proenkephalin. Nature tegmental area: studies by RT-PCR and in situ hybridization techniques.
383, 535–538. Neuropeptides 41 (6), 421–431.
Koob, G., Kreek, M.J., 2007. Stress, dysregulation of drug reward pathways, and the Levine, J.D., Feldmesser, M., Tecott, L., Lane, S., Gordon, N.C., 1984. The role of
transition to drug dependence. Am. J. Psychiatry 164 (8), 1149–1159. stimulus intensity and stress in opioid-mediated analgesia. Brain Res. 304,
Koob, G.F., Le Moal, M., 1997. Drug abuse: hedonic homeostatic dysregulation. 265–269.
Science 278 (5335), 52–58. Lewis, J.W., Cannon, J.T., Liebeskind, J.C., 1980. Opioid and nonopioid mechanisms
Koob, G.F., Mason, B.J., 2015. Existing and future drugs for the treatment of the of stress analgesia. Science 208, 623–625.
dark side of addiction. Annu. Rev. Pharmacol. Toxicol. 56. PubMed PMID: Lewis, J.W., Tordoff, M.G., Sherman, J.E., Liebeskind, J.C., 1982. Adrenal medullary
26514207. enkephalin-like peptides may mediate opioid stress analgesia. Science 217,
Koolhaas, J.M., Bartolomucci, A., Buwalda, B., de Boer, S.F., Flügge, G., Korte, S.M., 557–559.
Meerlo, P., Murison, R., Olivier, B., Palanza, P., Richter-Levin, G., Sgoifo, A., Lewis, J.W., Mansour, A., Khachaturian, H., Watson, S.J., Akil, H., 1987. Opioids and
Steimer, T., Stiedl, O., van Dijk, G., Wöhr, M., Fuchs, E., 2011. Stress revisited: pain regulation. In: Akil, H., Lewis, J.W. (Eds.), Neurotransmitters and Pain Control.
a critical evaluation of the stress concept. Neurosci. Biobehav. Rev. 35 (5), Karger, Basel, pp. 129–159.
1291–1301. Li, S., Zhu, J., Chen, C., Chen, Y.W., Deriel, J.K., Ashby, B., Liu-Chen, L.Y., 1993.
Kopin, I.J., Eisenhofer, G., Goldstein, D., 1988. Sympathomedullary system and stress. Molecular cloning and expression of a rat kappa opioid receptor. Biochem. J. 295
In: Chrousos, G.P., Loriaux, D.L., Gold, W. (Eds.), Mechanisms of Physical and (Pt 3), 629–633.
Emotional Stress. Plenum, New York, pp. 11–23. Li, D., Olszewski, P.K., Shi, Q., Grace, M.K., Billington, C.J., Kotz, C.M., Levine, A.S.,
Kostic, T., Andric, S., Maric, D., Kovacevic, R., 1998. The effect of acute stress and 2006. Effect of opioid receptor ligands injected into the rostral lateral hypothalamus
opioid antagonist on the activity of NADPH-P450 reductase in rat Leydig cells. on c-fos and feeding behavior. Brain Res. 1096 (1), 120–124.
J. Steroid Biochem. Mol. Biol. 66 (1–2), 51–54. Lightman, S.L., Young, W.S., 1989. Influence of steroids on the hypothalamic
Kreek, M.J., LaForge, K.S., 2007. Stress responsivity, addiction, and a functional corticotropin-releasing factor and preproenkephalin mRNA responses to stress.
variant of the human mu-opioid receptor gene. Mol. Interv. 2, 74–78. Proc. Natl. Acad. Sci. U.S.A. 86, 4306–4310.
Krishnan, V., Nestler, E.J., 2010. Linking molecules to mood: new insight into the Lim, A.T., Wallace, M., Oei, T.P., Gibson, S., Romas, N., Pappas, W.,
biology of depression. Am. J. Psychiatry 167, 1305–1320. Clements, J., Funder, J.W., 1982. Foot shock analgesia. Lack of correlation
Kulling, P., Frischknecht, H.R., Pasi, A., Waser, P.G., Siegfried, B., 1988. Social with pituitary and plasma immunoreactive-beta-endorphin. Neuroendocri-
conflict-induced changes in nociception and beta-endorphin-like immunoreac- nology 35, 236–241.
tivity in pituitary and discrete brain areas of C57BL/6 and DBA/2 mice. Brain Linton, E.A., Tilders, F.J.H., Hodgkinson, S., Berkenbosch, F., Vermes, I., Lowry, P.J.,
Res. 450, 237–246. 1985. Stress-induced secretion of adrenocorticotropin in rats is inhibited by
Kulling, P., Siegfried, B., Frischknecht, H.R., Messiha, F.S., Pasi, A., 1989. Beta- antisera to ovine corticotropin-releasing factor and vasopressin. Endocrinology 116,
endorphin-like immunoreactivity levels in the hypothalamus, the periaqueductal 966–970.
grey and the pituitary of the DBA mouse: determination by ELISA and relationship Liu, Q.S., Han, S., Jia, Y.S., Ju, G., 1999a. Selective modulation of excitatory
to nociception. Physiol. Behav. 46, 25–28. transmission by mu-opioid receptor activation in rat supraoptic neurons.
Kurumaji, A., Takashima, M., Shibuya, H., 1987. Cold and immobilization stress J. Neurophysiol. 82 (6), 3000–3005.
induced changes in pain responsiveness and brain met-enkephalin-like immuno- Liu, I.M., Niu, C.S., Chi, T.C., Kuo, D.H., Cheng, J.T., 1999b. Investigations of the
reactivity in the rat. Peptides 8, 355–359. mechanism of the reduction of plasma glucose by cold-stress in streptozotocin-
Kuzmin, A., Semenova, S., Zvartau, E.E., Van Ree, J.M., 1996. Enhancement of induced diabetic rats. Neuroscience 92, 1137–1142.
morphine self-administration in drug naive, inbred strains of mice by acute Lopez-Calderon, A., Ariznavarreta, C., Gonzalez-Quijano, M.I., Tresguerres, J.A.,
emotional stress. Eur. Neuropsychopharmacol. 6 (1), 63–68. Calderon, M.D., 1991. Stress induced changes in testis function. J. Steroid Bio-
LaBuda, C.J., Sora, I., Uhl, G.R., Fuchs, P.N., 2000. Stress-induced analgesia in mu- chem. Mol. Biol. 40, 473–479.
opioid receptor knockout mice reveals normal function of the delta-opioid receptor Lovallo, W.L., 1997. Stress and Health. Biological and Psychological Interactions. Sage
system. Brain Res. 869 (1–2), 1–5. Publications, Thousand Oaks, London, New Delhi.
Lalanne, L., Ayranci, G., Kieffer, B.L., Lutz, P.E., 2014. The kappa opioid receptor: Lovallo, W.R., 2006. Cortisol secretion patterns in addiction and addiction risk. Int. J.
from addiction to depression, and back. Front. Psychiatry 5, 170. Psychophysiol. 59 (3), 195–202.
Larsen, P.J., Mau, S.E., 1994. Effect of acute stress on the expression of hypothalamic Lucas, L.R., Wang, C.J., McCall, T.J., McEwen, B.S., 2007. Effects of immobilization
messenger ribonucleic acids encoding the endogenous opioid precursors pre- stress on neurochemical markers in the motivational system of the male rat. Brain
proenkephalin A and proopiomelanocortin. Peptides 15, 783–790. Res. 1155, 108–115.
Stress and Opioid Systems 255

Lucassen, P.J., Pruessner, J., Sousa, N., Almeida, O.F.X., Van Dam, A.M., McLaughlin, J.P., Li, S., Valdez, J., Chavkin, T.A., Chavkin, C., 2006. Social defeat
Rajkowska, G., Swaab, D.F., Czéh, B., 2014. Neuropathology of stress. Acta stress-induced behavioral responses are mediated by the endogenous kappa opioid
Neuropathol. 127, 109–135. system. Neuropsychopharmacology 31, 1241–1248.
Lutz, P.E., Kieffer, B.L., 2013. Opioid receptors: distinct roles in mood disorders. McLean, M., Thompson, D., Zhang, H.P., Brinsmead, M., Smith, R., 1994.
Trends Neurosci. 36, 195–206. Corticotrophin-releasing hormone and beta-endorphin in labour. Eur. J. Endocrinol.
Machado, A., Herrera, A.J., de Pablos, R.M., Espinosa-Oliva, A.M., Sarmiento, M., 131, 167–172.
Ayala, A., Venero, J.L., Santiago, M., Villarán, R.F., Delgado-Cortés, M.J., McQueen, D.S., 1983. Opioid peptide interactions with respiratory and circulatory
Argüelles, S., Cano, J., 2014. Chronic stress as a risk factor for Alzheimer’s systems. Br. Med. Bull. 39 (1), 77–82.
disease. Rev. Neurosci. 25, 785–804. Menendez, L., Andres-Trelles, F., Hidalgo, A., Baamonde, A., 1993. Involvement of
Mague, S.D., Pliakas, A.M., Todtenkopf, M.S., Tomasiewicz, H.C., Zhang, Y., spinal kappa opioid receptors in a type of footshock induced analgesia in mice.
Stevens Jr., W.C., Jones, R.M., Portoghese, P.S., Carlezon Jr., W.A., 2003. Brain Res. 611 (2), 264–271.
Antidepressant-like effects of kappa-opioid receptor antagonists in the forced swim Meng, F., Xie, G.X., Thompson, R.C., Mansour, A., Goldstein, A., Watson, S.J.,
test in rats. J. Pharmacol. Exp. Ther. 305, 323–330. Akil, H., 1993. Cloning and pharmacological characterization of a rat kappa opioid
Maier, S.F., Sherman, J.E., Lewis, J.W., Terman, G.W., Liebeskind, J.C., 1983. The receptor. Proc. Natl. Acad. Sci. U.S.A. 90 (21), 9954–9958.
opioid/nonopioid nature of stress-induced analgesia and learned helplessness. Mercer, M.E., Holder, M.D., 1997. Food cravings, endogenous opioid peptides, and
J. Exp. Psychol. Anim. Behav. Process. 9, 80–90. food intake: a review. Appetite 29, 325–352.
Majeed, N.H., Lason, W., Przewlocka, B., Przewlocki, R., 1986. Brain and peripheral Miczek, K.A., Thompson, M.L., Shuster, L., 1982. Opioid-like analgesia in defeated
opioids after changes in ingestive behaviour. Neuroendocrinology 42, 267–272. mice. Science 215, 1520–1522.
Makino, S., Asaba, K., Nishiyama, M., Hashimoto, K., 1999. Decreased type 2 Miczek, K.A., Thompson, M.L., Shuster, L., 1985. Naloxone injections into the peri-
corticotropin-releasing hormone receptor mRNA expression in the ventromedial aqueductal grey area and arcuate nucleus block analgesia in defeated mice.
hypothalamus during repeated immobilization stress. Neuroendocrinology 70 (3), Psychopharmacology (Berl.) 87, 39–42.
160–167. Millan, M.J., Przewlocki, R., Herz, A., 1980. A non-beta-endorphinergic adenohy-
Maley, B.E., 1996. Immunohistochemical localization of neuropeptides and neuro- pophyseal mechanism is essential for an analgetic response to stress. Pain 8,
transmitters in the nucleus solitarius. Chem. Senses 21 (3), 367–376. 343–353.
Mansi, J.A., Laforest, S., Drolet, G., 2000. Effect of stress exposure on the activation Millan, M.J., Przewlocki, R., Jerlicz, M.H., Gramsch, C., Hollt, V., Herz, A.,
pattern of enkephalin-containing perikarya in the rat ventral medulla. 1981a. Stress induced release of brain pituitary b-endorphin: major role of
J. Neurochem. 74, 2568–2575. endorphin in generation of hyperthermia, not analgesia. Brain Res. 208,
Mansour, A., Khachaturian, H., Lewis, M.E., Akil, H., Watson, S.J., 1987. Autora- 325–328.
diographic differentiation of mu, delta, and kappa opioid receptors in the rat Millan, M.J., Tsang, Y.F., Przewlocki, R., Hollt, V., Herz, A., 1981b. The influence of
forebrain and midbrain. J. Neurosci. 7 (8), 2445–2464. footshock stress upon brain pituitary and spinal cord pools of immunoreactive
Mansour, A., Khachaturian, H., Lewis, M.E., Akil, H., Watson, S.J., 1988. Anatomy of dynorphin in rats. Neurosci. Lett. 24, 75–79.
CNS opioid receptors. Trends Neurosci. 11 (7), 308–314. Miller, B.R., Hen, R., 2015. The current state of the neurogenic theory of depression
Marek, P., Yirmiya, R., Panocka, I., Liebeskind, J.C., 1989. Genetic influences on brain and anxiety. Curr. Opin. Neurobiol. 30, 51–58.
stimulation-produced analgesia in mice. I. Correlation with stress-induced anal- Minami, M., Toya, T., Katao, Y., Maekawa, K., Nakamura, S., Onogi, T., Kaneko, S.,
gesia. Brain Res. 489, 182–184. Satoh, M., 1993. Cloning and expression of a cDNA for the rat kappa-opioid
Marotti, T., Gabrilovac, J., Rabatic, S., Smejkal-Jagar, L., Rocic, B., Haberstock, H., receptor. FEBS Lett. 329 (3), 291–295.
1996. Met-enkephalin modulates stress-induced alterations of the immune Mizoguchi, H., Narita, M., Kampine, J.P., Tseng, L.F., 1997. [Met5]enkephalin and
response in mice. Pharmacol. Biochem. Behav. 54, 277–284. delta2-opioid receptors in the spinal cord are involved in the cold water swimming-
Marquez, P., Baliram, R., Kieffer, B.L., Lutfy, K., 2007. The mu opioid receptor is induced antinociception the mouse. Life Sci. 61 (7), PL81–PL86.
involved in buprenorphine-induced locomotor stimulation and conditioned place Mizoguchi, H., Watanabe, C., Osada, S., Yoshioka, M., Aoki, Y., Natsui, S.,
preference. Neuropharmacology 52, 1336–1341. Yonezawa, A., Kanno, S., Ishikawa, M., Sakurada, T., Sakurada, S., 2010.
Marti, O., Harbuz, M.S., Andres, R., Lightman, S.L., Armario, A., 1999. Activation of Lack of a rewarding effect and a locomotor-enhancing effect of the selective
the hypothalamic-pituitary axis in adrenalectomised rats: potentiation by chronic mu-opioid receptor agonist amidino-TAPA. Psychopharmacology (Berl.) 212,
stress. Brain Res. 821 (1), 1–7. 215–225.
Mason, J.W., 1971. A reevaluation of the concept of nonspecificity in stress theory. Mogil, J.S., Marek, P., O’Toole, L.A., Helms, M.L., Sadowski, B., Liebeskind, J.C.,
J. Psychiatr. Res. 8, 323–333. Belknap, J.K., 1994. Mu-opiate receptor binding is up-regulated in mice selectively
Mateus-Pinheiro, A., Pinto, L., Bessa, J.M., Morais, M., Alves, N.D., Monteiro, S., bred for high stress-induced analgesia. Brain Res. 653 (1–2), 16–22.
Patrício, P., Almeida, O.F.X., Sousa, N., 2013. Sustained remission from Mogil, J.S., Sternberg, W.F., Balian, H., Liebeskind, J.C., Sadowski, B., 1996. Opioid
depressive-like behavior depends on hippocampal neurogenesis. Transl. and nonopioid swim stress-induced analgesia: a parametric analysis in mice.
Psychiatry 3, e210. Physiol. Behav. 59, 123–132.
Matsuzawa, S., Suzuki, T., Misawa, M., Nagase, H., 1999. Different roles of mu-, Mogil, J.S., Richards, S.P., O’Toole, L.A., Helms, M.L., Mitchell, S.R., Kest, B.,
delta- and kappa-opioid receptors in ethanol-associated place preference in rats Belknap, J.K., 1997. Identification of a sex-specific quantitative trait locus medi-
exposed to conditioned fear stress. Eur. J. Pharmacol. 368, 9–16. ating nonopioid stress-induced analgesia in female mice. J. Neurosci. 17 (20),
Matthews, S.G., Challis, J.R., 1995. Developmental regulation of preproenkephalin 7995–8002.
mRNA in the ovine paraventricular nucleus: effects of stress and glucocorticoids. Molina, P.E., 2002. Stress-specific opioid modulation of haemodynamic counter-
Brain Res. Dev. Brain Res. 86 (1–2), 259–267. regulation. Clin. Exp. Pharmacol. Physiol. 29 (3), 248–253.
Matthews, R.T., German, D.C., 1984. Electrophysiological evidence for excitation of rat Molina, P.E., 2006. Opioids and opiates: analgesia with cardiovascular, haemodynamic
ventral tegmental area dopamine neurons by morphine. Neuroscience 11, and immune implications in critical illness. J. Intern. Med. 259 (2), 138–154.
617–625. Morley, J.E., Elson, M.K., Levine, A.S., Shafer, R.B., 1982. The effects of stress on
Matthews, S.G., Parrott, R.F., Sirinathsinghji, D.J., 1993. Isolation- and dehydration- central nervous system concentrations of the opioid peptide, dynorphin. Peptides 3,
induced changes in neuropeptide gene expression in the sheep hypothalamus. 901–906.
J. Mol. Endocrinol. 11, 181–189. Morley, J.E., Levine, A.S., Yim, G.K., Lowy, M.T., 1983. Opioid modulation of appetite.
McCubbin, J.A., Bruehl, S., Wilson, J.F., Sherman, J.J., Norton, J.A., Colclough, G., Neurosci. Biobehav. Rev. 7, 281–305.
1998. Endogenous opioids inhibit ambulatory blood pressure during naturally Mormede, P., Vincent, J.D., Kerdelhue, B., 1986. Vasopressin and oxytocin
occurring stress. Psychosom. Med. 60, 227–231. reduce plasma prolactin levels of conscious rats in basal and stress condi-
McEwen, B.S., Wingfield, J.C., 2003. The concept of allostasis in biology and tions. Study of the characteristics of the receptor involved. Life Sci. 39,
biomedicine. Horm. Behav. 43, 2–15. 1737–1743.
McGivern, R.F., Mousa, S., Couri, D., Berntson, G.G., 1983. Prolonged intermittent Moskowitz, A.S., Goodman, R.R., 1985. Autoradiographic analysis of mu1, mu2, and
footshock stress decreases met- and leu-enkephalin levels in brain with delta opioid binding in the central nervous system of C57BL/6BY and CXBK (opioid
concomitant decreases in pain threshold. Life Sci. 33, 47–54. receptor-deficient) mice. Brain Res. 360 (1–2), 108–116.
McLaughlin, J.P., Marton-Popovici, M., Chavkin, C., 2003. Kappa opioid receptor Moynihan, J.A., Karp, J.D., Cohen, N., Ader, R., 2000. Immune deviation following
antagonism and prodynorphin gene disruption block stress-induced behavioral stress odor exposure: role of endogenous opioids. J. Neuroimmunol. 102,
responses. J. Neurosci. 23 (13), 5674–5683. 145–153.
256 Stress and Opioid Systems

Mucha, R.F., Herz, A., 1985. Motivational properties of kappa and mu opioid receptor Okada, Y., Tsuda, Y., Bryant, S.D., Lazarus, L.H., 2002. Endomorphins and related
agonists studied with place and taste preference conditioning. Psychopharma- opioid peptides. Vitam. Horm. 65, 257–279.
cology (Berl.) 86 (3), 274–280. Okada, Y., Tsuda, Y., Fujita, Y., Yokoi, T., Sasaki, Y., Ambo, A., Konishi, R.,
Mucha, R.F., Herz, A., 1986. Preference conditioning produced by opioid active Nagata, M., Salvadori, S., Jinsmaa, Y., Bryant, S.D., Lazarus, L.H., 2003. Unique
and inactive isomers of levorphanol and morphine in rat. Life Sci. 38 (3), high-affinity synthetic mu-opioid receptor agonists with central- and systemic-
241–249. mediated analgesia. J. Med. Chem. 46, 3201–3209.
Mucha, R.F., Iversen, S.D., 1984. Reinforcing properties of morphine and naloxone Olszewski, P.K., Levine, A.S., 2007. Central opioids and consumption of sweet tast-
revealed by conditioned place preferences: a procedural examination. Psycho- ants: when reward outweighs homeostasis. Physiol. Behav. 91 (5), 506–512.
pharmacology (Berl.) 82 (3), 241–247. Omiya, Y., Goto, K., Ishige, A., Komatsu, Y., 2000. Changes in analgesia-producing
Munck, A., Guyre, P.M., Holbrook, N.J., 1984. Physiological functions of glucocorti- mechanism of repeated cold stress loading in mice. Pharmacol. Biochem.
coids in stress and their relation to pharmacological actions. Endocr. Rev. 5, Behav. 65, 261–266.
25–44. Osborne, H., Przewlocki, R., Hollt, V., Herz, A., 1979. Release of b-endorphin from rat
Nabeshima, T., Katoh, A., Hiramatsu, M., Kameyama, T., 1986. A role played by hypothalamus in vitro. Eur. J. Pharmacol. 55, 425–428.
dopamine and opioid neuronal systems in stress-induced motor suppression Osgood, P.F., Murphy, J.L., Carr, D.B., Szyfelbein, S.K., 1987. Increases in plasma
(conditioned suppression of motility) in mice. Brain Res. 398, 354–360. beta-endorphin and tail flick latency in the rat following burn injury. Life Sci. 40,
Nabeshima, T., Katoh, A., Kameyama, T., 1988. Inhibition of enkephalin degradation 547–554.
attenuated stress-induced motor suppression (conditioned suppression of motility). Otagiri, A., Wakabayashi, I., Shibasaki, T., 2000. Selective corticotropin-releasing factor type
J. Pharmacol. Exp. Ther. 244, 303–309. 1 receptor antagonist blocks conditioned fear-induced release of noradrenaline in the
Nakagawasai, O., Tadano, T., Tan, N.K., Niijima, F., Sakurada, S., Endo, Y., Kisara, K., hypothalamic paraventricular nucleus of rats. J. Neuroendocrinol. 12 (10), 1022–1026.
1999. Changes in beta-endorphin and stress-induced analgesia in mice after expo- Overstreet, D.H., Janowsky, D.S., Gillin, J.C., Shiromani, P.J., Sutin, E.L., 1986.
sure to forced walking stress. Methods Find. Exp. Clin. Pharmacol. 21, 471–476. Stress-induced immobility in rats with cholinergic supersensitivity. Biol. Psychiatry
Nakamura, M., Kamata, K., Inoue, H., Inaba, M., 1989. Effects of opioid peptides 21 (7), 657–664.
administered in conscious rats on the changes in blood adrenaline levels caused by Pan, B., Castro-Lopes, J.M., Coimbra, A., 1996. Activation of anterior lobe cortico-
immobilization stress. Jpn. J. Pharmacol. 50, 354–356. trophs by electroacupuncture or noxious stimulation in the anaesthetized rat, as
Nakamura, H., Nagase, H., Yoshida, M., Ogino, K., Seto, T., Hatta, K., Matsuzaki, I., shown by colocalization of Fos protein with ACTH and beta-endorphin and
1998. Opioid peptides mediate heat stress-induced immunosuppression during increased hormone release. Brain Res. Bull. 40, 175–182.
pregnancy. Am. J. Physiol. 274, 672–676. Pan, Y.-X., Xu, J., Bolan, E.A., Abbadie, C., Chang, A., Zuckerman, A., Rossi, G.C.,
Nakanishi, S., Inoue, A., Kita, T., Nakamura, M., Chang, A.C., Cohen, S.N., Numa, S., Pasternak, G.W., 1999. Identification and characterization of three new alterna-
1979. Nucleotide sequence of cloned cDNA for bovine corticotropin-beta-lipotropin tively spliced mu opioid receptor isoforms. Mol. Pharmacol. 56, 396–403.
precursor. Nature 278 (5703), 423–427. Pan, Y.-X., Xu, J., Bolan, E., Chang, A., Mahurter, L., Rossi, G., Pasternak, G.W.,
Nakata, Y., Chang, K.J., Mitchell, C.L., Hong, J.S., 1985. Repeated electroconvulsive 2000. Isolation and expression of a novel alternatively spliced mu opioid receptor
shock down regulates the opioid receptors in rat brain. Brain Res. 346, 160–163. isoform, MOR-1F. FEBS Lett. 466, 337–340.
Naleid, A.M., Grace, M.K., Chimukangara, M., Billington, C.J., Levine, A.S., 2007. Pan, Y.-X., Xu, J., Mahurter, L., Bolan, E.A., Xu, M.M., Pasternak, G.W., 2001.
Paraventricular opioids alter intake of high-fat but not high-sucrose diet depending Generation of the mu opioid receptor (MOR-1) protein by three new splice variants
on diet preference in a binge model of feeding. AJP Regul. Integr. Comp. Physiol. of the Oprm gene. Proc. Natl. Acad. Sci. U.S.A. 98, 14084–14089.
293 (1), R99–R105. Pan, Y.-X., Xu, J., Mahurter, L., Xu, M.M., Gilbert, A.-K., Pasternak, G.W., 2003.
Nankova, B., Kvetnansky, R., Hiremagalur, B., Sabban, B., Rusnak, M., Sabban, E.L., Identification and characterization of two new human mu opioid receptor splice
1996. Immobilization stress elevates gene expression for catecholamine biosyn- variants, hMOR-1O and hMOR-1X. Biochem. Biophys. Res. Commun. 301,
thetic enzymes and some neuropeptides in rat sympathetic ganglia: effects of 1057–1061.
adrenocorticotropin and glucocorticoids. Endocrinology 137 (12), 5597–5604. Pancheri, P., Zichella, L., Fraioli, F., Carilli, L., Perrone, G., Biondi, M., Fabbri, A.,
Narita, M., Kuzumaki, N., Miyatake, M., Sato, F., Wachi, H., Seyama, Y., Suzuki, T., Santoro, A., Moretti, C., 1985. ACTH, beta-endorphin and met-enkephalin:
2006. Role of delta-opioid receptor function in neurogenesis and neuroprotection. peripheral modifications during the stress of human labor. Psychoneur-
J. Neurochem. 97, 1494–1505. oendocrinology 10, 289–301.
Nestler, E.J., Carlezon Jr., W.A., 2006. The mesolimbic dopamine reward circuit in Panerai, A.E., Martini, A., Sacerdote, P., Mantegazza, P., 1984. Kappa-receptor
depression. Biol. Psychiatry 59, 1151–1159. antagonist reverse ‘non-opioid’ stress-induced analgesia. Brain Res. 304 (1),
Newton, S.S., Thome, J., Wallace, T.L., Shirayama, Y., Schlesinger, L., Sakai, N., 153–156.
Chen, J., Neve, R., Nestler, E.J., Duman, R.S., 2002. Inhibition of cAMP response Panerai, A.E., Bianchi, M., Brini, A., Sacerdote, P., 1987. Endogenous opioids and
element-binding protein or dynorphin in the nucleus accumbens produces an their receptors in stress-induced analgesia. Pol. J. Pharmacol. Pharm. 39,
antidepressant-like effect. J. Neurosci. 22 (24), 10883–10890. 597–607.
Nikolarakis, K.E., Almeida, O.F.X., Herz, A., 1986. Stimulation of hypothalamic Pasternak, G.W., Pan, Y.X., 2000. Antisense mapping: assessing functional signifi-
(3endorphin and dynorphin release) by corticotropin releasing factor (in vitro). Brain cance of genes and splice variants. Methods Enzym. 314, 51–60.
Res. 399, 152–155. Pasternak, G.W., Pan, Y.X., 2013. Mu opioids and their receptors: evolution of
Nikulina, E.M., Hammer Jr., R.P., Miczek, K.A., Kream, R.M., 1999. Social defeat a concept. Pharmacol. Rev. 65 (4), 1257–1317.
stress increases expression of mu-opioid receptor mRNA in rat ventral tegmental Pasternak, G.W., 2004. Multiple opiate receptors: déjà vu all over again. Neuro-
area. Neuroreport 10 (14), 3015–3019. pharmacology 47, 312–323.
Nikulina, E.M., Miczek, K.A., Hammer Jr., R.P., 2005. Prolonged effects of repeated Patchev, V.K., Almeida, O.F.X., 1999. Steroid Hormone-Dependent Organization of
social defeat stress on mRNA expression and function of mu-opioid receptors in the Neuroendocrine Function. R.G. Landes Company, Austin, TX.
ventral tegmental area of rats. Neuropsychopharmacology 30 (6), 1096–1103. Patchev, A.V., Rodrigues, A.J., Sousa, N., Spengler, D., Almeida, O.F.X., 2014. The
Nishi, M., Takeshima, H., Fukuda, K., Kato, S., Mori, K., 1993. cDNA cloning and future is now: early life events preset adult behaviour. Acta Physiol. (Oxf.) 210,
pharmacological characterization of an opioid receptor with high affinities for 46–57.
kappa-subtype-selective ligands. FEBS Lett. 330 (1), 77–80. Pechnick, R.N., Morgan, M.J., 1987. The role of endogenous opioids in footshock-
Noda, M., Furutani, Y., Takahashi, H., Toyosato, M., Hirose, T., Inayama, S., induced hyperthermia. Pharmacol. Biochem. Behav. 28, 95–100.
Nakanishi, S., Numa, S., 1982. Cloning and sequence analysis of cDNA for bovine Peckys, D., Landwehrmeyer, G.B., 1999. Expression of mu, kappa, and delta opioid
adrenal preproenkephalin. Nature 295 (5846), 202–206. receptor messenger RNA in the human CNS: a 33P in situ hybridization study.
Noel, M.B., Gratton, A., 1995. Electrochemical evidence of increased dopamine Neuroscience 88 (4), 1093–1135.
transmission in prefrontal cortex and nucleus accumbens elicited by ventral Peterson, P.K., Molitor, T.W., Chao, C.C., 1998. The opioid-cytokine connection.
tegmental mu-opioid receptor activation in freely behaving rats. Synapse 21 (2), J. Neuroimmunol. 83 (1–2), 63–69.
110–122. Petralgia, F., Vale, W., Rivier, C., 1986. Opioids act centrally to modulate stress
Nordin, M., Morat, P., Zainora, M., 1987. The effect of endogenous opioids on blood induced decrease in luteinizing hormone in the rat. Endocrinology 119,
pressure during stress. Clin. Exp. Pharmacol. Physiol. 14, 303–308. 2445–2450.
Nunes, E.V., Sullivan, M.A., Levin, F.R., 2004. Treatment of depression in patients with Pfeiffer, A., Brantl, V., Herz, A., Emrich, H.M., 1986. Psychotomimesis mediated by
opiate dependence. Biol. Psychiatry 56, 793–802. kappa opiate receptors. Science 233 (4765), 774–776.
Stress and Opioid Systems 257

Piazza, P.V., Le Moal, M., 1998. The role of stress in drug self-administration. Trends Quinones-Jenab, V., Jenab, S., Ogawa, S., Inturrisi, C., Pfaff, D.W., 1997. Estrogen
Pharmacol. Sci. 19 (2), 67–74. regulation of mu-opioid receptor mRNA in the forebrain of female rats. Brain Res.
Pietrzak, R.H., Naganawa, M., Huang, Y., Corsi-Travali, S., Zheng, M.Q., Stein, M.B., Mol. Brain Res. 47 (1–2), 134–138.
Henry, S., Lim, K., Ropchan, J., Lin, S.F., Carson, R.E., Neumeister, A., 2014. Raab, A., Seizinger, B.R., Herz, A., 1985. Continuous social defeat induces an increase
Association of in vivo k-opioid receptor availability and the transdiagnostic of endogenous opioids in discrete brain areas of the mongolian gerbil. Peptides 6,
dimensional expression of trauma-related psychopathology. JAMA Psychiatry 71, 387–391.
1262–1270. Ragnauth, A., Schuller, A., Morgan, M., Chan, J., Ogawa, S., Pintar, J., Bodnar, R.J.,
Pitman, R.K., van der Kolk, B.A., Orr, S.P., Greenberg, M.S., 1990. Naloxone- Pfaff, D.W., 2001. Female preproenkephalin-knockout mice display altered
reversible analgesic response to combat-related stimuli in posttraumatic stress emotional responses. Proc. Natl. Acad. Sci. U.S.A. 98, 1958–1963.
disorder. A pilot study. Arch. Gen. Psychiatry 47, 541–544. Ray, A., Henke, P.G., Sullivan, R.M., 1988. Opiate mechanisms in the central
Piva, F., Limonta, P., Dondi, D., Pimpinelli, F., Martini, L., Maggi, R., 1995. Effects of amygdala and gastric stress pathology in rats. Brain Res. 442, 195–198.
steroids on the brain opioid system. J. Steroid Biochem. Mol. Biol. 53, 343–348. Reynolds, D.G., Gurll, N.J., Vargish, T., Lechner, R.B., Faden, A.I., Holaday, J.W.,
Pliakas, A.M., Carlson, R.R., Neve, R.L., Konradi, C., Nestler, E.J., Carlezon Jr., W.A., 1980. Blockade of opiate receptors with naloxone improves survival and cardiac
2001. Altered responsiveness to cocaine and increased immobility in the forced performance in canine endotoxic shock. Circ. Shock 7 (1), 39–44.
swim test associated with elevated cAMP response element-binding protein Rivier, C., Rivier, J., Mormede, P., Vale, W., 1984. Studies on the nature of the
expression in nucleus accumbens. J. Neurosci. 21 (18), 7397–7403. interaction between vasopressin and corticotropin-releasing factor on adrenocortin
Plotnikoff, N.P., Miller, G.C., 1983. Enkephalins as immunomodulators. Int. J. release in the rat. Endocrinology 115, 882–886.
Immunopharmacol. 5, 437–441. Robins, H.I., Kalin, N.H., Shelton, S.E., Martin, P.A., Shecterle, L.M., Barksdale, C.M.,
Plotsky, P.M., 1986. Opioid inhibition of immunoreactive corticotropin-releasing factor Neville, A.J., Marshall, J., 1987. Rise in plasma beta-endorphin, ACTH, and
secretion into the hypophysial-portal circulation of rats. Regul. Pept. 16, 235–242. cortisol in cancer patients undergoing whole body hyperthermia. Horm. Metab. Res.
Pohorecky, L.A., Skiandos, A., Zhang, X., Rice, K.C., Benjamin, D., 1999. Effect of 19 (9), 441–443.
chronic social stress on delta-opioid receptor function in the rat. J. Pharmacol. Exp. Rodgers, R.J., Deacon, R.M., 1979. Effect of naloxone on the behaviour of rats
Ther. 290, 196–206. exposed to a novel environment. Psychopharmacology (Berl.) 65 (1), 103–105.
Pradhan, A.A., Smith, M.L., Kieffer, B.L., Evans, C.J., 2012. Ligand-directed signalling Rodgers, R.J., Deacon, R.M., 1981. Footshock-analgesia: prevention by behavioural
within the opioid receptor family. Br. J. Pharmacol. 167, 960–969. manipulation but not by naloxone. Physiol. Behav. 26 (2), 183–187.
Priest, C.A., Borsook, D., Pfaff, D.W., 1997. Estrogen and stress interact to regulate Ros-Bernal, F., Hunot, S., Herrero, M.T., Parnadeau, S., Corvol, J.C., Lu, L., Alvarez-
the hypothalamic expression of a human proenkephalin promoter-beta-galactosi- Fischer, D., Carrillo-de Sauvage, M.A., Saurini, F., Coussieu, C., Kinugawa, K.,
dase fusion gene in a site-specific and sex-specific manner. J. Neuroendocrinol. 9 Prigent, A., Höglinger, G., Hamon, M., Tronche, F., Hirsch, E.C., Vyas, S., 2011.
(4), 317–326. Microglial glucocorticoid receptors play a pivotal role in regulating dopaminergic
Primeaux, S.D., Wilson, M.A., Wilson, S.P., Guth, A.N., Lelutiu, N.B., Holmes, P.V., neurodegeneration in parkinsonism. Proc. Natl. Acad. Sci. U.S.A. 108, 6632–6637.
2003. Herpes virus-mediated preproenkephalin gene transfer in the ventral stria- del Rosario, C.N., Pacchioni, A.M., Cancela, L.M., 2002. Influence of acute or repeated
tum mimics behavioral changes produced by olfactory bulbectomy in rats. Brain restraint stress on morphine-induced locomotion: involvement of dopamine, opioid
Res. 988 (1–2), 43–55. and glutamate receptors. Behav. Brain Res. 134, 229–238.
Przewlocka, B., Vetulani, J., Lason, W., Dziedzicka, M., Silberring, J., Castellano, C., Rosecrans, J.A., Robinson, S.E., Johnson, J.H., Mokler, D.J., Hong, J.S., 1986.
Przewlocki, R., 1988. The difference in stress-induced analgesia in C57BL/6 and Neuroendocrine, biogenic amine and behavioral responsiveness to a repeated foot-
DBA/2 mice: a search for biochemical correlates. Pol. J. Pharmacol. Pharm. 40, shock-induced analgesia (FSIA) stressor in Sprague-Dawley (CD) and Fischer-344
497–506. (CDF) rats. Brain Res. 382, 71–80.
Przewlocka, B., Sumova, A., Lason, W., 1990. The influence of anticipation stress on Rossier, J., French, E.D., Rivier, C., Ling, N., Guillemin, R., Bloom, F.E., 1977. Foot-
opioid systems in rat. Pharmacol. Biochem. Behav. 37, 661–666. shock induced stress increases beta-endorphin levels in blood but not brain. Nature
Przewlocka, B., Turchan, J., Lason, W., Przewlocki, R., 1996. The effect of single and 270, 618–620.
repeated morphine administration on the prodynorphin system activity in the Rossier, J., Guillemin, R., Bloom, F.E., 1978. Foot-shock induced stress decreases
nucleus accumbens and striatum of the rat. Neuroscience 70 (3), 749–754. leuenkephalin immunoreactivity in rat hypothalamus. Eur. J. Pharmacol. 48,
Przewlocki, R., Höllt, V., Voight, K.H., Herz, A., 1979. Modulation of in vitro release 465–466.
of 3-endorphin from the separate lobes of the rat pituitary. Life Sci. 24, Roth, K.A., Katz, R.J., Schmaltz, K., Sibel, M., 1981. Reduced behavioral activity due
1601–1608. to opiate blockade: relations to stress. Int. J. Neurosci. 12, 59–62.
Przewlocki, R., Millan, J., Gramsch, C., Millan, M.H., Herz, A., 1982. The influence of Roth, B.L., Baner, K., Westkaemper, R., Siebert, D., Rice, K.C., Steinberg, S.,
selective adeno-neurointermedio-hypophysectomy upon plasma and brain levels of Ernsberger, P., Rothman, R.B., 2002. Salvinorin A: a potent naturally occurring
b-endorphin and their response to stress in rats. Brain Res. 242, 107–117. nonnitrogenous kappa opioid selective agonist. Proc. Natl. Acad. Sci. U.S.A. 99,
Przewlocki, R., Lason, W., Konecka, A., Gramsch, C., Herz, A., Reid, L., 1983a. The 11934–11939.
opioid peptide dynorphin, circadian rhythms, and starvation. Science 219, 71–73. Roth-Isigkeit, A., Dibbelt, L., Schmucker, P., Seyfarth, M., 2000. The immune-
Przewlocki, R., Shearman, G.T., Herz, A., 1983b. Mixed opioid/nonopioid effects of endocrine interaction varies with the duration of the inflammatory process in
dynorphin and dynorphin-related peptides after their intrathecal injection in rats. cardiac surgery patients. J. Neuroendocrinol. 12 (6), 546–552.
Neuropeptides 3, 233–239. Rubinstein, M., Mogil, J.S., Japon, M., Chan, E.C., Allen, R.G., Low, M.J., 1996.
Przewlocki, R., Lason, W., Hollt, V., Silberring, J., Herz, A., 1987. The influence of Absence of opioid stress-induced analgesia in mice lacking beta-endorphin by site-
chronic stress on multiple opioid peptide systems in the rat: pronounced effects directed mutagenesis. Proc. Natl. Acad. Sci. U.S.A. 93, 3995–4000.
upon dynorphin in spinal cord. Brain Res. 413, 213–219. Rush, A.J., Trivedi, M.H., Wisniewski, S.R., Nierenberg, A.A., Stewart, J.W.,
Przewlocki, R., Majeed, N.H., We˛ dzony, K., Przewlocka, B., 1988a. The effect of stress Warden, D., Niederehe, G., Thase, M.E., Lavori, P.W., Lebowitz, B.D.,
on the opioid peptide systems in the rat nucleus accumbens. In: Kvetnansky, R., McGrath, P.J., Rosenbaum, J.F., Sackeim, H.A., Kupfer, D.J., Luther, J., Fava, M.,
Van Loon, G.R., McCarty, R., Axelrod, J. (Eds.), Stress: Neurochemical and 2006. Acute and longer-term outcomes in depressed outpatients requiring one or
Humoral Mechanisms. Gordon and Breach, New York. several treatment steps: a STAR*D report. Am. J. Psychiatry 163, 1905–1917.
Przewlocki, R., Haarmann, I., Nikolarakis, K., Herz, A., Hollt, V., 1988b. Prodynorphin Sacerdote, P., Manfredi, B., Bianchi, M., Panerai, A.E., 1994. Intermittent but not
gene expression in spinal cord is enhanced after traumatic injury in the rat. Mol. continuous inescapable footshock stress affects immune responses and immunocyte
Brain Res. 4, 37–41. beta-endorphin concentrations in the rat. Brain Behav. Immun. 8 (3), 251–260.
Przewlocki, R., Przewlocka, B., Lason, W., 1991. Adaptation of opioid systems to Sacerdote, P., Limiroli, E., Gaspani, L., 2003. Experimental evidence for immuno-
stress. In: Almeida, O.F.X., Shippenberg, T.S. (Eds.), The Neurobiology of Opioid modulatory effects of opioids. Adv. Exp. Med. Biol. 521, 106–116.
Systems, pp. 229–243. Sacerdote, P., 2003. Effects of in vitro and in vivo opioids on the production of IL-12
Przewlocki, R., Hassan, A.H., Lason, W., Epplen, C., Herz, A., Stein, C., 1992. Gene and IL-10 by murine macrophages. Ann. N.Y. Acad. Sci. 992, 129–140.
expression and localization of opioid peptides in immune cells of inflamed tissue: Sadowski, B., Konarzewski, M., 1999. Analgesia in selectively bred mice exposed to
functional role in antinociception. Neuroscience 48 (2), 491–500. cold in helium/oxygen atmosphere. Physiol. Behav. 66 (1), 145–151.
Quinones-Jenab, V., Ogawa, S., Jenab, S., Pfaff, D.W., 1996. Estrogen regulation of Sadowski, B., Panocka, I., 1993. Cross-tolerance between morphine and swim
preproenkephalin messenger RNA in the forebrain of female mice. J. Chem. analgesia in mice selectively bred for high and low stress-induced analgesia.
Neuroanat. 12 (1), 29–36. Pharmacol. Biochem. Behav. 45 (3), 527–531.
258 Stress and Opioid Systems

Saitoh, A., Yamada, M., 2012. Antidepressant-like effects of d opioid receptor agonists Sinha, R., 2007. The role of stress in addiction relapse. Curr. Psychiatry Rep. 9 (5),
in animal models. Curr. Neuropharmacol. 10, 231–238. 388–395.
de los Santos-Arteaga, M., Sierra-Domínguez, S.A., Fontanella, G.H., Delgado- Sitsen, J.M., Van Ree, J.M., De Jong, W., 1982. Cardiovascular and respiratory effects
García, J.M., Carrión, A.M., 2003. Analgesia induced by dietary restriction is of beta-endorphin in anesthetized and conscious rats. J. Cardiovasc. Pharmacol. 4
mediated by the kappa-opioid system. J. Neurosci. 23 (35), 11120–11126. (6), 883–888.
Scatton, B., D’Angio, M., Driscoll, P., Serrano, A., 1988. An in vitro voltametric study Skoubis, P.D., Lam, H.A., Shoblock, J., Narayanan, S., Maidment, N.T., 2005.
of the response of mesocortical and mesoaccumbens dopaminergic neurons to Endogenous enkephalins, not endorphins, modulate basal hedonic state in mice.
environmental stimuli in strains of rats with differing levels of emotionality. Ann. Eur. J. Neurosci. 21 (5), 1379–1384.
N.Y. Acad. Sci. 537, 124–137. Skultetyova, I., Jezova, D., 1999. Dissociation of changes in hypothalamic
Schnur, P., Martinez, Y., Hang, D., 1988. Effects of stress on morphine-elicited corticotropin-releasing hormone and pituitary proopiomelanocortin mRNA levels
locomotor activity in hamsters. Behav. Neurosci. 102 (2), 254–259. after prolonged stress exposure. Brain Res. Mol. Brain Res. 68 (1–2), 190–192.
Schoffelmeer, A.N.M., Hogenboom, F., Mulder, A.H., 1988. Sodium dependent 3H- Slominski, A., Zbytek, B., Pisarchik, A., Slominski, R.M., Zmijewski, M.A.,
noradrenaline release from rat neocortical slices in the absence of extracellular Wortsman, J., 2006. CRH functions as a growth factor/cytokine in the skin. J. Cell
calcium: presynaptic modulation by p-opioid receptor and adenylate cyclase Physiol. 206 (3), 780–791.
activation. Naunyn Schmiedebergs Arch. Pharmacol. 338, 548–552. Slominski, A.T., 2006. Proopiomelanocortin signaling system is operating in mast cells.
Schoffelmeer, A.N., Hogenboom, F., Mulder, A.H., 1997. k1- and k2-opioid receptors J. Investig. Dermatol. 126 (9), 1934–1936.
mediating presynaptic inhibition of dopamine and acetylcholine release in rat Slugg, R.M., Hayward, M.D., Ronnekleiv, O.K., Low, M.J., Kelly, M.J., 2000. Effect of
neostriatum. Br. J. Pharmacol. 122 (3), 520–524. the mu-opioid agonist DAMGO on medial basal hypothalamic neurons in beta-
Schuller, A.G., King, M.A., Zhang, J., Bolan, E., Pan, Y.X., Morgan, D.J., Chang, A., endorphin knockout mice. Neuroendocrinology 72 (4), 208–217.
Czick, M.E., Unterwald, E.M., Pasternak, G.W., Pintar, J.E., 1999. Retention of Sofroniew, M.V., 1979. Immunoreactive beta-endorphin and ACTH in the same
heroin and morphine-6ß-glucuronide analgesia in a new line of mice lacking exon 1 neurons of the hypothalamic arcuate nucleus in the rat. Am. J. Anat. 154 (2),
of MOR-1. Nat. Neurosci. 2 (2), 151–156. 283–289.
Seeger, T.F., Sforzo, G.A., Pert, C.B., Pert, A., 1984. In vivo autoradiography: visu- Sotiropoulos, I., Cerqueira, J.J., Catania, C., Takashima, A., Sousa, N., Almeida, O.F.X.,
alization of stress induced changes in opiate receptor occupancy in the rat brain. 2008. Stress and glucocorticoid footprints in the brain – the path from depression
Brain Res. 305, 303–311. to Alzheimer’s disease. Neurosci. Biobehav. Rev. 32, 1161–1173.
Selye, H., 1936. A syndrome produced by diverse nocuous agents. Nature 138, 32. Sousa, N., Almeida, O.F.X., 2012. Disconnection and reconnection: the morphological
Shaham, Y., Stewart, J., 1994. Exposure to mild stress enhances the reinforcing basis of (mal)adaptation to stress. Trends Neurosci. 35, 742–751.
efficacy of intravenous heroin self-administration in rats. Psychopharmacology Spanagel, R., Herz, A., Schippenberg, T.S., 1990. The effects of opioid peptides on
(Berl.) 114 (3), 523–527. dopamine release in the nucleus accumbens: an in vivo microdialysis study.
Shaham, Y., Funk, D., Erb, S., Brown, T.J., Walker, C.D., Stewart, J., 1997. J. Neurochem. 55, 1734–1740.
Corticotropin-releasing factor, but not corticosterone, is involved in stress-induced Spanagel, R., Herz, A., Shippenberg, T.S., 1992. Opposing tonically active endogenous
relapse to heroin-seeking in rats. J. Neurosci. 17 (7), 2605–2614. opioid systems modulate the mesolimbic dopaminergic pathway. Proc. Natl. Acad.
Shaham, Y., Erb, S., Stewart, J., 2000. Stress-induced relapse to heroin and cocaine Sci. U.S.A. 89 (6), 2046–2050.
seeking in rats: a review. Brain Res. Rev. 33 (1), 13–33. Spanagel, R., Sillaber, I., Zieglgansberger, W., Corrigall, W.A., Stewart, J., Shaham, Y.,
Shavit, Y., Lewis, J.W., Terman, G.W., Gale, R.P., Liebeskind, J.C., 1984. Opioid 1998. Acamprosate suppresses the expression of morphine-induced sensitization
peptides mediate the suppressive effect of stress on natural killer cell cytotoxicity. in rats but does not affect heroin self-administration or relapse induced by heroin or
Science 223, 188–190. stress. Psychopharmacology (Berl.) 139 (4), 391–401.
Shavit, Y., Terman, G.W., Martin, F.C., Lewis, J.W., Liebeskind, J.C., Gale, R.P., 1985. Spencer, R.L., Ayres, E.A., Burks, T.F., 1985. Temperature responses in restrained
Stress, opioid peptides, the immune system, and cancer. J. Immunol. 135, 834s–837s. and unrestrained rats to the selective mu opioid agonist, DAGO. Proc. West.
Shavit, Y., Lewis, J.W., Terman, G., Gale, R.P., Liebeskind, C., 1986. Stress, opioid Pharmacol. Soc. 28, 107–110.
peptides and immune function. In: Frederickson, R.C.A., Hendrie, J.N., Starec, M., Nejedly, A., Malek, J., Rosina, J., Gojisova, E., Krsiak, M., 1997. Exposure
Hingtgen, H.C. (Eds.), Neuroregulation of Autonomic Endocrine and Immune to stress alters the effects of dynorphins in the hot plate test. Physiol. Res. 46,
Systems. Nijhoff, Boston, p. 343. 451–457.
Sher, L., 2004. The role of endogenous opioids in the placebo effect in post-traumatic Starke, K., Schoffel, E., Illes, P., 1985. The sympathetic axons innervating the sinus
stress disorder. Forsch. Komplementärmed. Klass. Naturheilkd. 11, 354–359. node of the rabbit possess presynaptic opioid x-, but not lx- or S-receptors. Naunyn
Sherman, T.G., Civelli, O., Douglas, J., Herbert, E., Watson, S.J., 1986. Coordinate Schmiedebergs Arch. Pharmacol. 329, 206–209.
expression of hypothalamic pro dynorphin and pro-vasopressin mRNA with osmotic Stein, C., Hassan, A.H., Przewlocki, R., Gramsch, C., Peter, K., Herz, A., 1990. Opioids
stimulation. Neuroendocrinology 44, 222–228. from immunocytes interact with receptors on sensory nerves to inhibit nociception
Shippenberg, T.S., Herz, A., 1986. Differential effects of mu and kappa opioid systems in inflammation. Proc. Natl. Acad. Sci. U.S.A. 87 (15), 5935–5939.
on motivational processes. NIDA Res. Monogr. 75, 563–566. Stein, E.A., Hiller, J.M., Simon, E.J., 1992. Effects of stress on opioid receptor binding
Shippenberg, T.S., Zapata, A., Chefer, V.I., 2007. Dynorphin and the pathophysiology in the rat central nervous system. Neuroscience 51 (3), 683–690.
of drug addiction. Pharmacol. Ther. 116 (2), 306–321. Stein, C., Schäfer, M., Machelska, H., 2003. Attacking pain at its source: new
Shirayama, Y., Ishida, H., Iwata, M., Hazama, G.I., Kawahara, R., Duman, R.S., 2004. perspectives on opioids. Nat. Med. 9, 1003–1008.
Stress increases dynorphin immunoreactivity in limbic brain regions and dynorphin Stohr, T., Almeida, O.F., Landgraf, R., Shippenberg, T.S., Holsboer, F., Spanagel, R.,
antagonism produces antidepressant-like effects. J. Neurochem. 90 (5), 1999. Stress- and corticosteroid-induced modulation of the locomotor response to
1258–1268. morphine in rats. Behav. Brain Res. 103 (1), 85–93.
Shmygalev, S., Damm, M., Weckbecker, K., Berghaus, G., Petzke, F., Sabatowski, R., Stuckey, J., Marra, S., Minor, T., Insel, T.R., 1989. Changes in mu opiate receptors
2011. The impact of long-term maintenance treatment with buprenorphine on following inescapable shock. Brain Res. 476, 167–169.
complex psychomotor and cognitive function. Drug Alcohol Depend. 117, Suarez-Roca, H., Silva, J.A., Arcaya, J.L., Quintero, L., Maixner, W., Pinerua-
190–197. Shuhaibar, L., 2006. Role of mu-opioid and NMDA receptors in the develop-
Shuster, S.J., Riedl, M., Li, X., Vulchanova, L., Elde, R., 2000. The kappa opioid ment and maintenance of repeated swim stress-induced thermal hyperalgesia.
receptor and dynorphin co-localize in vasopressin magnocellular neurosecretory Behav. Brain Res. 167 (2), 205–211.
neurons in guinea-pig hypothalamus. Neuroscience 96 (2), 373–383. Suaudeau, C., Costentin, J., 2000. Long lasting increase in nociceptive threshold
Sibinga, N.E., Goldstein, A., 1988. Opioid peptides and opioid receptors in cells of the induced in mice by forced swimming: involvement of an endorphinergic mecha-
immune system. Annu. Rev. Immunol. 6, 219–249. nism. Stress 3 (3), 221–227.
Silverman, M.B., Hermes, S.M., Zadina, J.E., Aicher, S.A., 2005. Mu-opioid receptor is Suda, T., Sato, Y., Sumitomo, T., Nakano, Y., Tozawa, F., Iwai, I., Yamada, M.,
present in dendritic targets of endomorphin-2 axon terminals in the nuclei of the Demura, H., 1992. Beta-endorphin inhibits hypoglycemia-induced gene expression
solitary tract. Neuroscience 135 (3), 887–896. of corticotropin-releasing factor in the rat hypothalamus. Endocrinology 130 (3),
Simonin, F., Valverde, O., Smadja, C., Slowe, S., Kitchen, I., Dierich, A., Le Meur, M., 1325–1330.
Roques, B.P., Maldonado, R., Kieffer, B.L., 1998. Disruption of the kappa-opioid Sumova, A., Jakoubek, B., 1989. Analgesia and impact induced by anticipation stress:
receptor gene in mice enhances sensitivity to chemical visceral pain, impairs involvement of the endogenous opioid peptide system. Brain Res. 13, 273–280.
pharmacological actions of the selective kappa-agonist U-50,488H and attenuates Szikszay, M., Benedek, G., Szekely, J.I., 1983. Thermoregulatory effects of D-met2-
morphine withdrawal. EMBO J. 17, 886–897. pro5-enkephalinamide. Neuropeptides 3, 465–475.
Stress and Opioid Systems 259

Szilagyi, J.E., 1991. Psychosocial stress elevates blood pressure via an opioid dependent Tong, Y., Pelletier, G., 1992. Role of dopamine in the regulation of proopiome-
mechanism in normotensive rats. Clin. Exp. Hypertens. A 13, 1383–1394. lanocortin (POMC) mRNA levels in the arcuate nucleus and pituitary gland of
Tache, Y., Garrick, T., Raybould, H., 1990. Central nervous system action of the female rat as studied by in situ hybridization. Brain Res. Mol. Brain Res. 15
peptides to influence gastrointestinal motor function. Gastroenterology 98, (1–2), 27–32.
517–528. Torregrossa, M.M., Folk, J.E., Rice, K.C., Watson, S.J., Woods, J.H., 2005. Chronic
Taha, S.A., Norsted, E., Lee, L.S., Lang, P.D., Lee, B.S., Woolley, J.D., Fields, H.L., administration of the delta opioid receptor agonist (þ)BW373U86 and antide-
2006. Endogenous opioids encode relative taste preference. Eur. J. Neurosci. 24 pressants on behavior in the forced swim test and BDNF mRNA expression in rats.
(4), 1220–1226. Psychopharmacology (Berl.) 183, 31–40.
Takahashi, M., Senda, T., Tokuyama, S., Kaneto, H., 1990. Further evidence for the Torregrossa, M.M., Jutkiewicz, E.M., Mosberg, H.I., Balboni, G., Watson, S.J.,
implication of kappa opioid receptor mechanism in the production of the stress Woods, J.H., 2006. Peptidic delta opioid receptor agonists produce
induced analgesia. Jpn. J. Pharmacol. 53, 487–494. antidepressant-like effects in the forced swim test and regulate BDNF mRNA
Takayama, H., Ota, Z., Ogawa, N., 1986. Effect of immobilization stress on neuro- expression in rats. Brain Res. 1069, 172–181.
peptides and their receptors in rat central nervous system. Regul. Pept. 15, Trujillo, K.A., Bronstein, D.M., Sanchez, I.O., Akil, H., 1995. Effects of chronic opiate
239–248. and opioid antagonist treatment on striatal opioid peptides. Brain Res. 698 (1–2),
Tanaka, M., Kohno, Y., Tsuda, A., Nakagawa, R., Ida, Y., Iimori, Y., Hoaki, Y., 69–78.
Nagasaki, N., 1983. Differential effect of morphine on noradrenaline release in Tsagarakis, S., Rees, L.H., Besser, N., Grossman, A., 1990. Opiate receptor subtype
brain regions of stressed and non-stressed rats. Brain Res. 275, 105–115. regulation of CRF-41 release from hypothalamus in vitro. Neuroendocrinology 51,
Tanaka, M., Tsuda, A., Ida, Y., Ushijima, I., Tsujimaru, S., Nagasaki, N., 1985. 599–605.
Methionine-enkephalin inhibits stress-induced increases in noradrenaline turnover Tsuda, A., Tanaka, M., Ida, Y., Tsujimaru, S., Ushijima, I., Nagasaki, N., 1986.
in brain regions of rats. Jpn. J. Pharmacol. 37, 117–119. Effects of preshock experience on enhancement of rat brain noradrenaline
Tanaka, M., Ida, Y., Tsuda, A., 1988. Naloxone, given before but not after stress turnover induced by psychological stress. Pharmacol. Biochem. Behav. 24 (1),
exposure, enhances stress-induced increases in regional brain noradrenaline 115–119.
release. Pharmacol. Biochem. Behav. 29 (3), 613–616. Tsuji, S., Nakai, Y., Fukata, J., Nakaishi, S., Takahashi, H., Usui, T., Imura, H., 1987.
Tanaka, M., Ida, Y., Tsuda, A., Tsujimaru, S., Shirao, I., Oguchi, M., 1989. Met- Effects of food deprivation and high fat diet on immunoreactive dynorphin A(1-8)
enkephalin, injected during the early phase of stress, attenuates stress-induced levels in brain regions of Zucker rats. Peptides 8, 1075–1078.
increases in noradrenaline release in rat brain regions. Pharmacol. Biochem. degli Uberti, E.C., Ambrosio, M.R., Vergnani, L., Portaluppi, F., Bondanelli, M.,
Behav. 32, 791–795. Trasforini, G., Margutti, A., Salvadori, S., 1993. Stress-induced activation of
Tanaka, M., Tsuda, A., Yokoo, H., Yoshida, M., Mizoguchi, K., Shimizu, T., 1991. sympathetic nervous system is attenuated by the delta-opioid receptor agonist
Psychological stress-induced increases in noradrenaline release in rat brain regions deltorphin in healthy man. J. Clin. Endocrinol. Metab. 77 (6), 1490–1494.
are attenuated by diazepam, but not by morphine. Pharmacol. Biochem. Behav. 39 Uhl, G.R., Sora, I., Wang, Z., 1999. The mu opiate receptor as a candidate gene for
(1), 191–195. pain: polymorphisms, variations in expression, nociception, and opiate responses.
Tanaka, K., Kersten, J.R., Riess, M.L., 2014. Opioid-induced cardioprotection. Curr. Proc. Natl. Acad. Sci. U.S.A. 96 (14), 7752–7755.
Pharm. Des. 20 (36), 5696–5705. Vale, W., Rivier, C., Yang, L., Minick, S., Guillemin, R., 1978. Effects of purified
Tanaka, M., 1999. Emotional stress and characteristics of brain noradrenaline release hypothalamic corticotropin releasing factor and other substances on the secretion
in the rat. Ind. Health 37 (2), 143–156. of adrenocorticotropin and b-endorphin immunoreactivities in vitro. Endocrinology
Tejedor-Real, P., Micó, J.A., Smadja, C., Maldonado, R., Roques, B.P., Gilbert- 103, 1911–1915.
Rahola, J., 1998. Involvement of delta-opioid receptors in the effects induced Valentino, R.J., Van Bockstaele, E., 2001. Opposing regulation of the locus coeruleus by
by endogenous enkephalins on learned helplessness model. Eur. J. Pharmacol. corticotropin-releasing factor and opioids. Potential for reciprocal interactions between
354, 1–7. stress and opioid sensitivity. Psychopharmacology (Berl.) 158 (4), 331–342.
Tenore, P.L., 2008. Psychotherapeutic benefits of opioid agonist therapy. J. Addict. Valentino, R.J., Van Bockstaele, E., 2015. Endogenous opioids: the downside of
Disord. 27, 49–65. opposing stress. Neurobiol. Stress 1, 23–32.
Terashvili, M., Wu, H.E., Leitermann, R.J., Hung, K.C., Clithero, A.D., Valverde, O., Ledent, C., Beslot, F., Parmentier, M., Roques, B.P., 2000. Reduction of
Schwasinger, E.T., Tseng, L.F., 2004. Differential conditioned place preference stress-induced analgesia but not of exogenous opioid effects in mice lacking CB1
responses to endomorphin-1 and endomorphin-2 microinjected into the posterior receptors. Eur. J. Neurosci. 12, 533–539.
nucleus accumbens shell and ventral tegmental area in the rat. J. Pharmacol. Exp. Van den Berg, C.L., Lamberts, R.R., Wolterink, G., Wiegant, V.M., Van Ree, J.M.,
Ther. 309 (2), 816–824. 1998. Emotional and footshock stimuli induce differential long-lasting behavioural
Terman, G.W., Shavit, Y., Lewis, J.W., Cannon, J.T., Liebeskind, J.C., 1984. Intrinsic effects in rats; involvement of opioids. Brain Res. 799, 6–15.
mechanisms of pain inhibition: activation by stress. Science 226, 1270–1277. Van der Kooy, D., Mucha, R.F., O’Shaughnessy, M., Bucenieks, P., 1982. Reinforcing
Terman, G.W., Penner, E.R., Liebeskind, J.C., 1985. Stimulation-produced and stress- effects of brain microinjections of morphine revealed by conditioned place pref-
induced analgesia: cross-tolerance between opioid forms. Brain Res. 360, erence. Brain Res. 243 (1), 107–117.
374–378. Van Loon, G.R., Pierzchala, K., Houdi, A.A., Kvetnansky, R., Zeman, P., 1990.
Terman, G.W., Lewis, J.W., Liebeskind, J.C., 1986a. Two opioid forms of stress Tolerance and cross-tolerance to stress-induced increases in plasma meten-
analgesia: studies of tolerance and cross-tolerance. Brain Res. 368, kephalin in rats with adaptively increased resting secretion. Endocrinology 126,
101–106. 2196–2204.
Terman, G.W., Morgan, M.J., Liebeskind, J.C., 1986b. Opioid and non-opioid stress Vanderah, T.W., Wild, K.D., Takemori, A.E., Sultana, M., Portoghese, P.S.,
analgesia from cold water swim: importance of stress severity. Brain Res. 372, Bowen, W.D., Hruby, V.J., Mosberg, H.I., Porreca, F., 1993. Modulation of
167–171. morphine antinociception by swim-stress in the mouse: involvement of
Teskey, G.C., Kavaliers, M., 1988. Effects of opiate agonists and antagonists on supraspinal opioid delta-2 receptors. J. Pharmacol. Exp. Ther. 267 (1),
aggressive encounters and subsequent opioid-induced analgesia, activity and 449–455.
feeding responses in male mice. Pharmacol. Biochem. Behav. 31, 43–52. Van’t Veer, A., Carlezon Jr., W.A., 2013. Role of kappa-opioid receptors in stress and
Thompson, R.C., Mansour, A., Akil, H., Watson, S.J., 1993. Cloning and anxiety-related behavior. Psychopharmacology (Berl.) 229, 435–452.
pharmacological characterization of a rat mu opioid receptor. Neuron 11 (5), Vargish, T., Reynolds, D.G., Gurll, N.J., Lechner, R.B., Holaday, J.W., Faden, A.I.,
903–913. 1980. Naloxone reversal of hypovolemic shock in dogs. Circ. Shock 7 (1),
Tian, M., Broxmeyer, H.E., Fan, Y., Lai, Z., Zhang, S., Aronica, S., Cooper, S., 31–38.
Bigsby, R.M., Steinmetz, R., Engle, S.J., Mestek, A., Pollock, J.D., Lehman, M.N., Vaswani, K.K., Richard, C.W., Tejwani, G.A., 1988. Cold swim stress-induced changes
Jansen, H.T., Ying, M., Stambrook, P.J., Tischfield, A., Yu, L., 1997. Altered in the levels of opioid peptides in the rat CNS and peripheral tissues. Pharmacol.
hematopoiesis, behavior, and sexual function in mu opioid receptor-deficient mice. Biochem. Behav. 29, 163–168.
J. Exp. Med. 185 (8), 1517–1522. Vellucci, S.V., Parrott, R.F., 1997. Vasopressin and oxytocin gene expression in the
Tiligada, E., Wilson, J.F., 1990. Ionic, neuronal and endocrine influences on the porcine forebrain under basal conditions and following acute stress. Neuropeptides
proopiomelanocortin system of the hypothalamus. Life Sci. 46, 81–90. 31, 431–438.
Till, M., Gati, T., Rabai, K., Szombath, D., Szekely, J.I., 1988. Effect of [D-Met2, Pro]- Ventura, C., Guarnieri, C., Vaona, I., Campana, G., Pintus, G., Spampinato, S., 1994.
enkeph, alinamide on gastric ulceration and transmucosal potential difference. Eur. Dynorphin gene expression and release in the myocardial cell. J. Biol. Chem. 269
J. Pharmacol. 15, 325–330. (7), 5384–5386.
260 Stress and Opioid Systems

Vergura, R., Balboni, G., Spagnolo, B., Gavioli, E., Lambert, D.G., McDonald, J., Yamauchi, N., Shibasaki, T., Wakabayashi, I., Demura, H., 1997. Brain beta-endorphin
Trapella, C., Lazarus, L.H., Regoli, D., Guerrini, R., Salvadori, S., Caló, G., 2008. and other opioids are involved in restraint stress-induced stimulation of the
Anxiolytic- and antidepressant-like activities of H-Dmt-Tic-NH-CH(CH2-COOH)-Bid hypothalamic-pituitary-adrenal axis, the sympathetic nervous system, and the
(UFP-512), a novel selective delta opioid receptor agonist. Peptides 29, 93–103. adrenal medulla in the rat. Brain Res. 777, 140–146.
Vidal, C., Suaudeau, C., Jacob, J., 1984. Regulation of body temperature and noci- Younes, A., Pepe, S., Barron, B.A., Spurgeon, H.A., Lakatta, E.G., Caffrey, J.L., 2000.
ception induced by non-noxious stress in rat. Brain Res. 297, 1–10. Cardiac synthesis, processing, and coronary release of enkephalin-related
Vitale, G., Arletti, R., Sandrini, M., 2005. Acute noise stress analgesia in relation to 5- peptides. AJP Heart Circ. Physiol. 279 (4), H1989–H1998.
HT2 and mu-opioid receptor changes in the frontal cortex of young mice. Life Sci. Young, E.A., Akil, H., 1985. Corticotropin-releasing factor stimulation of adrenocorti-
77 (20), 2500–2513. cotropin and b-endorphin release: effect of acute and chronic stress. Endocri-
Viveros, D.H., Diliberto Jr., E.J., Hazum, E., Chang, K.J., 1979. Opiate-like materials in nology 117, 23–30.
the adrenal medulla: evidence for storage and secretion with catecholamines. Mol. Young, W.S., Lightman, S.L., 1992. Chronic stress elevates enkephalin expression in the
Pharmacol. 16, 1101–1108. rat paraventricular and supraoptic nuclei. Brain Res. Mol. Brain Res. 13, 111–117.
Wager, T.D., Scott, D.J., Zubieta, J.K., 2007. Placebo effects on human mu-opioid Young, E.A., Bronstein, D., Akil, H., 1993a. Dopamine regulation of swim stress
activity during pain. Proc. Natl. Acad. Sci. U.S.A. 104 (26), 11056–11061. induction of the pituitary intermediate lobe proopiomelanocortin system. Neuro-
Walker, J.M., Berntson, G.G., Paulucci, T.S., Champney, T.C., 1981. Blockade of endocrinology 58, 294–302.
endogenous opiates reduces activity in the rat. Pharmacol. Biochem. Behav. 14, Young, E.A., Przewlocki, R., Patel, P., Watson, S.J., Akil, H., 1993b. Altered ratios of
113–116. beta-endorphin: beta-lipotropin released from anterior lobe corticotropes with
Wang, X.Q., Imaki, T., Shibasaki, T., Yamauchi, N., Demura, H., 1996. Intra- increased secretory drive. I. Effects of diminished glucocorticoid secretion.
cerebroventricular administration of beta-endorphin increases the expression of J. Neuroendocrinol. 5, 115–120.
c-fos and of corticotropin-releasing factor messenger ribonucleic acid in the Young, E.A., 1990. Induction of the intermediate lobe pro-opiomelanocortin system
paraventricular nucleus of the rat. Brain Res. 707 (2), 189–195. with chronic swim stress and b-adrenergic modulation of this induction. Neuro-
Wang, X.M., Zhou, Y., Spangler, R., Ho, A., Han, J.S., Kreek, M.J., 1999. Acute edocrinology 52, 405–414.
intermittent morphine increases preprodynorphin and kappa opioid receptor mRNA Yukhananov, R.Y., Handa, R.J., 1996. Alterations in kappa opioid receptor mRNA
levels in the rat brain. Brain Res. Mol. Brain Res. 66 (1–2), 184–187. levels in the paraventricular nucleus of the hypothalamus by stress and sex
Wang, Q.P., Zadina, J.E., Guan, J.L., Shioda, S., 2003. Morphological evidence of steroids. Neuroreport 7, 1690–1694.
endomorphin as an agonist for the mu-opioid receptor in the rat spinal cord. Yukhananov, R.Y., Handa, R.J., 1997. Estrogen alters proenkephalin RNAs in the par-
Neurosci. Lett. 341 (2), 107–110. aventricular nucleus of the hypothalamus following stress. Brain Res. 764, 109–116.
Ward, H.G., Nicklous, D.M., Aloyo, V.J., Simansky, K.J., 2006. Mu-opioid receptor Zacharko, R.M., Maddeaux, C., Hebb, A.L., Mendella, P.D., Marsh, N.J., 1998.
cellular function in the nucleus accumbens is essential for hedonically driven Vulnerability to stressor-induced disturbances in self-stimulation from the dorsal
eating. Eur. J. Neurosci. 23 (6), 1605–1613. and ventral A10 area: differential effects of intraventricular D-Ala2-Met5-
Watanabe, Y., Weiland, N.G., McEwen, B.S., 1995. Effects of adrenal steroid enkephalinamide, D-Ala2, N-Me-Phe4, Gly-Ol5-enkephalin, and D-Pen2, D-
manipulations and repeated restraint stress on dynorphin mRNA levels and Pen5-enkephalin administration. Brain Res. Bull. 47, 237–248.
excitatory amino acid receptor binding in hippocampus. Brain Res. 680, 217–225. Zadina, J.E., Hackler, L., Ge, L.J., Kastin, A.J., 1997. A potent and selective
Watson, S.J., Akil, H., Ghazarossian, V.E., Goldstein, A., 1981. Dynorphin immuno- endogenous agonist for the mu-opiate receptor. Nature 386, 499–502.
cytochemical localization in brain and peripheral nervous system: preliminary Zadina, J.E., Martin-Schild, S., Gerall, A.A., Kastin, A.J., Hackler, L., Ge, L.J.,
studies. Proc. Natl. Acad. Sci. U.S.A. 78 (2), 1260–1263. Zhang, X., 1999. Endomorphins: novel endogenous mu-opiate receptor agonists in
Weber, M.M., Emrich, H.M., 1988. Current and historical concepts of opiate treatment regions of high mu-opiate receptor density. Ann. N.Y. Acad. Sci. 897, 136–144.
in psychiatric disorders. Int. Clin. Psychopharmacol. 3, 255–266. Zadina, J.E., 2002. Isolation and distribution of endomorphins in the central nervous
Weber, M.M., 1987. The opium cure in psychiatry. A contribution to the history of system. Jpn. J. Pharmacol. 89, 203–208.
psychopharmacotherapy. Sudhoffs Arch. 71, 31–61. Zangen, A., Shalev, U., 2003. Nucleus accumbens beta-endorphin levels are not
Wideman, C.H., Murphy, H.M., McCartney, S.B., 1996. Interactions between vaso- elevated by brain stimulation reward but do increase with extinction. Eur. J.
pressin and food restriction on stress-induced analgesia. Peptides 17 (1), 63–66. Neurosci. 17, 1067–1072.
Wiedenmayer, C.P., Barr, G.A., 2000. Mu opioid receptors in the ventrolateral peri- Zangen, A., Ikemoto, S., Zadina, J.E., Wise, R.A., 2002. Rewarding and psychomotor
aqueductal gray mediate stress-induced analgesia but not immobility in rat pups. stimulant effects of endomorphin-1: anteroposterior differences within the ventral
Behav. Neurosci. 114, 125–136. tegmental area and lack of effect in nucleus accumbens. J. Neurosci. 22, 7225–7233.
Will, M.J., Watkins, L.R., Maier, S.F., 1998. Uncontrollable stress potentiates Zeman, P., Alexandrova, M., Kvetnansky, R., 1988. Opioid lx and b and dopamine
morphine’s rewarding properties. Pharmacol. Biochem. Behav. 60 (3), 655–664. receptor number changes in rat striatum during stress. Endocrinol. Exp. 22,
Wittert, G., Hope, P., Pyle, D., 1996. Tissue distribution of opioid receptor gene 59–66.
expression in the rat. Biochem. Biophys. Res. Commun. 218 (3), 877–881. Zhang, H., Shi, Y.G., Woods, J.H., Watson, S.J., Ko, M.C., 2007. Central kappa-opioid
Wittmann, W., Schunk, E., Rosskothen, I., Gaburro, S., Singewald, N., Herzog, H., receptor-mediated antidepressant-like effects of nor-Binaltorphimine: behavioral
Schwarzer, C., 2009. Prodynorphin-derived peptides are critical modulators of anxiety and BDNF mRNA expression studies. Eur. J. Pharmacol. 570, 89–96.
and regulate neurochemistry and corticosterone. Neuropsychopharmacology 34, Zhao, B.G., Chapman, C., Bicknell, R.J., 1988. Functional kappa-opioid receptors on
775–785. oxytocin and vasopressin nerve terminals isolated from the rat neurohypophysis.
Woolfolk, D.R., Holtzman, S.G., 1995. Rat strain differences in the potentiation of Brain Res. 462, 62–66.
morphine-induced analgesia by stress. Pharmacol. Biochem. Behav. 51 (4), Zhou, Y., Spangler, R., Maggos, C., Wang, X.M., Han, J.S., Ho, A., Kreek, M.J., 1999.
699–703. Hypothalamic-pituitary-adrenal activity and pro-opiomelanocortin mRNA levels in
Woolley, J.D., Lee, B.S., Kim, B., Fields, H.L., 2007a. Opposing effects of intra- the hypothalamus and pituitary of the rat are differentially modulated by acute
nucleus accumbens mu and kappa opioid agonists on sensory specific satiety. intermittent morphine with or without water restriction stress. J. Endocrinol. 163,
Neuroscience 146 (4), 1445–1452. 261–267.
Woolley, J.D., Lee, B.S., Taha, S.A., Fields, H.L., 2007b. Nucleus accumbens opioid Zhu, X., Desiderio, D.M., 1994. Effects of space flight stress on proopiomelanocortin,
signaling conditions short-term flavor preferences. Neuroscience 146 (1), 19–30. proenkephalin A, and tachykinin neuropeptidergic systems in the rat posterior
Wybran, J., Appelboom, T., Famaey, J.P., Govaerts, A., 1979. Suggestive evidence for pituitary. Life Sci. 55, 347–350.
receptors for morphine and methionine-enkephalin on normal human blood T Zimmer, A., Valjent, E., Konig, M., Zimmer, A.M., Robledo, P., Hahn, H., Valverde, O.,
lymphocytes. J. Immunol. 123 (3), 1068–1070. Maldonado, R., 2001. Absence of delta-9-tetrahydrocannabinol dysphoric effects
Wybran, E., 1985. Enkephalins and endorphins as modifiers of the immune system: in dynorphin-deficient mice. J. Neurosci. 21, 9499–9505.
present and future. Fed. Proc. 44, 92–96. Zimprich, A., Simon, T., Höllt, V., 1995. Cloning and expression of an isoform of the rat
Xie, G.X., Han, J.S., Höllt, V., 1983. Electroacupuncture analgesia blocked by mu opioid receptor (rMOR1B) which differs in agonist induced desensitization from
microinjection of anti-beta endorphin antiserum into periaqueductal grey in rabbit. rMOR1. FEBS Lett. 359 (2–3), 142–146.
Int. J. Neurosci. 18, 287–292. Zubieta, J.K., Bueller, J.A., Jackson, L.R., Scott, D.J., Xu, Y., Koeppe, R.A.,
Yamaguchi, H., Aiba, A., Nakamura, K., Nakao, K., Sakagami, H., Goto, K., Nichols, T.E., Stohler, C.S., 2005. Placebo effects mediated by endogenous opioid
Kondo, H., Katsuki, M., 1996. Dopamine D2 receptor plays a critical role in cell activity on mu-opioid receptors. J. Neurosci. 25 (34), 7754–7762.
proliferation and proopiomelanocortin expression in the pituitary. Genes Cells 1 Zurita, A., Molina, V., 1999. Prior morphine facilitates the occurrence of immobility and
(2), 253–268. anhedonia following stress. Physiol. Behav. 65, 833–837.
1.09 Social Stress: Concepts, Assumptions, and Animal Models
Sally P Mendoza, University of California, Davis, Davis, CA, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by C.R. McKittrick, D.C. Blanchard, M.P. Hardy, R.J. Blanchard, volume 1, pp. 333–366,
Ó 2009, Elsevier Inc.

1.09.1 Introduction 261


1.09.1.1 The Stress Response: What Is It and How Is It Measured? 262
1.09.1.2 Social Systems 263
1.09.1.2.1 Limitations of Inclusion 263
1.09.1.2.2 What Constitutes a Social Stressor? 264
1.09.2 Aggression, Intruders, Social Defeat 264
1.09.2.1 Resident/Intruder Models of Social Stress 264
1.09.2.2 Application and Limitation of Resident/Intruder Model 265
1.09.3 Aggression and Dominance in Social Groups 266
1.09.3.1 Case Study: Visible Burrow System in Rats 266
1.09.3.2 Mice in Groups 267
1.09.3.3 Social Dominance in Primates 268
1.09.3.3.1 Case Study: Physiological Processes Associated with Dominance in Baboons 269
1.09.3.3.2 Case Study: Social Influences on Stress Response Systems in Squirrel Monkeys 270
1.09.4 Social Instability 271
1.09.5 Disruption of Established Relationships: Separation, Bereavement, and Loneliness 273
1.09.5.1 Comparative Case Study: Polygynous Squirrel Monkeys versus Monogamous Titi Monkeys 274
1.09.5.2 Comparative Case Study: Voles 276
1.09.5.3 Comparative Case Study: Guinea Pigs 277
1.09.5.4 Social Isolation in Humans 278
1.09.6 Conclusions 279
References 281

1.09.1 Introduction used and what they have contributed to our understanding of
social stress in animals and humans.
Social stress is by definition a hybrid field, combining concepts We will begin with examination of the stress concept and its
in endocrinology and social processes into a single investigation. origins. In our everyday language, and in much of the literature,
Two contributions stand out as facilitating the shift from an stress refers both to the conditions that create discomfort in our
occasional mention of social stress to a field of inquiry in its lives and to the physiological reactions to those conditions. The
own right. The first was provided by Henry and Stephens most commonly evoked metaphor for understanding the phys-
(1977) in their treatment of personality types expressed within iological reactions to stressful conditions is the encounter
and created by the social environment. This work was based between predator and prey – from the perspective of the
on a long history of animal research organized to deal with prey. Nothing is more important than escape from the certain
a problem posed by human research – differences between folks death that awaits. All physiological systems involved with
with Type-A or Type-B personality. The second was an article by mobilizing resources are activated while others concerned
House et al. (1988) that posited that social stress, particularly with immediately nonessential processes are shut down.
social isolation, should be considered a risk factor of comparable Collectively this concerted physiological reaction to an emer-
magnitude to cigarette smoking. This work combined expertise gency situation is referred to as the stress response. How apt
in epidemiological studies in human populations with experi- is this metaphor for explaining the kinds of reactions to the
mental studies, largely conducted in animals. social and psychological conditions that are referred to as
From its inception social stress has been an interdisciplinary stressful in the modern condition? Our understanding of stress
effort, and most of the investigators look to studies in animals responses has grown immensely in the time since studies of
to model aspects of human biology. Recent years have seen social stress emerged. A particularly important distinction,
a blossoming of studies of social stress into investigations of that was not considered important initially, is between acute
neuroscience, immunology, health-care practices, and more. and chronic responses to stress.
Given the diversity of expertise that is currently required to Similarly, our understanding of social systems has
conduct studies of social stress and the rapidity within which increased enormously. Relationships first became a target of
the field has developed, it is timely to examine the underlying research with Bowlby (1969) focusing on mother–infant
assumptions and concepts both with respect to the models relationships and elaborated to more diverse relationships

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00009-2 261


262 Social Stress: Concepts, Assumptions, and Animal Models

with Hinde’s (1974) contribution to understanding social Most animal studies of stress focus on the alarm reaction
dynamics in human and nonhuman primates. This was fol- and the stage of resistance which together comprise the acute
lowed by a flurry of studies investigating the proximate mech- stress response. Acute stress has a predicable temporal
anisms that contribute to animal societies necessitating new sequence: corticotropin-releasing hormone (CRH) activates
conceptual understanding of social systems (Mason, 1976). SNS, including adrenomedullary release of epinephrine (Epi),
From this work we know that social organization is a charac- and stimulates adrenocorticotropin (ACTH) release; ACTH, in
teristic of the species which can vary within limits for any turn, stimulates adrenocortical release of corticosteroid
given species but changes dramatically between species. (cortisol in primates; corticosterone in most rodents); finally,
Animal studies of social stress need to be evaluated with corticosteroids acting through negative feedback mechanisms
a keen understanding of the social system. Moreover, studies inhibit further activation of the system. The temporal character-
of social stress have helped to illuminate social systems istics of the stress cascade are predictable. With the onset of the
(Mendoza and Mason, 1989). For these reasons emphasis in stressor, CRH release from the hypothalamus occurs within
this chapter will be placed on systematic investigations of milliseconds, and the sympathetic response follows within
particular research programs with particular species that seconds; conversion of norepinephrine (NE) to Epi occurs
were influenced by and influence the field of social stress as rapidly, and Epi released into the bloodstream augments and
it has developed and emerged as a field of inquiry. extends SNS effects; increased ACTH levels become detectable
within 1–2 min; cortisol takes longer time to increase appre-
ciably (5–10 min) in the general circulation and does not reach
1.09.1.1 The Stress Response: What Is It and How Is It
peak values for at least 20 min after stressor onset. At this point
Measured?
there are hypothetically two opposing processes acting within
Walter B. Cannon (1871–1945) and Hans Selye (1907–82) the HPA and SNS system. Activational mechanisms are stimu-
together provide the historical view of stress that we currently lating release of CRH and negative feedback mechanisms are
employ. Both Cannon and Selye were influenced by another operating to restore homeostasis. Corticosteroids are both the
physiologist – Claude Bernard (1813–78), a French physiolo- signal that activational mechanisms are still operating and
gist, who introduced the concept of milieu interieur. Bernard the means by which they are shut off or rendered ineffective.
recognized the importance of maintaining a constant internal Negative feedback sites regulating the HPA system include
environment – temperature, chemical balance, fluids, etc. – the hippocampus, amygdala, hypothalamus, pituitary, and
in the face of a continually changing external environment. adrenal. The multiplicity of feedback sites and their efficacy
He viewed physiology as compensating for even the smallest ensure that the HPA system is not activated continuously
deviations in the external environment in order to maintain even under circumstances when the stressor is sustained
constancy in the internal environment. Regulation of the milieu (de Kloet, 2003; Dallman, 2007). In addition to inhibitory
interieur, Bernard (1872) also recognized, was the purview of effects from corticosteroids, the other branch of the autonomic
the brain creating the possibility that mental imagery could nervous system (ANS), the parasympathetic nervous system
initiate physiological processes, such as those involved with (PNS), opposes many SNS effects at target organs, and it is
eating or thermogenesis. intimately involved in mediating social processes (Porges,
Cannon (1932) elaborated Bernard’s notion that physiolog- 2007, 2009).
ical systems exist to regulate internal processes in relation to Because of the rapidity of response in SNS and CRH and
external events, processes Cannon collectively referred to as ACTH, technological sophistication is required to measure
homeostasis. He too was concerned with anticipatory activa- early stages of the stress cascade especially in freely moving
tion of metabolic processes and coined the phrase ‘fight or and interacting animals. Measurement of corticosteroid
flight’ to characterize the emergency metabolic state underlying responses is relatively easy in comparison. Blood samples, for
the preparation for sudden action (Cannon, 1929). Whether example, can be collected within 5–10 min time delay from
responding fearfully or aggressively was determined by evalua- activation of the stress response to initial detection of stress-
tion of the circumstance, an appraisal process subsequently induced elevations in corticosteroid concentrations. Corticoste-
elaborated by Lazarus (1999). Cannon’s focus in these studies roid levels are often used to monitor the magnitude and
was the sympathetic nervous system (SNS). duration of the stress response, both of which are considered
The fight-or-flight response largely comprised the initial to reflect the intensity of the stressor (Hennessy and Levine,
stage (Alarm) of Selye’s (1956) General Adaptation Syndrome. 1978). Most procedures used to collect blood samples,
In addition to high levels of SNS activity, the Alarm stage, however, do constitute a stressor, and physiological events
according to Selye, is characterized by activation of the hypo- subsequent to collection of an initial blood sample would
thalamic–pituitary–adrenal (HPA) system. The second stage reflect the response to the experimental procedures as well as
of the General Adaptation Syndrome is Resistance during antecedent conditions (Coe et al., 1978). This has led people
which elevations in glucocorticoids reach their peak response. to turn to less invasive methods for inferring corticosteroid
Glucocorticoids during this stage serve multiple functions dynamics. As steroid hormones corticosteroids can pass lipid
including freeing stored energy in the form of glucose, barriers unless prevented from doing so by binding to its carrier
preserving use of energy for brain, and, most importantly, protein, corticosteroid-binding globulin (CBG), or albumin.
serving as a negative feedback signal to inhibit further activa- Only a small percentage of the circulating corticosteroid is
tion of the HPA and SNS, restoring homeostasis (Dallman, not bound to large carrier proteins. The free component can
2007). Responses to most stressors, according to Selye, are pass into saliva, urine, or feces directly or in metabolized
effectively over at the second stage. form. Of these salivary concentrations of corticosteroids mimic
Social Stress: Concepts, Assumptions, and Animal Models 263

the temporal cascade most directly (Hellhammer et al., 2009). principles governing the response to acute stress provide
Urine and feces are not instantly available and corticosteroid adequate explanatory value. The initiation of response can be
concentrations in these biological specimens reflect all events equated with the perception and identification of the intruder
since last void. and the physiological response followed, at least theoretically,
Measurement of Epi and NE are possible in both plasma from initiation to reestablishing homeostasis. That is, the acute
and urine, but capturing the dynamics of early stress events is response to stress provides an adequate conceptual framework
not possible with either measure. This has led to the use of to understand isolated incidences or contrived events. But in
known effects of the ANS system on heart rate (HR), blood most species, including humans, individuals are inextricably
pressure, and the like. HR, in particular, has been a useful embedded within the social system, and its constant demands,
measure of both SNS and PNS activity (Porges et al., 2007). potentials, benefits, and hazards are ongoing and difficult to
In summary, the SNS and the HPA system are often referred isolate from one another. Without being able to identify stress
to as the stress-response systems, and their end-product onset or isolate one stressor from another, more complex
hormones, Epi (adrenalin) and cortisol (or corticosterone), are models of social stress are needed than those provided by
the stress hormones. So profound has been the influence of our understanding of the acute stress response.
Cannon and Selye that we often equate levels of these hormones To further complicate studies of social stress, the type of
with the degree of stress so that higher hormone levels social structure varies from species-to-species. While we can
mean greater stress therefore greater adverse health outcomes. recognize common features among many different species
It is important to remember that these hormones are activated not all types of relationships are equally relevant to each. For
for reasons other than stress. Cortisol, for example, displays example, dominance relations which primarily govern interac-
circadian activity which rises just before awakening. In humans tions between same-sex individuals are not relevant for under-
there is an additional activation of the HPA system upon awak- standing social stress for monogamous species. Conversely, the
ening, the magnitude and form of which is similar to an acute adult emotional bond which is characteristic of monogamous
stress response (Hellhammer et al., 2007). Similarly, as a primary species is not a feature of adult relations in polygynous species.
metabolic hormone, cortisol is also released in large quantities Moreover, social systems while species-typical are not invariant
surrounding meals (Warne et al., 2009). Exercise activates both and adaptations within and between generations are
HPA system and SNS. We would not want to suggest that any commonly reported. No element of animal societies can be
of these activities – awakening, exercise, and eating – are stressful said to follow a phylogenetic progression, yet rapid phyloge-
and detrimental to health. The principal way we have differenti- netic changes in form of society do occur (Mendoza and
ated the stress response from activation for other purposes is by Mason, 1989).
the nature of the stressor. It is our value judgment, if you will,
that the nature of the eliciting stimulus defines the response as 1.09.1.2.1 Limitations of Inclusion
stressful or simply metabolic adjustment. In this chapter we will consider social stress to be any condition
Selye incorrectly believed that under conditions of extreme emanating from the social environment that significantly alters
stress the homeostatic mechanisms would be overwhelmed activity in the SNS and/or HPA system for normally socialized
and the Stage of Exhaustion initiated. We now know that the mammalian individuals. This pursuit will include changes that
Stage of Exhaustion does not happen as Selye envisioned. are both protective and detrimental to health. To truly fit the
Negative feedback mechanisms are too potent to permit definitions of stress intended by Cannon and Selye negative
continued activation of the HPA system even in the presence health outcomes are expected to result from too much social
of a persistent stressor (Dallman, 2007). Likewise, repeated stress. Too often, however, the necessary steps to link stress
stressors lose their ability to activate the HPA system even and disease have not been identified. Consequently, social situ-
when habituation to the stressor does not occur (Ruys et al., ations may have been incorrectly categorized as beneficial or
2004). Increasingly it appears that chronic, persistent, or detrimental based on incomplete information. Mistakes have
repeated stressors lead to altered regulation and not activation been made.
to the point of adrenal depletion (Mendoza et al., 2000; Munck Behaviorally and physiologically our approach is too
et al., 1984). Dysregulation of HPA system is increasingly being limiting to capture the full essence of studies of social stress.
considered a sign of chronic stress. Dysregulation can take the Many other physiological systems contribute to the outcome
form of excessive activation, lack of negative feedback regula- of social stress or provide evidence for its effects. Oxytocin
tion, excessive feedback regulation, or flattened circadian regu- and vasopressin, for example, are intimately involved in medi-
lation. Importantly, dysregulation can take the form of an ating social relationships, and they impinge on other physio-
overactive system (e.g., diminished negative feedback) or an logical processes, including HPA and SNS, in ways that can
underactive one (e.g., enhanced negative feedback), and corti- obscure or reveal instances of social stress (Insel et al., 1993;
costeroid concentrations, likewise, can be either higher or lower Carter et al., 2008). These hormones receive some scant atten-
(Lupien et al., 1995). tion in this chapter, and they are limited to a discussion of
prairie voles. Focusing on normally socialized animals leaves
out a wealth of information on developmental consequences
1.09.1.2 Social Systems
of early social manipulations (Mason, 2008). Our focus will
The problems of evaluating social stress are further com- be on mammals leaving aside birds and reptiles. Within
pounded by the ongoing nature of social stress. For animals mammals, we are further limited by the typical choice of study
living in simple social systems where the appearance of subjects. The most studied laboratory animals are mice, rats,
a conspecific is either a potential mate or potential rival, the guinea pigs, and voles. Systematic studies of social stress have
264 Social Stress: Concepts, Assumptions, and Animal Models

also been conducted extensively in several primate species interanimal aggression when food was restricted or space
including tree shrews, titi monkeys, squirrel monkeys, severely reduced. Instead it was discovered that novel animals,
macaques, baboons, and humans. In order to preserve our intruders, entering the living area defined physically or socially,
focus on species-typical social systems, discussion of each were the most effective aggression eliciting stimuli (Bernstein
will be accomplished in succession and more or less separately. and Gordon, 1974).
The HPA-axis response to an agonistic interaction appears
1.09.1.2.2 What Constitutes a Social Stressor? to depend, in part, on the outcome of the encounter. After
By virtue of their reproductive systems all mammals are at least fighting between male rats, corticosterone levels are increased
moderately social in order to breed and nurture offspring. in the losers (Koolhaas et al., 1983), while corticosterone
Perhaps the most typical social system among mammals is levels decline in the winner (Haller et al., 1996). The SNS is
one of overlapping territories. In these species females typically likewise altered in defeat. For example, if a smaller rat (S/JR)
have smaller territories which overlap with one or more larger experiences defeat by a rat of a larger strain (Long-Evans), the
territories maintained by males. Other adults are not usually defeated, smaller rat exhibits decreases in systolic BP that
encountered and when they are they are treated either as an persists for up to 2 h (Adams et al., 1987).
intruder or a potential mate. Only about 5% of mammals Most animals show an affinity not only for particular indi-
live in permanent social groups. Some are reliant on interper- viduals but also for place. Territoriality is most common in
sonal relationships; other species respond to the category animals with social structures defined by overlapping territories
another represents (male/female, young/mature) rather than or monogamy (Davis and Marler, 2003). For many animals
differentiating among individuals within category. The forms defense of territory is an important social task and, with the
of relationships in each society and their importance vary possible exception of mating, the primary social interaction
from loose associations in herds or colonies without individual especially for males. The resident/intruder test capitalizes on
recognition; monogamous groups characterized by a single that tendency. Miczek (1979) is credited with developing the
male and a single female and their offspring; harems with resident/intruder model used extensively in studies of aggres-
a single male and multiple females; to complex societies with sion in rodents.
individuals of all ages and both sexes and within which all indi-
viduals are uniquely identified.
1.09.2.1 Resident/Intruder Models of Social Stress
What constitutes a social stressor for mammals needs to be
filtered through the social system of the test subject. We would Due to the sex difference in proclivity to fight upon encoun-
expect isolation to be a stressor for a macaque that ordinarily tering another like-sex animal, resident/intruder studies in
lives in very large social groups with complex and highly indi- rodents are almost exclusively performed using male subjects.
vidualized relationships, but not for orangutans which live an Residents in this model are generally favored in outcomes. In
isolated existence normally. Current circumstances are also some cases the scales are further tipped by having the resident
important. Consider the monogamous titi monkey, for or intruder preselected for size or aggressiveness (Davis and
example. When living with a mate, a stranger is treated as an Marler, 2003). If focus is on the winner of the encounter,
intruder to be expelled or avoided regardless of sex of intruder; commonly referred to as the challenge hypothesis (Wingfield
when living without a mate, a stranger represents the potential et al., 1990), one typically finds that corticosteroids are not
for a new mate if it is the right sex. altered. Repeated defeat, on the other hand, leads to reduction
Studies of social stress can also inform our understanding in body weight and adrenal hypertrophy with elevations in
of a particular social system. For example, if presence of corticosteroids (Archer, 1970; Kudryavtseva and Avgustinovich,
a stranger but not a familiar social companion elicits a stress 1998). The importance of territory is underscored by compar-
response, then we can infer at least that the test subject can ison of rates of aggression in the resident/intruder model
recognize the difference and the stranger represents a need with aggression if the same two males meet in an environment
to mobilize resources. If we further determine that separation equally novel to both animals. When the encounter takes place
from one familiar animal induces a stress response and sepa- in the resident/intruder situation, latency to attack is faster,
ration from another does not, we can conclude that the test more frequent, and more intense than are aggressive bouts
subject perceives absence of one as more important socially when the males meet in a place equally unfamiliar to both
than the other. (Takahashi et al., 2012).
Rats placed with a resident from a highly aggressive strain of
rats show increases in NE, Epi, and corticosterone to defeat
1.09.2 Aggression, Intruders, Social Defeat (Sgoifo et al., 1996). In this study males were fitted with an
indwelling cardiac catheter which permitted collection of
Aggression is typically the social behavior that immediately blood samples throughout the test period without additional
comes to mind when thinking about a stressor emanating stress introduced with the collection of samples. Results from
from within the social environment (McKittrick et al., 2009). social defeat were compared to introduction of a shock prod
Early studies of aggression noted that spontaneous aggression to males while in the home cage. Within 1 min Epi and NE
occurs during mate competitions, territorial encounters, were significantly elevated in response to social defeat and
defense of young particularly by mothers, resource competi- shock. By 5 min Epi and NE had reached peak values and
tion, and so forth (Hinde, 1966). Competition for incentives by 15 min levels of Epi plateaued and NE decreased even
as the primary motivating source of aggression was questioned though the test was still ongoing. Following the 15-min test
when Southwick (1967) failed to find sustained increases in period, both catecholamines declined to pretest basal levels
Social Stress: Concepts, Assumptions, and Animal Models 265

15–45 min following removal from the test situation. Cortico- exposure to the conditions of social defeat did not produce
sterone, as expected, was slower to react to the test situation evidence of HPA dysfunction (such as increased adrenal
reaching significant elevations at 5 min, but not reaching weight, thymus involution, altered circadian rhythms, or basal
peak until 15–30 min following test initiation with peak levels corticosterone outside the test situation) as has been found for
occurring after the cessation of the stressor. Even though the other repeated daily stressors (Bhatnagar and Vining, 2003).
time course of the response of each of the hormones to each Repeated exposure to the conditions of social defeat does sensi-
stressor was the same, the magnitude of response to social tize the response to a novel stressor, physical restraint, indi-
defeat was much exaggerated in comparison to shock. Pretests cating at least some long lasting consequences.
of latency to attack an intruder placed in the subject’s home Studies pharmacologically manipulating HPA axis suggest
cage were used to determine trait aggressiveness. Aggressiveness that corticosteroids promote aggressive behavior in the
did not alter the corticosterone response to either stressor. In residents during resident/intruder tests (Mikics et al., 2004).
contrast, males with shorter attack latencies in the pretest Metyrapone blocks synthesis of cortisol, and administration 2
were considered more aggressive, and these animals showed or 20 min prior to resident/intruder tests decreased initiation
a greater NE and Epi response to both shock and social defeat of aggression by residents directed to smaller opponents.
that could not be explained by different behavioral reactions Central or peripheral injections of corticosterone reinstated
to defeat. aggressive reactions. Blocking protein synthesis by administra-
These results illustrate how closely the response to defeat tion of cycloheximide prevents glucocorticoid effects if admin-
mimics the classic stress response, but it is a substantially istered immediately (2 min) before the test but is ineffective in
greater response than another stressor, electric shock. At least modulating glucocorticoid effects if administered 20 min prior
a possible explanation for this is that the two stress situations, to the test. These findings imply that cortisol acts centrally to
shock and social defeat, afford the mice differing degrees of facilitate aggressive responses and that it acts through a rapid
control. The shock was delivered by a shock prod which mechanism – cell membrane changes perhaps – that does
afforded the rats an opportunity to avoid subsequent exposure not require the inevitable time delay for genomic-mediated
to the shock device. The mice spent considerable amounts of mechanisms.
time burying the shock prod (Sgoifo et al., 1996). Engagement
in this behavior could be considered a coping response as Weiss
1.09.2.2 Application and Limitation of Resident/Intruder
(1972) noted providing a modicum of predictability and
Model
control in the situation could reduce the stress response to elec-
tric shock. The notion that coping responses provide a total Given the involvement of SNS and the long-term changes
explanation for the differential response to shock prod and produced by social defeat on cardiovascular function, the resi-
social defeat is somewhat qualified by the finding that those dent/intruder model has proven useful for study of early stage
animals that exhibited the most attempts to bury the shock cardiovascular disease, including hypertension, determinants
prod also exhibited the greatest response to social defeat. The of arrhythmias, and cardiac myopathy (Sgoifo et al., 2012).
authors suggest that animals that are inclined to bury the shock The resident/intruder model is extremely useful in identifying
prod are generally more likely to respond to stressful circum- characteristics of the acute stress response, its dynamics, and
stances by adopting an active coping style (Sgoifo et al., its regulatory control (Koolhaas et al., 1997). Hamsters show
1996). The authors further note that if this is true it did not similar responses to the resident/intruder paradigm as
lead to observed differences in the behavior of defeated rats. described for mice and rats, except the defeated animal is
The response to a single exposure to social defeat and the equally likely to be the resident or intruder (Kollack-Walker
attendant physiological changes can leave lasting changes et al., 1997). Examination of neural activity, as measured by
(Koolhaas et al., 1997). Long after the stressful experience, expression of c-fos mRNA, reveals that defeated hamsters
these authors note that binding capacity of Type I and Type II exhibited enhanced activity in regions typically associated
receptors for glucocorticoids is reduced in hippocampus with acute stress including, cingulate cortex, lateral septum,
3 weeks later; circadian temperature rhythms take 8–10 days bed nucleus of the stria terminalis, medial preoptic area, central
to recover; and growth is suppressed for 5 days, but body amygdala, and locus coeruleus. Moreover, most of these
weight may be permanently altered. The consequences of regions have also been implicated in the behavior of stressed
a severe attack leave the attacked individual altered. animals. The resident/intruder model is not a good model, as
Experiencing multiple defeat episodes does not produce illustrated in the next section for spontaneous formation of
habituation to the procedure (Sgoifo et al., 2005). Victors in dominance–subordinate relationships in complex social
a series of resident/intruder episodes (i.e., the residents from animals.
those encounters) who are placed in the cage of another aggres- For a few, modestly social species, social behavior can be
sive resident display long-term changes to the circadian rhythm modeled effectively using the resident/intruder model. Tree
of HR. In this circumstance the response of the defeated animal shrews are one such species. Tree shrews, considered a rodent
is determined by his behavior rather than that of the attacker. If by most taxonomies but a primate by some, exhibit a particu-
the defeated animal responds to being placed in the resident’s larly virulent response to an intruder. Resident males will
cage with environmental exploration and responds to being immediately attack another male and defeat them within
attacked with counterattack (i.e., an actual fight occurs), then a few minutes. Shortly after the fight has ended the resident
the effect of being the intruder after having been the victorious (victor) shows no further signs of arousal and generally ignores
resident is minimized. In spite of the lack of habituation to the defeated male. The defeated tree shrew typically withdraws
social defeat in the resident/intruder paradigm, repeated as completely as possible from the purview of dominant and
266 Social Stress: Concepts, Assumptions, and Animal Models

may even die within a few days (von Holst, 1972). Those assumption is safest to make in the common intruder para-
subordinates that survive the defeat, engage in active avoidance digms that prevent physical aggression by introducing animals
of the dominant, quitting the field anytime the dominant to one another in circumstances that preclude direct interaction
approaches. For these subordinates, high levels of SNS activity (Sgoifo et al., 1998). In these circumstances the response to
have been reported (Fuchs, 1984) with concomitant loss of threat is attenuated in comparison to direct interactions. In
body weight and reproductive function (Fischer et al., 1985). group-living situations threats to attack are more common
For studies wanting to examine the consequences of unbri- than attacks even when there are limiting circumstances –
dled attack, perhaps the best animal model might be found in such as confinement in small spaces that preclude escape.
the response of female rats, protecting their litter. Presenting Also in contrast to controlled dyadic encounters such as in
a male intruder to a lactating female rat elicited maternal aggres- the resident/intruder paradigm, aggression or the threat of
sion which rarely included elements of ritualized attack and aggression within social groups is part of an ongoing stream
submission typical of male–male encounters. Instead females of behavior which includes multiple agonistic encounters as
responded to intruders with bites directed to the most vulnerable well as feeding, sexual, and affiliative behaviors which may
body regions. Defeat in this circumstance induces increased mitigate any effects of stress by social or nonsocial means.
blood pressure, catecholamines, and corticosteroids (Koolhaas In most group-living primates and rodents it is evident that
and Bohus, 1991). For the female, engaging in maternal aggres- all-out fighting is rare. Within group aggression is generally
sion (15 min) induced increased HR with evidence of higher mild, ritualized, and also infrequent – most of the time, in
SNS activity. Defeat (of the intruder) induced elevations in Epi most circumstances, for most species. When aggression does
and NE. If the intruder were presented behind a mesh barrier, occur, it is usually unidirectional. Dominance relationships
effectively eliminating direct fighting but perhaps enhancing are a particularly common and often prominent feature of
the threat of aggression, elevations in Epi and NE were evident male–male relationships in large group-living species and in
but not to the same extent as with direct aggression. Elevations some species can describe aspects of female–female relations
in HR were likewise noted in the intruder, but did not differ in as well. It is important to note, however, that male–male rela-
conditions in which direct contact was or was not permitted. tionships in many species, particularly primates, are more
HR variability indicative of heightened PNS activity positively complex than can be described only by the direction and
correlated with passive behavior (immobility, on the back, frequency of quasi-aggressive interactions. Nonetheless, it is
upright defense) but did not change during testing and instead generally assumed that the subordinate member of the dyad
increased during recovery from aggressive encounters (Sgoifo is inhibited and socially suppressed in a way that is largely
et al., 1998). detrimental to well-being. “Subordination constitutes a natural
The resident/intruder model is an excellent means of and often chronic stress condition for males of virtually all
inducing and studying the acute stress response. The question species of social mammals” (Blanchard et al., 1998: p. 307).
must be asked, however, to what extent the resident/intruder
situations model the natural behavior of the animals. It would
1.09.3.1 Case Study: Visible Burrow System in Rats
virtually never be the case that animals appear in the core of
another animal’s territory as an initial encounter. Instead Wild rats, including the progenitor of most laboratory strains
such initial encounters occur at the periphery of territories, (Rattus norvegicus), live in colonies in which multiple adults
and a vigorous attempt to expel another is likely to be met share nesting sites and feeding grounds (Barnett, 1975). Such
with flight on the part of the intruder (Barnett, 1975). More- colonies can number more than 100 individuals. Most early
over, most studies utilize methods to exaggerate the incidence laboratory studies of dominance and aggression in group-
of attack by using larger residents, selecting the most aggressive housed rats used an open arena in which previously unfamiliar
animals to be residents, or by enhancing aggression in residents animals were introduced to one another and the novel environ-
by prior social isolation using various taunting methods to ment simultaneously. In these circumstances rats are often in
induce aggression. We would not expect to witness the proximity to each other and will typically huddle together in
complete resident/intruder consequences to occur in nature groups during sleep. Attacks are usually reserved for noncolony
even for the species typically used in these experiments. With members and often results in death of the intruder or, if the
the possible exception of tree shrews, the resident/intruder situation allows, expulsion of the stranger. Aggressive episodes
paradigm, therefore, is not a good model of stress emanating between adults within the colony, natural or artificial, are
spontaneously from the social environment. restricted to ritualized male–male encounters and females pro-
tecting the young (Barnett, 1975).
Rats are normally burrowing animals and, since feeding
1.09.3 Aggression and Dominance in Social Groups sites are above ground, they are removed from sleeping or nest-
ing sites. In order to study dominance in an environment that
While we might expect the physiological response to threat to permits the full range of behaviors generally employed by rats,
share some similar response elements with an actual bout of a visible burrow system was devised (Blanchard and Blanchard,
injurious aggression, the stressor in the instance of threat is 1990). This apparatus includes an open arena containing food
purely psychological which, obviously, is not the case with and water which is lighted for 12 h per day simulating daytime;
injurious aggression. We reason that animals respond to threat the arena during the dark phase and remainder of the apparatus
by preparing for the possibility of injury. While possibly true is lit by red light that permits observation but is perceived as
this assumes that we know the animals are processing a threat dark by the rats. The open arena is attached to burrows via
as a situation that will inevitably lead to attack. This tunnels. Rats must move from the burrows to the open arena
Social Stress: Concepts, Assumptions, and Animal Models 267

to forage. This is accomplished during the dark phase and subordinate animals indicative of hypersecretion of cortico-
provides an opportunity to engage with others or presents sterone. The high levels of cortisol were not accompanied by
a challenge to obtain food and water while avoiding others. significant changes in brain or pituitary glucocorticoid
Thus the visible burrow system provides an elegant solution receptor populations or organ weights (adrenal, thymus,
to problems posed by nocturnal animals which spend the spleen) although directional shifts suggest that even longer
majority of their time below ground. Within the visible burrow time in the burrow may have produced such effects. These
system rats can organize their daily activities in a species-typical animals did show a large reduction in CBG in subordinate
fashion closely approximating natural colonies while permit- (70%) and dominant (40%) males relative to nonsocial
ting investigators to observe virtually all social interactions. control males (Spencer et al., 1996). The drastic reduction
Small colonies formed with six rats (three males, three in CBG combined with the finding that there were fewer
females) quickly adapt daily schedules to the visible burrow Type II glucocorticoid receptors in spleen suggests dysregula-
system remaining in the burrows during the light portion of tion of HPA function, particularly in subordinate males.
the day and emerging to the open arena during the dark phase This interpretation is further supported by the finding that
(Blanchard and Blanchard, 1990). Rats do not form linear adrenalectomy, a necessary precondition for receptor studies,
dominance hierarchies, but instead a single male will emerge led to the death of 9 of 24 subordinate animals. Dominant
as dominant to the others within a few days. Dominant males and control animals did not suffer mortally from the surgical
account for 75–100% bites within the colony and engage in intervention perhaps due to sustained presence of the pool of
offensive behaviors and few if any defensive behaviors. Domi- cortisol preserved in circulation by CBG.
nant males also have freer movement within the burrow Comparison of groups that are selected for aggressiveness
system; typically emerging into the open arena first within and those that are formed by random assignment differ in
the first hour of the dark phase, they engage in more eating the response of the dominant. When groups are preselected
and drinking bouts, and in more sexual behavior. Following for aggression, the dominant male shows physiological
exposure to a predator (a docile cat) dominant males engage changes that are indicative of social stress, but not to the
in significant risk assessments – peeking out of the tunnel same degree as subordinates. This group of investigators was
and surveying the open arena, rapid forays to the corners, trying to establish animal models of human health conditions
and concomitant reductions in offensive behaviors directed that also show HPA dysregulation such as depression, anxiety,
to subordinates and sexual overtures to females (Blanchard and autism (Blanchard et al., 2013). Since the most profound
and Blanchard, 1989). Thus, it may be supposed that dominant effects of dominance/subordination only occur in groups that
rats provide an important role in risk assessment for the entire would not likely be found in nature – more males than females
colony. The tendency to emerge first into open areas is consis- in the colony and colonies composed of only the most aggres-
tent with this role, and the suppression of subordinate sive males – the translational value of studies focusing on
behavior serves to limit the risk of predation for subordinate aggression or dominance in rat colonies is minimal.
males and females while increasing risk for dominant males.
Thus, dominance in rats develops from a series of aggressive
1.09.3.2 Mice in Groups
encounters which include at least minor injurious aggression
and may be likened to a contest of strength. High status also Like rats, mice live in colonies that can number over 100 indi-
confers a role of vigilance for the entire colony. viduals and dominance relations best described as a single
Studies of the physiological response of male rats to the dominant with all others assuming subordinate status. Early
visible burrow system were conducted by placing four to studies of mice suggest that circumstances that lead to social
five males and two females into the burrow system (Blanchard instability are likely to produce greater indices of physiological
et al., 1995). In most animals this disparity in sex ratio would stress reactions than dominance relations per se (Henry et al.,
result in heightened competition among the males for access 1971). Formerly isolated animals have difficulty in meeting
to females, thus aggression in these studies would be expected demands of prolonged social interaction and failed to develop
to occur at a higher rate than would occur naturally. In each of a stable hierarchy. In a stable group, levels of wounding are
the groups a single male emerged as dominant. After 2 weeks relatively low and the dominant male has higher blood pres-
of colony living, subordinate males showed elevated cortico- sure than subordinate males. In poorly socialized groups
sterone as compared to dominant animals. If colonies were everyone has higher blood pressure.
formed using males preselected for aggressiveness, clear Stable laboratory groups were formed by simultaneously
dominance status was settled more quickly. Changes in defen- placing 5 males and 10 females (approximating the natural
sive behavior was evident on day 1, and status differences in sex bias) into a colony cage which consisted of eight cages
offensive behavior, quite variably expressed on day 1, interconnected by tunnels (Ely and Henry, 1978). Stabiliza-
emerged by day 2. Wounding was largely restricted to subor- tion of dominance relations took 45 days and was signaled
dinate males and generally was found on the less vulnerable by decrease in wounding rates and threat gestures replaced
regions of the body. Subordinate males experienced weight biting or other forms of aggression. As with rats, then, domi-
loss by day 6, and somewhat surprisingly dominant males nance status appears to be determined by a contest of strength
experienced significant weight loss by day 13. Corticosterone which is only partially ritualized. Behavior of the dominant
levels were also elevated in both dominant and subordinate male included patrolling behavior characterized by rapid
males, and subordinate males had significantly higher cortico- movement between the interconnected boxes. In contrast to
sterone levels than dominant males. Thus preselection for the antipredator behavior described for rats, the patrolling
aggressiveness resulted in changes in dominant as well as behavior of mice seems more concerned with intragroup
268 Social Stress: Concepts, Assumptions, and Animal Models

processes such as seeking and expelling competitors. This thus function to ritualize aggressive tendencies between
behavior was very frequent early following group formation potential rivals (Bernstein, 1981).
but stabilized at a much lower level (but still higher than
subordinate males) around the time wounding decreased.
1.09.3.3 Social Dominance in Primates
Tyrosine hydroxylase and PNMT (rate-limiting enzymes for
catecholamine synthesis) increased in all animals after Unlike mice and rats, formation of dominance relationships in
14 days, continued increasing until day 42, and then declined primates is not based on outcomes of contests of strength
precipitously by day 105. The increased concentrations of the (Mendoza, 1993). Studies of initial encounters in macaques
enzymes necessary for production of NE and Epi were most among previously unfamiliar, like-sex animals have shown
pronounced in dominant animals and remained higher in that while agonistic behavior is frequent initially it is highly
dominant animals even through day 105. Subordinate ritualized and orderly. Notably absent in studies of initial
animals while exhibiting substantially lower levels of tyrosine encounters is attack followed by counterattack – or actual
hydroxylase and PNMT were still elevated above control fighting. Indeed, most contact aggression is mild – grabbing,
animals living in social isolation. Evaluation of HPA activity hitting, or comprised of ritualized threat gestures such as stare
revealed that corticosterone levels in subordinate animals or open-mouth threat. This is the case if dyadic encounters
were elevated substantially at day 14, but declined precipi- occur in isolation of other social partners (Maxim, 1976) or
tously by day 42 and again slightly by day 105. Corticosterone if they occur within the context of larger group formations
levels for dominant males also showed a slight increase that (Bernstein and Mason, 1963; Hawkes, 1970). It is also not
was not observed until Day 42 and by day 105 corticosterone likely that animals assess fighting ability in their opponents
levels of dominant and subordinate did not differ and both and concede dominance in the face of obviously superior part-
were slightly elevated relative to control levels. Dominance ners. Reducing the ability of animals to predict outcome by
reversal resulted in a reversal in physiological profile as well carefully matching individuals for characteristics associated
(Ely and Henry, 1978). with status such as age, sex, weight, and aggressiveness leads
The mice in these studies did not show a pattern of physio- to less, not more aggression (Mendoza, 1993). At first both
logical activation that could simply be described as activation animals in the new dyad are equally likely to initiate agonistic
of the acute stress response to differing degrees in dominant interactions, but very quickly the individuals limit themselves
and subordinate animals. Dominant animals responded to to behaviors consistent with the dominant or the subordinate
group formation with an increased evidence of SNS activity role within the dyad – without anything closely approximating
whereas subordinate animals responded with a greater HPA a contest of strength.
activation. It was this distinction that led Henry and Stephens Group level processes are also involved in formation of
(1977) to conclude that there are two different stress- dyadic status relationships in many animals (Chase, 1984)
response profiles, one emphasizing adrenomedullary or SNS including macaques (Barchas and Mendoza, 1984). In
activity and one emphasizing adrenocortical activity. Although contrast to rats and mice, linear hierarchies are a frequent
the generality of these findings beyond mice does not suggest feature of primate societies. In triads of rhesus macaques,
that this distinction is applicable to other species, it did spawn for example, three dyadic relationships must be resolved.
a great deal of future work investigating individual differences The outcome of dyadic status resolution can be transitive
in stress responsiveness. (A / B / C; A / C) or intransitive (A / B / C; C / A).
The studies in rats and mice support the notion that domi- Intransitive or circular dominance relations would be inimical
nance status is imposed on one animal by another. In either to a linear hierarchy. A study of 13 like-sex triads of rhesus
case, fighting and wounding happens but in a kind of monkeys revealed that in both male and female triads the
controlled manner that is generally not life-threatening. sequence and direction of dyadic agonistic interactions during
A reasonable question from these studies is whether the phys- the first 45 min of initial encounters occur in an orderly
iological changes noted are due to dominance per se and how fashion which promotes linearity in the resultant hierarchy.
much is due to an untenable social circumstance. The increase Moreover, some interaction sequences are suppressed in
in sympathetic activity in dominant animals early in the group newly formed groups (Barchas and Mendoza, 1984). Given
formation might be expected to be supportive of the hypervig- an initial encounter – A threatens B, there was a high proba-
ilant state and increased activity displayed by them. The bility (90.2%) the next aggressive overture conformed to
increased adrenocortical activity of the subordinate males is one of three sequences: A threatening B again (Repeat), A
a response similar to the intruder’s response in the resident/ threatening C (Double Attack), or C threatening B (Double
intruder paradigm and may be a consequence of directed Receive). Each of these patterned sequences support the
aggression. formation of transitive relationships. Surprisingly, three
Traditionally, it has been assumed that the formation of possible sequences were infrequent (9.8% of interactions): B
dominance relationships is a simple case of mutual shaping. threatening A (Reverse), C threatening A (Attack the Attacker),
These aggressive encounters, typical of rats and mice during or B threatening C (Pass-on). These interaction sequences do
initial stages of group formation, are likened to contests of not inevitably lead to intransitive patterns of interaction, but
strength. The loser of fights learns that the strength and they have a 50% theoretical probability of doing so. The
fighting ability of the other supersedes its own. The subordi- predominance of Repeat, Double Attack, Double Receive
nate role, according to this view, is established when the patterns of interaction held for all behaviors including those
loser responds at the outset of each agonistic or competitive that are not considered to be concerned with dominance
encounter with submissive responses. Dominance relations interactions such as sniffing or grooming. It should also be
Social Stress: Concepts, Assumptions, and Animal Models 269

noted that the patterning of successive acts according to the With regard to the SNS similar differences were found.
Repeat, Double Initiate, and Double Receive patterns need Dominant males had the largest and fastest response to Epi,
not imply that dyadic interactions are unidirectional. For greatest HR increase and peak systolic pressure, and the most
dominance and submissive behaviors, directionality was not rapid recovery (Sapolsky and Share, 1994). Interestingly,
immediately apparent, but emerged in most dyads by the administration of an alpha blocker (proterenol) did not
end of the 45-min observation period, and all triads presented equalize the differences in blood pressure nor did administra-
a transitive hierarchy. tion of a beta blocker eliminate status differences in HR.
As relationships in macaques stabilize, patterns of interac- Administration of metyrapone which blocks synthesis of
tions change somewhat and the frequency of redirected aggres- cortisol did eliminate status differences in both blood pressure
sion (Pass-on pattern) and defending an ally (Attack the and HR. Thus it appears that status differences in HPA function
Attacker) increase and become a part of the rich repertoire of has an impact on the actions of the SNS and confers a physio-
expressing and controlling aggression in groups (Chase, logical advantage for dominant individuals.
1984; de Waal, 1993). For large groups it is worth noting Other glucocorticoid-related differences between dominant
that dominance relations are most commonly formed in and subordinate baboons include diminished high-density
within-sex dyads and not in male–female dyads and that lipoprotein cholesterol with elevated cortisol levels character-
a middle ranking animal performs the dominant role with istic of subordinate monkeys (Sapolsky and Mott, 1987).
some group members and the subordinate role with others. Likewise, circulating lymphocytes are higher in dominant indi-
Examination of physiological correlates of dominance viduals but were reduced to the same level as seen in subordi-
status reveals considerable species variability in studies of nate males when cortisol levels were pharmacologically raised
nonhuman primates (Abbott et al., 2003). Nonetheless where in dominant males to that observed in subordinate males
differences are found it is generally in the direction of domi- (Sapolsky, 1993). One rank-related physiological effect that
nant animals exhibiting lower cortisol and lower indices of was not attributable to differential glucocorticoid levels was
SNS activity than subordinates. insulin-like growth factor which is suppressed in subordinate
monkeys (Sapolsky and Spencer, 1997).
1.09.3.3.1 Case Study: Physiological Processes Associated All of the above studies with baboons were conducted
with Dominance in Baboons during periods of social stability when rates of aggression and
The most detailed studies regarding the physiological under- other indices of dominance status were relatively low. Sapolsky
pinning of dominance are provided by studies of free-ranging also studied his troop of baboons during periods of instability
baboons (Papio anubis) (Sapolsky, 1993). Baboons live in large – following ousting of a dominant male by a cohort of young
social groups, and as is the case with most species of primates males who themselves failed to establish a clear hierarchy once
living in large groups, group members can be ranked within the dominant animal was gone. With social instability the
a pecking order or dominance hierarchy. Using darting tech- dominant animals lost the physiological benefit of high status,
niques to anesthetize baboons and using elegant physiological and their HPA system looked like that of subordinate males
probes Sapolsky has demonstrated status includes physiolog- (Sapolsky, 1993). When a highly aggressive male entered the
ical differentiation of HPA and SNS systems. In his initial troop he became dominant, but unlike previous, less aggressive
studies, Sapolsky (1982, 1989) found that dominant males dominant animals in the troop, the aggressive dominant
have lowest initial concentrations of plasma cortisol and the male had the highest cortisol and lowest lymphocyte levels
most rapid rise in circulating levels in response to the stress (Sapolsky, 1995).
of darting and anesthetization. Administration of labeled Individual differences in sociability also altered status
cortisol (3H-cortisol) indicated that dominant and differences in HPA regulation. A subset of subordinate males
subordinate were equally efficient in metabolizing and had high rates of prolonged association with estrous females,
removing cortisol from the circulation. The low HPA activity or consortships, comparable to dominant males. These subor-
characteristic of dominant individuals was due to a low dinates also had larger stress responses, similar to that
secretion rate of cortisol rather than enhanced clearance rates described for dominant males, but they also exhibited elevated
(Sapolsky, 1983). The possibility that the adrenal of basal cortisol typical of a subordinate male. Similarly, furtive
subordinate animals is more sensitive was eliminated by the males with high rates of stolen copulations did not show
comparable response of dominant and subordinate animals elevated basal cortisol, and subordinate males who initiated
to ACTH. Paradoxically, subordinate animals were found to aggression or who exhibited redirected aggression (usually
be less sensitive to CRH, suggesting that central mechanisms threatening a bystander) also show lower cortisol typical of
are even more active in stimulating HPA activity in a dominant individual (Virgin and Sapolsky, 1997). As noted
subordinate than in dominant animals (Sapolsky, 1989, above redirected aggression is virtually nonexistent during rela-
1990). Dexamethasone, a synthetic glucocorticoid that can be tionship formation when all strangers meet for the first time.
biochemically distinguished from cortisol, but presents a very The data from Sapolsky’s studies show that redirected aggres-
potent negative feedback signal to the HPA system, was more sion does occur in stable groups and that it also can serve
effective in suppressing endogenous HPA activity in a protective function for the animals that engage in this
dominant animals than in subordinate animals (Sapolsky, behavior pattern.
1983). In short, dominant animals have an efficient HPA A particularly interesting set of changes in one troop of
system in which basal levels of HPA activity are relatively baboons occurred when an outbreak of bovine tuberculosis
low, and the response to acute stressors is rapid and rapidly from spoiled meat at a garbage dump led to the death of all
terminated. adult males that frequented the site, but spared those that
270 Social Stress: Concepts, Assumptions, and Animal Models

did not (Sapolsky and Share, 2004). Aggressiveness of animals the lowest cortisol level (Mendoza, 1991). Cortisol levels for all
competing for food at the garbage site was much higher than males, including the alpha male, were still lower than individ-
aggressive tendencies in the animals that did not visit. The ually housed males.
result was a baboon troop comprised of the least aggressive Although the methods used to perform group formations,
males. When new males transferred to the troop, the lower collect blood samples, and cage configurations were similar
levels of aggression and higher levels of affiliation were main- between the studies, several differences in subjects and
tained well after the troop consisted only of new males. This methods could account for the discrepant results. One such
altered pattern of interactions among troop members did not difference is subspecies variation. Male Bolivian squirrel
alter the overall composition of the new troop nor the expres- monkeys, the subject of the earlier studies (Coe et al., 1979;
sion or stability of the hierarchy. The subordinate males in the Mendoza et al., 1979), are considerably larger than other
new troop, however, no longer exhibited hypercortisolemia. subspecies, and they are dominated by the much smaller
These studies in baboons illustrate a few principles perti- females acting in concert to displace males from preferred
nent to the study of social stress. The social system is capable resting sites, which is not the case for other subspecies
of exerting an influence on the physiology of its members. (Mendoza et al., 1978; Lyons et al., 1992). Cortisol concentra-
Whether the society is stable or unstable, more aggressive over- tions are overall higher for Bolivian males, and their HPA
all or more affiliative, all contribute to rank-related differences system is more reactive than other subspecies of squirrel
in stress physiology. When cortisol levels are sustained at high monkeys (Coe et al., 1979, 1978). Perhaps the most influential
levels, there are strong reasons to suppose that they extract difference between the studies was timing relative to the
a cost to the animals in terms of physical health and probably breeding season. In the Mendoza et al. (1979) study, group
psychological well-being (Sapolsky et al., 2000). We also know formations were timed to occur 6 months out of phase with
from this body of research that heightened cortisol is not the breeding season; group formation induced breeding in
simply the result of being subordinate even within the same each of the three groups. In contrast, the triad formations in
baboon troop. Highly aggressive animals, even though domi- the Lyons et al. (1994) study occurred just prior to the breeding
nant, exhibit heightened cortisol and highly affiliative animals season and by 3 months following formation of male triads
may not, even though subordinate. It is also important to colony females were cycling (Mendoza, 1991). Perhaps differ-
note that even in the troops with stratified cortisol related to ential HPA activity only occurs in new groups in which
dominance status, high or low position in hierarchy is not breeding is also happening. Heterosexual groups were formed
a life-long commitment. Male tenure in a given group, and 5–7 months following triad formation. At this time most
hence occupation in a given position in the hierarchy, is rela- colony females had ceased cycling. Male cortisol was not
tively short, generally a year or two. Therefore, it is entirely altered by the addition of females to their groups and breeding
likely that adverse consequences of heightened cortisol levels was not induced. Heterosexual group formation, however, did
when they occur are not sustained long enough to cause irrep- affect the timing of the subsequent breeding season which
arable damage. occurred 12 months following group formation, considerably
later than that predicted by timing of heightened gonadal
1.09.3.3.2 Case Study: Social Influences on Stress Response hormone activity in the previous year (Schiml et al., 1996).
Systems in Squirrel Monkeys Female squirrel monkeys also show a reduction in cortisol
Studies of captive squirrel monkeys present a quite different upon formation of female triads (Mendoza and Mason,
picture than Sapolsky describes for wild baboons. When 1991). The reduction in cortisol did not happen right away.
rank-related differences in HPA activity are found, they occur Cortisol levels 24 h following triad formation were comparable
in newly formed and relatively unstable social groups. Domi- to cortisol titers when females were housed individually. In this
nant male squirrel monkeys in newly formed same-sex dyads study a single male was added to each female triad, and
or triads exhibited the highest levels of cortisol and subordi- breeding was induced within the first week of heterosexual
nates exhibited the lowest (Coe et al., 1979; Mendoza et al., group formation. Somewhat surprisingly, cortisol levels
1979). Examination of hormonal change from living alone to remained low in the females in spite of all females undergoing
living in male triads to living in male–female groups (through massive changes in gonadal hormone activity. Cortisol levels
the addition of five females to each of three male triads) sug- do show circannual changes (Schiml et al., 1996). For females
gested that each stage of group formation was responded to living in established groups, elevations in cortisol coincide with
by an increase in plasma cortisol that was greatest in the the onset of ovarian cyclicity typical of the breeding season. In
most dominant male, but in this study dominance effects males the seasonal elevation in cortisol coincides with annual
were not significant until the females were added to the male weight gain – the so-called fatting response – both of which
triads. It is not always the case that male squirrel monkeys peak in the month prior to seasonal elevations in testosterone.
respond to group formation with elevated HPA activity. In Both males and females show clear and linear within-sex domi-
one study, formation of male triads did not result in an imme- nance relations based on the direction and frequency of
diate rank-related difference in cortisol, but simultaneous agonistic interactions; neither males nor females exhibit rank-
introduction of males, all strangers to one another, in a novel related differences in cortisol levels or in the magnitude and
cage led to an overall reduction in cortisol in all group timing of circannual changes once the groups have stabilized.
members (Lyons et al., 1994); 3 months following triad forma- The reduction in cortisol upon the formation of like-sex
tion cortisol levels did reflect dominance status, and once groups of squirrel monkeys could suggest an alleviation of
again, the dominant male had significantly higher cortisol level stress associated with isolation. This seems unlikely for several
than the other two males and the most subordinate males had reasons. First, the reduction takes time to develop. In our
Social Stress: Concepts, Assumptions, and Animal Models 271

studies, more than 24 h and less than 6 days is required for One notable absence in all of our studies of group forma-
cortisol levels to reach their socially induced low level. This tion in squirrel monkeys was any evidence of dominance
slowly developing time frame in response to separation and contests. Dominance relations were detectable early following
pair formation is of the type delineated by Hofer (1987) for relationship formation in both males and females. In all of
removal or provision of homeostatic regulation possibly the group formations conducted in squirrel monkeys only
induced by a shift in receptor populations. Once developed one reports any instances of actual fighting (Gonzalez et al.,
the hypocortisolemia is apparent at all points in the circadian 1981). That fighting was severe is evidenced by the fact that
rhythm (Mendoza et al., 2000). Secondly, the acute responses the most subordinate males in two of four groups in this study
to isolation and pair formation are distinctly different than died within a week of group formation. Why severe fighting
the prolonged effects (Mendoza et al., 1992). Female squirrel occurred in this study and fighting rarely, if at all, in other
monkeys, living in established female–female pairs, responded studies is related to methods employed in forming groups. In
to separation from their cage mate with reduced cortisol 1 h the Gonzalez et al. study heterosexual groups were formed by
later; by 6–8 days following separation cortisol levels were combining subgroups that were familiar with one another,
significantly elevated. Subsequent formation of new female some of which were living together and transferred as a group
dyads resulted in further cortisol elevations at 1 h followed to the new heterosexual group. These are the conditions
by significant reductions 6–8 days later. Thus, long-term conse- that lead to increased aggression during group formations
quences of separation and pair formation are opposed to the (Bernstein et al., 1974).
acute response to these social manipulations and cannot be In combination the studies of dominance and stress-
considered an extension or continuation of the acute response. response systems reveal some generalizations and lack thereof.
Finally, cortisol levels in established groups do not differ from Fights are rare. Contrary to popular assumptions, even in
animals housed individually (Schiml et al., 1999). Following newly formed groups dominance relationships are established
the increase in cortisol associated with the first full breeding quickly without contests of strength. Once established domi-
season as cortisol declines to nonbreeding levels, the social nance relationships are stable. Subordinate animals are not
effect on HPA activity disappears and does not reemerge in continually striving for dominance. In contrast to mice and
stable groups. rats, the dominance relations in most polygynous primates
HR in male squirrel monkeys shows a similar reduction are linear, implying some group level processes control the
upon formation of new relationships (Mendoza, 1991). Within behavior of the individuals therein. Early in primatological
the first hour of pair formation HR was considerably reduced research, KRL Hall (1964) suggested that aggression within
compared to HR in the same testing situation while living primate societies is most often the result of violating social
alone. Following 2–3 weeks of continuous cohabitation, HR order either by acting at variance with their status and role
was further reduced. It is possible that the reduction in HR within the group or in response to attempts to gain access to
was attributable partially to sympathetic withdrawal, but the the group by outsiders. As Sapolsky’s work, reviewed above,
evidence suggests that the inhibitory influence of parasympa- suggests there are occasions when the subordinate animal
thetic activity was more influential. HR variability, a measure displays a physiological profile consistent with a chronically
of parasympathetic activity, increased with pair formation stressful state, which if sustained might result in health prob-
and, like HR, was further increased after the males had lived lems. It is also the case that dominant baboons can exhibit
continuously together. Female squirrel monkeys, tested simi- the same stress-related pathologies during times of instability
larly, also showed the lowest HR when living in established or that all animals are saved from experiencing chronic stress
pairs, but this was not significant nor was there any indication effects if the larger group culture is one characterized by higher
of enhanced parasympathetic activity (Saltzman et al., 1991). than usual affiliation and lower conflict. How dominance
This finding of increased parasympathetic activity in male affects stress response systems is species specific. Physiological
squirrel monkeys is particularly interesting since squirrel differentiation of status in squirrel monkeys when it appears
monkeys living without like-sex companions show very little suggests the dominant male is most at risk, whereas in
parasympathetic activity in the regulation of HR (Mendoza baboons the subordinate male is most at risk. Rank-related
and Mason, 1997). differences in stress physiology in squirrel monkeys are also
In summary, the hypocortisolemia characteristic of newly transitory, appearing only in situations in which social insta-
formed squirrel monkey groups is evident only during the bility and breeding readiness coincide, and thus unlikely to
period following group formation and preceding the first pose risks to health. Social instability, rather than rank, in
breeding season of the new group. At least the initial a well-ordered hierarchy appears to be key in understanding
response to formation of within-sex relationships is also social stress.
accompanied by an increase in parasympathetic activity in
males but not in females. The tendency to ascribe the socially
induced reduction in cortisol to something akin to stress 1.09.4 Social Instability
buffering does not conform to the data. Instead the group
formation studies in squirrel monkeys suggest a chronic The impact of social instability on stress response systems and
response to the stress of encountering strange animals for health is provided by a study of rhesus macaques (Capitanio
the first time, but it is in the opposite direction from that et al., 1998). In this study male rhesus monkeys were allowed
usually considered indicative of chronic stress. The question social interaction for 100 min day 1 for 3–5 days week 1. All
of greatest import is whether the reduced cortisol is beneficial other times the animals were housed individually, but within
or detrimental to health. sight and sound of other monkeys. During the social
272 Social Stress: Concepts, Assumptions, and Animal Models

interaction periods animals were either placed in triads with of HPA activity following dexamethasone administration
the same other animals each day (Stable condition) or they (Capitanio et al., 1998). The lower cortisol levels exhibited
were placed in groups of two to four with the identity of the by squirrel monkeys and rhesus macaques during periods
other group members changing from session to session of social instability are reminiscent of findings of patients
(Unstable condition). The rationale for this study was that in suffering from posttraumatic stress disorder (PTSD)
the Stable group, in spite of relatively limited daily interaction, (Daskalakis et al., 2013). Also similar to PTSD (Yehuda
the animals would quickly learn the identity and proclivities of et al., 2004), there appears to be a heightened sensitivity of
one another and form stable relationships. The Unstable GR receptors leading to increased negative feedback evident
groups would afford the same amount of interaction time for both in the unmanipulated HPA system and in the HPA
each male but since formation of stable dyadic relationships response to dexamethasone suppression.
is facilitated by stability at the group level, it was reasoned PTSD does not occur in all individuals exposed to a trau-
that males from the Unstable group would not be able to matic stressor, and work is beginning to suggest that ante-
form stable relationships. Instead, each time they met a new cedent to a PTSD-inducing trauma, differences exist which
social order, in particular new dominance relations, would increase the risk of developing PTSD (Yehuda and Bierer,
need to be established. After three interaction days half of the 2008; Yehuda et al., 2002). The possibility that individual
animals were inoculated with simian immunodeficiency virus, differences in genotype or personality contribute to stress-
SIVmac251, and stress-related physiology and disease progres- induced susceptibility to SIV was investigated in macaques.
sion were monitored. Early in the disease process as the immune system is
Aggression was rare in all groupings, but males from the activated to contain the virus and the virus is replicating
Unstable condition were more likely to be the recipients of rapidly, the two biological processes reach a set point in
threats, contact aggression, and chasing; males in the Stable which viral replication is maintained at a static level
condition were not observed to engage in either physical throughout the remainder of the infection until the
aggression or chases. Animals in the Unstable condition development of full-blown AIDS immediately preceding
exhibited a more rapid disease course and a shorter survival death. Viral set point is established 6 weeks postinfection,
time than animals in the Stable condition. The ability to mount is measured by viral RNA (vRNA), and is the single best
an antibody response to the virus was significantly related to predictor of survival. In two studies, for example, survival
survival. However, animals experiencing stable social encoun- duration was correlated .79 and .73 with viral set point
ters did not differ in formation of SIV-specific antibody from (Capitanio et al., 1998; Capitanio et al., 2008). Immune
males experiencing unstable social encounters. Therefore, processes early in infection include interferons, produced by
antibody formation could not account for social condition infected cells within hours, and their effector molecules
effects on survival. whose effects on viral replication are processed via receptors
As with squirrel monkeys during the initial period following on other immune cells. Each of these processes influences
triad formation, cortisol levels declined for male rhesus viral set point, and each is influenced by behavior and
monkeys in both the Unstable and Stable social conditions behavioral tendencies created by personality and genetic
during the first 12 weeks of social experience. After this point polymorphisms (Capitanio et al., 2008).
cortisol levels for monkeys in the Stable condition recovered, Specifically, animals were selected as subjects based on
whereas cortisol levels in the monkeys experiencing unstable a personality assessment conducted with animals in their natal
social conditions continued to decline through 24 weeks of field cages. Males scoring in the highest or lowest third of the
testing. After this time the cortisol levels for males experiencing colony on sociability, a trait associated with affiliative tenden-
Unstable social conditions rose once again, and cortisol no cies (Capitanio, 2004), were tested for their allelic variation of
longer differed between Stable and Unstable social experiences. the promoter region for the serotonin transporter (5-HTTLPR).
Thus monkeys in both conditions experienced an initial drop A short allelic variant, is associated with aggressiveness, anxiety,
followed by recovery in basal cortisol levels, but for animals and neuroticism (Lesch et al., 1997; Barr et al., 2003; Caspi and
in the Unstable group the decline in cortisol was greater and Moffett, 2006). Animals received socialization opportunities
persisted for a longer period of time. Blood samples were for 100 min day 1, 3–5 days per week. Triads of animals in
collected in this study on days when no social interaction the Stable social condition met with the same animals each
testing occurred so the changes in cortisol are in response to socialization period, whereas animals in the Unstable condi-
long-term socially induced alterations in HPA regulation and tion met with animals of varying number (2–4) and identity.
not an acute response to social experience. Perhaps because All groupings were performed within personality type (High
of the intermittent nature of social encounters in the rhesus or Low Sociable) and all were inoculated with SIV following
or perhaps due to known species differences in HPA regulation three such pairings. None of the categorical variables in this
(Mendoza and Moberg, 1985), the time to fully develop hypo- study (social condition, high or low sociability, or short versus
cortisolemia in the rhesus monkeys (weeks) was considerably long 5-HTTLPR) directly influenced viral set point. However,
longer than in squirrel monkeys (days). Nonetheless, survival each was found to influence viral set point indirectly largely
differences between Stable and Unstable animals in the face through altering actions on IFN-a which is more directly
of a lethal virus implicates low cortisol as a health risk or at involved in establishing viral set point. Animals in the Unstable
the very least associated with a more lethal physiological state. group exhibited sustained aggression and reduced affiliation.
The reduction in cortisol during the early weeks of stable Sustained aggression performed predominantly by animals in
and unstable conditions was accompanied by a dampened the Unstable group with the short 5-HTTLPR genotype was
HPA response to physical restraint and a greater suppression associated with decreased expression of CXCR3 (receptor for
Social Stress: Concepts, Assumptions, and Animal Models 273

both IFN-a and IFN-g effectors). Recipients of sustained aggres- Urinary analysis of catecholamine metabolites indicated that
sion had reduced SIV antibody (anti-SIV IgG). Sustained social instability was associated with higher catecholamine
submission, displayed most prominently by Low Sociable output. Enhanced expression of proinflammatory genes
animals, was correlated reductions in cortisol and with (IFN-g, IL-6, and TNF-a) was induced with social instability.
increased ISG (an IFN-a effector). Aggression initiated in Stable Behavioral, endocrine, and immune consequences of social
groups, performed by animals with long 5-HTTLPR genotype, instability were reversed when social stability was reinstated.
was associated with increased ISG activity. Reduced anti-SIV Switching from Stable to Unstable social conditions did not
IgG, increased expression of ISG, and decreased expression of affect measures of disease progression – anti-SIV IgG increased
CXCR3 were all related to increased viral set point and thus at each stage of the study and vRNA did not change.
point to a more rapid disease course. Interestingly the reduc- In another study (Capitanio and Cole, 2015) administra-
tions in cortisol found earlier to be related to survival was tion of methamphetamine to animals socialized in Stable or
only found to be related to increased ISG levels and to sus- Unstable social conditions was used to enhance the apparent
tained submission. Whether hypocortisolemia is an inter- SNS contribution to viral set point. Interestingly, administra-
vening variable between the association of submissive tion of methamphetamine did not affect lymph architecture
expression and viral set point is not known. It is clear, however, even though clear elevations in plasma EPi and NE were
that HPA function is not clearly related to establishing viral set accomplished. Animals from the Unstable social condition
point, the best predictor of disease course available during early exhibited greater innervation of lymph tissue than animals in
stage disease. Low levels of cortisol may, however, permit many the Stable condition and neither were different whether meth-
disease processes to go unchecked. amphetamine was administered or not.
In addition to low cortisol, PTSD is also characterized by It is clear that the stress response systems, HPA and SNS, are
high levels of catecholamines, CRH, and proinflammatory both altered by the same circumstances – social instability –
cytokines suggestive that the low cortisol is insufficient to that alter disease processes and the body’s effort to combat
inhibit these aspects of stress response (Daskalakis et al., them. It is less clear that measures of global activity as repre-
2013). In keeping with this suggestion, inquiry into the role sented by circulating levels of the hormones of each system
of SNS in understanding differences in SIV disease progression can be linked to specific disease processes. For the SNS system,
between animals in Stable and Unstable social conditions was in particular, local changes in lymph tissue appear to be altered
conducted. Examination of neural fibers enervating lymphoid by social conditions which, in turn, alter the ability to deal with
tissues suggest a role for SNS in altered disease progression. At disease processes independently of circulating hormone levels.
37 weeks postinoculation, lymph nodes for 20 animals were It is also clear that the HPA response to social instability in
biopsied. Replication of the SIV virus, as indicated by local squirrel monkeys and rhesus macaques results in lower circu-
SIV RNA expression, was nearly four times higher in proximity lating levels of cortisol than in social contexts that enable
to axonal varicosities of sympathetic fibers (Sloan et al., animals to form stable social relationships. Thus in both
2006). Although the number of sympathetic fibers was systems, looking for consequences of social stress can be
reduced in SIV-infected animals, for surviving animals of the misleading, in the SNS because tissue effects do not mimic
Unstable social condition innervation was greater than for circulating hormone levels and in the HPA system because
animals in the Stable social condition and viral replication our tendency is to use high cortisol as a marker for what is
was nearly 60% greater for Unstable males (Sloan et al., and what is not stressful.
2007). The enhanced SNS innervation of lymph tissue was
associated with altered interferon activity – reduced INF-
b and increased INF-a. Both greater innervation and altered 1.09.5 Disruption of Established Relationships:
INF expression account for enhanced SIV gene expression. Separation, Bereavement, and Loneliness
Social instability was also associated with increased expres-
sion of neurotrophic factors, BDNF and NGF, in lymph tissue Structured and recurrent interchanges between individuals
(Sloan et al., 2008b). NGF was particularly influential, within relatively stable social units are a general feature of life
inducing more SNS innervation providing more varicosities for social mammals. When such interchanges occur regularly
and greater local release of NE in Unstable animals, which and predictably, it is customary to refer to them as relationships
moderated the INF response ultimately increasing viral repli- (Hinde, 1976, 1983). Another aspect of social mammals is that
cation (Sloan et al., 2008a). each species tends to display characteristic modal form of social
The role of the SNS in mediating the enhanced SIV disease organization. At a gross and superficial level this is reflected in
progression with social instability was examined in another set the size and composition of the group. Social organization is
of studies (Capitanio and Cole, 2015). Selection of animals for generally considered to reflect the combined nature and quality
this study balanced groups for sociability. Animals were social- of relationships among the members of the group (Hinde,
ized in stable groups (male triad with same animals each day) 1976; Mendoza, 1984). This is not to say that relationships
for 5 weeks, inoculated with SIV, followed by 5–15 additional create the existing social structure. The most obvious evidence
weeks of stable groupings, then switched to 5 weeks of unstable that relationships alone cannot account for group structure is
groups in which animals met in groups of two to four with that for most social systems the group structure is defined in
number and identity of interactants changed daily. Animals part from the individuals excluded from the core of social inter-
were then replaced in their original stable groups and testing actions. One-male groups or monogamous groups, for
continued for at least 5 weeks. When in Unstable groups, males example, are defined by the absence of potential relationships
groomed less, spent more time alone, and aggressed more. as much as those which are actually present. Comparative
274 Social Stress: Concepts, Assumptions, and Animal Models

studies aid in understanding generic aspects as well as species- involuntary separation of 3-month-old infants from their
typical variants in relationships. mothers elicits an increase in plasma cortisol of up to 100%
As indicated in the previous section, formation of new of baseline values in both mother and infant within 30 min,
relationships is a prolonged process and is accompanied by and cortisol levels return to baseline levels 1–6 h following
periods of vulnerability, behaviorally and physiologically. reunion (Mendoza et al., 1980). When infants are cared for
Cooperation or protection is as much of a mainstay of social by a familiar adult female during separation from the mother,
life as competition. What happens when stable relationships they show as great an increase in cortisol as when they are left
are disrupted? Some of the answer to this question depends alone. One of the features of the above separation studies
on the nature of the relationship and the society within which includes a disturbance control condition. Establishing this
the relationship is embedded. Removal of a bully might be condition includes capture of the pair, brief separation, and
responded to positively by other group members. Removal immediate return to familiar living conditions. This procedure
of a mother can have devastating consequences for the infant, was anticipated to elicit a modest stress response albeit not as
even for infants beyond the stage of nutritional dependency. substantial as separation per se. Indeed, capture and handling
Friendships describe a tendency to affiliate with particular have been used as a stressor to elicit an acute stress response
others and to engage infrequently with others – a common (Mendoza et al., 1978). Surprisingly, neither mother nor infant
feature of large polygynous groups. Do these friendships shows a response to the disturbance control procedures. This
confer behavioral or physiological advantages? Virtually finding has been used to argue that the counterpart to the sepa-
all adult humans have experienced a friend moving to ration response is stress buffering.
another geographical location preventing regular interaction. The acute pituitary–adrenal response to disruption of the
Is the response to loss of a friend of many years at all compa- mother–infant bond is remarkably resistant to habituation.
rable to separation from a lover even if the relationship is Hennessy (1986b) found that over the course of 80 mother–
relatively new? infant separations conducted between 12 and 31 weeks of
age, each 2-h separation reliably elicited an increase in cortisol
levels. Inasmuch as basal cortisol levels in these repeated sepa-
1.09.5.1 Comparative Case Study: Polygynous Squirrel
rated infants remained unchanged and did not differ from
Monkeys versus Monogamous Titi Monkeys
basal values in nonseparated control infants. These recurrent
Titi monkeys live in family groups consisting of an adult male reactions to mother–infant separation in squirrel monkeys
and female and one to three offspring (Mason, 1966). The occur around a roughly stable physiological set point – regula-
adults are monogamous, and like-sex encounters are generally tion of the HPA system was not altered. The HPA response to
restricted to highly ritualized territorial displays at the separation for infant squirrel monkeys does not occur if infants
boundary of adjacent territories (Van Belle et al., 2016). Fathers are within sight of their mother but unable to contact her (Vogt
provide the majority of infant care, typically infants transfer and Levine, 1980). Behavioral agitation, however, is most
from the father to the mother for brief periods surrounding extreme under these circumstances. When visually isolated
nursing (Mendoza and Mason, 1986b). from mother, the cortisol response to separation is sustained
Squirrel monkeys, in contrast, live in large groups and, for at least a week, in spite of a lack of sustained behavioral
with the exception of infants, primarily associate with agitation and an absence of depressive symptoms noted under
animals of the same age and sex. Females tend to cluster similar circumstances in macaques (Coe et al., 1985).
into smaller social units or cliques and may act in concert These findings, together with data from other species,
with clique members to actively exclude other group confirm the initial interpretation the effective social dimension
members, including males (Leger et al., 1981), and are often triggering the acute physiological response to separation is not
credited with maintaining the sexually segregated social removal of maternal behaviors, but rather it symbolized the
structure (Mendoza, 1991). Interactions among females disruption of a well-integrated and specific psychosocial attach-
within a clique are generally affiliative, but agonistic interac- ment between mother and infant (Hennessy, 1997). Many
tions are also more frequent within cliques. Membership in investigators proceed as if all affinitive social relationships
cliques is transitory, and changes in clique size and member- resemble the attachment bond in form, structure, and conse-
ship are frequent in laboratory populations and field data quences, differing only in the intensity of participant involve-
indicate that females periodically transfer between groups ment. Surprisingly, this is not the case.
(Boinski, 1987). Male relationships are generally less affilia- Our initial study designed to extend our findings to other
tive than female relationships but are more stable. Both relationships focused on the response of female squirrel
males and females form within-sex linear dominance hierar- monkeys to involuntary separation from like-age, same-sex
chies that are more clearly demarcated among males companions, relationships which might be expected to
(Mendoza et al., 1978). Infants are cared for almost exclu- conform most closely to mother–infant relationships
sively by mothers, although occasional ‘aunting’ by other (Hennessy et al., 1982). To our surprise, the results indicated
adult females occurs (Mendoza, 1991). that juvenile females did not show a specific response to sepa-
Social separation studies have helped to elucidate the rela- ration from isosexual companions, nor did familiar compan-
tional dynamics in these species. The first studies of social sepa- ions reduce the adrenocortical response to stressful events
ration focused on the HPA response and the mother–infant such as capture, handling, or exposure to a novel environment.
relationship (Mendoza et al., 1978). Mother and infant squirrel These findings were extended to adult females removed from
monkeys react to the disruption of their relationship with large group cages and placed in novel cages with a preferred
a substantial and rapid increase in HPA activity. Brief or not preferred female (Hennessy, 1986a). The HPA response
Social Stress: Concepts, Assumptions, and Animal Models 275

of adult females to a novel environment was not influenced by Surprisingly, the only time we have found the response to
the presence of a familiar like-sex cage mate, and the degree of parental separation in young adult titi monkeys truncated by
affiliation displayed by the companions in the home cage did anything other than reunion with the father was if they were
not influence the response to a novel environment. Similarly, provided a potential mate (Mendoza et al., 2000). In the
disruption of established heterosexual relationships does not process of forming 10 new pairs, most of the new partners
induce behavioral or physiological indications of stress (six males and seven females) were removed from their natal
(Mendoza and Mason, 1986a). groups and hence experienced separation from their fathers
Similar studies in titi monkeys, some of which were per- (and other family members) immediately prior to pair forma-
formed in direct comparison with squirrel monkeys, show tion. Blood samples were collected twice per week for 3 weeks
quite different patterns of responses to separation from group before and 4 weeks following new pair formation. Cortisol
members. Adult males and females living in established hetero- levels were elevated 24 h following pair formation but there-
sexual pairs respond to separation from their mate with a rapid after returned to preseparation levels. The response to the
and substantial increase in HPA activity (Mendoza and Mason, formation of new pairs was the same for monkeys who were
1986a,b). When parental response to separation was tested, removed from their natal groups and for monkeys who were
neither titi monkey mothers nor fathers responded to removal not currently living with either a mate or their parents at the
of their infant. Disturbance control procedures are not different time of pair formation. We do not know from these studies
from basal values of cortisol and for parents not different than when titi monkeys would begin to exhibit a separation
when separated from their infant. Infants respond to separation response from their new mate, but clearly a new mate, even
from their fathers even with their mother present; when sepa- though a stranger, is capable of buffering the potent parental
rated from both parents (remaining alone in the home cage), separation response. Further underscoring the unique response
cortisol levels for infant titi monkeys reach their highest levels of the young adult titis to pair formation, is the finding that for
(Hoffman et al., 1996). For infants, removal of the mother does animals living in established pairs provision of a potential
not elicit a separation response as long as the father is present. mate does not ameliorate the response to separation from
Titi monkeys continue to respond to separation from their the actual mate and may even augment it (Cubicciotti and
fathers into adulthood and do so with prolonged activation Mason, 1975).
of the HPA axis (Mendoza et al., 2000). The response to sepa- There is no indication that the persistent elevations in
ration was compared in infant (6–12 months of age), juvenile cortisol pose a health risk in titi monkeys. It is likely that the
(1–2 years of age), and adult offspring (2–3 years of age). sustained cortisol in response to separation facilitates the
Cortisol concentrations were determined from blood samples motoric output deemed necessary to find the object of attach-
collected on day 1 and day 5 of separation from the parents. ment or a replacement. Titi monkeys are one of the truly
The results from the separation condition were compared to monogamous primate species. It is likely that the unique
a separation–reunion condition in which the parents were response to social separation supports their equally unique
separated from their offspring for 1–2 h the day before the first social system. Other species for which paternal behavior is
blood sample was collected. All offspring responded to separa- common occasionally have nonrelated adults or adult
tion from their parents with an increase in plasma cortisol offspring contribute to infant care, neither of which are avail-
levels, even though all were tested in the familiar home cage able for titi monkeys (Fernandez-Duque et al., 2009; Van Belle
and 20 of 21 subjects were housed with siblings throughout et al., 2016).
the separation period. The separation response was sustained Separation studies have been a critical element in
through the 5-day period without attenuation. In addition to categorizing relationships as attachment bonds or friendships
demonstrating that the response to parental separation (Hennessy, 1997; Mendoza et al., 1980; Mendoza, 1991). In
continues into adulthood, these data importantly show that addition to evidence of agitation upon separation, recognition
stress buffering for titi monkeys is provided only by parents, of and selective preference for the object of attachment, prox-
and together with the results from the previous studies only imity seeking, and stress buffering which is expressed behavior-
effectively by the father. ally by using the object of attachment as a secure base are all
Further extensions of the separation response in adult titi important elements of the attachment relationship in human
monkeys were performed in order to determine if longer time- and nonhuman primates (Ainsworth, 1972). In squirrel and
frames would lead to an attenuated response and if reduced titi monkeys several relationships meet the criteria for attach-
cortisol would be reinstated after a prolonged absence from ment. These include the mother–infant relationship in squirrel
the family group (Mendoza et al., 2000). Adult daughters monkeys (bidirectional); the pair bond of adult titi monkeys
were removed from their groups and placed in a new cage (bidirectional); and the relationship of young titi monkeys to
(identical to the home cage) in the same colony room for the father (unidirectional). Relationships that are affiliative
30 days and then reunited with their families and monitored behaviorally but do not meet the criteria of an attachment
for the ensuing 30 days. Blood samples were collected at weekly bond include female–female relationships in squirrel
intervals in both phases beginning 24 h following separation monkeys; male–male relationships in squirrel monkeys;
and 4 h (with the second sample collected at 48 h) following sibling relationships in titi monkeys; and titi mother’s and
reunion. Plasma cortisol was elevated 24 h following separa- father’s relationship with their offspring of any age. All other
tion compared to basal samples collected in the previous dyadic potential relationships in both species are not generally
week and remained at high levels throughout the month long affiliative and/or do not occur with any great frequency and
separation period. Reunion with the parents led to return to thus cannot be categorized as either friendships or attachment
the preseparation levels within 48 h. bonds.
276 Social Stress: Concepts, Assumptions, and Animal Models

The separation studies of these species have helped to clarify and the other chamber contains unfamiliar potential mate.
the relational aspects of social organization and their possible Pair bonding is operationally defined by spending more time
functional consequences. Titi parents are never confronted in contact and proximity to the familiar stimulus animal.
with a situation in which their baby can be mixed up with Females typically require 3 h of cohabitation to show a clear
another baby, whereas this is a daily occurrence for squirrel partner preference, whereas males require longer (up to
monkey mothers particularly once the infants begin to leave 24 h). In addition to partner preference, the pair bonding expe-
mother and interact in playgroups. The bidirectional attach- rience also leads to increased aggression directed to strange
ment relationship in squirrel monkeys may facilitate a mother’s animals (potential intruders) (Carter et al., 1995).
reunion with her own infant in sometimes chaotic group activ- The main focus of this research has been the roles of
ities during travel and foraging. If visual contact is retained, oxytocin and vasopressin in the formation of pair bonds, oper-
then agitated behavior on the part of either mother or infant ationally defined as selective preference for partner over other
is sufficient to signal to the other that it is time for reunion. potential mates. Generally, vasopressin facilitates development
If visual contact is broken, the activation of the stress response of partner preference in males and oxytocin in females (Devries
would provide the necessary metabolic resources to initiate et al., 2007). Centrally, oxytocin and vasopressin are believed
a search and find operation in both mother and infant. Short to act via dopaminergic reward system to promote social inter-
term, the equivalent response in both mother and infant would action and recognition of mate as mate (DeVries et al., 2007).
be considered adaptive, but long term the mother should The powerful role these peptides can play in partner preference
consider cutting her losses, whereas the infant needs the is illustrated by a study comparing the effects of centrally
mother for survival for some time following nutritional inde- administered oxytocin and vasopressin on partner preference
pendence and must sustain efforts to reestablish contact. formation and social contact in male and female prairie voles
For titi monkeys, all group members form an attachment (Cho et al., 1999). After only 1 h of cohabitation and pretreat-
bond with the father. It is the seeking of proximity with the ment with either AVP or OT, both males and females exhibited
father by all group members and the father’s attraction to the increased social contact and significant preference for the
mother that accounts for remarkable group unity characteristic familiar partner. If either oxytocin receptors or vasopressin
of titis. It is common to see all group members sitting in huddle receptors were blocked, partner preferences were not evident
groups with tails entwined with the father at the center, or in either males or females and neither oxytocin nor vasopressin
maintaining close proximity during travel and foraging. The was effective in eliciting partner preference. Thus, both receptor
infant is not a particularly cherished member of the group. populations must be available for pair preference to develop,
The father is tolerant of infant contact but other group but either of these closely related peptides is effective in elicit-
members are not. Except for short periods surrounding nursing ing pair formation. Sex differences in the roles of oxytocin and
most titi mothers are intolerant of prolonged infant contact vasopressin in pair bond formation are clear, and their interac-
and attempts by the infant to maintain contact with her or tions with HPA may help to understand differing impact of
even brief contact with siblings are met with active efforts to HPA activity in regulating the social system of prairie voles.
dislodge the infant. Indeed, when the father is carrying the Vasopressin stimulates release of ACTH thus potentially aug-
infant all other family members stay a little further away menting release of corticosteroids, whereas oxytocin tends to
from him (Van Belle et al., 2016). suppress ACTH and corticosteroids and these conditions
influence pair bonding in a sex-specific fashion (DeVries
et al., 2007).
1.09.5.2 Comparative Case Study: Voles
Like titi monkeys, monogamous prairie voles exhibit
Another monogamous species that has received a lot of atten- responses to separation and reunion that are indicative of an
tion in physiological studies of social stress is the prairie vole. emotional bond or attachment. Following 2 weeks of cohabita-
Many studies have been done comparing this species with its tion, ovariectomized adult females and intact males were sepa-
very closely related cousin species the nonmonogamous rated for 24 h and then either left alone, reunited with familiar
montane vole. In nature, a variety of behavioral mechanisms mate for 30 min, or placed with a stranger for 30 min. A sepa-
facilitate monogamy in the prairie vole. Females are selectively ration of 24 h leads to elevated corticosterone levels. Reunion
aggressive toward strange females, sexually naïve males are with the mate leads to reductions in corticosterone, below
nonaggressive unless they are met with aggression, and sexually basal values for females, and exposure to an unfamiliar partner
experienced males are highly aggressive to unfamiliar animals instead of the mate during the reunion period results in
(Carter et al., 1995). This means that females will not associate elevated levels of corticosterone (Carter et al., 1995). Socially
readily in multifemale groups and experienced males actively inexperienced males exposed to a strange naïve female and
exclude any strangers. Forming new pairs is more likely to occur males that had been living in a pair bond had exactly the
with a naïve male since experienced males are generally too same response to a novel female, suggesting that for males
aggressive even toward potential mates. In the laboratory, the reduction in cortisol with pairing is not selective or at least
studies of pair bond formation are tested by allowing a period does not change the male’s subsequent response to another
of cohabitation between a male and a female who are repro- female. Males exposed to another male in the same testing
ductively naïve and without prior pairing experience. Testing paradigm failed to reduce adrenocortical responses, so the buff-
proceeds with the experimental animal allowed to choose ering response is to a category – female rather than to a specific
between three chambers – an empty middle chamber and female (DeVries et al., 1997). Similarly, females living with
two side chambers with tethered stimulus animals. One a female sibling also respond to isolation with increased corti-
chamber contains their now familiar cohabitation partner costerone and reunion with the cage mate leads to reduction of
Social Stress: Concepts, Assumptions, and Animal Models 277

corticosterone. For female siblings the reduction in corticoste- bacteria killing ability in both males and females (Scotti
rone is also apparent when just the bedding the sibling has et al., 2015).
used is returned. This may be a specific response to a specific There are many parallels in the titi monkey and prairie voles
individual, but it certainly indicates that, at least for siblings, related to their monogamous lifestyle. Research with the prairie
contact is not required. Similar findings for peer effectiveness voles focuses on pair bonding, and research with the titi
in reducing the response to social isolation has been found monkey has focused more on stable family units. Nonetheless
in sheep with exposure to simply the face of a familiar partner parallels exist including suggestions that prairie voles, like titi
(da Costa et al., 2004). monkeys, develop an attachment bond between adults.
Exogenous administration of corticosteroids to females Imaging studies in titi monkeys and more invasive studies of
immediately before the cohabitation period leads to an brain circuits important to pair bond formation in prairie voles
inability to bond with the familiar partner and is reported to indicate that a neural circuit beginning with olfactory input and
actually induce preference for the stranger (Carter et al., involving the dopaminergic reward circuit (ventral pallidum,
1995; DeVries et al., 1996). Similarly, if females are exposed nucleus accumbens, ventral tegmental area) and the social
to a stressor, such as forced swim test, immediately before recognition circuits in the medial amygdala and lateral septum
cohabitation with a naïve male, pair bonding does not happen. are critically involved in pair bonding (Bales et al., 2007; Young
Adrenalectomy leads to greater efficiency in forming partner et al., 2005).
preference and, without corticosterone, females require much
less time in the cohabitation phase to induce partner prefer-
1.09.5.3 Comparative Case Study: Guinea Pigs
ence. Timing of corticosterone effects is critical. Administration
of corticosterone following cohabitation but before testing is Guinea pigs are perhaps the most studied rodent from the
not disruptive of partner preference formation in females. In perspective of social separation and stress buffering for any
males, the effects of corticosterone treatment and exposure to polygynous rodent. When housed in large heterosexual groups
a stressor were opposite to that in females. Artificially elevating with multiple animals of each age and sex categories, guinea
corticosterone prior to the cohabitation phase facilitates forma- pigs form harem groups consisting of one male and several
tion of partner preference, whereas adrenalectomy abolished females. Adult males exposed to a novel environment exhibit
partner preference formation even with extended periods of reduced cortisol response to the procedure if also placed with
cohabitation. an adult female from his harem, but not a familiar female
Autonomic activity using HR as the signal also shows signif- outside the harem or an unfamiliar female (Sachser et al.,
icant change with social isolation. In the first study of auto- 1998). Similarly, the male buffers the female’s response to
nomic balance, female prairie voles living with like-sex novelty. For females, however, the response to novelty is best
siblings exhibited a predominant vagal tone (Grippo et al., attenuated by the harem male, but other familiar males and
2007a). Pharmacologically blocking the PNS significantly familiar females also provide a modicum of buffering the
increased HR, reduced HR variability, and reduced the ampli- HPA response (Hennessy et al., 2008; Kaiser et al., 2003).
tude of respiratory sinus arrhythmia. Pharmacologically block- Guinea pig mothers left in the home cage when their litters
ing the sympathetic input to the heart significantly reduced HR. were removed showed an increase in cortisol (Ritchey and Hen-
Combined PNS and SNS blockade significantly increased HR, nessy, 1987); however, like squirrel monkeys (see above), if
and reduced HR variability and respiratory sinus arrhythmia pups were within auditory range, mothers do not exhibit an
amplitude. Placing young naïve females in social isolation for HPA response. In a novel environment mothers are capable
4 weeks reverses autonomic tone from predominantly para- of buffering the response to novelty for their infants, but
sympathetic to predominantly sympathetic; HR increases and another adult female and siblings are not effective in reducing
HR variability decreases (Grippo et al., 2007b). In another HPA response to stressful circumstances (Graves and Hennessy,
study, adult female prairie voles were exposed to social isola- 2000; Ritchey and Hennessy, 1987; Sachser et al., 1998). Infant
tion or continued pairing with a female sibling for 4 weeks. guinea pigs allowed to stay in their familiar environment (in
During weeks 3 and 4 of this period, animals were adminis- this case a large mixed sex group), unlike squirrel monkeys,
tered oxytocin daily for a total of 14 days. As in the previous show activity and vocal behaviors indicative of distress, but
experiment, isolation significantly increased basal HR and do not show heightened pituitary–adrenal activity. Infant
reduced HR variability and vagal regulation of the heart; these guinea pigs do not show the full stress buffering effect if the
changes in isolated animals were prevented with oxytocin mother and infant are separated by wire mesh (and cannot
administration (Grippo et al., 2009). make full contact), and HPA responses are greatest for the
Disruption of an established bond can also lead to altered infant when confronting the novel environment completely
regulation of HR in male prairie voles (McNeal et al., 2014). alone (Hennessy, 1988).
This study monitored males living initially with siblings then For young guinea pigs raised with their mother and siblings,
formed into new pairs for 5 days followed by removal of the the mother continues to be a source of stress buffering for her
female and the males subjected to at least 8 days of social isola- offspring still living in the natal nest into adulthood. However,
tion. Moving from living with siblings to formation of new the ability to provide stress buffering is no longer selective, and
pairs did not produce a change in HR. Isolation resulted in for offspring beyond the infancy period other adult females are
increased HR largely due to enhanced sympathetic input to also capable of buffering the response to novelty (Graves and
the heart as previously shown for females. Recently, social Hennessy, 2000; Hennessy et al., 2000). This picture is some-
isolation has also been shown to result in impaired immune what more complicated if young guinea pigs are colony raised
functions as indicated by reduced hemolytic complement and and, hence, have a multitude of social partners to select or
278 Social Stress: Concepts, Assumptions, and Animal Models

avoid in interactions during development. One obvious differ- providing pair-living males with extra-social interactions
ence is that males can now choose to interact with females during the early stage of adolescence. In wild cavies, the
other than the mother and may even begin to prefer to do so. ancestor species for the guinea pig, comparisons of adolescent
Studies of colony-raised guinea pigs reveal interesting males from high and low population densities show similar
changes that occur surrounding achievement of sexual and behavioral differences between adolescents reared in pairs
social maturity which in guinea pigs, like humans, occurs and those reared in large colonies. These findings suggest that
somewhat later than reproductive maturity (Hennessy et al., the shifting male responsiveness, both with respect to behavior
2006). Animals in this study were tested with their favored and the organization of physiological processes, occurs in
female social partner, a strange female, or alone during a 4 h response to the density of population and the shifting resources
exposure to a novel environment. Infant guinea pigs’ favored populations rely upon.
adult female was always the mother and, when with her, they
showed reduced adrenocortical response novelty. When alone
1.09.5.4 Social Isolation in Humans
or with an unfamiliar female, cortisol levels were higher than
with the mother and were sustained for the full 4 h, whereas, Separation and stress buffering studies in animals have
when with the mother, cortisol levels returned to basal values provided important insights in human stress and vulnerability
by 4 h. Beyond the period of infancy none of the males showed to disease. The most striking example is bereavement (Stroebe
an HPA response that persisted through the 4-h period. This is et al., 2007). Although details vary from study to study, the
not likely to reflect maturation of negative feedback mecha- overwhelming result of the research to date suggests that
nisms, since infants with mothers are capable of terminating following bereavement the surviving spouse is at greater risk
the HPA response within the 4-h period. As juveniles, some of mortality. Excess mortality is greatest in early months
males (1/3) still preferred their mothers, while others prefer following bereavement and decreases with duration of bereave-
to interact with another adult female. The preferred females, ment. Bereaved individuals are more likely to have new or
however, are not effective in buffering juvenile males’ response worsened illness, increased pain, and weight loss and corre-
to a novel environment; instead, unfamiliar females provide spondingly higher rates of disability, medication use, and
a modicum of stress buffering. At sexual maturity all males hospitalization. Heightened ACTH levels and resistance to
shifted preference from mother to other females. Sexually dexamethasone suppression indicating impaired feedback
mature (but not socially mature) males appear to go through mechanisms have been found for bereaved individuals,
a stress hyporesponsive period. For these males, cortisol levels although cortisol did not differ except following treatment
remained low throughout the testing period, rising only with dexamethasone (Gerra et al., 2003).
slightly above basal levels at 1 h; by 4 h levels were below basal There are many changes that occur in the immediate after-
values. Cortisol levels for sexually mature males were not influ- math of bereavement, and it is no surprise that physical and
enced by either favored or unfamiliar adult females. With social psychological processes need time to fully adapt. For our
maturity the reduced response to novelty was retained when purposes it is difficult to compare the human response to
novelty was encountered with a favored female. When alone sudden separation from spouse by death with the findings
response to novelty was as high at 60 min as for juvenile males, from animals simply because of the number of changes and
but for socially mature males cortisol levels returned to base the extent of differences in the extended social circumstances
more quickly. The response to unfamiliar females for socially within which the bereaved individual is embedded. The clarity
mature males was intermediate and not clearly different of animal studies is suggestive that mechanisms involved with
from the response to novelty when either alone or with isolation are similar to those involved in bereavement
a favored female. (Karelina and DeVries, 2011), but we cannot with certainty
The distinctive response profile shown by sexually, but not suggest the mechanisms of social isolation and bereavement
yet socially, mature guinea pigs comes at a time in the male are the same. It is almost certainly the case, however, that
guinea pig’s life during which it might leave the natal colony studies of humans during bereavement or otherwise are not
altogether (Hennessy et al., 2006). The lack of HPA responsive- studying the complete social isolation typical of animal studies.
ness to novel environments may induce a tendency to range In research on social isolation in humans the social network
further from the nest in sexually mature guinea pigs than would size is operationalized as indicating the degree of social isola-
be the case for younger or even older guinea pigs. Adolescence tion. This, in turn, is associated with greater awakening
is also a critical period in determining how a male guinea pig response to cortisol and higher cortisol at the trough of the
will behave and react to social challenge later in life (Sachser cycle which is accompanied by insensitivity of glucocorticoid
et al., 2013). In laboratory studies of guinea pigs, males that receptor to cortisol if isolation is prolonged. Together with
experience adolescence in large colonies apparently learn social enhanced SNS activity, the physiological changes associated
skills associated with formation of stable hierarchies and can with relative social isolation in humans create a proinflamma-
integrate themselves in new social groups with relatively little tory immune profile with attendant risks for inflammatory
aggression and in the absence of stress profiles sometimes asso- disease (Hawkley et al., 2012).
ciated with subordination. For males that live with only a single As Cacioppo et al. (2015) note a given type of relationship
female from early adolescence, interactions with other males may be supportive or dismissive, protective or threatening, and
become increasingly aggressive and long-term associations the perception of the relationship by its participants is likely
difficult. Stress responsiveness to novelty in males living in a determinant of the physiological consequences of engaging
pairs is also greater than that in colony-raised males. Moreover, or disrupting the relationship. Moreover, they argue that the
these profiles can be shifted in the expected direction by number of others in the network or frequency of interactions
Social Stress: Concepts, Assumptions, and Animal Models 279

with others is not as important as is the perception of loneli- by mothers protecting their nest, the resident/intruder test
ness. Indeed, individual differences in the degree to which indi- does not model an aspect of the natural biology of most
viduals seek social stimulation are not considered a risk factor, rodents. Aggressive encounters in colonies of mice and rats
but if an individual’s social environment or interaction generally occur at the periphery rather than the core of the terri-
network does not provide the level of stimulation desired, tory, or in neutral foraging areas. Therefore, the advantage of
loneliness ensues with attendant risks to health. Health risks the resident is likely to result in flight of the intruder rather
associated with loneliness cannot be explained by simple avail- than inescapable conflict. Moreover, even in the circumstances
ability of family and friends or by changes imposed by others in of the resident/intruder tests, considerable care must be taken
one’s network to improve healthful behaviors. Thus, loneliness to ensure expression of aggression in the test situation. Resi-
is a risk factor for morbidity and mortality beyond that which dents are often selected to be larger, more aggressive, spend
can be ascribed to isolation, which in humans is virtually never prolonged periods in isolation, and even teased to elicit the
complete in any event. attack behavior of the resident. Social defeat might happen in
Studies of the physiological processes attendant to loneli- nature, but it would be rare. Thus, rather than modeling sources
ness are just beginning. Early results indicate that both SNS of normative social stress, the resident/intruder model may be
and HPA systems are apparently responsive to perceived lone- more likened to an unusual traumatic experience outside the
liness (Cacioppo et al., 2015). Enhanced sympathetic activity purview of ordinary social experience. Modification of the resi-
particularly in tissue, elevated daily cortisol output, and dent/intruder model has proven useful to identify a shift in
decreased glucocorticoid receptor sensitivity have all been aggressiveness or attraction directed to an unfamiliar animal.
found to be associated with loneliness. Moreover, loneliness This is illustrated, for example, in the shift of voles’ response
may increase negative evaluations of social potential – others to unfamiliar animals from nonaggressive to more aggressive
are perceived as more dangerous and less supportive. These as a consequence of mate formation.
authors argue that the psychological dampening of perceived Prolonged periods of aggressive and more ritualized forms
benefits of social interaction prevents stress buffering mecha- of agonistic behavior are seen in newly formed colonies of
nisms. These findings have led to the conclusion that animal mice and rats. Particularly informative of behavior in natural
studies of repeated social threat are a better model for the social situations are models such as the visible burrow system
neuroendocrine changes and the immunological consequences (Blanchard and Blanchard, 1990) or other means of providing
than are animal studies of separation or isolation. a laboratory simulation of burrowing and foraging typical of
mice and rats. In these circumstances, normally socialized
rats and mice form groups in which one male is dominant
1.09.6 Conclusions and the others submissive. Submissive animals are vulnerable
and typically experience greater and earlier mortality than
In this chapter we have concentrated on presenting several dominants. Altered regulation of SNS and HPA can accompany
influential attempts to study social stress largely using animal formation of dominance relations perhaps contributing to
models. To evaluate the efficacy of the models two major mortality. When colonies are allowed to stabilize, the physio-
criteria should be considered. First is the extent to which the logical differences are minimal, but believed to reemerge
research illuminates aspects of the social biology of the species when stressors are encountered (Henry and Stephens, 1977).
investigated. Does the paradigm permit natural expression of It is important to note that the dominant animal can also be
social proclivities? As a result of the research do we come to viewed as more vulnerable to predation by virtue of his status.
a better understanding about the animals being investigated? Dominant mice and rats tend to be hypervigilant and more
Second, the justification for conducting most social stress often exposed to the risk of predation (Blanchard and
research is its translation potential. How aptly do the studies Blanchard, 1989). While dominance/subordination in groups
capture aspects of human biology? How might understanding of mice and rats have great potential for understanding social
sociophysiological processes in animals contribute to under- influences on physiological regulation, their generality to
standing vulnerability to disease processes in humans and processes in common with human and nonhuman primates
inform medical practice? must be treated cautiously. Establishing dominance/submis-
Starting with the notion that aggression is the key behavior sive relations in mice and rats are generally dyadic in contrast
mediating negative consequences of sociality, studies were to most primates for which linear dominance hierarchies are
initiated, largely with rodents to explore the physiological formed implying at a minimum a triadic process (Chase,
underpinnings of aggression. The resident/intruder paradigm 1984). Also in contrast to large group-living primates, agonistic
was central to early explorations. This approach has the virtue encounters are prolonged when new colonies of mice and rats
of being able to describe stress onset and follow physiological are formed and include a fair amount of contact aggression and
changes throughout the ensuing response. The defeated animal wounding. Status relations in mice and rats take days and
experiences a substantial acute stress response that is compa- weeks to be evident, whereas, for primates formation of domi-
rable to the response to other stressors in form and temporal nance relations in comparable circumstances does not include
sequence and does not habituate (Bhatnagar and Vining, prolonged periods of agonistic interactions; dominance rela-
2003; Sgoifo et al., 2005). As a translational tool to study the tions are settled within minutes and typically without fighting.
acute stress response the resident/intruder model has much Sapolsky’s studies of feral groups of baboons provide a truly
to recommend. However, the resident/intruder model does excellent model for engaging in studies of social stress. His
not fulfill its promise as a means of studying social stress. combination of behavioral observation of a natural population
With the possible exception of maternal aggression displayed and physiological probes is unique in animal studies of social
280 Social Stress: Concepts, Assumptions, and Animal Models

stress. Clearly his work has enriched our understanding of both another initially. Whether the instability induced by the forma-
social processes in baboons and endocrine physiology. tion of new laboratory groups, models spontaneous occurrence
A particularly important outcome of his research is the clear of periods of instability in established or feral groups is debat-
finding that physiological responses to social dominance can able. Unfortunately, we do not have the methods to describe
be altered by group level processes. A concern with this research relative degrees of stability or instability in large groups other
is the extent to which generalizations beyond baboons is advis- than spontaneous outbreaks of aggression which may or may
able. The nature of physiological processes associated with not be sensitive measures of cohesion or the lack thereof.
dominance is not the same in baboons as in other species If dominance relations are one aspect of sociality important
and often not even in the same direction. Squirrel monkeys, to understanding social stress, availability of emotional bonds
for example, show status differentiation of stress response is another that has received a great deal of research attention.
systems only when instability and breeding coincide; when it Some relationships can be characterized by an attachment
does appear, status differentiation is opposite to that found bond. Filial bonds are the most commonly studied relation-
in baboons. Thus, while the approach Sapolsky used is an ship of this type and dependent offspring usually, but not
ideal approach, the results are at present best considered always, form a strong and abiding emotional bond with the
species specific. mother. Disruption of the bond induces an acute stress
The focus on aggression and dominance relations as key to response and engagement in the bond can be stress buffering
understanding social stress must also be questioned. The (Hennessy, 1997). Considerable insight into the characteristic
studies of group formations in primates indicate that domi- social structure can be provided by studies that explore the
nance relationships are an effective means of limiting aggres- extent to which relationships conform to the requirements
sion and not a manifestation of unregulated aggression. Is of an attachment bond as delineated by Bowlby (1969),
the subordinate’s lot in life as bad as often depicted? Almost Ainsworth (1972), and others. Our understanding of the social
certainly not. Moreover, in most primate societies, duration systems of voles, guinea pigs, and titi monkeys have benefitted
in a particular status is delimited by processes of emigration from application of concepts developed to understand the
and immigration. Dominance relations provide an easy means filial bond in humans – a valuable instance where transla-
of role differentiation and enhanced predictability particularly tional research occurs in the reverse direction.
during periods of instability, but a subordinate role is not a life- In the last decade we have improved our technical ability to
time sentence. One of the problems with the research on domi- pursue the physiological and neurobiological mechanisms
nance and aggression is that it is only one aspect of social life, necessary to link processes originating in the social environ-
particularly in primates. Clear testament to the myopia in this ment to health and disease. If we are to improve our
research is that females, often considered the cohesive force in approaches to social stress, better behavioral tools are required.
primate societies, are completely neglected in research on In particular, objective measures of cohesion and stability in
aggression and dominance. groups, the rules that govern individual behavior within
Studies of social instability provide promising avenues for society, and the degree of embeddedness of an individual
examining the impact of group level processes. Work by Capi- within its society are needed. Recent studies of loneliness
tanio and colleagues have shown clear linkages between stress- suggest the perception of loneliness is a risk factor but the
response systems, immune processes, and disease progression. number of family and friends is not (Cacioppo et al., 2015).
In addition to providing another excellent example of pursuit Modeling loneliness or other aspects of human sociality is
of understanding social processes, behavior, and physiology not currently possible, but promising methods are being devel-
in a disease context, these studies offer caution in the assump- oped (McCowan et al., 2011) which link individual proclivities
tions that we bring to studies of social stress. SNS processes play and experience with group processes.
an important role in disease progression but seem to do so To appropriately model human social stress with animal
independently of circulating levels of Epi and NE. This means studies, we also need to be clear that the processes we study
that correlational studies of social processes and activity in in animals are relevant to human societies. As we have seen
SNS are in danger of being misleading rather than informative. social structure is species specific. More concern needs to be
Similarly, rhesus monkeys and squirrel monkeys respond to paid to what human society means to humans. The attach-
formation of new social groups with an initial reduction in ment processes are probably the human relationship best
cortisol and evidence of dysregulation of HPA by excessive modeled in animal research, and animal studies have helped
negative feedback. Rather than conferring advantage, reduced to generalize processes involved in filial attachment to other
cortisol is associated with more rapid disease course through primate relationships including adult–adult emotional bonds
as yet unidentified mechanisms. The clear message from these (Mason and Mendoza, 1998). The extension of attachment
findings is cortisol levels cannot be equated with stress as is processes in humans to health issues currently includes adoles-
so often the case. Lower than optimal levels of cortisol can be cent and adult romantic attachment and parental behavior in
disadvantageous and even on occasion lethal (Heim et al., addition to the more commonly considered filial attachment
2000). A more general cautionary note these studies provide as a unique relational system (Mikulincer and Shaver, 2012).
is that assumptions regarding disease processes and vulnerabil- This work parallels work on monogamous species, such as titis
ities based on correlational studies should be considered a start- or voles, which also show adult emotional bonds and similar
ing point rather than a conclusion in studies of social stress. neurobiological mechanisms. This suggests humans are
In fairness, it should be noted that group formation studies monogamous. But unlike titi monkeys or voles, human
are also a laboratory artifact. It is only in the lab that new groups monogamy is embedded in a social nexus that more closely
are ever formed de novo with all animals unfamiliar to one resembles the complex societies exhibited by baboons or
Social Stress: Concepts, Assumptions, and Animal Models 281

macaques. At present we do not have adequate and similar Capitanio, J., Abel, K., Mendoza, S., Blozis, S., McChesney, M., Cole, S.M., 2008.
tools for comparing human and nonhuman societies and Personality and serotonin transporter genotype interact with social context to affect
immunity and viral set-point in simian immunodeficiency virus disease. Brain
these need to be developed if truly translational research on
Behav. Immun. 22, 676–689.
social stress is to be implemented. Capitanio, J., 2004. Personality factors between and within species. In: Thierry, B.,
Singh, M., Kaumanns, W. (Eds.), Macaque Societies: A Model for the Study of
Social Organizations. Cambridge University Press, Cambridge, pp. 169–177.
Carter, C., DeVries, A., Getz, L., 1995. Physiological substrates of mammalian
References monogamy: the prairie vole model. Neurosci. Biobehav. Rev. 19, 303–314.
Carter, C., Grippo, A., Pournajafi-Nazarloo, H., Ruscio, M., Porges, S., 2008. Oxytocin,
Abbott, D., Keverne, E., Berkovitch, F., Shively, C., Mendoza, S., Saltzman, W., vasopressin and sociality. Prog. Brain Res. 170, 331–336.
Snowdon, C., Zeigler, T., Banjevic, M., Garland Jr., T., Sapolsky, R., 2003. Are Caspi, A., Moffett, T., 2006. Gene-environment interactions in psychiatry: joining
subordinates always stressed? A comparative analysis of rank differences in forces with neuroscience. Nat. Rev. Neurosci. 7, 583–590.
cortisol levels among primates. Horm. Behav. 43, 67–82. Chase, I., 1984. Social process and hierarchy formation in small groups: a comparative
Adams, N., Lins, M.D., Blizard, D.A., 1987. Contrasting effects of social stress and perspective. In: Barchas, P. (Ed.), Social Hierarchies: Essays toward a Socio-
foot-shock on acute cardiovascular response in salt-sensitive rats. Behav. Neural physiological Process. Greenwood Press, Westport, Connecticut, pp. 45–80.
Biol. 48, 368–381. Cho, M., DeVries, A., Williams, J., Carter, C., 1999. The effects of oxytocin and
Ainsworth, S., 1972. Attachment and dependency: a comparison. In: Gewirtz, J. (Ed.), vasopressin on partner preferences in male and female prairie voles (Microtus
Attachment and Dependency. VH Winston & Sons, Washington DC, pp. 97–137. ochrogaster). Behav. Neurosci. 113, 1071–1079.
Archer, J., 1970. Effects of aggressive behavior on the adrenal cortex in male labo- Coe, C., Mendoza, S., Davidson, J., Smith, E., Dallman, M., Levine, S., 1978.
ratory mice. J. Mammol. 51, 327–332. Hormonal response to stress in the squirrel monkey (Saimiri sciureus). Neuroen-
Bales, K., Mason, W., Catana, C., Cherry, S., Mendoza, S., 2007. Neural correlates of docrinology 26, 367–377.
pair-bonding in a monogamous primate. Brain Res. 1184, 245–253. Coe, C., Mendoza, S., Levine, S., 1979. Social status constrains the stress response in
Barchas, P., Mendoza, S., 1984. Emergent hierarchical relationships in rhesus the squirrel monkey. Physiol. Behav. 23, 633–638.
macaques: an application of Chase’s model. In: Barchas, P. (Ed.), Social Hierar- Coe, C., Weiner, S., Rosenberg, L., Levine, S., 1985. Physiological consequences of
chies. Greenwood Press, Westport, Connecticut, pp. 81–95. maternal separation and loss in the squirrel monkey. In: Rosenblum, L., Coe, C. (Eds.),
Barnett, S., 1975. The Rat. University of Chicago Press, Chicago. Handbook of Squirrel Monkey Research. Plenum Press, New York, pp. 127–148.
Barr, C., Newman, T., Becker, M., Parker, C., Champoux, M., Lesch, K., Higley, J., Cubicciotti, D., Mason, W., 1975. Comparative studies of social behavior in Callicebus
2003. The utility of the non-human primate model for studying gene by environ- and Saimiri: male-female emotional attachments. Behav. Biol. 16, 185–197.
ment interactions in behavioral research. Genes Brain Behav. 2, 336–340. da Costa, A., Leigh, A., Man, M., Kendrick, K., 2004. Face pictures reduce behav-
Bernard, C., 1872. Des fonctions du cerveau. Rev. Des. Deux. Mondes 98, 373–385. ioural, autonomic, endocrine and neural indices of stress and fear in sheep. Proc.
Bernstein, I., Gordon, T., 1974. The function of aggression in primate societies. Am. Biol. Sci. 271, 2077–2084.
Sci. 62, 302–311. Dallman, M., 2007. Modulation of stress responses: how we cope with excess
Bernstein, I., Mason, W., 1963. Group formation by rhesus monkeys. Anim. Behav. glucocorticoids. Exp. Neurol. 206, 179–182.
11, 28–31. Daskalakis, N., Lehner, A., Yehuda, R., 2013. Endocrine aspects of post-traumatic
Bernstein, I., Gordon, T., Rose, R., 1974. Factors influencing the expression of stress disorder and implications for diagnosis and treatment. Endocrinol. Metab.
aggression during introductions to rhesus monkey groups. In: Holloway, R. (Ed.), Clin. N. Am. 42, 503–513.
Primate Aggression, Territoriality, and Xenophobia. Academic Press, New York, Davis, E., Marler, C., 2003. The progesterone challenge: steroid hormone changes
pp. 211–240. following a simulated territorial intrusion in female Peromyscus californicus. Horm.
Bernstein, I., 1981. Dominance: the baby and the bathwater. Behav. Brain Sci. 4, Behav. 44, 185–198.
419–457. DeVries, A., DeVries, M., Taymans, S., Carter, C., 1996. The effects of stress on social
Bhatnagar, S., Vining, C., 2003. Facilitation of hypothalamic-pituitary-adrenal preferences are sexually dimorphic in prairie voles. Proc. Natl. Acad. Sci. U.S.A.
responses to novel stress following repeated social stress using the resident/ 93, 11980–11984.
intruder paradigm. Horm. Behav. 43, 158–165. DeVries, A., Taymans, S., Carter, C., 1997. Social modulation of corticosteroid
Blanchard, R., Blanchard, D., 1989. Antipredator defensive behaviors in a visible responses in male prairie voles. Ann. N.Y. Acad. Sci. 807, 494–497.
burrow system. J. Comp. Psychol. 103, 70–82. DeVries, A., Craft, T., Glasper, E., Neigh, G., 2007. Social influences on stress
Blanchard, D., Blanchard, R., 1990. Behavioral correlates of chronic dominance- responses and health. Psychoneuroendocrinology 32, 587–603.
subordination relationships of male rats in a seminatural situation. Neurosci. Edwards, D., Rowe, F., 1975. Neural and endocrine control of aggressive behavior. In:
Biobehav. Rev. 14, 455–462. Hormonal Correlates and Behavior. Plenum Press, New York, pp. 275–303.
Blanchard, D., Spencer, R., Weiss, S., Blanchard, R., McEwen, B., Sakai, R., 1995. Ely, D., Henry, J., 1978. Neuroendocrine response patterns in dominant and subor-
Visible burrow system as a model of chronic social stress: behavioral and dinate mice. Horm. Behav. 10, 156–169.
neuroendocrine correlates. Psychoneuroendocrinology 20, 117–134. Fernandez-Duque, E., Valeggia, C., Mendoza, S., 2009. The biology of paternal care in
Blanchard, R., Hebert, M., Sakai, R., McKittrick, C., Henrie, A., Yudko, E., human and nonhuman primates. Annu. Rev. Anthropol. 38, 115–130.
Blanchard, D., 1998. Chronic social stress: changes in behavioral and physiological Fischer, H., Heinzeller, T., Raab, A., 1985. Gonadal responses to psychosocial stress in
indices of emotion. Aggress. Behav. 24, 307–321. male tree shrews (Tupaia belangeri): morphometry of testes, epididymis and
Blanchard, D., Summers, C., Blanchard, R., 2013. The role of behavior in translational prostrate. Andrologia 17, 262–275.
models for psychopathology: functionality and dysfunctional behaviors. Neurosci. Fuchs, E., 1984. Activity of the sympathoadrenomedullary system in male Tupaia
Biobehav. Rev. 37, 1567–1577. belangeri under control and stress conditions. In: Usdin, E., Kvetnansky, R.,
Boinski, S., 1987. Birth synchrony in squirrel monkeys (Saimiri oerstedi): a strategy to Axelrod, J. (Eds.), Stress: The Role of Catecholamines and Other Neurotransmit-
reduce neonatal predation. Behav. Ecol. Sociobiol. 21, 383–400. ters, Proceedings of the Third International Symposium on Catecholamines and
Bowlby, J., 1969. Attachment. In: Attachment and Loss, vol. 1. Basic Books, Other Neurotransmitters in Stress, vol. 1. Gordon and Breach Science Publishers,
New York. pp. 595–602.
Cacioppo, J., Cacioppo, S., Capitanio, J., Cole, S., 2015. The neuroendocrinology of Gerra, G., Monti, D., Panerai, A., Sacerdote, P., Anderlini, R., Avanzini, P.,
social isolation. Annu. Rev. Psychol. 66, 733–767. Franceschi, C., 2003. Longterm immune-endocrine effects of bereavement:
Cannon, W., 1929. Bodily Changes in Pain, Hunger, Fear, and Rage. Appleton- relationships with anxiety levels and mood. Psychiatry Res. 121, 145–158.
Century-Crofts, New York. Gonzalez, C., Hennessy, M., Levine, S., 1981. Subspecies differences in hormonal and
Cannon, W., 1932. The Wisdom of the Body. W.W. Norton, New York. behavioral responses after group formation in squirrel monkeys. Am. J. Primatol. 1,
Capitanio, J., Cole, S., 2015. Social instability and immunity in rhesus monkeys: the 432–439.
role of the sympathetic nervous system. Philos. Trans. R. Soc. Lond. B. Biol. Sci. Graves, F., Hennessy, M., 2000. Comparison of the effects of the mother and an
370, 1–11. unfamiliar adult female on cortisol and behavioral responses of pre- and post-
Capitanio, J., Mendoza, S., Lerche, N., Mason, W., 1998. Social stress results in weaning guinea pigs. Dev. Psychobiol. 36, 91–100.
altered glucocorticoid regulation and shorter survival in simian acquired immune Grippo, A., Lamb, D., Carter, C., Porges, S., 2007a. Cardiac regulation in the socially
deficiency syndrome. Proc. Natl. Acad. Sci. U.S.A. 95, 4714–4719. monogamous prairie vole. Physiol. Behav. 90, 386–393.
282 Social Stress: Concepts, Assumptions, and Animal Models

Grippo, A., Lamb, D., Carter, C., Porges, S., 2007b. Social isolation disrupts autonomic Koolhaas, J., Bohus, B., 1991. Coping strategies and cardiovascular risk: a study of
regulation of the heart and influences negative affective behaviors. Biol. Psychiatry rats and mice. In: Appels, A. (Ed.), Behavioral Observations in Cardiovascular
62, 1162–1170. Research. Swets and Zeitlinger, Amsterdam, pp. 45–60.
Grippo, A., Trahanas, D., Zimmerman, R., Porges, S.C., 2009. Oxytocin protects Koolhaas, J., Schuuman, T., Fokkema, E., 1983. Social behavior of rats as a model for
against negative behavioral and autonomic consequences of long-term social the psychophysiology of hypertension. In: Dembroski, T., Schmidt, T., Blumchen, G.
isolation. Psychoneuroendocrinology 34, 1542–1553. (Eds.), Biobehavioral Bases of Coronary Heart Disease. Karger, Basel, pp. 391–400.
Hall, K., 1964. Aggression in monkey and ape societies. In: Carthy, J., Ebling, F. (Eds.), Koolhaas, J., Meerlo, S., De Boer, J., Strubbe, J., Bohus, B., 1997. The temporal
The Natural History of Aggression. Academic Press, New York, pp. 51–64. dynamics of the stress response. Neurosci. Biobehav. Rev. 6, 775–782.
Haller, J., Kiem, D., Makaram, G., 1996. The physiology of social conflict in rats: what Kudryavtseva, N., Avgustinovich, D.F., 1998. Behavioral and physiological markers of
is particularly stressful? Behav. Neurosci. 110, 353–359. experimental depression induced by social conflicts (DISC). Aggress. Behav. 24,
Hawkes, P., 1970. Group Formation in Four Species of Macaques. University of 271–286.
California, Davis (Unpublished doctoral dissertation). Lazarus, R., 1999. Stress and Emotion: A New Synthesis. Springer Pub Co, New York.
Hawkley, L., Cole, S., Capitanio, J., Norman, G., Cacioppo, J., 2012. Effects of social Leger, D., Mason, W., Fragaszy, D., 1981. Sexual segregation, cliques and social
isolation on glucocorticoid regulation in social mammals. Horm. Behav. 74, 907–914. power in squirrel monkey (Saimiri) groups. Behaviour 76, 163–181.
Heim, C., Ehlert, U., Hellhammer, D., 2000. The potential role of hypocortisolism in the Lesch, K., Meyer, J., Glatz, K., Flugge, G., Hinney, A., Hebebrand, J., Heils, A., 1997.
pathophysiology of stress-related bodily disorders. Psychoneuroendocrinology 25, The 5-HT transporter gene-linked polymorphic region (5-HTTLPR) in evolutionary
1–35. perspective: alternative biallelic variation in rhesus monkeys. J. Neural Transm.
Hellhammer, J., Fries, E., Schweisthal, O., Scholtz, W., Stone, A., Hagemann, D., 2007. 104, 1259–1266.
Several daily measurements are necessary to reliably assess the cortisol rise after Lupien, S., Lecours, A., Schwartz, G., Sharma, S., Hauger, R., Meaney, M., Nair, N.,
awakening: state and trait components. Psychoneuroendocrinology 32, 80–86. 1995. Longitudinal study of basal cortisol levels in healthy elderly subjects:
Hellhammer, D., Wust, S., Kudielka, B., 2009. Salivary cortisol as a biomarker in stress evidence for subgroups. Neurobiol. Aging 17, 95–105.
research. Psychoneuroendocrinology 34, 163–171. Lyons, D., Mendoza, S., Mason, W., 1992. Sexual segregation in squirrel monkeys
Hennessy, M., Levine, S., 1978. Sensitive pituitary-adrenal responsiveness to varying (Saimiri sciureus): a transactional analysis of adult social dynamics. J. Comp.
intensities of psychological stimulation. Physiol. Behav. 21, 295–297. Psychol. 106, 323–330.
Hennessy, M., Mendoza, S., Kaplan, J., 1982. Behavior and plasma cortisol following Lyons, D., Mendoza, S., Mason, W., 1994. Psychosocial and hormonal aspects of hierarchy
brief peer separation in juvenile squirrel monkeys. Am. J. Primatol. 3, 143–151. formation in groups of male squirrel monkeys. Am. J. Primatol. 32, 109–122.
Hennessy, M., Maken, D., Graves, F., 2000. Consequences of the presence of the Mason, W., Mendoza, S., 1998. Generic aspects of primate attachments: parents,
mother or unfamiliar adult female on cortisol, ACTH, testosterone and behavioral offspring and mates. Psychoneuroendocrinology 23, 765–778.
responses of periadolescent guinea pigs during exposure to novelty. Psycho- Mason, W., 1966. Social organization of the South American monkey, Callicebus
neuroendocrinology 25, 619–632. moloch: a preliminary report. Tulane Stud. Zool. 13, 23–28.
Hennessy, M., Hornschuh, G.K., Sachser, N., 2006. Cortisol responses and social Mason, W., 1976. Primate social behavior: pattern and process. In:
buffering: a study throughout the life span. Horm. Behav. 49, 383–390. Masterton, R., Bitterman, M., Campbell, C., Hotton, N. (Eds.), Evolution of
Hennessy, M., Zate, R., Maken, D., 2008. Social buffering of the cortisol response of Brain and Behavior in Vertebrates. Lawrence Erlbaum Associates, Hillsdale,
adult female guinea pigs. Physiol. Behav. 93, 883–888. New Jersey, pp. 425–455.
Hennessy, M., 1986a. Effects of social partners on pituitary-adrenal activity during Mason, W., 2008. Social development: a psychobiological perspective. Integr. Psychol.
novelty exposure in adult female squirrel monkeys. Physiol. Behav. 38, 803–807. Behav. Sci. 42, 389–396.
Hennessy, M., 1986b. Multiple, brief maternal separations in the squirrel monkey: Maxim, P., 1976. An interval scale for studying and quantifying social relations in pair
changes in hormonal and behavioral responsiveness. Physiol. Behav. 36, 245–250. of rhesus monkeys. J. Exp. Psychol. Gen. 105, 123–147.
Hennessy, M., 1988. Both prevention of physical contact and removal of distal cues McCowan, B., Beisner, B., Capitanio, J., Jackson, M., Cameron, A., Seil, S.,
mediate cortisol and vocalization responses of guinea pig pups to maternal Fushing, H., 2011. Network stability is a balancing act of personality, power and
separation in a novel environment. Physiol. Behav. 43, 729–733. conflict dynamics in rhesus macaque societies. PLoS One 6, e22350.
Hennessy, M., 1997. Hypothalamic-pituitary-adrenal responses to brief social sepa- McKittrick, C., Blanchard, D., Hardy, M., Blanchard, R., 2009. Social stress effects on
ration. Neurosci. Biobehav. Rev. 21, 11–29. hormones, brain, and behavior. In: Hormones, Brain and Behavior, vol. 2. Elsevier,
Henry, J., Stephens, P., 1977. Stress, Health and the Social Environment. Springer, New York, pp. 333–366.
New York. McNeal, N., Scotti, M.W., Chandler, D., Bates, S., Larocca, M., Trahanas, D.,
Henry, J., Stephens, P., Axelrod, J., Mueller, R., 1971. Effect of psychosocial stim- Grippo, A., 2014. Disruption of social bonds induces behavioral and physiological
ulation on the enzymes involved in the biosynthesis and metabolism of dysregulation in male and female prairie voles. Auton. Neurosci. 180, 9–16.
noradrenaline and adrenaline. Psychosom. Med. 33, 227–237. Mendoza, S., 1984. The psychobiology of social relationships. In: Barchas, P.,
Hinde, R.A., 1966. Animal Behaviour: A Synthesis of Ethology and Comparative Mendoza, S. (Eds.), Social Cohesion: Essays Toward a Sociophysiological
Psychology, second ed. McGraw-Hill, New York. Perspective. Greenwood Press, Westport, CT, pp. 3–29.
Hinde, R.A., 1974. Biological Basis of Human Social Behavior. McGraw-Hill, New York. Mendoza, S., 1991. Behavioural and physiological indices of social relationships:
Hinde, R.A., 1976. Interactions, relationships, and social structure. Man 11, 1–17. comparative studies of New World monkeys. In: Box, H. (Ed.), Primate Responses
Hinde, R.A., 1983. A conceptual framework. In: Hinde, R.A. (Ed.), Primate Social to Environmental Change. Chapman and Hall, London, pp. 311–335.
Relationships. Sinauer Associates, Sunderland, MA, pp. 1–7. Mendoza, S., Lowe, E., Levine, S., 1978. Social organization and social behavior in two
Hofer, M., 1987. Early social relationships: a psychobiologist’s view. Child. Dev. 58, subspecies of squirrel monkeys. Folia Primatol. 30, 126–144.
633–647. Mendoza, S., Mason, W., 1986a. Contrasting responses to intruders and involuntary
Hoffman, K., Mendoza, S., Hennessy, M., Mason, W., 1996. Responses of infant titi separation by monogamous and polygynous new world monkeys. Physiol. Behav.
monkeys, Callicebus moloch, to removal of one or both parents: evidence for 38, 795–801.
paternal attachment. Dev. Psychobiol. 28, 399–407. Mendoza, S., Mason, W., 1986b. Parental division of labour and differentiation of
House, J., Landis, K., Umberson, D., 1988. Social relationships and health. Science attachments in a monogamous primate (Callicebus moloch). Anim. Behav. 34,
241, 540–545. 1336–1347.
von Holst, D., 1972. Renal failure as a cause of death in Tupaia belangeri exposed to Mendoza, S., Mason, W., 1989. Primate relationships: social dispositions and physiological
persistent social stress. J. Comp. Psychol. 78, 236–273. processes. In: Seth, P., Seth, S. (Eds.), Perspectives in Primate Biology, Neurobiology,
Insel, T., Winslow, J., Williams, J., Hastings, N., Shapiro, L., Carter, C., 1993. The role vol. 2. Today and Tomorrow’s Printers and Publishers, New Delhi, pp. 129–143.
of neurohypophyseal peptides in the central mediation of complex social processes Mendoza, S., Mason, W., 1991. Breeding readiness in squirrel monkeys: female-
– evidence from comparative studies. Regul. Pept. 45, 127–131. primed females are triggered by males. Physiol. Behav. 49, 471–479.
de Kloet, E., 2003. Hormones, brain and stress. Endocr. Regul. 37, 51–68. Mendoza, S., Mason, W., 1997. Autonomic balance in Saimiri sciureus and Callicebus
Kaiser, S., Kirtzeck, G.H., Sachser, N., 2003. Sex-specific difference in social support moloch: relation to life-style. Folia Primatol. 68, 307–318.
– a study in female guinea pigs. Physiol. Behav. 79, 297–303. Mendoza, S., Moberg, G., 1985. Species differences in adrenocortical activity in new world
Karelina, K., DeVries, A., 2011. Modeling social influences on human health. Psy- primates: response to dexamethasone suppression. Am. J. Primatol. 8, 215–224.
chosom. Med. 73, 67–74. Mendoza, S., Smotherman, W., Miner, M., Kaplan, J., Levine, S., 1978. Pituitary-
Kollack-Walker, S., Watson, S., Akil, H., 1997. Social stress in hamsters: defeat adrenal response to separation in mother and infant squirrel monkeys. Dev. Psy-
activates specific neurocircuits within the brain. J. Neurosci. 17, 8842–8855. chobiol. 11, 169–175.
Social Stress: Concepts, Assumptions, and Animal Models 283

Mendoza, S., Coe, C., Lowe, E., Levine, S., 1979. The physiological response to group Sapolsky, R., 1995. Social subordinance as a marker of hypercortisolism. Some
formation in adult male squirrel monkeys. Psychoneuroendocrinology 3, 221–229. unexpected subtleties. Ann. N.Y. Acad. Sci. 771, 626–639.
Mendoza, S., Coe, C., Smotherman, W., Kaplan, J., Levine, S., 1980. Functional Schiml, P., Mendoza, S., Saltzman, W., Lyons, D., Mason, W., 1996. Seasonality in
consequences of attachment: a comparison of two species. In: Bell, R., Smoth- squirrel monkeys (Saimiri sciureus): social facilitation by females. Physiol. Behav.
erman, W. (Eds.), Maternal Influences and Early Behavior. Spectrum Publications, 60, 1105–1113.
New York, pp. 235–252. Schiml, P., Mendoza, S., Saltzman, W., Lyons, D., Mason, W., 1999. Annual physi-
Mendoza, S., Lyons, D., Saltzman, W., 1991a. The sociophysiology of squirrel ological changes in individually housed squirrel monkeys (Saimiri sciureus). Amer.
monkeys. Am. J. Primatol. 23, 795–801. J. Primat. 47, 93–103.
Mendoza, S., Saltzman, W., Lyons, D., Schiml, P., Mason, W., 1991b. Within-sex Scotti, M., Carlton, E., Demas, G., Grippo, A., 2015. Social isolation disrupts innate
relationships in squirrel monkeys regulate pituitary-adrenal activity. In: Ehara, A., immune responses in both male and female prairie voles and enhances agonistic
Kimura, T., Tanenaka, O., Iwamoto, M. (Eds.), Primatology Today. Elsevier, behavior in female prairie voles (Microtus ochrogaster). Horm. Behav. 70, 7–13.
Amsterdam, pp. 443–446. Selye, H., 1956. The Stress of Life. McGraw-Hill, New York.
Mendoza, S., Hennessy, M., Lyons, D., 1992. Distinct immediate and prolonged Sgoifo, A., De Boer, S., Haller, J., Koolhaas, J., 1996. Individual differences in plasma
effects of separation on plasma cortisol in adult female squirrel monkeys. catecholamine and corticosterone stress responses of wild-type rats: relationship
Psychobiology 20, 300–306. with aggression. Physiol. Behav. 60, 1403–1407.
Mendoza, S., Capitanio, J., Mason, W., 2000. Chronic social stress: studies in non- Sgoifo, A., Stilli, D., De Boer, S., Koolhaas, J., Musso, E., 1998. Acute social stress
human primates. In: Moberg, G., Mench, J., Moberg, G., Mench, J. (Eds.), The and cardiac electrical activity in rats. Aggress. Behav. 24, 287–296.
Biology of Animal Stress: Basic Principles and Implications for Animal Welfare. Sgoifo, A., Costili, T., Meerlo, P., Buwalda, B., Pico-Alfonso, M., De Boer, S.,
CABI Publishing, New York, pp. 227–248. Koolhaas, J., 2005. Individual differences in cardiovascular response to social
Mendoza, S., 1991. Behavioural and physiological indices of social relationships: challenge. Neurosci. Biobehav. Rev. 29, 59–66.
comparative studies of new world monkeys. In: Box, H. (Ed.), Primate Responses to Sgoifo, A., Carnevali, L., Grippo, A., 2012. The social stressed heart: insights from
Environmental Change. Chapman & Hall, London, pp. 311–335. studies in rodents. Neurosci. Biobehav. Rev. 39, 51–60.
Mendoza, S., 1993. Social conflict on first encounters. In: Mason, W., Mendoza, S. (Eds.), Sloan, E., Tarara, R., Capitanio, J., Cole, S., 2006. Enhanced replication of simian
Primate Social Conflict. State University of New York Press, Albany, NY, pp. 85–107. immunodeficiency virus adjacent to catecholaminergic varicosities in primate lymph
Miczek, K., 1979. A new test for aggression in rats without aversive stimulation: differential nodes. J. Virol. 80, 4326–4335.
effects of d-amphetamine and cocaine. Psychopharmacology 60, 253–259. Sloan, E., Capitanio, J., Tarara, R., Mendoza, S., Mason, W., Cole, S., 2007. Social
Mikics, E., Kruk, M., Haller, J., 2004. Genomic and non-genomic effects of glucocorticoids stress enhances sympathetic innervation of primate lymph nodes: mechanisms and
on aggressive behavior in male rats. Psychoneuroendocrinology 29, 618–635. implications for viral pathogenesis. J. Neurosci. 27, 8857–8865.
Mikulincer, M., Shaver, P., 2012. An attachment perspective on psychopathology. Sloan, E., Capitanio, J., Cole, S., 2008a. Stress-induced remodeling of lymphoid
World Psychiatry 11, 11–15. innervation. Brain Behav. Immun. 22, 15–21.
Munck, A., Guyre, P., Holbrook, N., 1984. Physiological functions of glucocorticoids in Sloan, E., Nguyen, C., Cox, B., Tarara, R., Capitanio, J., Cole, S., 2008b. SIV infection
stress and their relation to pharmacological actions. Endocr. Rev. 5, 25–44. decreases sympathetic innervation of primate lymph nodes: the role of neuro-
Porges, S., Heilman, K., Bazhenova, O., Bal, E., Doussard-Roosevelt, J., Koledin, M., trophins. Brain Behav. Immun. 22, 185–194.
2007. Does motor activity during psychophysiological paradigms confound the Southwick, C., 1967. An experimental study of intragroup agonistic behavior in rhesus
quantification and interpretation of heart rate and heart rate variability measure in monkeys (Macaca mulatta). Behaviour 28, 182–209.
young children? Dev. Psychobiol. 49, 485–494. Spencer, R., Miller, A., Moday, H., McEwen, B., Blanchard, R., Blanchard, D.,
Porges, S.W., 2007. The polyvagal perspective. Biol. Psychol. 74, 116–143. Sakai, R., 1996. Chronic social stress produces reductions in available splenic
Porges, S.W., 2009. The polyvagal theory: new insights into adaptive reactions of the Type II corticosteroid receptor binding and plasma corticosteroid binding globulin
autonomic nervous system. Clevel. Clin. J. Med. 76, S86–S89. levels. Psychoneuroendocrinology 21, 95–97.
Ritchey, R., Hennessy, M., 1987. Cortisol and behavioral responses to separation in Stroebe, M., Schut, H., Stroebe, W., 2007. Health outcomes of bereavement. Lancet
mother and infant guinea pigs. Behav. Neural Biol. 48, 1–12. 370, 1960–1973.
Ruys, J., Mendoza, S., Capitanio, J., Mason, W., 2004. Behavioral and physiological Takahashi, A., Schilit, A., Kim, J., DeBold, J., Koide, T., Miczek, K.A., 2012. Behavioral
adaptation to repeated chair restraint in rhesus macaques. Physiol. Behav. 82, characterization of escalated aggression induced by GABA B receptor activation in
205–213. the dorsal raphe nucleus. Psychopharmacology 224, 155–166.
Sachser, N., Durschlag, M., Hirzel, D., 1998. Social relationships and the management Van Belle, S., Fernandez-Duque, F., Di Fiore, A., 2016. Demography and life history
of stress. Psychoneuroendocrinology 23, 891–904. of wild red titi monkeys (Callicebus discolor) and equatorial sakis (Pithecia
Sachser, N., Kaiser, S., Hennessy, M., 2013. Behavioural profiles are shaped by social aequatorialis) in Amazonian Ecuador: a 12-year study. Am. J. Primatol. 78,
experience: when, how and why. Philos. Trans. R. Soc. B Biol. Sci. 368, 1–11. 204–215.
Saltzman, W., Mendoza, S., Mason, W., 1991. Sociophysiology of relationships in Virgin, C.J., Sapolsky, R., 1997. Styles of male social behavior and their endocrine
squirrel monkeys. I. Formation of female dyads. Physiol. Behav. 50, 271–280. correlates among low-ranking baboons. Am. J. Primatol. 42, 25–39.
Sapolsky, R., Mott, G., 1987. Social subordinance in a wild primate is associated with Vogt, J., Levine, S., 1980. Response of mother and infant squirrel monkeys to
suppressed HDL cholesterol concentrations. Endocrinology 121, 1605–1610. separation and disturbance. Physiol. Behav. 24, 829–832.
Sapolsky, R., Share, L., 1994. Rank-related differences in cardiovascular function among de Waal, F., 1993. Reconciliation among primates: a review of empirical evidence and
wild baboons: role of sensitivity to glucocorticoids. Am. J. Primatol. 32, 261–275. unresolved issues. In: Mason, W., Mendoza, S. (Eds.), Primate Social Conflict.
Sapolsky, R., Share, L., 2004. A pacific culture among wild baboons: its emergence State University of New York Press, Albany, NY, pp. 111–144.
and transmission. PLoS Biol. 2, 534–541. Warne, J., Akana, S., Ginsberg, A., Horneman, H., Pecoraro, N., Dallman, M.,
Sapolsky, R., Spencer, E., 1997. Insulin-like growth factor I is suppressed in socially 2009. Disengaging insulin from corticosterone: roles of each on energy intake
subordinate male baboons. Am. J. Physiol. Regul. Integr. Comp. Physiol. 42, and disposition. Am. J. Physiol. Regul. Integr. Comp. Physiol. 296, R1366–
R1346–R1351. R1375.
Sapolsky, R., Romero, L., Munck, A., 2000. How do glucocorticoids influence stress Weiss, J., 1972. Influence of psychological variables on stress-induced pathology.
responsiveness? Integrating permissive, suppressive, stimulatory, and preparative Physiol. Emot. Psychosom. Illn. 8, 253–265.
actions. Endocr. Rev. 21, 55–89. Wingfield, J., Hegner, R., Dufty Jr., A.M., Ball, G., 1990. The “challenge hypothesis”:
Sapolsky, R., 1982. The endocrine stress-response and social status in the wild theoretical implications for patterns of testosterone secretion, mating systems, and
baboon. Horm. Behav. 16, 279–292. breeding strategies. Am. Nat. 136, 829–846.
Sapolsky, R., 1983. Individual differences in cortisol secretory patterns in the wild Yehuda, R., Bierer, L., 2008. Transgenerational transmission of cortisol and PTSD risk.
baboon: role of negative-feedback sensitivity. Endocrinology 113, 2263–2267. Prog. Brain Res. 167, 121–135.
Sapolsky, R., 1989. Hypercortisolism among socially-subordinate wild baboons origi- Yehuda, R., Halligan, S., Bierer, L., 2002. Cortisol levels in adult offspring of Holocaust
nates at the CNS level. Arch. Gen. Psychiatry 46, 1047–1051. survivors: relation to PTSD symptom severity in the parent and child. Psycho-
Sapolsky, R., 1990. Adrenocortical function, social rank, and personality among wild neuroendocrinology 27, 171180.
baboons. Biol. Psychiatry 28, 862–878. Yehuda, R., Golier, J., Halligan, S., Meaney, M., Bierer, L., 2004. The ACTH response
Sapolsky, R., 1993. The physiology of dominance in stable versus unstable social to dexamethasone in PTSD. Am. J. Psychiatry 161, 1397–1403.
hierarchies. In: Mason, W., Mendoza, S. (Eds.), Primate Social Conflict. State Young, L., Murphy Young, A., Hammock, E., 2005. Anatomy and neurochemistry of
University of New York Press, Albany, NY, pp. 171–204. the pair bond. J. Comp. Neurol. 493, 51–57.
This page intentionally left blank
1.10 Communication by Chemical Signals: Physiological Mechanisms,
Ontogeny and Learning, Function, Evolution, and Cognition
MH Ferkin, University of Memphis, Memphis, TN, USA
J delBarco-Trillo, Liverpool John Moores University, Liverpool, UK
A Petrulis, Georgia State University, Atlanta, GA, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by R.E. Johnston, J. delBarco-Trillo, volume 1, pp. 395–441, Ó 2009, Elsevier Inc.

1.10.1 Introduction 286


1.10.1.1 Brief Overview of Odor Cues and Scent Marking 286
1.10.1.1.1 Sources of Scents and Odors 287
1.10.1.1.2 The Attractiveness of Scent Marks and Conspecific Odor Preferences 287
1.10.2 Physiological Mechanisms 289
1.10.2.1 Neural Substrates 289
1.10.2.2 Mechanisms of Communication 289
1.10.2.2.1 Scent Marking 290
1.10.2.2.2 Ultrasonic Vocalizations 292
1.10.3 Development and Learning 293
1.10.3.1 Reproductive States and Odors 293
1.10.3.2 Individual Discrimination 294
1.10.3.2.1 HabituationDishabituation 295
1.10.3.2.2 Two Primary Polymorphic and Multigenic Complexes 296
1.10.3.3 Kin Recognition 297
1.10.3.3.1 Mechanisms of Kin Recognition 297
1.10.3.4 Group Discrimination 298
1.10.3.5 Species Discrimination: Competitors and Predators 298
1.10.3.6 Sex Identification 299
1.10.3.6.1 Endocrine Effects 300
1.10.3.7 Social Effects 300
1.10.3.7.1 Role of Odors in Social Dominance and Aggression 300
1.10.3.7.2 Food and Odors: Diet and Food Deprivation 302
1.10.3.7.3 Effect of Health and Environmental Stressors on Odors 303
1.10.3.7.4 Age and Odors: From Infancy to Senescence 303
1.10.4 Functions of Odor Communication 304
1.10.4.1 Scent Marking 304
1.10.4.1.1 Hypotheses about Function 304
1.10.4.2 Over-marking 306
1.10.4.2.1 Top- and Bottom-Scent Donors Are Same-Sex Conspecifics 306
1.10.4.2.2 Mixed-Sex Over-marks and the Effects of Familiarity 307
1.10.4.2.3 Ten Hypotheses about the Function of Over-marks 309
1.10.4.3 An Over-mark versus a Single Scent Mark 310
1.10.4.4 Self-Grooming as a Form of Scent Marking 311
1.10.4.4.1 Mate Attraction 311
1.10.4.5 Scent Marks Provide Public Information 311
1.10.5 Evolution 312
1.10.5.1 Basic Concepts 312
1.10.5.2 Comparative Approaches to the Study of Olfactory Communication 312
1.10.5.3 Genetic Variation, Natural Selection, and Odors 313
1.10.5.4 Odors and Sexual Selection 313
1.10.6 Cognition and the Assessment of Scent Marks 314
1.10.6.1 Processing Scent Marks 315
1.10.6.2 Cognitive Ability, Numerosity, and Scent Marks 315
1.10.7 Concluding Thoughts 316
Acknowledgments 316
References 316

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00010-9 285


286 Communication by Chemical Signals

1.10.1 Introduction marks may affect subsequent interactions with conspecifics.


Presumably, individuals may spend more time investigating
This chapter focuses on scent marking and odor communica- the scent marks of more attractive than of less attractive
tion. We provide examples of odor communication in terres- opposite-sex conspecifics. Similarly, individuals in better
trial vertebrates but focus most of our efforts on mammals, physical condition may scent mark and over-mark more
the class of vertebrates most familiar to the authors. Such an often than would individuals in worse condition (Ferkin,
endeavor was previously carried out in a text by Brown and 2011, 2015; Hurst and Beynon, 2004; Johnston and
Macdonald (1985) and by Wyatt (2014). Because of space limi- delBarco-Trillo, 2009).
tations in this chapter our focus will be less comprehensive. For many terrestrial mammals, scent marks convey informa-
However, much of the chapter will focus on scent marking tion about the donor to nearby conspecifics and heterospecifics
and, to a lesser extent, on other forms of olfactory communica- (Johnston and delBarco-Trillo, 2009; Kaur et al., 2014; Roberts
tion such as self-grooming in terrestrial mammals, which are et al., 2014; Wyatt, 2014). These scent marks are viewed as
excellent subjects for the study of such topics. This is because honest signals of the donor’s quality or condition (Ferkin,
olfaction is a major sensory modality for terrestrial mammals, 2011; Roberts, 2007; Thonhauser et al., 2013a) because
most of which display scent marking (Biben, 1980; Brashares many of the substances used as scent marks are digestive
and Arcese, 1999; Drea, 2015; Ferkin, 2015; Hurst, 1990a,b,c; exudates (Albone, 1984). Thus, scent marks from sources
Heymann, 1998; Johnson, 1973; Johnston and delBarco- such as the urine (Drickamer, 1995; Zala et al., 2004; Charlton,
Trillo, 2009; Klailova and Lee, 2014; Lewis, 2005; Macdonald, 2015), saliva (Block et al., 1981), feces (Tinnesand et al., 2015),
1980; McClintock, 2002; Sliwa and Richardson, 1998). and those from specialized glands, such as the preorbital gland
To understand odor communication in terrestrial (Ceacero et al., 2015; Pluhácek et al., 2015), anal gland (Bills
mammals, it is necessary to differentiate between studies et al., 2013), foot pads (Owen et al., 2015), submandibular
that examine proximate causation and ultimate consequences glands (Camacho-Arroyo et al., 1999; Mykytowycz, 1965),
of scent marking and then integrate the findings of these and the integument (Martin et al., 2014; McLean, 2014) accu-
disparate studies into a comprehensive picture of this rately reflect the condition and phenotype (Sabau and Ferkin,
behavior. In this chapter, our approach is to discuss advances 2013a), height (Sharpe, 2015) or genotype of the sender
and controversies in odor communication among terrestrial (Green et al., 2015). Scent marks from these multiple sources
mammals. Our approach is based on Tinbergen’s (1963) likely provide unique and overlapping information about the
four levels of behavioral analysis. Specifically, we categorized sender (Ferkin and Johnston, 1995a; Hurst and Beynon,
and discussed studies that deal with proximate causation, 2004; Johnston, 2003, 2009; Kaur et al., 2014). As a result,
such as the physiological mechanisms that underlie and the response of terrestrial mammals to scent marks and odors
mediate odor communication and the role of ontogeny and of conspecifics has remained of interest to researchers in
learning in the development of odor communication. We anthropology, biology, chemistry, psychology, neuroscience,
also identify and discuss studies that pertain to ultimate and medicine (Apps, 2013; Drea, 2015; Ferkin, 2015; Grimm,
consequences, such as the function and adaptive significance 2014; Johnston and delBarco-Trillo, 2009; Logan, 2014;
of odor communication, the benefits and costs of scent Petrulis, 2013a,b, 2015; Roberts, 2007; Roberts et al., 2014;
marking in terms of survival and fitness, and its evolution in Schulte et al., 2013; Wyatt, 2014).
terrestrial mammals. We also discuss the role of cognition in The fact that researchers from different fields are interested
odor communication. We selected papers that are recent in olfactory communication has led to confusion in the termi-
and provide new information and detail and also ‘classics,’ nology to describe chemical signals. Johnston and delBarco-
many of which provided the initial hypotheses and theoretical Trillo (2009) created a means to identify different types of
framework for current researchers studying odor communica- signals involved in odor communication. The researchers
tion in terrestrial mammals. argued that chemical signals may be categorized as being
a pheromone, a pheromone blend, or a mosaic. Accordingly,
a pheromone would be a single chemical compound, which
1.10.1.1 Brief Overview of Odor Cues and Scent Marking
would be sufficient to induce one or more responses in
In most terrestrial mammals scent marks are typically depos- the receiver. A pheromone blend would be comprised of
ited on prominent objects or along paths that are shared with several compounds in relatively specific proportions. These
conspecifics (Brown and Macdonald, 1985; Clapham et al., compounds, in the proper amount, would be necessary to stim-
2013; Sharpe, 2015). Therefore, individuals will enter areas ulate responses in the receiver. A mosaic signal contains a large
that contain single scent marks and overlapping scent marks number of chemical compounds, which are necessary to evoke
(over-marks) which are comprised of scent marks from two responses in the receiver. It is not known if a mosaic signal
or more conspecifics (Ferkin et al., 2011a,b). These scent creates an individually distinctive odor, sexually distinctive
marks may be a signal that provides particular information odor, or group (species) odors based on differences in the
about the donors to individuals that encounter such marks. proportions of these compounds across individuals. It is
These, in turn, may affect how the receivers respond to the possible that most odor signals produced by terrestrial
donors (Wyatt, 2014). Typically, individuals spend more mammals are mosaic signals as they often involve many
time investigating the scent marks of opposite-sex conspe- different compounds from multiple sources of scent. The ratio-
cifics than of same-sex conspecifics (Ferkin and Seamon, nale for this conjecture is that odor signals contain many chem-
1987). The response of individuals to scent marks as well ical compounds (Apps, 2013; Schulte et al., 2013) and are
as the location where individuals deposit their own scent derived from multiple sources of scent, including by-products
Communication by Chemical Signals 287

of hormone breakdown, excreta, and specialized tissues and meadow voles spent self-grooming in response to the odors
glands in the integument (Johnston and delBarco-Trillo, of opposite-sex conspecifics. Specifically, male and female
2009; Petrulis, 2013). meadow voles that were food deprived for either 6 or 24 h
spent less time self-grooming compared to voles that had
1.10.1.1.1 Sources of Scents and Odors continuous access to food (Hobbs and Ferkin, 2012b). It is
Some sources of scent produce marks that convey sexual infor- not clear at this time why food deprivation and restriction
mation about the donor. had different effects on self-grooming and scent marking in
For example, meadow voles, Microtus pennsylvanicus, have voles. Perhaps, the difference lies in the fact that voles self-
a highly localized pattern of sexual information on their bodies groom when they encounter the scent marks of conspecifics
during the breeding season (Ferkin and Johnston, 1995a). The that are in close proximity, whereas voles scent mark when
feces, anogenital area, and urine scent marks of meadow voles they encounter the scent marks of conspecifics that may be
are attractive to opposite-sex conspecifics but not to same-sex nearby and when they are distant (Hobbs and Ferkin, 2012b;
conspecifics. However, saliva/mouth secretions of female voles Hobbs et al., 2008).
were attractive to male but not to female conspecifics, whereas Given the importance of finding potential mates of the
the posterolateral region scent marks of male voles were attrac- opposite sex and competing with rivals of the same sex, identi-
tive to both male and female conspecifics. For meadow voles, fication of males and females is probably universal in all
the secretions from the mouth and posterolateral region likely species that reproduce sexually. Chemical signals often provide
provide different information than that from the feces, anogen- crucial information about the sex of conspecifics; a few exam-
ital area, and urine (Ferkin and Johnston, 1995a). ples of species in which individuals have been shown to be
Similarly, the chemical signals for discrimination and recog- more attracted to odors of opposite-sex conspecifics than those
nition of individuals can come from many different specific of same-sex conspecifics include many species of terrestrial
sources, such as a variety of specialized scent glands (e.g., seba- mammals (Eisenberg and Kleiman, 1972; Ferkin and Johnston,
ceous glands, apocrine and eccrine sweat glands) as well as 1995a,b; Johnston, 1983). Male mice (Mus musculus) respond
urine and feces. In Syrian hamsters, Mesocricetus auratus, for with a surge in luteinizing hormone when exposed to female
example, there are five different sources of individually distinc- mouse odors but not when exposed to male mouse odors or
tive scents, namely flank gland, vaginal secretions, ear glands female hamster odors (Maruniak and Bronson, 1976). Such
(inside the pinna), urine, and feces (Johnston et al., 1991). hormonal responses indicate that males discriminate sex
Six other potential sources of odors in Syrian hamsters were from odors even if a behavioral preference cannot be detected.
tested but were not found to be individually distinctive, as In most species, there may be several distinct sources of odor
measured by habituationdishabituation tests (fur from the that provide information about sex and/or reproductive state,
midline ventral surface, fur from the dorsal surface between although not all odors necessarily contain such information.
the shoulders, saliva, feet, fur behind the ears, and the flank- For example, in Djungarian hamsters, males showed a prefer-
gland area from flank-glandectomized males) (Johnston ence for the odors of receptive females over the odors of males
et al., 1991). Similar results were found with Djungarian when the odor source was urine, anogenital secretions, saliva,
hamsters, Phodopus campbelli. Male hamsters were able to or midventral gland secretion but not when the odor source
discriminate individual differences between other males using was feces or secretions from the feet (Lai et al., 1996).
scents from the midventral gland, urine, feces, mouth, and Several studies have shown that the attractiveness of the
the sacculi from the corner of the mouth (Lai and Johnston, scent marks of spontaneous ovulators also varies temporally
1994). These different odor sources that provide information (Brown and Macdonald, 1985). The changes in the attractive-
about individual identity may contain some redundant infor- ness of a sender’s scent mark appear to be concomitant with
mation, but the combined information from of all of these changes in the responses of receivers to such marks (Ferkin,
sources may facilitate the creation of stronger memories about 2011; Johnston, 2009; Roberts, 2007; Sabau and Ferkin,
the identity of a given conspecific. 2013a). The scent marks of female house mice and Syrian
hamsters are attractive to male conspecifics only when the
1.10.1.1.2 The Attractiveness of Scent Marks and Conspecific female is in estrus (delBarco-Trillo et al., 2009c; Johnston,
Odor Preferences 1983; Kavaliers et al., 1994; Figure 1). Postpartum estrus
The attractiveness of scent marks from the above sources can female mammals produced odors and scent marks that were
vary according to, for example, the diet and reproductive condi- more attractive to males than were those produced by females
tion of senders (Boonstra, 1994; Wade et al., 1996). This can be not in postpartum estrus (Ferkin and delBarco-Trillo, 2014;
reflected in the attractiveness of its scent marks to receivers Ferkin and Johnston, 1995b; Lai et al., 1996; Witt et al.,
(Pierce et al., 2005, 2007; Sabau and Ferkin, 2013a; Sabau 1990; Zeigler et al., 1993). Postpartum estrus female voles, rela-
et al., 2014). Scent marks also provide honest signals of health tive to females not in postpartum estrus, also spent more time
(Zala et al., 2004) and nutritional status of the sender to the and showed increases in scent marking, over-marking, and self-
receivers (Hobbs and Ferkin, 2011a,b; Pierce et al., 2005). grooming when they encountered the scent marks of males
Nutritional stresses such as food deprivation and food restric- (Ferkin and Leonard, 2010; Ferkin, 2011).
tion are an ecological challenge faced by small herbivores In addition to showing a preference for odors of opposite-
that live in transitional grasslands, where food sources are sex individuals, male mammals usually show a preference for
patchy and vary in quality across the territories of female odors of females that are sexually receptive over those of
conspecifics (Batzli, 1985; Bronson, 1989; Getz, 1985). Food females that are in other reproductive states (Brown, 1985).
availability affected the amount of time male and female For example, male meadow voles prefer the odors of females
288 Communication by Chemical Signals

Figure 1 Habituationdishabituation test. A male Syrian hamster was exposed to vaginal secretions of a donor female during four habituation trials
(‘Hab 1’ to ‘Hab 4’) and consequently to the vaginal secretions of a second donor female during the dishabituation trial (‘Test’). The four panels
represent four scenarios in which: (a) both female donors were in estrus; (b) both female donors were in diestrus-2 (i.e., not in estrus); (c) the first
female was in diestrus-2 and the second female in estrus; (d) the first female was in estrus and the second female in diestrus-2. A dishabituation
response (a significant increase in investigation time in the dishabituation trial compared to the last habituation trial) was observed only when the
second donor female was in estrus. E, estrus; Di, diestrus-2; *p < 0.05; **p < 0.01; ***p < 0.001. Reproduced from delBarco-Trillo, J., LaVenture,
A.B., Johnston, R.E., 2009. Male hamsters discriminate estrous state from vaginal secretions and individuals from flank marks. Behav. Process. 82,
18–24, with permission from Elsevier.

in postpartum estrus (when females are in a state of heightened the important point that the ability to discriminate between
sexual receptivity) than those of females in induced estrus or odors of males and females does not necessarily lead to func-
undergoing pregnancy (Ferkin and delBarco-Trillo, 2014). tionally relevant responses.
Sexually experienced female rats, Rattus norvegicus, prefer odors The preference for opposite-sex conspecifics is also often
of normal males over the odors of castrated males (Carr et al., dependent on the reproductive state of the receiver. In domestic
1965). Similarly, male rats prefer odors of receptive females rats, males prefer the odors of estrous females to those of
over odors of nonreceptive females. These preferences, females in other reproductive states but castrated males do
however, may depend on knowledge that males have learned not show such preference (Carr et al., 1965; Randall, 1986).
during previous interactions with females. For example, sexu- Similarly, the preference that diestrous, estrous, and lactating
ally experienced male rats prefer odors of receptive females female Syrian hamsters show for the odors of intact male
over odors of nonreceptive females but sexually naïve males hamsters over castrated males is not shown by pregnant
do not show this preference (Carr et al., 1965). Yet, the ability females (Johnston, 1979).
of males to discriminate between male and female and the Familiarity of a female with a male and/or his odors can
male preference for female odors may be two distinct mecha- also be a determining factor in her odor preference. A female
nisms. Experiments with house mice showed that males with repeatedly exposed to odors of a male may show a preference
the vomeronasal organ removed did not show a preference for that male over a male that is unfamiliar, possibly because
for the odors of estrous females over odors of males (Pankevich familiarity indicates the ability to defend a territory or living
et al., 2004). However, these males were still able to discrimi- area for some period of time. Female pygmy loris, Nycticebus
nate between odors of males and estrous females when tested pygmaeus, in captivity were first exposed to urine marks from
in a habituationdishabituation method. These results make one male over 14–20 weeks. When the female was close to
Communication by Chemical Signals 289

ovulation, males confined in small cages were placed in the


much larger living areas of females. Using three different behav-
ioral measures, females showed strong preferences for the
familiar male (Fisher et al., 2003a). In another example, female
collared lemmings were first housed with a male for 30 days
and were then separated for 1, 12, or 24 days. Females showed
a strong preference for their partners’ odors over the odors of an
unfamiliar male 12 days after separation, but they did not
show such a preference after 24 days of separation (Huck and
Banks, 1979). Female rats also show a preference for the odors
of a previous, familiar mate over the odors of a novel male. In
contrast, male rats show a preference for the odors of a novel
female over the odors of a previous mate (Carr et al., 1980).
In socially monogamous species one predicts that mated indi-
viduals will not only be able to recognize their partner from
other conspecifics but also that they will show a preference
for the partner’s odors compared to those of unfamiliar,
opposite-sex individuals (Duarte et al., 2015).

1.10.2 Physiological Mechanisms


1.10.2.1 Neural Substrates
Most social odors are processed by two complimentary chemo-
sensory systems, the main (MOS) and accessory olfactory
(AOS) system (Baum and Kelliher, 2009) as represented in
Figure 2. AOS sensory neurons are located in the vomeronasal Figure 2 Abbreviated schematic of main olfactory (MOS; gray) and
organ (VNO), a blind-ended tube that primarily takes up con- accessory olfactory or vomeronasal (AOS; white) systems along with
integrative areas (black). For clarity, only unidirectional olfactory bulb
tacted, nonvolatile, chemosignals (Chamero et al., 2012;
connections are presented and several areas without known relevance
Halpern and Martinez-Marcos, 2003; Meredith, 1994; Zufall
to social odor processing are omitted. AOB, accessory olfactory bulb;
and Leinders-Zufall, 2007). In contrast, sensory neurons of ACo, anterior cortical amygdala; BNST, bed nucleus of stria terminalis;
the MOS (main olfactory epithelium: MOE) are located within END, endopiriform nucleus; ENT, entorhinal cortex; HIPP, hippo-
the nasal turbinates and respond primarily to volatile chemo- campus; MA, medial amygdala; MDth, mediodorsal thalamus; MOB,
signals via sniffing (Murthy, 2011; Wachowiak, 2011). These main olfactory bulb; MOE, main olfactory epithelium; MPOA, medial
systems also have separate projection zones within the olfac- preoptic area; OFC, orbitofrontal cortex; PIR, piriform cortex; PMCo,
tory bulbs (OB): MOE neurons project to main olfactory bulbs posteromedial cortical amygdala; VMH, ventromedial hypothalamus;
(MOB) and the VNO neurons project to the accessory olfactory VNO, vomeronasal organ; VPH, ventral premammilary hypothalamus.
bulbs (AOB) (Scalia and Winans, 1975). The separation of AOS Reproduced from Petrulis, A., 2013. Chemosignals, hormones and
mammalian reproduction. Horm. Behav. 63, 723–741, with permission
and MOS is extended further into the brain systems such that
from Elsevier.
there are few areas of overlap, primarily in the anterior part
of the medial amygdala (MA) (Fan and Luo, 2009; Kang
et al., 2009; Mohedano-Moriano et al., 2012) or through indi- Newman, 1999; Wood, 1997) that provide a substrate where
rect connections in the cortical amygdala (MOB input, projects steroid hormones and chemosensory systems may interact
to MA) and posterior bed nucleus of the stria terminalis (BNST; (Been and Petrulis, 2011; Maras and Petrulis, 2010a). Impor-
AOB input, projects to MA) (Maras and Petrulis, 2010a; tantly, these interconnected brain regions form the core of
Martinez-Marcos, 2009). Indeed, the MOS can alter how the the “social behavior neural network” (SBNN), a highly
AOS processes social odors. For example, MOB inputs to the conserved system of interconnected, steroid-sensitive and sexu-
anterior MA alter how the AOB responds to volatile phero- ally dimorphic limbic structures that control multiple social
mones (Martel and Baum, 2009) and the detection of volatile behaviors (Newman, 1999; O’Connell and Hofmann, 2012).
odors by the MOS triggers VNO sampling, thereby increasing
chemosignal access to the VNO (Meredith, 1998).
The MA is not only the primary area of chemosensory inte-
1.10.2.2 Mechanisms of Communication
gration, it is also a steroid-sensitive and sexually dimorphic
structure (Cooke, 2006) as are its connected structures, such Chemosensory cues from conspecifics guide almost every kind
as the BNST, medial preoptic area (MPOA), anterior hypothal- of mammalian social behavior studied to date, including
amus (AH), and ventromedial hypothalamus (VMH) (Segovia aggressive, sexual, and parental behaviors, and the physiolog-
and Guillamon, 1993; Simerly, 1990; Wood, 1997). Detailed ical control of these behaviors is described elsewhere (Petrulis,
analysis of MA and its connected structures shows that steroid 2013). Here we focus on the hormonal and neural control of
and chemosensory information are represented by partially behaviors representing productive (scent marking, vocaliza-
separate and parallel systems (Maras and Petrulis, 2010a,b; tions) aspects of odor communication. The vast majority of
290 Communication by Chemical Signals

detailed information on this topic comes from the study of lesions have no effect on the ability of AVP injections in AH
only a few species: the Syrian hamster, the Mongolian gerbil, to drive flank marking (Ferris et al., 1994). This strongly
the house mouse, the Norway rat, and the rabbit. Conse- suggests that AH is downstream of LS control and is the
quently, our coverage will be constrained to these species, but primary regulator of AVP-driven flank marking, perhaps via
it should be noted that neural studies with similar findings midbrain structures (Hennessey et al., 1992).
have been reported in other species, such as domestic cats Initial reports demonstrated that both flank marking and
(Hart and Voith, 1978; Lischka et al., 2008) and dogs (Hart, V1a receptors (but not AVP itself) in MPOA/AH are dependent
1974; Gadbois and Reeve, 2014; Jia et al., 2014). on gonadal steroids, especially androgens (Albers and
When rodents of either sex investigate conspecific odor cues, Rowland, 1989; Albers and Prishkolnik, 1992; Albers et al.,
they often deposit their own odor cues through stereotyped 1988, 1996; Huhman and Albers, 1993; Young et al., 2000).
scent-marking responses and/or vocalize, often in the ultra- However, more recent work suggests a more complicated
sonic range (Brown and Macdonald, 1985; Petrulis, 2013; picture. Although male hamsters in a nonbreeding photope-
Pierce et al., 1989; Wyatt, 2014). Both of these behaviors serve riod have regressed testes and low levels of circulating testos-
to facilitate localization of mates over different temporal and terone, flank marking levels are testes-independent and not
spatial ranges (Pfaff et al., 2008). Although the details may reduced during social encounters, despite steroid-dependent
vary as to exact dominant vocal frequencies or the postural reductions in V1a receptors (Caldwell et al., 2008).
and chemical nature of deposited scent marks, there is remark- The AVP manipulations within AH alter flank marking irre-
able consistency in the chemosensory, hormonal, and neural spective of social status, but they do not have lasting effects on
substrates of these behaviors across model species. Indeed, dominance interactions (Ferris et al., 1986b), suggesting that
the aggregate evidence, described below, suggests that both AH AVP primarily regulates behavioral output rather than stim-
scent marking and vocalizations are driven by a steroid- ulus evaluation or dominance/subordination itself. So, what
dependent neural network composed of the chemosensory then is the source of social odor information to the AH?
inputs to the extended medial amygdala (MA, BNST), its Initially, this information comes from both the MOS and
outputs to basal forebrain (MPOA, AH, VMH) and connected AOS representations in the olfactory bulb as olfactory bulb
midbrain elements (periaqueductal gray, PAG). removal eliminates flank marking (Kairys et al., 1980). Periph-
eral lesions of the MOS reduce male and female flank marking
1.10.2.2.1 Scent Marking in arenas scented with the odors of both male and female
1.10.2.2.1.1 Syrian Hamster: Flank Marking conspecifics (Johnston, 1992; Johnston and Mueller, 1990).
Unlike species discussed below, both male and female Although VNO removal does not reduce flank marking in
hamsters deposit products of dorsal flank sebaceous scent this context, excising the VNO in females reduces preferential
glands on vertical surfaces, a behavior termed flank marking targeting of this behavior toward other females, without
(Johnston, 1975a, 1977). Flank marking is thought to reflect altering their low baseline response to male odors (Petrulis
territorial or competitive motivation as it occurs primarily in et al., 1999). As the MA serves as the main point of convergence
response to same-sex odors and following establishment of between MOS and AOS, it is perhaps unsurprising that
dominantsubordinate relationships (Johnston, 1975b, complete lesions of MA virtually eliminate flank marking in
1977). This behavior has been extensively investigated at the females (Petrulis and Johnston, 1999); smaller lesions of either
neural level following the accidental discovery that MPOA anterior or posterodorsal MA result in reductions rather than
injections of arginine-vasopressin (AVP) injections elicit excep- outright elimination of this behavior (A.P., unpublished obser-
tional levels of flank marking in both males and females vations). As the MA has substantial inputs to MPOA/AH, it is
(Albers, 2015; Ferris et al., 1984). Both AVP- and odor- likely that odor modulation of flank marking is mediated by
induced flank marking is blocked by MPOA injections of AVP MA (and perhaps cortical amygdala; (Petrulis et al., 2000) effer-
V1a receptor antagonists (Albers et al., 1986). The key locations ents to AVP-sensitive regions of AH; this awaits further
within MPOA that trigger AVP-induced marking are primarily investigation.
in the ventral parts of the anterior hypothalamus (AH) (Ferris
et al., 1986a) and the source of the AVP appears to be from 1.10.2.2.1.2 Syrian Hamster: Vaginal Marking
nonpituitary projecting nucleus circularis (NC) cells within Female hamsters scent mark (vaginal mark) to advertise
AH and the medial supraoptic nucleus (mSON) (Delville impending sexual receptivity (Johnston, 1977) similar to rats,
et al., 1998; Ferris et al., 1990b, 1992; Mahoney et al., 1990). house mice, and rabbits (Birke, 1984; Chang et al., 2000;
Indeed, subordinate hamsters (that do not mark) have lower González-Mariscal et al., 1990; Rich and Hurst, 1999). Vaginal
levels of AVP in NC (Ferris et al., 1989) and these cells (and marking is a stereotyped behavior that deposits sexually attrac-
mSON) show increased activity during aggressive interactions tive vaginal fluid on the surface (Been and Petrulis, 2008; Been
(Delville et al., 2000). Further studies have indicated that et al., 2012) and is specifically targeted toward male odors
AVP action within the AH requires excitatory glutamatergic (Johnston and Brenner, 1982; Johnston, 1977; Petrulis and
neurotransmission (Bamshad et al., 1996) and that it is Johnston, 1997). This behavior changes substantially across
antagonized by serotonin action (Albers et al., 2002). Input the female’s estrous cycle (Figure 3) such that females mark
to the AH from the broader AVP-sensitive brain also regulates most on the night prior to sexual receptivity (during high circu-
flank marking as LS/BNST lesions reduce marking and AVP lating estrogen levels) and then show no vaginal marking
injections in lateral septum (LS), or in nearby BNST, trigger it during sexual receptivity (during high progesterone levels)
(Ferris et al., 1990a). Lesions of AH block the induction of (Johnston, 1977; Lisk and Nachtigall, 1988). This remarkable
flank marking by AVP injections in LS/BNST. In contrast, LS hormone-dependent shift in vaginal marking is likely regulated
Communication by Chemical Signals 291

(a) Diestrous day 2 (b) Proestrus


30
Male
*

Number of vaginal marks


25 Female

20

15 *
10

0
SHAM BNST-X SHAM BNST-X

Figure 3 Mean (SEM) number of vaginal marks to male and female odors. SHAM, but not BNST-X, females preferentially marked at higher levels
to male odors compared to female odors on both (a) diestrous day 2, and (b) proestrus. *p < 0.01, male versus female odor condition.

by the steroid action on MPOA and VMH: E implants in these and its estrogenic metabolites (Juntti et al., 2010; Kimura and
areas restore vaginal marking in OVX females (Takahashi et al., Hagiwara, 1985; Wu et al., 2009). These steroid effects appear
1985). However, the VMH may not be critical for vaginal to be exerted on MPOA/AH, BNST, or VMH neurons as testos-
marking, as VMH lesions do not eliminate the behavior terone or estradiol implants near or in these structures can
(Floody, 2002). The role of the MPOA is presently unclear as maintain urine marking in gonadectomized males (Matochik
large, fiber-damaging MPOA lesions disrupt vaginal marking et al., 1994; Nyby et al., 1992), perhaps through interactions
(Malsbury et al., 1977), whereas smaller, fiber-sparing lesions with midbrain dopamine systems (Sipos and Nyby, 1996).
limited to MPOA do not (Martinez and Petrulis, 2013). This The available evidence suggests that the MA is not a substrate
discrepancy is likely due to the concomitant damage to BNST for steroid effects on urine marking (Sipos and Nyby, 1998;
present in the earlier study, as fiber-sparing lesions confined Unger et al., 2015). Recent evidence suggests that central
to the posterior BNST impair vaginal marking (Martinez and oxytocin decreases urine marking and thereby provides
Petrulis, 2011) without altering the cyclicity of this behavior a possible chemical mechanism through which marking can
(Figure 3). be decreased in subordinate animals (Arakawa et al., 2015).
As is the case for flank marking, olfactory bulbectomy Surprisingly, little is known as to how chemosensory informa-
reduces vaginal marking (Kairys et al., 1980) and the disruptive tion is transmitted to steroid-sensitive structures. What
effects of MOS peripheral lesions on vaginal marking are much evidence exists suggests that urine marking is partially medi-
more pronounced than those following VNO removal (John- ated by the AOS (MOS contribution not yet assessed) as evi-
ston, 1992; Petrulis et al., 1999). MOS inputs to the MA, rather denced by decreased marking in males following VNO
than to other MOS targets (Petrulis et al., 1998, 2000), appears removal (Labov and Wysocki, 1989; Maruniak et al., 1986).
to regulate vaginal marking as MA lesions lead to significant As in many other social contexts, removal of VNO has a greater
reductions in vaginal marking (Petrulis and Johnston, 1999; effect in sexually naïve animals than in males with sexual expe-
Takahashi and Gladstone, 1988). Interestingly, females with rience (Clancy et al., 1984; Labov and Wysocki, 1989; Maru-
MA lesions still show differentially high marking rates to niak et al., 1986).
male odors and still retain normal cyclic variation in vaginal
marking. This suggests that MA is not critical for hormonal
1.10.2.2.1.4 Mongolian Gerbil: Ventral Marking
control of marking (likely mediated by hypothalamic struc-
Dominant male gerbils engage in more scent marking than do
tures) or differential marking responses. In contrast, the BNST
subordinate male gerbils. Much of this scent marking involves
is critical for differential marking to male odors (Martinez
the ventral gland. Briefly, individuals rub odors from this
and Petrulis, 2011). This effect is likely mediated by oxytocin
ventral sebaceous scent gland onto the substrate (Thiessen
action because blockade of oxytocin receptors in BNST and
et al., 1968, 1970; Whitsett and Thiessen, 1972; Yahr, 1977).
MPOA reduces male-directed vaginal marking (Martinez
Adult gonadectomy reduces both male and female marking
et al., 2010, 2013).
in gerbils and can be reinstated with administration of testos-
terone or its metabolites in males (Yahr and Stephens, 1987;
1.10.2.2.1.3 House Mouse: Urine Marking Yahr et al., 1979) or estradiol plus progesterone in females
Urine marking by socially dominant male mice is generally (Owen and Thiessen, 1974; Yahr and Thiessen, 1975). The
used in the context of territorial defense (Arakawa et al., sex difference in overall levels of marking is due to organiza-
2008a,b; Desjardins et al., 1973; Lumley et al., 1999). However, tional effects of gonadal steroids as neonatal testosterone and
male urine marking does not appear to be differentially tar- subsequent adult T in females will increase female marking
geted toward males versus females (Hurst, 1990a,b,c; Labov to male levels (Ulibarri and Yahr, 1988, 1996). Testosterone
and Wysocki, 1989), suggesting a more general advertising appears to exert its effect via the MPOA; implants near the
function. Nevertheless, males do urine mark at much greater most rostral or caudal limits of MPOA are most effective in
levels than do females, an effect largely driven by sex differ- restoring marking in gonadectomized males (Thiessen and
ences in adult and developmental exposure to testosterone Yahr, 1970; Thiessen et al., 1973; Yahr et al., 1982). Indeed,
292 Communication by Chemical Signals

large MPOA lesions that include these areas reduce ventral (USV) specifically during investigation of novel females or their
marking in males (Yahr et al., 1982) particularly if these lesions urine; males or their urine do not elicit USVs from other males
remove the sexually dimorphic cell groups in caudal MPOA or females (Guo and Holy, 2007; Nyby et al., 1977; Nyby,
and BNST (Commins and Yahr, 1984). However, small fiber- 1983b; Wang et al., 2008; Warburton et al., 1989; White
sparing lesions of these cell groups had no effect on ventral et al., 1998) and so these USVs are often considered to be
marking (Yahr and Gregory, 1993) indicating that previous male-typical promoters of opposite-sex interactions (Chabout
effects were due to damage to passing axon fibers. Further et al., 2015; Musolf et al., 2010; Pomerantz et al., 1983; Sugi-
attempts at identifying brain regions that regulate ventral moto et al., 2011). Female mice do produce a modest amount
marking have not been successful: small, fiber-sparing lesions of USVs during female–female mutual investigation (Moles
of posterodorsal MA, medial preoptic nucleus, or posterome- et al., 2007) but, unlike female-directed calling by males, this
dial BNST were without effect on ventral marking (Heeb and female-directed calling habituates rapidly (Maggio and
Yahr, 2000; Sayag et al., 1994). Whitney, 1985; Warburton et al., 1989).
Removal of olfactory bulbs in males eliminates USVs to
1.10.2.2.1.5 Rabbits: Chinning female odors and similar or lesser effects are also observed after
Studies have examined the mechanisms underlying chinning, AOS damage, especially in sexually naïve animals (Bean, 1982;
a type of scent marking in rabbits. For example, González- Wysocki et al., 1982). Females with genetically induced VNO
Mariscal et al. (1990) found that the frequency of chinning dysfunction display increased USVs to males suggesting that
varied with the female’s reproductive cycle. Moreover, during the VNO may normally suppress male-typical behavior in
estrous there were more acini/field than during pregnancy female mice (Kimchi et al., 2007; Stowers et al., 2002, but
and lactation (Cerbón et al., 1996). Gonadectomy reduced see Pankevich et al., 2004). Data on the role of the MOS in
the number of acini/field and increased their diameter in USV generation is more equivocal: peripheral lesions of MOS
females but had the opposite effect in males (Cerbón et al., are reported to either have no effect (Bean, 1982) or to reduce
1996). Hudson et al. (1990) found that estradiol treatment USVs (Sipos et al., 1995).
was necessary to increase chinning in ovariectomized rabbits. Given the sex difference in USV production, it is perhaps not
Interestingly, progesterone treatment given to estradiol- surprising that male USVs are strongly steroid-dependent. Male
primed, ovariectomized females induced an immediate USVs are eliminated by adult gonadectomy and restored, and
suppression of chin marking and lordosis, but progesterone even induced in females, by circulating testosterone or its estro-
therapy alone had no effect on these measures in ovariecto- genic/androgenic metabolites (Bean et al., 1986; Juntti et al.,
mized female rabbits (Hudson et al., 1990). González- 2010; Matochik and Barfield, 1991; Nunez et al., 1978;
Mariscal et al. (1993) found that chinning in males depends Pierman et al., 2008; Wu et al., 2009). The male-biased pattern
on testicular steroids, as castration decreased the frequency of of USV production does not appear to require organizational
chinning. This behavior was nearly restored to precastration hormone action, as neonatal castration does not alter male-
levels by treatment with testosterone propionate, whereas the typical patterns of calling (Warburton et al., 1989). The identi-
administration of estradiol and dihydrotestosterone increased fied brain sites of gonadal steroid action on USVs (MPOA/AH,
chinning above precastration levels (Gonzalez-Mariscal et al., BNST, or VMH) are the same ones identified for urine marking
1993). Melo et al. (2008) examined the brain sites where as testosterone or estradiol implants around these structures
steroids act to stimulate chinning. They did so by implanting maintains USVs in gonadectomized males (Matochik et al.,
testosterone propionate or estradiol benzoate into the ventro- 1994; Nyby et al., 1992), perhaps interacting with midbrain
medial hypothalamus (VMH) or the medial preoptic area dopamine systems (Sipos and Nyby, 1996). However, small
(MPOA) of gonadectomized male and female rabbits, respec- lesions to MPOA do not change USVs (Bean et al., 1981) and
tively. Melo et al. (2008) found that, for female rabbits, so suggests that MPOA steroid implants are effective because
chinning was stimulated by EB implants into the VMH or of steroid spread to nearby areas, such as BNST. The available
MPOA. In contrast, for males, chinning was stimulated by evidence suggests that the MA is not a substrate for steroid
testosterone implants into the MPOA, but not into the VMH effects on USVs (Sipos and Nyby, 1998; Unger et al., 2015),
(Melo et al., 2008). Camacho-Arroyo et al. (1999) discovered but MA’s broader role in regulating USVs has not been assessed.
progesterone and estrogen receptors in the nucleus of acini
cells of the submandibular (chin) glands of rabbits of both 1.10.2.2.2.2 Syrian Hamster
sexes. Intact males had a smaller number of estrogen Both estrous female and male hamsters (Floody et al., 1977,
receptor-immunoreactive cells and progesterone receptor- 1979a; Johnston and Kwan, 1984) produce USVs in response
immunoreactive cells than did estrous females. Progesterone to opposite-sex conspecifics or their odors; this response is
receptor-immunoreactive cells were more numerous in steroid-dependent in both sexes (Floody and Petropoulos,
estrous than they were in ovariectomized rabbits. Estradiol 1987; Floody et al., 1979a,b, 1987). Hamster USVs to odors
therapy increased the number of progesterone receptor- requires intact olfactory bulbs (Kairys et al., 1980) and either
immunoreactive cells in ovariectomized female rabbits a functioning MOS or AOS (Johnston, 1992). Lesions of the
(Camacho-Arroyo et al., 1999). MA leads to deficits in USV production by female hamsters,
suggesting that MOS/AOS chemosensory information
1.10.2.2.2 Ultrasonic Vocalizations converge in this structure (Kirn and Floody, 1985). It is likely
1.10.2.2.2.1 House Mouse that MA projections to MPOA/AH or BNST are critical for this
In mice, reproductively active and territorial/dominant adult behavior as damage to MPOA/AH impairs USVs (Floody,
males produce high amounts of ultrasonic vocalizations 1989) as do knife cuts that damage BNST connections
Communication by Chemical Signals 293

(Miceli and Malsbury, 1982). Similar to studies in other Postpartum estrus occurs in many species but it is especially
species (Jürgens, 2009), hamster vocalizations require prevalent in small-sized rodents (Ferkin and delBarco-Trillo,
processing by the periaqueductal gray (PAG) region of 2014) and in lagomorphs (i.e., rabbits, hares, pikas). Female
the midbrain (Floody and DeBold, 2004; Floody and meadow voles in postpartum estrus are more sexually receptive
O’Donohue, 1980) via a steroid-sensitive mechanism (Floody and likely to become pregnant than females in other estrous
et al., 1986). states (Ferkin and delBarco-Trillo, 2014). These females in
postpartum estrus also deposit more scent marks, advertising
1.10.2.2.2.3 Norway Rat their heightened sexual state to males, who show a preference
Both male and estrous female rats emit USVs (50 kHz) during for the odors of postpartum-estrus females compared to those
interactions with opposite-sex individuals or their odors (Geyer of females in other estrous states (Ferkin and delBarco-Trillo,
and Barfield, 1978; White et al., 1991). Male USVs are depen- 2014). In contrast, in mice and prairie voles (Microtus
dent on both androgen- and estrogen-receptor activity ochrogaster), females do not increase scent marking when in
(Matochik and Barfield, 1991; Vagell and McGinnis, 1998). postpartum estrus (Coquelin, 1992; Ferkin and delBarco-
Testosterone action within the AH and/or VMH appears to be Trillo, 2014), possibly because females in these species nor-
necessary for USVs in males (Harding and McGinnis, 2004) mally mate with the same partner, who may be familiar with
even though it is not sufficient (Harding and McGinnis, the female’s state of sexual receptivity.
2003). Indeed, small lesions in VMH inhibit the ability of testos- Different sources of female odor may contain information
terone to restore USVs in gonadectomized male rats (Harding about her sexual receptivity. Male Syrian hamsters show high
and McGinnis, 2005) suggesting that the VMH is one of the crit- levels of copulatory behavior when all of a female’s odors are
ical steroid-sensitive brain regions for USVs. Other structures present but they show declining levels of copulatory behavior
may also be involved in regulating USVs: pharmacological stim- as specific odor sources are removed (Johnston, 1986). Elimi-
ulation of the BNST or MPOA/AH increase 50 kHz USVs in nation of vaginal secretions significantly reduced male copula-
males (Burgdorf et al., 2007; Fu and Brudzynski, 1994; Wintink tory behavior and removal of flank, ear, and Harderian
and Brudzynski, 2001), likely via their direct connections with secretions caused additional decreases in copulatory behavior.
midbrain vocal centers (Kyuhou and Gemba, 1998). Therefore, these four different odor sources carry information
about the attractivity of females, influencing male sexual
arousal and performance (Johnston, 1986). Similarly, male
1.10.3 Development and Learning Djungarian hamsters show a preference for the urine, saliva,
and vaginal secretion of estrous females over similar odors of
1.10.3.1 Reproductive States and Odors
diestrous females, whereas a preference is not shown when
In many mammalian species females approaching or in estrus males investigate other types of body odor, such as feces and
may release odors or increase their scent marking to advertise midventral gland secretions, even though these odors are
their sexual receptivity and thus attract mates. For example, important in other aspects of communication (Lai et al.,
female Syrian hamsters increase their vaginal and flank 1996). Also in Djungarian hamsters, the attractiveness of three
marking immediately prior to sexual receptivity, concentrating different odors of females varied across the estrous cycle, each
those scent marks around the dominant male territories and odor being maximally attractive at a different stage of the cycle
also creating a trail of scent marks between the burrow of (Lai and Johnston, 1994). That is, different odors from the
a dominant male and her own burrow (Huck et al., 1985a). same female may be most attractive at different times during
Several mechanisms may be used for advertising sexual recep- the estrous cycle. This type of pattern is especially interesting
tivity. The quantity of secretions produced and available for because it might allow males to accurately track and predict
scent marking may be increased, the frequency of scent the timing of estrous.
marking may increase, the composition of odors may change It has already been mentioned that males show a preference
with the female’s reproductive state, or a combination of these for the odors of sexually receptive females. Such preference for
mechanisms may be employed. With the first two mechanisms odors of receptive females may be universal and has indeed
there need not be a change in the quality of the odor, but in been shown to exist in many species, including the house
most cases both the quality and the quantity of odors may mice (Hayashi and Kimura, 1974), brown lemmings, Lemmus
change. Indeed, given that metabolic products of sexual trimucronatus, and collared lemmings, Dicrostonyx groenlandicus
hormones are likely to be contained in the female’s odors, (Huck and Banks, 1984), meadow voles (Ferkin and Johnston,
such odors can be a direct reflection of the reproductive state 1995a), Indian desert gerbils, Meriones hurrianae (Kumari and
of the female and the attractiveness of female odors. Prakash, 1984), Mongolian gerbils (Block et al., 1981), wood-
As a further example of how females advertise their sexual rats, Neotoma lepida (Fleming et al., 1981), Columbian ground
receptivity through odors, we can consider those species in squirrels, Spermophilus columbianus (Harris and Murie, 1984),
which females undergo postpartum estrus. Postpartum estrus dogs, Canis lupus familiaris (Beach and Gilmore, 1949; Dunbar,
is a relatively short period of heightened sexual receptivity 1977), rams, Ovis aries (Lindsay, 1965), and pygmy marmosets,
that occurs immediately after parturition. Females that mate Cebuella pygmaea (Converse et al., 1995). A lack of a preference
and become pregnant during postpartum estrus gestate a set for odors of estrous females over odors of nonestrous females is
of offspring while they are nursing the previous litter. As difficult to interpret and may be due to methodological short-
a consequence, postpartum estrus allows females to be simul- comings, but it has been reported in some species such as
taneously pregnant and lactating, maximizing the number of deer mice, Peromyscus maniculatus (Dewsbury et al., 1986), and
litters that they can produce during the breeding season. guinea pigs, Cavia porcellus (Nyby, 1983a). Indeed, in species
294 Communication by Chemical Signals

that have been studied by many investigators, such as mice, rats, males, but not to water containing females or males without
and Syrian hamsters, some studies report male preferences for breeding glands. Males are not attracted to any of these odors,
estrous females (Carr et al., 1965; Johnston, 1980; Lydell and indicating that the attractant is specific to females and not
Doty, 1972), whereas other studies report a lack of such a pref- a general signal that promotes aggregation (Pearl et al.,
erence (Brown, 1977; Kwan and Johnston, 1980; Landauer 2000). Sexually mature, terrestrial female toadlets, Pseudo-
et al., 1978; Taylor and Dewsbury, 1990). Such differing results phryne bibronii, tested in a two-choice Y-maze, were attracted
are probably due to differences in methodology (delBarco-Trillo to the odors deposited by males compared to the clean arm
et al., 2009c; Taylor and Dewsbury, 1990). For example, in one of the Y-maze (Byrne and Keogh, 2007). Similarly, male red-
set of experiments with female Syrian hamsters, six different bellied newts, Cynops pyrrhogaster, produce a peptide
methods were compared using measures of investigation of pheromone in a cloacal gland that has a female-attracting
a single stimulus and five similar methods were used to measure function (Kikuyama et al., 1995). In contrast, female palmate
preference for one of two odor stimuli. Results showed that tests newts, Triturus helveticus, are equally attracted to male and
in the subject’s home cage or in a moving air stream were rela- female chemical cues (Secondi et al., 2005). In this species,
tively poor, whereas tests in which the stimulus (or stimuli) males and females aggregate on breeding sites at high
were in the center of a relatively large arena or were outside densities. Attraction to odors of conspecifics, irrespective of
the arena yielded data that often demonstrated differences in sex, may serve to locate a breeding site and then short-range
attraction (Johnston, 1981). chemical or visual signals may be used to locate mates
In addition to showing a preference for the odors of recep- (Secondi et al., 2005). In terrestrial salamanders, Plethodon
tive females, males may increase their scent-marking rates when jordani, the mental gland of males produces a proteinaceous
in areas marked by sexually receptive females. For example, secretion that operates as a ‘courtship pheromone,’
male meadow voles over-mark more the scents of females in increasing female receptivity and easing insemination
a heightened state of sexual receptivity, i.e., females in post- (Feldhoff et al., 1999). There are also several mammalian
partum estrus, than those of females in other estrous states examples, including mice, Syrian hamsters, dogs, and pigs,
(Ferkin and delBarco-Trillo, 2014). Males can also respond Sus scrofa, in which females show a preference for the odors
physiologically to odors of receptive females; for example, of intact males over those of castrated males, indicating that
more erections were elicited by exposing males to odors of male attractivity may be important during sexual interactions
receptive females than to odors of unreceptive females (Sachs, across many species (Brown, 1977; Johnston, 1981; Lisberg
1999). Males can also use female odors to discriminate and Snowdon, 2009; Signoret, 1976).
between mated and unmated females (Thomas, 2011).
In several species, sexually experienced males show a prefer-
ence for odors of receptive females but sexually naïve males do
1.10.3.2 Individual Discrimination
not (Carr et al., 1965; Hayashi and Kimura, 1974; Huck and
Banks, 1984; Johnston, 1980; Lydell and Doty, 1972). In other Individual discrimination involves the ability of a subject to
studies, however, both sexually naïve and sexually experienced detect the odors of different donors as being different. Recogni-
males show a similar preference for odors of estrous females tion does not only involve the ability to discriminate between
over odors of nonestrous females, e.g., in male mice (Rose the cues/signals from one individual and another, but it also
and Drickamer, 1975). implies that the subject has some previous knowledge about
Changes in seasonality may result in changes in the compo- the stimulus animal (Halpin, 1986). At the simplest level,
sition of odors and the scent-marking rates of individuals, this knowledge may just involve familiarity with one or more
which may affect the preference of individuals for the odors signals from another individual and the ability to categorize
of conspecifics. When meadow voles were tested in short-day a stimulus into one of two categories (familiar or unfamiliar).
conditions, females showed a preference for the odors of other For example, using the knowledge about the odors of one’s
short-photoperiod females over those of males, probably asso- own group may be sufficient to accept a given individual into
ciated with the habit of often nesting with other females in the nest or not. True individual recognition, however, implies
winter to maximize warmth (Ferkin and Johnston, 1995a). more thorough knowledge of the characteristics of other
Short-photoperiod males did not show a preference for the individuals.
odors of short-period females over those of other short- Individual recognition requires the existence of distinct
period males. Furthermore, short-photoperiod voles (both signals and the ability to detect and discriminate between these
sexes) did not show sex-specific preferences for long- signals. Most animals may combine a broad spectrum of
photoperiod conspecifics, and vice versa (Ferkin and Johnston, signals to communicate individual identity, including a combi-
1995a). Some of these seasonal changes in odor preferences nation of chemical, visual, and auditory cues, producing multi-
may depend on the endocrine state of individuals. For modal signals (Higham and Hebets, 2013). Individuals are the
example, during long photoperiods, the attractiveness of fundamental units of social interaction and social organiza-
male scent to females depends on high titers of both testos- tion. Thus discrimination and recognition of individuals, or
terone and prolactin (Ferkin et al., 1997a). classes of individuals, are fundamental to the understanding
Mate attraction is not universally driven by females. There of animal behavior. Even though individual recognition medi-
are species in which males produce odors to attract females. ated by odors is not restricted to vertebrates (Caldwell, 1985),
For example, female frogs of the genus Hymenochirus are most of such research has been conducted on vertebrates,
attracted to secretions produced by male ‘breeding glands’ including many mammalian species (Kent and Tang-
(Pearl et al., 2000). Females are attracted to water containing Martínez, 2014).
Communication by Chemical Signals 295

Studies on rodents such as Mongolian gerbils, Meriones phase should investigate odor B longer than odor A during the
unguiculatus, ground squirrels, Syrian hamsters, meadow voles, last dishabituation trial. This result would suggest that the indi-
otters (Lontra canadensis), koalas (Phascolarctos cinereus), vidual views odor A and odor B as being different from one
raccoons (Procyon lotor), and house mice have also provided another. The habituationdishabituation technique is much
evidence that these animals can distinguish between the scent more naturalistic and much easier to implement than reinforc-
marks of different individuals (Charlton, 2015; Ferkin et al., ing techniques. A variation on this method is to present two
2016; Halpin, 1986; Kean et al., 2015; Kent and Tang- stimuli on the test trial after the habituation phase, namely,
Martínez, 2014; Thom and Hurst, 2004), responding preferen- the stimulus that the subject has been habituated to and a novel
tially or displaying a selective memory for the scent mark of stimulus. Simultaneous presentation of the familiar and novel
a familiar conspecific compared to that of an unfamiliar stimuli is an easier task and may allow the visualization of
conspecific (Heth et al., 1998; Johnston, 1993; Kaur et al., differences in response to the novel stimulus when a single-
2014; Mateo and Johnston, 2000a,b). Recent work on house stimulus test does not (Brown et al., 1987). The optimal dura-
mice has shown that females can learn the individual identity tion of the interval between trials will depend on the species. In
of males from their scent marks and females can use this infor- hamsters, varying the intertrial interval from 1 s to 2 days led to
mation to select potential mates (Roberts et al., 2014). similar results (Johnston, 1993). In contrast, dogs and captive
In a series of elegant experiments, Johnston and colleagues wolves would not even approach the stimulus on the second
(Heth et al., 1999; Johnston, 1993, 2003; Johnston and trial after an interval of 15–30 min, apparently because they
Bullock, 2001; Mayeaux and Johnston, 2002; Todrank et al., could determine from a distance that the odor presented on
1998) used a habituation/dishabituation paradigm to show the second trial was the same as that on the first trial; when
that Syrian hamsters could distinguish between scent marks the interval between trials was shifted to 24 h, both dogs and
of two same-sex conspecifics that were similar in condition, wolves showed the typical habituationdishabituation
treating each scent mark as if it were individually distinct. John- response (Brown and Johnston, 1983).
ston and coworkers found that the amount of time that Syrian Some studies have used across-odor habituation approaches
hamsters spent sniffing the scent mark of an individual donor to determine if animals can remember familiar individuals
decreased with each successive exposure to that individual’s using several different distinctive signals and integrate these
marks, indicating that the hamsters habituated to the scent separate memories into integrated representations of others.
marks, and viewed them as being familiar. The hamsters were For example, male hamsters first had a series of brief interac-
then exposed to a scent mark of a different conspecific and tions with females on four successive days. During these
that of a familiar donor. During this exposure, the hamsters 4 days males had the opportunity to investigate several odor
spent more time sniffing the novel scent mark than the familiar sources from those females. After habituation to one individu-
scent mark, suggesting that scent marks convey individually ally distinctive odor of a stimulus animal (e.g., that from vaginal
distinct information to conspecifics (Johnston, 2009). secretions), males were also habituated to other odors from the
same individual (e.g., sebaceous flank glands). That is, subjects
1.10.3.2.1 HabituationDishabituation showed an across-odor habituation (Johnston and Jernigan,
Different methodological approaches can be used to demon- 1994). Since the odors themselves are composed of very
strate individual discrimination and recognition. The first different chemical compounds, it is not likely that this effect
studies to demonstrate discrimination of odors in mammalian was due to chemical similarities in the two odors. Additional
species used training procedures with food reinforcement to evidence for this conclusion comes from results of control
demonstrate such abilities (Bowers and Alexander, 1967; experiments in which subjects were not familiar with the stim-
Rasa, 1973). During positive reinforcement the subject is pre- ulus animals. If there was similarity in odor quality across
sented with odors from two different donors but rewarded different odor sources, subjects should show across-odor habit-
only when responding to only one of those two odors. If uation without familiarity with the scent donor. However, if
over a series of trials the subject animal learns to discriminate subjects had not previously interacted with the scent donors,
between the two odors we can assume that the animal has no cross-odor habituation was observed (Johnston and Bullock,
the ability to distinguish between them. Positive reinforcement 2001; Johnston and Jernigan, 1994). Subsequent experiments
is a useful approach to determine the discriminating potential showed that the same effects were observed with other pairs
of animals but it has a limited use to understand individual of odors (Johnston and Bullock, 2001).
recognition under normal, nontaught conditions. In virtually all laboratory studies aimed at individual recog-
The most commonly used approach to study individual nition, it has proven difficult to provide evidence for ‘true’
discrimination involves variations of the habituationdishabi- individual recognition. That is, the unique significance of an
tuation technique (Halpin, 1986). A subject animal is first pre- individual, not just the significance of categories of individuals
sented with the odor of one donor (odor A) over several trials such as familiar versus unfamiliar or dominant versus subordi-
(this is the habituation phase), and then the same subject is nate (Halpin, 1986). In particular, it has been difficult to
presented with the odor of a second donor (odor B; this is demonstrate the emotional significance of another individual
the discrimination phase). In each trial, the time the subject without possible confounding interpretations, such as
spends investigating the odor is recorded. During the habitua- differences in familiarity or dominance status (Martin and
tion phase, with repeated presentations of new samples of the Beauchamp, 1982). In one attempt to disentangle the effect
same odor stimulus, the behavioral response declines signifi- of familiarity during individual discrimination, male hamsters
cantly, indicating that habituation has occurred. In this case, were exposed to two different males. One stimulus male beat
individuals habituated to odor A at the end of the habituation the subject in a series of three brief fights, whereas the subject
296 Communication by Chemical Signals

became familiar with a second male by interacting across In fact, the components involved in individual discrimination
a wire-mesh screen. This latter type of exposure is sufficient either are the MUPs themselves or the MUPligand complexes,
for males to develop multicomponent memories of the stim- rather than the volatiles emanating from a scent mark (Nevison
ulus animal (Johnston and Peng, 2008). After both of these et al., 2003).
experiences, male subjects were then tested for their responses Studies have also shown that some subsets of MUPs
in a Y-maze with odors and other cues from the two stimulus found in scent marks of male house mice and rats provide
animals. Subjects were attracted to the odors and other cues information about the identity of the sender, essentially
from a familiar neutral male but were hesitant to approach a ‘bar code’ (Gómez-Baena et al., 2014; Hurst et al., 2001).
the odors of the familiar winner or stay near him (Lai et al., This information is stable over time and individual as well
2004, 2005). These results indicate specific types of response as species-specific and appears to be based on the individ-
to two different individuals that have different significance to ual’s genotype and not its phenotype (Cheetham et al.,
the subject but have equivalent (or at least very similar) 2007; Green et al., 2015; Hurst et al., 2001). Recently, Stow-
levels of familiarity. These results are based on a relatively ers and colleagues discovered that wild male house mice
simple set of procedures and experiences compared to the expressed their own unique combination and ratio of
number and potential complexity of experiences in the wild. MUPs in their urine and that the subsets of MUPs were indi-
One disadvantage of habituation tasks is that a failure to vidually distinct (Kaur et al., 2014). For example, Kaur et al.
show dishabituation to a novel stimulus may have two (2014) discovered that wild male house mice learn their own
different interpretations: (1) the animal did not discriminate, subset of MUPs that provide individually distinct informa-
or (2) the animal did not investigate the stimulus because of tion and distinguish it from those found in the urine of other
other motivational/emotional reasons, for example, if the male house mice. Male house mice used this information to
novel stimulus came from an animal that had beaten the avoid countermarking their own scent marks and those of
subject in a fight the subject might be reluctant to investigate male conspecifics whose subset of MUPs is similar to their
an odor from the familiar winner. Thus, failure to discriminate own. In contrast, male house mice countermarked the scent
in habituationdishabituation tasks may be difficult to marks of conspecifics whose composition of MUPs was
interpret. unlike their own (Kaur et al., 2014). Male house mice,
Both trained discrimination and the basic habitua- however, did not countermark the scent marks of male
tiondishabituation tests are primarily useful for determining conspecifics that they had learned to be socially dominant
whether animals discriminate between and remember indi- to them (Kaur et al., 2014). In addition, in a recent study
vidual signatures. If one wants to determine other types of Green et al. (2015) found that MUPs, which act as a genetic
information, such as the content of the memory or the marker, can also be used by female house mice to select
emotional salience of a memory for an individual, other closely related females as nest partners to help look after
methods of testing are necessary. After individuals interact their offspring and that they can do so without being familiar
with one another, what do they remember about each other? with those close relatives.
Two types of information may be valuable to remember: (1) Mice were shown to spontaneously notice differences in the
memories that incorporate several different characteristics of odors of conspecific mice with different MHC types (Penn and
the individual, such as several separate odors or other cues Potts, 1998b). Norway rats also discriminated body odors
(sound of voice, visual features) and (2) the emotional or func- based on slight genetic differences in the MHC (Brown et al.,
tional significance of the individual to the subject. 1987, 1989, 1990). These results demonstrate that genes in
the MHC complex do influence body odors and mice and
1.10.3.2.2 Two Primary Polymorphic and Multigenic rats can discriminate between the odors of individuals with
Complexes different MHC types. However, later studies that specifically
There are two primary polymorphic and multigenic complexes tested whether mice can use MHC-related odors to recognize
that are important in studies of olfactory communication that the owner of a given scent found negative results (Cheetham
contribute to or determine individual differences in odors. et al., 2007; Hurst et al., 2005). In these studies, MUPs were
These complexes are the major histocompatibility complex involved in individual discrimination but the MHC was not
(MHC) and the major urinary proteins (MUPs). MHC genes (Cheetham et al., 2007). It is important to notice that MHC-
produce highly polymorphic glycoproteins involved in immune related odors are affected by such factors as status and diet
system function (Yamazaki et al., 1980). MUPs are mostly (Schellinck et al., 1997) and thus are not a good candidate to
produced in the liver and become concentrated in the urine in offer a stable individual chemical fingerprint (Hurst et al.,
mice. MUPs are nonvolatile molecules, but they bind smaller, 2005). Therefore, there seems to be no clear evidence that the
volatile molecules and release them slowly (Hurst and Beynon, MHC of an animal offers an individual signature that is
2004), thus prolonging the effectiveness of volatile compounds perceived by conspecifics. In addition, Green et al. (2015)
contained in scent marks (Hurst et al., 1998). Without such found that the vertebrate-wide MHC was not involved in kin
binding the volatile compounds might be lost from the scent recognition in house mice.
mark relatively quickly (in minutes), which would decrease MUPs appear to be a better candidate for individually
the information contained in the scent mark (Hurst and distinctive information, at least in mice (Hurst and Beynon,
Beynon, 2004). By being bound to MUPs, these signaling vola- 2004), because the pattern of MUPs expressed by an individual
tiles can emanate from the scent mark for up to 24 h is unchanged and thus provides a constant individual signature
(Humphries et al., 1999). MUPS also contain individually not affected by factors such as diet or social status (Nevison
distinctive information on their own (Hurst et al., 2001). et al., 2003). For example, when two individuals share the
Communication by Chemical Signals 297

same pattern of MUPs, odors of one mouse do not trigger focus aggressive behavior toward nonkin as a means of
competitive behaviors in the other (Hurst et al., 2001). MUPs increasing its inclusive fitness. Many animals use chemical
from other conspecifics have also been shown to elicit more signals from conspecifics to differentiate between kin and non-
countermarking than self-MUPs (Kaur et al., 2014). Also, kin. For example, odors of kin elicit lower levels of agonistic
when a purified MUP is added to the urine of a male, he treats scent marking (flank marking) in hamsters than did odors of
his own modified urine as if it belonged to an intruder male nonkin (Heth et al., 1998). Recognizing kin also allows an indi-
(Hurst et al., 2001). vidual to avoid mating with closely related individuals,
A recent study challenges the fingerprint properties of MUPs achieving optimal outbreeding, and thus reducing inbreeding
(Thoß et al., 2015). In this study the MUP profiles were more depression in its offspring (Nunes, 2007).
similar between brothers than between unrelated males. A special aspect of kin recognition is that of parent
However, the MUP profile of a given individual changed over offspring recognition. Mutual recognition between a mother
the course of several weeks, supporting more a ‘dynamic and her offspring is likely to be beneficial to both parties under
expression’ hypothesis than a ‘barcode’ hypothesis. Further most scenarios. Most if not all senses may be involved in paren-
studies will need to determine if mice in natural conditions, toffspring communication, but olfaction is especially impor-
i.e., having the ability to investigate the scent marks of neigh- tant, particularly in rodents (Porter, 1983) but also in other
boring conspecifics in a daily basis, adjust their memories of mammalian groups such as ungulates (Grau, 1976) and carni-
individual neighbors according to the slowly changing compo- vores (Pitcher et al., 2011). In altricial species, neonates remain
sition of their scent marks. That is, it is possible that animals blind and deaf during the first days, whereas they can already
recognize individuals by investigating relatively dynamic process scents from the mother and from other pups (Porter,
signals instead of fixed ones. 1983). Rodent pups may rely on olfaction to locate the mother’s
Although MUPs have been described and thoroughly nipples as shown by olfactory bulbectomy of pups impairing
studied in the house mice, similar proteins can also have equiv- nipple location (Porter, 1983). In rabbits, a pheromone
alent semiochemical functions in other species (Gómez-Baena emanating from the mother’s ventrum triggers a stereotyped
et al., 2014). A particular MUP, Darcin, also found in the urine behavior in the pups that guides them toward the mother’s
of male house, plays a particular role in chemical communica- nipples and suckling (Hudson and Distel, 1983). This volatile
tion. Roberts et al. (2010) found that Darcin stimulated very pheromone is sensed by the main olfactory system of the young
rapid learning of spatial cues associated with its location in (Hudson and Distel, 1986) and it is also involved in reinforced
male and female house mice. Female mice recalled the location learning (Coureaud et al., 2006; Schaal et al., 2003). The emis-
where they encountered Darcin and later returned to that area, sion of such a pheromone is hormonally controlled (González-
presumably to locate that male donor. Similarly, male mice Mariscal et al., 1994; Hudson et al., 1990). The source of the
also rapidly learn and remember the location of rival male pheromone 2-methylbut-2-enal is milk (Schaal et al., 2003).
scents and drive off these rivals or over-mark their scent marks Yet, even females that are not producing milk, i.e., estrous
(Roberts et al., 2010). Mice that encountered Darcin scent only or pregnant, can still produce an olfactory cue that triggers
once would return to that location 2 weeks later even if the Dar- the same behaviors as those seen following exposure to
cin scent was absent (Roberts et al., 2010). 2-methylbut-2-enal (González-Mariscal et al., 2016).
Some species of male mammals do not produce large quan- Mothers can identify the odors produced by their offspring.
tities of MUPs. Instead, some animals such as Syrian hamsters For example, human mothers can distinguish the odors on T-
produce a proteinaceous compound referred to as Aphrodisin shirts of their children from those of unfamiliar children of
(Singer et al., 1986). Exposure to Aphrodisin from the vaginal the same age and sex (Porter and Moore, 1981). Using
secretions of female hamsters was sufficient to increase copula- a similar methodological approach mothers, but not fathers,
tory behavior among male hamsters (Macrides et al., 1986). In were able to recognize the odor of their children better than
a recent paper, Stopková et al. (2010) found that Aphrodisin by chance (Ferdenzi et al., 2010). Many studies on goats and
like proteins are produced by prostate, preputial, and salivary sheep, two precocial species, have established that mothers
glands, and liver and uterus in male and female bank voles, use olfactory information to discriminate between their own
Myodes glareolus. Stopková et al. (2010) also identified three newborns and those of other females, although olfaction is
novel odorant-binding proteins, Obp1, Obp2, and Obp3, in not the only sensory modality involved in such discrimination
urine and saliva. Stopková et al. (2010) suggested that due to (Lévy et al., 2004; Romeyer et al., 1994; Viviers et al., 2014).
their high sequence homology (i.e., Obp1, Obp2, and Obp3 Mothers may also mark their offspring for later recognition.
with Aphrodisin) Obps may have capacity to bind ligands Female Mongolian gerbils use their ventral gland to scent
similar to those bound by Aphrodisin like proteins. If so, mark their own pups. Females will scent mark their pups if
Aphrodisin-like proteins and their ligands binding proteins they have been licked and groomed, and females will
are likely to play a role in chemical communication similar preferentially retrieve pups marked with their own secretion
to that of MUPs in attracting mates (Humphries et al., 1999; compared to unmarked pups (Wallace et al., 1973).
Kaur et al., 2014; Roberts et al., 2010).
1.10.3.3.1 Mechanisms of Kin Recognition
There are at least three mechanisms by which an individual can
1.10.3.3 Kin Recognition
use odors to recognize kin: familiarity-based recognition,
The main functions of kin recognition involve nepotism and phenotype matching, and self-referent phenotype matching.
inbreeding avoidance. Kin recognition allows an individual In familiarity-based recognition, animals learn the characteris-
to focus its cooperative behavior toward extended kin and to tics of others that they grow up with and later in life treat only
298 Communication by Chemical Signals

these individuals as kin. This type of learned recognition can retrieve pups with a similar MHC (Yamazaki et al., 2000).
only be useful for actual kin recognition in species in which Pups also show a preference for the odors of the mother and
litters of one female’s pups are physically separated from the siblings, which supposedly share a similar MHC, over the odors
litters of other females so that developing pups are only of individuals with a dissimilar MHC (Yamazaki et al., 2000).
exposed to kin, and in which future kin interactions are Little is known about the specific mechanisms beyond the role
restricted to the mother and siblings. This type of recognition of MUPs and MHC genes. Nonetheless, it is clear that genetic
can be demonstrated by using a cross-fostering design in which similarity does translate into odor similarity. Many genes
pups grow up with siblings or foster siblings from another must have roles in producing odors, since many different meta-
litter. When these individuals are adult, results showing that bolic processes can be reflected in the output of scent glands
individuals treat nonsiblings that shared their nest like kin indi- and other secretory and excretory products.
cate that the mechanism underlying such recognition is the
association in the nest.
1.10.3.4 Group Discrimination
In phenotype matching, animals learn the phenotype of the
mother and siblings and later compare those phenotypes with Recognition of group members is important in social species in
the phenotypes of unfamiliar individuals, treating unfamiliar which agonistic behaviors are directed preferentially to alien-
individuals as kin if there is a close match between phenotypes. group individuals. Group discrimination may occur by individ-
Phenotype matching in which features of mothers can be used ually recognizing each member of the own group and/or by
as a template occurs in Belding’s ground squirrels, Spermophilus means of a shared group odor. Not in all social species,
beldingi (Mateo, 2009). Young were exposed to odors from an however, there may be a need to discriminate between the
unrelated mother during early stages of development and scents of other groups. For example, in banded mongooses,
before natal emergence. These young chose as play partners Mungos mungo, there is no difference in the composition of
not only their littermates but also juveniles from the unrelated chemical signals from different groups, and individuals do
mother, indicating that the odors of the unrelated mother were not discriminate between the scents of different groups of
incorporated into the kin template of the subject young. similar familiarity, i.e., either two familiar neighboring groups
A third mechanism is self-referent phenotype matching, in or two unfamiliar groups (Jordan et al., 2010).
which an individual compares its own phenotype (e.g., odors) The scent of a group can be conferred by a high degree of
to that of other conspecifics. If there is a high correlation genetic similarity, either due to kin relationships (e.g., family
between these characteristics, other individuals are treated as group) or to high levels of inbreeding. If odors are diet-
kin. This method of kin recognition may be found in species related and all individuals from a group share the same food
with multiple paternity or maternity, where full-siblings and source, this can also create a shared group odor (Halpin,
half-siblings may share a nest. It also may occur in species in 1986). A group odor can also be produced by mixing the scent
which pups of unrelated females can mix early in life. In this of all individuals together on each individual by scent marking
type of situation, recognition by association may not be a reli- one another (allomarking), or merely by being in contact with
able method to establish kinship. Self-referent phenotype one another or by scent marking on the same sites. For
matching can also facilitate optimal inbreeding and example, European badgers, Meles meles, produce a secretion
outbreeding. An example of self-referent phenotype matching in the subcaudal pouch which contains distinctive group
has been shown in female Syrian hamsters. Estrous females membership information (Buesching et al., 2003). This secre-
that had been cross-fostered shortly after birth were more sexu- tion is partly produced by bacterial flora. Group members
ally attracted to unfamiliar nonkin than to unfamiliar kin transfer such flora between one another by pressing the subcau-
(Mateo and Johnston, 2000a). dal pouch against the body of another group member. In some
Unless using a complex experimental design, it may be cases, two individuals press both pouches together, so that the
impossible to determine whether kin recognition is based on bacterial flora and/or secretions are transferred between the
phenotype matching or self-referent phenotype matching. For subcaudal pouches of the two individuals. The authors
example, female meerkats, Suricata suricatta, exposed to odors observed thousands of instances of allomarking between 40
of unfamiliar individuals discriminated between the scent of individuals in natural conditions, indicating the regularity
unfamiliar kin and unfamiliar nonkin. However, it could not and importance of this behavior (Buesching et al., 2003).
be determined whether meerkats used information from known Another example is the European rabbit, which lives in close-
kin or from themselves as templates (Leclaire et al., 2013). knit social groups (Mykytowycz, 1968, 1970). Dominant males
The basis for chemical recognition of kinship must be mark all members of the group with their chin glands. If a rabbit
provided by odor similarities based on genetic similarities from one group is experimentally scented with scent from
between close kin (Heth and Todrank, 2000; Heth et al., a dominant male of another group, and then introduced
1998, 1999, 2001; Todrank and Heth, 2003; Todrank et al., back into its natal group, it is attacked (Mykytowycz, 1968).
1998). In ring-tailed lemurs, Lemur catta, for example, there is Dominant males even attack females of other groups and
a positive association between chemical and genetic differences females of his own group that are scented with the odors of
between individuals (Boulet et al., 2009), and males respond a male or female from another group (Mykytowycz, 1968).
more to the odor of less-related females than to those of
more-related females, although only during the breeding
1.10.3.5 Species Discrimination: Competitors and Predators
season (Charpentier et al., 2010). Both the MHC and the
MUPs seem to be involved in kin discrimination (Thoß et al., Discriminating between individuals of one’s own species and
2015; Yamazaki et al., 2000). Female mice are more likely to individuals of a closely related species is common across taxa
Communication by Chemical Signals 299

since individuals who mate with other species usually do not predators. A flexible solution to this trade-off by many prey
produce young or may produce infertile young. Female prefer- species is to decrease scent-marking rates under high predation
ence for odors of conspecific males and male attraction only to risk (Hughes et al., 2010; Roberts et al., 2001; Rosell and Sanda,
odors of conspecific females may serve as a precopulatory 2006), or to avoid the areas or the paths containing those pred-
isolating mechanism resulting in the avoidance of interspecific ator odors (Gorman, 1984; Je˛ drzejewski et al., 1993; Roberts
mating. Indeed, there are many species that show female pref- et al., 2001; Rosell and Sanda, 2006; Vlautin and Ferkin,
erences for odors of conspecific males over odors of closely 2012). Mice do not change their scent-marking rate in the pres-
related heterospecific males (Johnston, 1983), including deer ence of predator cues, but they tend to cluster their scent marks
mice (Doty, 1973; Moore, 1965), collared and brown when there are predator cues (Hughes and Banks, 2010).
lemmings (Huck and Banks, 1980), the fossorial mole rat, Spa- However, some prey species that are heavily preyed upon
lax ehrenbergi (Nevo et al., 1976), and two species of sac-winged may not change their behavior in response to the odors of pred-
bats, Saccopteryx bilineata and Saccopteryx leptura (Caspers et al., ators. The reason for this is that the risk of predation may
2009). always be high. It may be impossible for these prey species to
In pairs of closely related species in which hybridization is avoid areas marked by predators and counterproductive to
possible, individuals may not only show a preference for decrease scent marking. Instead, prey species may change
conspecific over heterospecific odors, they may even show foraging locations (Orrock et al., 2004; Wolff, 2004a) and
a preference for conspecific odors over those of hybrids. Such increase vigilance (Buchanan-Smith et al., 1993; Monclús
preferences has been shown in the well-studied hybrid zone et al., 2005). Finally, males and females may respond differ-
between Mus musculus musculus and Mus musculus domesticus ently to the odors of predators in a way that reflects their
across Europe (Latour et al., 2014). Male musculus mice around different mating strategies and the level of risk that they are
the hybrid zone showed a preference for the urine of conspe- prepared to undertake. Normally, it is likely that males will
cific females over the urine from hybrid females (Latour take riskier approaches if their reproductive fitness would be
et al., 2014). Interestingly, such preference did not occur negatively affected otherwise. For example, female field voles,
when subject males were from allopatric populations (where Microtus agrestis, avoid areas containing the scent marks of pred-
hybridization does not take place), suggesting that in the ators (Ronkainen and Ylönen, 1994), whereas males do not
sympatric populations there has been a process of character avoid such areas if they also contain the scent marks of females
displacement involving signal perception or processing, or (Norrdahl and Korpimäki, 1998).
preference response (Latour et al., 2014). Recognition of odors from heterospecific species can be
Further support for the notion that female preferences for innate, learned during the first stages of development, or
odors of conspecific males may have the ultimate function of learned during adulthood (Griffin et al., 2000). Male brown
avoiding interspecific mating comes from several studies and collared lemmings that were raised by their mothers
showing that preference for conspecific odors occurs only preferred odors of conspecific females in adulthood. However,
when females are sexually receptive. This effect has been when pups of these two species were cross-fostered, males
shown, for example, in fossorial mole rats (Nevo et al., showed a preference for the female odors of the cross-
1976), and deer mice (Doty, 1972). In the striped mouse, fostered species, i.e., heterospecific odors (Huck and Banks,
Rhabdomys pumilio, females of three populations showed a pref- 1980). Females that were not cross-fostered also showed
erence for the odors of males of the same population over those a strong preference for conspecific male odors, but cross-
of other populations, but only when females were in estrus fostered females did not show any longer a preference between
(Pillay, 2000). conspecific and heterospecific male odors (Huck and Banks,
Closely related species may not only pose the risk of intra- 1980). It is however important to notice that any preference
specific mating. Given that closely related species may still between the odors of conspecific and heterospecific opposite-
share many similarities in their ecology (e.g., feeding habits), sex individuals may not necessarily relate to mating decisions.
they may be direct competitors between closely related species. Syrian hamster females showed an innate preference for the
For example, two sympatric species of Acomys spiny mice have odors of conspecific males over those of heterospecific males
similar foraging needs. As a result, one species (A. russatus) (delBarco-Trillo et al., 2009a). However, female hamsters that
shifts its foraging activity from nocturnal to a less optimal have never interacted with heterospecific males will respond
diurnal foraging activity when A. cahirinus is present in the sexually in a similar way to both conspecific and heterospecific
same area. Odors of A. cahirinus in a particular area also reduce males (delBarco-Trillo et al., 2009b). Interestingly, when adult
the time that A. russatus will spend in that area (Baudoin et al., females were exposed to a heterospecific male during 4 or
2013). 8 days (one or two estrous cycles) behind a wire-mesh barrier,
Preypredator relationships can also promote the need for they stopped mating with heterospecific males. These female
the recognition of odors of different heterospecific species. hamsters started to behave in a way that reflects their innate
Both predators and prey can benefit by recognizing the odors odor preference for conspecific males (delBarco-Trillo and
from each other. Predators will benefit by recognizing and Johnston, 2011; delBarco-Trillo et al., 2010).
tracking down the odors of prey, whereas prey may benefit
by recognizing the odors of predators and avoiding areas
1.10.3.6 Sex Identification
recently marked by predators.
From the prey perspective, scent marking may suppose Sex identification based on chemical signals must rely on differ-
a trade-off between the benefits accrued by intraspecific ences in the composition of specific scents from male and
communication and the cost of advertising its presence to female donors. However, information about the chemical basis
300 Communication by Chemical Signals

of such differences is surprisingly limited. There are, however, each odor is affected by steroid hormones can vary. In meadow
a few cases in which active compounds involved in sex discrim- voles, odors from feces, mouth, and the posterolateral region
ination have been characterized. In mice, two farnesene were clearly affected by testosterone concentrations in the
compounds (E,E-a-farnesene and E-b-farnesene) are found in blood of males and by estradiol in females. Gonadectomy of
male preputial glands, and sexually experienced female mice scent donors eliminated preferences for these odors by
are attracted to these farnesenes when added to bladder urine opposite-sex subjects and these preferences were restored by
or water (Jemiolo et al., 1991). Another compound (methyl- replacement therapy with testosterone for male donors and
thio) methanethiol is found in male urine but not in female estradiol for female donors. In comparison, the attractiveness
urine. This compound is attractive to females and may be of urine and anogenital odors were reduced but not eliminated
important in the attraction of estrous females to adult males by gonadectomy (Ferkin and Johnston, 1993).
(Lin et al., 2005). The MUP Darcin is also only found in
male urine and females are attracted to it. Darcin is also
1.10.3.7 Social Effects
involved in odor learning, reinforcing the attraction of females
to male volatile compounds that might not elicit otherwise an 1.10.3.7.1 Role of Odors in Social Dominance and Aggression
innate attractive response in females (Roberts et al., 2010). Mammals can also learn the identity of dominant males, which
Several studies have shown that females show a preference affects the location and frequency of scent marking by subordi-
for odors of males with dissimilar MHC types compared to nate males. Dominant male rodents mark more often, along
those of males with their own MHC type (Penn and Potts, the boundaries of territories and on prominent objects that
1999). By mating with males that have a different MHC type, serve as sign posts. Subordinate male house mice and snow
females may produce MHC-heterozygous offspring and such voles, Chionomys nivalis, however, scent mark at a much lower
offspring should be able to respond more effectively to rate, and diffusely through the territory and usually not at
a wider range of pathogens (Penn and Potts, 1998a). Also, sign posts (Drickamer, 1995; Luque-Larena et al., 2001,
a preference for a male with a different MHC type reduces 2002). Similarly, socially dominant female primates deposited
inbreeding and produces offspring that are more more scent marks than did socially subordinate female
heterozygous across all of the genome, not just in the MHC primates (Kappeler, 1998; Pochron et al., 2005). Several studies
(Brown and Eklund, 1994; Potts and Wakeland, 1993). Most on house mice have shown that their response to the scent
of the studies on the role of the MHC in mate choice have marks of conspecifics depends on their ability to distinguish
been conducted using inbred laboratory mice that are between dominant and subordinate individuals. For example,
genetically identical except at the MHC locus (Brown et al., Hurst et al. (1993) tested the hypothesis that the scent marks
1990; Yamazaki et al., 1980). In these studies, male mice of subordinate mice maintain social tolerance between familiar
show the ability to discriminate between the odors of two mice living in the same group. Hurst et al. (1993) found that
conspecific females that have a dissimilar MHC (Yamazaki after investigating the scent mark of a familiar subordinate,
et al., 1980). Studies with laboratory rats show similar results the resident male mouse would attack that individual. If the
(Brown et al., 1990). Female mice in estrus also show scent mark was from an unfamiliar, subordinate male, the resi-
a preference for the odors of males with a dissimilar MHC dent dominant male would investigate the scent marks of all
when tested in a Y-maze (Egid and Brown, 1989). the subordinates in his territory. If, however, the scent mark
Interestingly, females that were not in estrus did not show was from an unfamiliar, dominant male, the resident over-
such a preference (Egid and Brown, 1989). Women in the marked it and would flee if it encountered that male. If the
fertile phase of their menstrual cycle also find the odors of scent mark was from a familiar, dominant male that was
men that are the most dissimilar in their MHC the most a neighbor, the resident male would ignore the scent mark
attractive and pleasant (Wedekind et al., 1995). When (Hurst et al., 1993). This suggests a ‘dear enemy’ effect, in
women are taking oral contraceptives, such preferences are which residents ignore the cues and signals of neighbors to
reversed, i.e., women then prefer the odors of men with reduce confrontations with them (Rosell and Bjorkoyli, 2002;
a similar MHC type. Contradicting some of the previous Wyatt, 2014). In addition, male house mice that deposited
literature on mice, studies using wild mice living in large more scent marks had higher reproductive success than other
enclosures have shown that MHC is not a relevant marker males (Thonhauser et al., 2013b). This observation indicates
that animals use for inbreeding avoidance, whereas MUPs that scent marking is maintained by sexual selection, as it
were sufficient to explain inbreeding avoidance (Sherborne enhanced males’ reproductive success. Studies on a variety of
et al., 2007). terrestrial mammals suggest that scent marks appear to be
a secondary sexual trait, which conspecifics use to assess the
1.10.3.6.1 Endocrine Effects quality of a potential mate and competitors (Ferkin, 2015;
Hormones such as testosterone and estradiol influence the Roberts et al., 2014; Thonhauser et al., 2013a,b; Tinnesand
attractiveness of odors of the opposite sex. Gonadectomy et al., 2015).
usually reduces the attractiveness of opposite-sex odors and In species that live in permanent social groups, there are
the attractive properties of such odors can be restored by often well-established dominance relationships between males
replacement therapy. Similarly, ovariectomy often reduces in a local area and dominance rank is often related to the ability
responses of males to female odors and replacement treatment of one male to monopolize or have preferential access to
with estradiol often restores the attractiveness of these odors resources. Females should prefer dominant males as mates
(Ferkin and Johnston, 1993). When several body odors contain because at least some of the qualities of dominant males may
information about the sex of an individual, the degree to which be based on genetic characteristics that may be passed on to
Communication by Chemical Signals 301

offspring. If odor parameters (e.g., testosterone metabolites) (Desjardins et al., 1973), Syrian hamsters (Ferris et al., 1987;
are related to male dominance, females may be able to distin- Johnston, 1975a,b), Mongolian gerbils (Thiessen et al.,
guish between dominant and subordinate males just by inves- 1971), stoats, Mustela erminea (Erlinge et al., 1982), European
tigating their odors. For example, female brown lemmings rabbits (Mykytowycz, 1965), sugar gliders, Petaurus breviceps
showed a preference for odors of dominant males over odors (Schultze-Westrum, 1969), marmosets, Callithrix jacchus (Ralls,
of subordinate males, even when the females were not familiar 1971), and ring-tailed lemurs (Kappeler, 1990).
with the two male stimulus animals before testing (Huck and Instead of or in addition to differential marking rates, the
Banks, 1982; Huck et al., 1981). Female brown lemmings dominant status of an individual may be coded by odors
also preferred odors of socially naïve males that later became having a chemical composition that differs from that of subor-
dominant over the odors of socially naïve males that later dinate individuals. For example, the chemical profiles of anal
became subordinate, suggesting that the differences between scent gland secretions from female spotted hyenas differ
males were at least partially determined by genetic differences depending on whether females have a low social ranking or
(Huck et al., 1981). Female European rabbits also show a pref- a high ranking (Burgener et al., 2009). The functional impor-
erence for the odors of dominant males (Engel, 1990). These tance of these chemical differences is suggested by the fact
types of results all indicate that there are some constituents that adult females respond to secretions of other females faster
of male odors that correlate with social status, possibly related and more often and are more likely to anoint themselves with
to levels of testosterone, cortisol, or other stress-related those secretions if they were produced by a high-ranking female
hormones (Engel, 1990; Huck et al., 1981). (Burgener et al., 2009). In beavers the chemical composition of
Subordinate individuals will also benefit by discriminating anal gland secretions also differs between dominant and subor-
between subordinate and dominant individuals and respond- dinate individuals, and beavers can use those chemical differ-
ing accordingly in order to reduce aggression from dominant ences to distinguish between dominant and subordinate
individuals. In many species, including Syrian hamsters, mice, unfamiliar intruders (Tinnesand et al., 2013). Similarly, in
rats, lagomorphs, and canids, individuals avoid areas marked house mice, the more aggressive females excreted a higher
by another individual that defeated them in a dyadic encounter, amount of MUPs in their urine (Stockley et al., 2013).
whereas winners do not normally avoid areas marked by indi- Normally, larger males are dominant to smaller males
viduals they defeated in such encounters (Drickamer, 2001; because size gives them an advantage during competitive
Petrulis et al., 2004). Reactions to odors of dominant or subor- displays and fights. However, other factors may also influence
dinate males may be determined by information in the odor which male becomes dominant. Smaller male mice, for
that identifies an animal as having a dominant status or not. example, may initially become dominant over larger males
Before the establishment of a polarized status relationship by investing more in preputial glands and in scent marking
between two male snow voles, both males investigated areas than larger males (Gosling et al., 2000). However, this strategy
scent marked by the other male similarly. However, after the is energetically very costly – a high investment in scent marking
establishment of a dominantsubordinate relationship, the leads to slower growth and lower adult body weight, so these
subordinate male investigated the area scented by the dominant small males are vulnerable to reversal of dominance status later
male much less than the dominant male investigated the area in life (Gosling et al., 2000).
scented by the subordinate male (Luque-Larena et al., 2002). The odors that a male normally deposits in his territory,
These results can be explained either by individual recognition home range or in the vicinity of a burrow can serve as a confi-
or by a reaction to information in the odors about status. Other dence-building context in aggressive interactions. For example,
results show that cues to status do influence the behavior of male Syrian hamsters took more time to enter an enclosure
animals. For example, group-housed male mice avoid areas con- when another male’s odors were present compared to a clean
taining urine of a dominant male, whereas urine of a subordi- area or an area scented by a flank-glandectomized male; subjects
nate male or water in the area results in much more time also spent less time in an arena scented by an intact male
spent in a similar test chamber (Jones and Nowell, 1973). compared to a flank-glandectomized male (Alderson and
Also, urine of a dominant male spread on a castrated male trig- Johnston, 1975). The effect of an ‘own-odor’ or ‘home-odor’
gers aggressive behavior in other conspecifics, whereas subordi- context should be maximal when the odor in one location is
nate urine spread on a castrated male does not promote consistent with other information about location (familiar
aggression in other conspecifics (Jones and Nowell, 1973). landmarks, etc.). In experiments in which agonistic interactions
After a subordinatedominant relationship is established, take place in an unfamiliar, neutral area, however, odors may
odors can be used to advertise dominant status. In many take on an especially important role and give an advantage to
species, dominant individuals scent mark more than subordi- the male whose odors are present. For example, when two
nate individuals (Ralls, 1971). A larger number of marks or European rabbits were placed in a neutral arena containing feces
a greater density of marks by one male may signal dominant from one of the two contestants, the owner of the feces was
status within the area that was marked (Gosling, 1990; Hurst, more aggressive, initiated more interactions and was more likely
1987; Rich and Hurst, 1999). Even in the absence of qualitative to win the contest (Mykytowycz, 1973). Thus, the factor deter-
differences in the chemical composition of their odors between mining dominance was not any intrinsic feature of the winner,
dominant and subordinate individuals, quantitative differ- but the fact of being in an environment containing the odors of
ences in marking may be used by conspecifics to determine if one of the males. When two female Syrian hamsters that are
a particular individual has a dominant status or not. Some unfamiliar with each other are placed in a neutral arena scented
examples of species in which dominant individuals scent with vaginal secretion of one of the females, the female whose
mark more than subordinates include the house mice odor is in the arena is more likely to become dominant;
302 Communication by Chemical Signals

subsequently the winner does all of the vaginal marking and to associate with males that fed on a high-quality diet
most of the flank marking (Fischer and McQuiston, 1991). (termites) compared to a low-quality diet (ants) and were
When two females with an established dominant–-subordinate attracted to the feces of males on the high-quality diet (Jaeger
relationship are placed in a neutral arena scented with vaginal and Wise, 1991; Walls et al., 1989). To the extent that a male
secretion from the subordinate female, a reversal in the domi- can defend or obtain high-quality food resources and to the
nance relationship was observed in 8 of 12 pairs (Fischer and extent that this ability is heritable, females who choose such
McQuiston, 1991). Similarly, when the encounter between males should have higher quality offspring and greater repro-
two male Syrian hamsters takes place in the territory or home ductive success than females that do not discriminate between
cage of male A or in a neutral arena that contains odors of males on the basis of the diet they consume.
male A, that male is more likely to win the initial encounter Phytoestrogens are nonsteroidal plant compounds that can
and become dominant. The results of the initial encounter cause estrogenic or antiestrogenic effects on the animals that
have a strong effect on later encounters between the same two consumed phytoestrogen-rich plants (Pierson and Ferkin,
individuals, even if a later encounter is in a neutral arena (Lai 2015). Despite the fact that such estrogenic and antiestrogenic
and Johnston, 2002; Lai et al., 2004). In house mice, an intruder effects could include changes in scent marking, over-marking,
is more submissive when there is a match between the odors on and the response of individuals to the odors of other conspe-
the substrate and the other individual (e.g., a resident), than cifics, the impact of phytoestrogens on olfactory communica-
when there is not a match between the substrate odors and tion requires further investigation (Pierson and Ferkin, 2015).
the other male (Gosling and McKay, 1990). Food restriction and food deprivation can have dramatic
After the establishment of a status relationship, there are effects on the information contained in the odors of affected
usually differences between the dominant and subordinate individuals. For example, long-term dietary restriction leads
individuals in how they respond to the odors of the other indi- to a reduction in the amount of MUPs in male mouse urine
vidual. For example, subordinate males may avoid odors of (Giller et al., 2013). Female swordtail fish, Xiphophorus birch-
dominant males, whereas dominant males usually do not manni, show a preference for odors of males that are well fed
avoid odors of subordinate males. Male cavies, Cavia aperea, over the odors of males that were food deprived for 5 days
that were dominant to one male and subordinate to another (Fisher and Rosenthal, 2006). Food restriction also affects the
male spent more time in the side of an arena containing odors highly sexually attractive odors of females in postpartum
of the subordinate male than in the side containing odors of estrus. Food deprivation or restriction prior to the beginning
the dominant male (Martin and Beauchamp, 1982). A male of postpartum estrus renders the postpartum-estrous odors of
Syrian hamster avoids odors of the male that beat him in a fight, those females less attractive than the odors of postpartum-
but this subject does not avoid the odors of either a familiar estrous females that were not food deprived or food restricted
male with whom he did not fight or an unfamiliar dominant (Sabau and Ferkin, 2013a). In contrast, the odors of male
male (Lai et al., 2004, 2005; Lai and Johnston, 2002; Petrulis meadow voles that were food deprived for 24 h were still attrac-
et al., 2004). These latter results indicate that the identity of tive to females (Pierce et al., 2005). Such sex differences in
the individual and the memories associated with a specific indi- meadow voles can be attributed to the different energetic
vidual are crucial factors that determine the type of response demands placed on female and male meadow voles during
observed toward an individual’s odor. the breeding season (Pierce et al., 2005, 2007).
The response of dominant and subordinate individuals to The loss of attractivity of odors may not only result from
the same unfamiliar odors may also differ. For example, domi- food restriction during adulthood but also from food restric-
nant male meerkats had a stronger response than did subordi- tion during development. Female rat-like hamsters, Cricetulus
nate males to intruder scent marks (Mares et al., 2011). triton, that were food restricted during pregnancy produced
male offspring whose odors during adulthood were less attrac-
1.10.3.7.2 Food and Odors: Diet and Food Deprivation tive to female conspecifics than those of males reared by
Across a population, different individuals may differ in their untreated dams (Liang et al., 2004; Meikle et al., 1995). A
particular diet. This may be the case even in species with similar experimental approach has been used in meadow voles,
a very specialized diet, given that a particular food source comparing the offspring reared by either untreated dams or
may vary in quantity and quality across an area. If the distribu- dams that were food restricted during early, middle, or late
tion of food is heterogeneous, dominant individuals are likely lactation (Sabau and Ferkin, 2013b, 2014; Sabau et al.,
to defend a territory that contains better higher-quality food 2014). Male offspring from dams food-restricted during mid-
resources and thus will consume a higher-quality diet than lactation produced scent marks that were less attractive to
subordinate males. A higher-quality diet should lead to better opposite-sex conspecifics than those of male offspring reared
condition and odors that reflect such a higher condition. In by control dams or dams food-restricted during early or late
several species, subjects have been shown to prefer the odors lactation (Sabau and Ferkin, 2014; Sabau et al., 2014). In
of opposite-sex conspecifics that have been feeding on contrast, female offspring from dams that were food-
a high-quality diet compared to those on a lower-quality diet. restricted during early lactation produced odors that were not
For example, both male and female meadow voles are more as attractive as those produced by female offspring from
attracted to odors of opposite-sex conspecifics that were fed control dams or dams that were food restricted during middle
a high-protein diet compared to those fed a diet containing or late lactation (Sabau et al., 2014). These differences between
the standard level of protein in commercial rodent food or males and females indicate that there may be different sensitive
a low level of protein in the food (Ferkin et al., 1997b). Simi- periods for the development of scent-producing tissues in male
larly, gravid female salamanders, Plethodon cinereus, preferred and female meadow voles (Sabau et al., 2014).
Communication by Chemical Signals 303

Food-restricted and food-deprived animals can also modify to heavy metals decreased their fright response to the odors of
their normal responses to conspecific odors. For example, predators (Lefcort et al., 1998). The endocrine system can be
female meadow voles that were food deprived for 6 h or longer involved in the production and perception of scents. Therefore,
stopped showing a preference for the odors of a male over exposure to compounds affecting the endocrine system can have
those of a female (Pierce et al., 2005). a direct effect on olfactory communication. This includes
substances that unintentionally enter in the animals, but it
1.10.3.7.3 Effect of Health and Environmental Stressors also includes hormone-based contraceptives. In ring-tailed
on Odors lemurs, a contraceptive altered the composition of odors, elim-
Individuals may use odors as honest signals to assess the health inating individual-specific olfactory information, and rendered
status of conspecifics and choose mates accordingly (Kavaliers these odors less interesting for males than were odors from
et al., 2005; Penn and Potts, 1998a). A high parasite load or control females (Crawford et al., 2011).
active disease processes can affect the information contained
in odors, due to changes in the composition of commensal 1.10.3.7.4 Age and Odors: From Infancy to Senescence
microbes, the production of metabolites related to immuno- Fell et al. (2006) studied social integration in European badger
logical responses and a decrease in hormonal metabolites cubs from their time of emergence to full independence. They
due to a reduction in androgen levels during infection (Penn discovered that as cubs grew older they interacted more with
and Potts, 1998a). Infected individuals can also reduce their adults, initiating a greater number of social interactions with
scent-marking rates, which may reduce their attractiveness to adults. These cubs would mark themselves with the subcaudal
opposite-sex conspecifics (Zala et al., 2004). In several species, gland secretions of adult group members. Fell et al. (2006)
females have been shown to be more attracted to the odors of referred to this behavior as scent-theft and concluded that
unparasitized males over the odors of parasitized males. For cubs do so to carry a group scent. Such a group scent would
example, female mice avoid the urine of males infected with allow the cubs to become integrated into the social group
lice, compared to the urine of males that do not have lice and be recognized by adults.
(Kavaliers et al., 2003). Female mice and rats also show a pref- Due to their particular endocrine state, juvenile individuals
erence for urine of males without endoparasites over the urine may produce odors that are not yet attractive to adult conspe-
of males infected with diverse species of such parasites cifics. Some scent glands may not even have developed and
(Kavaliers et al., 1998; Willis and Poulin, 2000). In addition, started to produce attracting compounds. On the other hand,
female mice are as attracted to water as to urine of males a lack of adult, aggression-eliciting odors may provide juveniles
infected with the influenza virus, whereas the same females with protection against aggression from adults. In Syrian
are more attracted to urine of uninfected males than to urine hamsters, adult males paired with a juvenile male in a neutral
of infected males, which indicates that viral infection can arena spent most of their time investigating the anogenital area
reduce the attractiveness of male odors (Penn et al., 1998). of the juvenile and were not aggressive against that juvenile,
Sick animals can also be more prone to predation or para- while the juvenile male raised his tail and assumed
sites. For example, mice and rats parasitized by Toxoplasma a lordosis-like position (two behaviors that are typical in
will suffer a behavioral change induced by the parasite so estrous females). Adult males in this species spend little time
that instead of avoiding feline odors will be attracted to investigating the anogenital area of other adult males, so it is
them, increasing their risk of predation and the transfer of possible that the anogenital odors of juvenile males share
the parasite to the predator gut where it can complete its repro- some similarity with those of adult females or at least contain
ductive cycle (Vyas et al., 2007). Ticks show a preference for the some compounds that elicit a nonaggressive response from
odors of sick hedgehogs, Erinaceus europaeus, possibly because adult males (delBarco-Trillo et al., 2011b; Figure 4).
of the compound indole being produced in higher proportions Females in many species show a preference for the odors of
in the scents of sick hedgehogs (Bunnell et al., 2011). adult males over those of males around puberty. For example,
Many species are exposed to anthropogenic substances that the chemical composition of urine from prepubescent and
can affect the synthesis of chemical signals, signal detection, adult male rats differs in the quantities of three volatile
and the responses of conspecifics to those signals (Blocker and compounds, and female rats are more attracted to the urine
Ophir, 2013; Lürling and Scheffer, 2007; Tuomainen and of adult males than the urine of prepubescent males (Osada
Candolin, 2011). For example, female palmate newts, Lissotriton et al., 2009). Female meadow voles also prefer odors of older
helveticus, preferred odors of unexposed males to those of males males, possibly because such males tend to be heavier, have
exposed to nitrates (Secondi et al., 2009). Similarly, pesticides larger testes, and be in better condition (Ferkin, 1999).
disrupt sexual olfactory communication and mate-choice However, as males start to senesce and their condition starts
behaviors in red spotted newts, Notophthalmus viridescens to decrease, the attractivity of their odors may also decrease.
(Lürling and Scheffer, 2007). Agricultural runoff and sewage This may be the case because a decrease in general condition
effluents can alter mate choice in the swordtail fish, Xiphophorus gets reflected in the composition of their odors, or because sen-
birchmanni, leading to interspecies mating and hybridization esced males cannot invest as many resources into the produc-
with the closely related species Xiphophorus malinche (Fisher tion of costly sexual signals as younger males can. In mice,
et al., 2006). Chemicals can also interfere with the perception urine of senescent males contains a lower concentration of
of chemical signals; male Atlantic salmon exposed to anti- MUPs and of the androgen-dependent volatile compounds
androgenic insecticides suffer a reduced response to female brevicomin and thiazole compared to middle-aged males
pheromones (Tierney et al., 2010). In regards to predation, (Garratt et al., 2011). When urine samples of senesced males
tadpoles of the Columbia spotted frog, Rana luteiventris, exposed and middle-age males are freshly deposited, females do not
304 Communication by Chemical Signals

Figure 4 Effect of juvenile age on the time that adult males investigate anogenital and flank areas in Syrian hamsters. Data represent the percentage
of sniffing time during which the adult male sniffed the anogenital area of the young male. Sniffing time is an aggregate measure of anogenital area
sniffing and flank sniffing. The 50% line represents equal anogenital and flank sniffing. Values over the line represent a major percentage of anogen-
ital area sniffing, whereas values below the line represent a major percentage of flank area sniffing. Black circles indicate trials without aggression
and white circles indicate trials with adult aggression. Juveniles did not show any aggression against adult males. Reproduced from delBarco-Trillo,
J., McPhee, M.E., Johnston, R.E., 2011. Syrian hamster males below an age threshold do not elicit aggression from unfamiliar adult males. Aggres-
sive Behav. 37, 91–97, with permission from Wiley.

show a preference for the urine of middle-age males. However, avoidance of agonistic neighboring individuals may reduce
when the urine samples had been deposited for 12 h, females the potential danger of physical conflicts (delBarco-Trillo
were less attracted to the urine of senesced males than to the et al., 2009c). In social species, individual recognition is neces-
urine of middle-age males, indicating that the reduced content sary for the establishment and maintenance of social interac-
of MUPs in the urine of senesced males affects the longevity tions (Doty, 1986). In pair-bonding species, recognition of
and attractivity of the scent signals (Garratt et al., 2011). the partner is obviously important to maintain close and
The sensitivity to odors and the ability to discriminate long-term relationships. In promiscuous species, an individual
between odors of conspecifics can also decrease in aged must discriminate between a recent mate and other opposite-
animals (Doty and Kamath, 2014). Such decrease in olfactory sex individuals, in order to mate preferentially with new indi-
sensitivity can be due to changes in several structures, including viduals (Coolidge effect) (Carr et al., 1970). Most terrestrial
the olfactory epithelium, the olfactory bulb, and central brain mammals must be able to distinguish between the myriad of
regions, and several processes, including loss of selectivity of scent marks they encounter as they move their habitat to find
receptor cells, changes in neurotransmitters, and neuronal mal- mates, secure resources, and avoid predation. How animals
function due to neurodegenerative disease (Doty and Kamath, use such information has been a topic of discussion for many
2014; Mobley et al., 2014). In a recent study in meadow voles, years. The adaptive significance of scent marking has been pre-
individuals of up to 13 months of age were able to distinguish sented in a multitude of papers (Ferkin, 2015; Hurst and
among odors of different conspecifics and they could also Beynon, 2004; Johnston and delBarco-Trillo, 2009; Roberts,
distinguish between the odors of males and females (Ferkin 2007; Roberts et al., 2014). Several hypotheses have been sug-
et al., 2016). However, 15- to 18-month-old meadow voles gested by authors (Brown and Macdonald, 1985; Ralls, 1971;
could not differentiate between two same-sex individuals, Thiessen and Rice, 1976; Roberts, 2007).
although these older voles were still able to distinguish
between male and female odors, showing a preference for the
odors of opposite-sex conspecifics (Ferkin et al., 2016). 1.10.4.1.1 Hypotheses about Function
Here, we list 15 putative functions of scent marking and its role
in communication. We provide representative citations of
recent work which relate directly to the functions (Table 1).
1.10.4 Functions of Odor Communication The first and most popular hypothesis is that scent marking
serves a role in spacing behavior, particularly territoriality.
1.10.4.1 Scent Marking
Several studies on a variety of species, particularly involving
Individual recognition is functionally relevant across different males, have suggested that scent marks provide a chemical
social and mating systems. In asocial species, recognition and fence or deterrent that prevents intrusions in a territory by
Communication by Chemical Signals 305

Table 1 Fifteen hypotheses that explain the role scent marking

Hypothesis Support/Species Recent References

1. Scent marks function in territoriality Eurasian beaver (Castor fiber); wolf (Canis lupus); Cross et al. (2014), Llanenza et al. (2014), and
European badger (Meles meles) Tinnesand et al. (2015)
2. Scent marks function in mate attraction House mice (Mus musculus); lynx (Lynx lynx); Roberts et al. (2014), Thonhauser et al.,
sifaka (Propithecus candidus) 2013a,b, Vogt et al. (2014), Drea (2015), and
Greene and Drea (2014)
3. Scent marks function in providing social House mice; snow voles (Chionomys nivalis) Hurst et al. (1993), Stockley et al. (2013), and
status information Luque-Larena et al. (2001, 2002)
4. Scent marks function in providing Meadow voles (Microtus pennsylvanicus); otters Ferkin et al. (2016), Kean et al. (2015), Charlton
individually distinct information (Lontra canadensis); koalas (Phascolarctos (2015), and Kaur et al. (2014)
cinereus); house mice
5. Scent marks function to indicate a food Hog badger (Arctonyx collaris); canids (Canis Zhou et al. (2015), Potts et al. (2012), and
source spp.); wildcats (Felis silvestris) Piñeiro and Barja (2015)
6. Scent marks function during interspecies Black-tailed prairie dogs (Cynomys ludovicianus) Eads et al. (2016)
competition
7. Scent marks allow for scent matching Meriones unguiculatus, Meriones tamariscincus, Gromov (2015)
Meriones meridianus
8. Scent marks provide a chemical bulletin Lynx; puma (Felis concolor); white-footed Vogt et al. (2014), Allen et al. (2014), and
board sportive lemurs (Lepilemur leucopus) Dröscher and Kappeler (2014)
9. Scent marks provide an information center Ocelots (Leopardus pardalis) Rodgers et al. (2015)
10. Scent marks allow group cohesion Red foxes (Vulpes vulpes) Soulsbury and Fawcett (2015)
11. Scent marks provide information about African wild dogs (Lycaon pictus) Jordan et al. (2014)
a social bond
12. Scent marks provide information that affects C57BL/6J mice (Mus domesticus); Rusa deer Arakawa et al. (2015), Ceacero et al. (2015), and
aggression in social encounters (Rusa timorensis); barasingha (Rucervus Pluhácek et al. (2015)
duvaucelii)
13. Scent marks of prey associated with those of Cabrera vole (Microtus cabrerae) Gomes et al. (2013)
predators reduces prey activity in that area
14. Scent marks provide information about Strepsirrhine primates delBarco-Trillo and Drea (2014)
species identity
15. Scent marks are used to facilitate parent House mice; humans (Homo sapiens); sheep Al Aïn et al. (2013), Corona and Lèvy (2015), and
offspring bond (Ovis aires); rats (Rattus norvegicus) Lonstein et al. (2015)

conspecifics (Brown and Macdonald, 1985; Thiessen and Rice, (Gromov, 2015; Gosling, 1982; Gosling and Roberts, 2001).
1976). Many recent studies have shown partial support for this Another function for scent marking is to provide a chemical
hypothesis, in that scent marks reduce intrusions by conspe- bulletin to indicate one’s presence in an area among lynx
cifics (Cross et al., 2014; Llaneza et al., 2014; Tinnesand (Vogt et al., 2014), puma, Felis concolor (Allen et al., 2014),
et al., 2015). Another frequently proposed function is that scent and lemurs (Dröscher and Kappeler, 2014). A ninth function
marking is used to attract mates (Johnston, 1983; Thiessen and suggests that scent marks act as an information center (Rodgers
Rice, 1976). Many studies provide data that support this et al., 2015). A tenth function of scent marking is to provide
hypothesis, particularly from those on house mice (Roberts cohesion among group members (Soulsbury and Fawcett,
et al., 2014; Thonhauser et al., 2013a,b), lynx, Lynx lynx 2015). Scent marks may also provide signals of a social bond
(Vogt et al., 2014) and sifaka, Propithecus candidus (Drea, between conspecifics to group members (Jordan et al., 2014).
2015; Greene and Drea, 2014). A third hypothesis is that scent A twelfth hypothesis is that scent marks provide information
marks provide information about the social status of the sender that affects the aggression displayed during paired encounters
(Hurst et al., 1993; Luque-Larena et al., 2001, 2002; Stockley between conspecifics (Arakawa et al., 2015; Ceacero et al.,
et al., 2013). A fourth hypothesis is that scent marks provide 2015; Pluhácek et al., 2015). A thirteenth function suggests
individually distinct information about the sender (Halpin, that scent marks from prey that are associated with those depos-
1986; Thom and Hurst, 2004) in a variety of animals, including ited by a predator serve as alarm signals that decrease activity in
meadow voles (Ferkin et al., 2016), otters (Kean et al., 2015), nearby conspecifics of the prey (Gomes et al., 2013). The 14th
koalas (Charlton, 2015), and house mice (Kaur et al., 2014). hypothesis is that scent marks provide information about species
A fifth function is related to defending or indicating a food identity (delBarco-Trillo and Drea, 2014). The last hypothesis is
resource among hog badgers, Arctonyx collaris (Zhou et al., that scent marks and odors produced by the parent and the
2015), canids (Potts et al., 2012), and wildcats, Felis silvestris offspring are used to facilitate a bond or attachment (Al Aïn
(Piñeiro and Barja, 2015). et al., 2013; Corona and Lévy, 2015; Lonstein et al., 2015).
It has also been hypothesized that scent marks are involved It is important to note that the 15 hypotheses listed above
in competition between species (Eads et al., 2016). A seventh may not be mutually exclusive. Some hypotheses may overlap
function is related to scent matching of conspecifics and may be necessary for other functions to be possible. For
306 Communication by Chemical Signals

example, it is difficult to imagine that a terrestrial mammal can the top- and bottom-scent marks as being distinct from one
identify the scent marks of conspecifics and learn features about another but also from a novel scent mark. These findings sug-
the donor’s phenotype, genotype, space use, and social biology gested that over-marks provide receivers with a chemical
(Ferkin, 2011; Green et al., 2015; Halpin, 1986; Johnston, bulletin board and on this bulletin board the top-scent mark
2009; Kaur et al., 2014). may have more value attached to it or salience compared to
the bottom-scent mark or to a scent mark that was not part
of the over-mark (Johnston et al., 1995; Woodward et al.,
1.10.4.2 Over-marking
1999, 2000). This finding has been repeated in subsequent
Over-marks are typically formed when the scent marks of two studies on these rodents (Ferkin et al., 2010, 2011a,b;
or more donors overlap (Ferkin and Pierce, 2007; Johnston Johnston, 2003, 2009) as well as in some primates (Fisher
et al., 1994; Macdonald, 1980). Our knowledge about how et al., 2003b; Heymann et al., 1998; Kappeler, 1998). The liter-
animals respond to over-marks, how they distinguish between ature is also replete with observations of other rodents, insecti-
the top- and bottom-scent donors of such marks, and what vores, ungulates, carnivores, and primates depositing their
kinds of information individuals can glean about the top- scent marks on or near the scent marks of conspecifics (Biben,
and bottom-scent donors has been discussed over the past 1980; Brashares and Arcese, 1999; Heymann et al., 1998; Hurst,
two decades and still continues in the present (Jezierski et al., 1990a,b,c; Johnson, 1973; Johnston et al., 1994, 1997a; Lewis,
2015; Jordan et al., 2014; Kaur et al., 2014; Tinnesand et al., 2005; Lisberg and Snowdon, 2009; Macdonald, 1980; Sliwa
2015; Vlautin and Ferkin, 2014). The pioneering work about and Richardson, 1998).
over-marking was carried out by Robert Johnston and co-
workers, who were interested in the perceptual mechanisms 1.10.4.2.1 Top- and Bottom-Scent Donors Are Same-Sex
that an animal may use to assess and differentiate between Conspecifics
the top-scent mark and bottom-scent mark of an over-mark. Our knowledge about how individuals respond to same-sex
Johnston (1994) provided essentially two hypotheses to over-marks is growing. The first wave of studies examined
explain how an individual may perceive an over-mark and its how conspecifics responded to same-sex over-marks in which
constituent components, the top-scent mark and the bottom- the top- and bottom-scent donors were similar in features of
scent mark. One hypothesis stated that the over-mark became their quality and were unfamiliar to the investigating conspe-
a mixture of two or more scent marks that were no longer cific. These studies on pygmy lorises, Syrian hamsters, prairie
distinct from one another. Johnston (1994) considered this voles, and meadow voles showed that after individuals were
mixture of two or more over-marks could be characterized as exposed to same-sex over-marks, they later displayed a selective
new or group scent which was made up of features that re- memory for the mark of the top-scent donor (Johnston et al.,
flected the identity of the group members rather than the iden- 1994, 1995) or they preferred that mark to the mark of the
tity of the individual members themselves. The other bottom-scent donor (Ferkin and Pierce, 2007; Fisher et al.,
hypothesis was that the scent marks in the over-mark did not 2003a; Woodward et al., 1999).
mix. Rather, the over-mark retains features of the individuals Ferkin (2011, 2015) has extended the three original hypoth-
that contribute to it (Johnston, 1993). In a series of elegant eses of Johnston (1994) to explain how individuals may
experiments Johnston et al. (1994, 1995, 1997a,b) determined perceive over-marks and respond to the top- and bottom-
whether Syrian hamsters and meadow voles treated the over- scent marks of two donors that are the same sex. One
lapping scent marks of conspecifics as if the top- and bottom- hypothesis is that individuals do not necessarily discriminate
scent mark mixed or remained distinct. To do so, Johnston the top-scent mark from the bottom-scent mark. Instead, the
and colleagues used a habituation/dishabituation paradigm individuals respond selectively to the top-scent mark because
to show that Syrian hamsters exposed to an over-mark of two it was the last mark to be deposited. In essence, the preferences
same-sex conspecifics later behaved as if they had a selective for the top-scent mark would be based on how recently it was
memory for the top-scent mark but not for the bottom-scent deposited. This simple rule of thumb would work if individuals
marks (Johnston et al., 1994, 1995). Similar results were ob- encounter an over-mark in which the top- and bottom-scent
tained when meadow voles were exposed to the over-mark of marks do not differ in features of their phenotype (Johnston
two same-sex conspecifics (Johnston et al., 1997a,b). These et al., 1994, 1995; Woodward et al., 1999) or if the top-scent
early papers suggested indicated that the two scent marks did donor is socially dominant to the bottom-scent donor (Ferkin
not mix but remained distinct. These data led to the hypothesis and Pierce, 2007; Hurst and Rich, 1999; Rozenfeld et al., 1987).
that by over-marking a scent mark the actor is attempting to The preference for the scent mark of the top-scent donor over
physically cover the bottom-scent mark or behaviorally block that of the bottom-scent donor suggests that many rodents
its information to subsequent receivers. and some primates exposed to same-sex over-mark behave as
This hypothesis was tested in studies on Syrian hamsters if they have a placed a higher value on the top-scent mark rela-
(Johnston and Bullock, 2001) and meadow voles (Ferkin tive to that of the bottom-scent mark (Ferkin and Pierce, 2007;
et al., 1999; Woodward et al., 1999). The results of these studies Fisher et al., 2003a; Johnston, 2009). This top-scent preference
were striking in that both meadow voles and Syrian hamsters may be rules of thumb for these animals. The first rule appears
were able to also discriminate the top-scent mark from a novel to be to prefer or have a selective memory for the top-scent
scent mark that was not part of an over-mark and the bottom- donor if the overlapping scent marks come from donors that
scent mark from a novel scent mark. This suggested that the do not differ sufficiently in some feature of their phenotype,
top-scent does not block the information contained in the such as their age, reproductive condition, social status, sex,
bottom-scent mark but also that the voles and hamsters viewed and diet (Ferkin and Pierce, 2007). Thus, we would expect to
Communication by Chemical Signals 307

see a preference for the top-scent donor of a same-sex over- conspecifics is affected by 4 days of olfactory experience with
mark if higher quality donor is the top-scent donor, the resi- those conspecifics. They found that female meadow voles spent
dent in an area rather than an intruder, or socially dominant more time investigating the scent mark of the novel male
to the bottom-scent donor. However, research on a variety of conspecific than that of the familiar male donor, whereas
rodents suggests that discriminating between and responding male voles spent similar amounts of time investigating the
to the top-scent and the bottom-scent mark of an over-mark scent mark of the familiar female and a novel female conspe-
may be more complex than simply preferring or having a selec- cific (Figure 5). Ferkin et al. (2010) also found that voles
tive memory for the top-scent mark. It appears the response exposed to a mixed-sex over-mark in which subjects did not
chosen by the individual depends on features of its genotype have 4 days of olfactory experience with either the top-scent
and phenotype, those of the top- and bottom-scent mark donor or the bottom-scent donor spent more time investi-
donors, the association between the investigating individual gating the scent mark of the opposite-sex conspecific that
and the donors, and context. And, most importantly, the provided the top-scent mark than that of a novel, opposite-
response chosen represents a trade-off in the benefits and costs sex conspecific. Male and female voles spent similar amounts
associated with over-marking and fitness and survival (Ferkin, of time investigating the scent mark of the bottom-scent donor
2015). and that of a novel opposite-sex conspecific (Figure 6). They
Because over-marking may occur in high-traffic areas also discovered that male voles exposed to a mixed-sex over-
(Brown and Macdonald, 1985; Ferkin et al., 2004a,b; Hurst, mark that contained the scent mark of a female conspecific
1990a,b,c; Hurst et al., 1993), individuals may encounter with which they had 4 days of olfactory experience spent
over-marks in which the top- and bottom-scent donors differ more time investigating the mark of the familiar, top-scent
in features of their phenotype and genotype. For example, female than the scent mark of a novel female donor but spent
they may encounter an over-mark in which a subordinate similar amounts of time investigating the mark of the familiar,
conspecific or an individual that is in poor nutritional health bottom-scent female and that of a novel female donor. In
over-marks the scent mark of a socially dominant conspecifics contrast, female voles spent more time investigating the mark
or one that is in good nutritional health. However, it is possible of a novel male donor than that of either the familiar, top-
that lower quality individuals may over-mark the scent marks scent male or that of the familiar, bottom-scent male
of higher quality individuals. If this occurs, do animals still (Figure 7).
maintain their preference for the top-scent donor over the
bottom-scent donor? If so, over-marking may not be an honest
signal but an alternative tactic that allows all top-scent donors
to gain an edge in the transfer of their olfactory information to
Olf exp
conspecifics, increase the value of their scent mark, or lower the
Novel 1
value of the bottom-scent mark. Interestingly, in those situa- Novel 2
tions when the top-scent donor is lower in quality relative to
the bottom-scent donor of a same-sex over-mark, investigating *
30
individuals no longer follow the rule of thumb to prefer the
top-scent donor to that of the bottom-scent donor or the alter-
25
native rule of thumb to prefer the marks of the higher quality
Investigation time (s)

scent donor to those of the lower quality scent donor.


20
Individuals may alter their response depending on features
of the top- and bottom-scent donor. For example, meadow
15
voles exposed to same-sex over-marks in which the top- and
bottom-scent donors differed in gonadal steroid hormone
10
titers, male voles preferred the marks of females that had higher
titers of estradiol than those that had lower titers of estradiol;
5
independently of whether they were the top- or the bottom-
scent donor. Female voles, however, preferred the scent marks
0
of males with higher testosterone titers than those of males Males Females
with lower testosterone titers but only when the former males
were the top-scent donors of the same-sex over-mark (Leonard Figure 5 The amount of time (mean  SEM) that male and female
et al., 2001). Similar results were obtained when meadow voles meadow voles spent investigating the scent mark of an opposite-sex
preferred the top-scent mark if the donor was fed a diet high in conspecific with which they had 4 days of olfactory experience (olf exp)
protein content but not if top-scent donor was fed a diet low in and that of a novel, opposite-sex conspecific as well as the time they
protein content. Voles preferred the bottom-scent mark if the spent investigating the scent marks of two different novel, opposite-sex
donor was fed a diet with a higher protein-content (Hobbs conspecifics. The scent marks used in the pairings were encountered
separately and were not part of a mixed-sex over-mark. * indicates
and Ferkin, 2011a, 2012a).
a statistical difference in investigation time between scent-mark pairings
(p < 0.05; multiple paired comparisons, Holm-Sidák method). Repro-
1.10.4.2.2 Mixed-Sex Over-marks and the Effects duced from Ferkin, M.H., Ferkin, D.A., Ferkin, B.D., Vlautin, C.T., 2010.
of Familiarity Olfactory experience affects the response of meadow voles to the
Ferkin et al. (2010) determined whether the amount of opposite-sex scent donor of mixed-sex over-marks. Ethology 116, 821–
time individuals investigate the scent marks of opposite-sex 831, with permission from Elsevier.
308 Communication by Chemical Signals

Top scent Top scent


Novel scent Novel scent
Bottom scent Bottom scent
* * *
30 30 * *
25 25
Investigation time (s)

Investigation time (s)


20 20

15
15

10
10
5
5
0
Males Females 0
Males Females
Figure 6 The amount of time (mean  SEM) that male and female
meadow voles spent investigating the scent mark of the top-scent, Figure 7 The amount of time (mean  SEM) that male and female
opposite-sex donor of the mixed-sex over-mark and that of a novel, meadow voles spent investigating the scent mark of the opposite-sex,
opposite-sex conspecific as well as the time they spent investigating the top-scent donor of a mixed-sex over-mark with which they had 4 days
bottom-scent, opposite-sex donor of a mixed-sex over-mark and that of of olfactory experience and that of a novel, opposite-sex conspecific as
a novel, opposite-sex conspecific. * indicates a statistical difference in well as the time they spent investigating the scent mark of the
investigation time between scent-mark pairings (p < 0.05; multiple opposite-sex, bottom-scent donor of a mixed-sex over-mark and that of
paired comparisons, Holm-Sidák method). Reproduced from Ferkin, a novel, opposite-sex conspecific. * indicates a statistical difference in
M.H., Ferkin, D.A., Ferkin, B.D., Vlautin, C.T., 2010. Olfactory experience investigation time between scent mark pairings (p < 0.05; multiple
affects the response of meadow voles to the opposite-sex scent donor paired comparisons, Holm-Sidák method). Reproduced from Ferkin,
of mixed-sex over-marks. Ethology 116, 821–831, with permission M.H., Ferkin, D.A., Ferkin, B.D., Vlautin, C.T. 2010. Olfactory experience
from Elsevier. affects the response of meadow voles to the opposite-sex scent donor
of mixed-sex over-marks. Ethology 116, 821–831, with permission
from Elsevier.

Collectively, these observations suggest that familiarity with


an opposite-sex conspecific has different effects on the behavior These results augment those obtained by Woodward et al.
of male and female meadow voles. Male voles may be spending (2000), who found that after encountering a mixed-sex over-
more time with the familiar female donor because she was the mark, prairie voles, and meadow voles later preferred the scent
top-scent donor in the mixed-sex over-mark and not because of mark of the opposite-sex donor to that of the same-sex donor,
any value that she may have gained by being familiar to that independent of whether the opposite-sex donor was encoun-
male. Being the top-scent female of a mixed-sex over-mark tered first as the top-scent donor or the bottom-scent mark in
may provide a signal to males that she is seeking mates the over-mark. Interestingly, male and female voles showed
(Woodward et al., 2000). Thus, she may somehow be more no preference between the mark of the bottom-scent donor
attractive to investigating males because of her prior associa- and the mark of an opposite-sex conspecific donor that was
tion with the bottom-scent male (Ferkin, 2011). However, not part of the over-mark. Thus, the position of the opposite-
male voles behave as if the scent marks of females that are sex donor’s mark in the mixed-sex over-mark determines how
bottom-scent donors are similar to those of novel female conspecifics will respond to its scent mark relative to the scent
donors in their attractivity. Thus, the bottom position in the mark of a novel, opposite-sex conspecific.
over-mark may signal to males that she has already found Unlike males, female voles spent more time during the test
a mate because of her association with the top-scent male phase investigating the mark of the novel male donor than the
(Woodward et al., 2000). Alternatively, by not being part of scent mark of the familiar male conspecific, independent of
a mixed-sex over-mark, and as a result not being associated whether he was the top- or bottom-scent donor. Female voles
with the scent mark of a male, the novel female donor’s scent may not respond preferentially to a familiar male if his scent
mark could indicate that she may or may not have mated mark was part of an over-mark with the scent mark of another
with another male. In any case, male voles may not respond female (Woodward et al., 2000), which may make that male
favorably to the scent marks of the bottom-scent female less attractive to a female seeking a mate. The preference of
because he is attempting to reduce competition with the top- a female meadow vole for a novel male is similar to the partner
scent male, who may have mated the bottom-scent female. preference found among female house mice (Cheetham et al.,
Similar conclusions about the role of familiarity and responses 2007). By preferring novel males to familiar males, female
to scent marks have been adduced for house mice (Arakawa voles may be encouraging sperm competition between males,
et al., 2015; Kaur et al., 2014) and the white-footed sportive increasing paternity confusion, or avoiding repeated copula-
lemur, Lepilemur leucopus (Dröscher and Kappeler, 2014). tions with the same partners.
Communication by Chemical Signals 309

1.10.4.2.3 Ten Hypotheses about the Function of Over-marks Lewis, 2005; Mertl-Millhollen et al., 1986; Palagi et al., 2004;
The literature is replete with observations of mammals over- Smith, 2006).
marking the scent marks of conspecifics and at least 10 hypoth- The second hypothesis is that over-marking provides
eses have been attributed to its role in odor communication a bulletin board that allows individuals to place their mark
(Ferkin and Pierce, 2007; Table 2). Many of these hypotheses along paths traversed by conspecifics or on a large feature in
suggest functions that are similar to those we presented for the area, such as a rock, tree, or mound (Johnston et al.,
scent marks earlier in this chapter. Briefly, the 10 hypotheses 1994). In this way, individuals may indicate their presence in
state that over-marking may be a form of competitive marking, an area (Ferkin, 2015). Direct and indirect evidence for this
a bulletin board, a form of self-advertisement, a territorial hypothesis can be drawn from studies on prairie voles, red-
behavior, a form of mate attraction, a means to increase bellied tamarins (Saguinus labiatus), marmosets, and meadow
foraging efficiency, a guide for navigation, the formation of voles (Lazaro-Perea et al., 1999; Smith and Gordon, 2002;
a group/colony scent, a threat behavior, a means to identify Thomas and Wolff, 2002; Woodward et al., 1999).
conspecifics, and an artifact of laboratory studies (Table 2). The third hypothesis is that an individual over-marks the
Some of these hypotheses are not mutually exclusive or may scent marks of intruders but is less likely to do so when it is
occur under very special conditions (Ferkin and Pierce, 2007). an intruder. Support for this hypothesis can be inferred from
The first hypothesis is that over-marking is a type of counter- studies on hyena (Crocuta crocuta), Eurasian beavers (Castor
marking that provides the top-scent donor with some advan- fiber), Ethiopian wolves (Canis simensis), rabbits, Alpine
tage in information transfer over the bottom-scent donor. marmots (Marmota marmota), aardwolves (Proteles cristata),
The advantage may be as simple as physically masking the pres- konik stallions (Konik polski), and klipspringers (Oreotragus
ence of the bottom-scent mark, as a means to incite competi- oreotragus) (Drea et al., 2002; Jezierski et al., 2015; Roberts
tion, as an indication of social rank or as a threat. Support and Dunbar, 2000; Rosell and Sanda, 2006; Rosell et al.,
for the competition hypothesis comes from studies on a variety 2000; Sillero-Zubiri and Macdonald, 1998; Sliwa and
of terrestrial mammals (Ferkin et al., 2004a,b; Hurst, 1990a,b; Richardson, 1998; Theis et al., 2008).

Table 2 Ten hypotheses to explain the role of over-marking

Hypothesis Support/Species Recent citations

1. Over-marking is a type of counter-marking Meadow voles; house mice; sifaka; wolf; Ferkin et al. (2004a,b), Hurst (1990a,b,c), Lewis
that provides the top-scent donor with lemurs; common marmosets (Callithrix (2005), Palagi et al. (2004), and Smith
some advantage in information transfer jacchus) (2006)
over the bottom-scent donor
2. Over-marking provides a chemical bulletin Prairie voles; red-bellied tamarins (Saguinus Lazaro-Perea et al. (1999), Thomas and Wolff
board labiatus); common marmosets; meadow (2002), Smith (2006), and Woodward et al.
voles (1999)
3. An individual over-marks the scent marks of Hyena (Crocuta crocuta); Eurasian beavers Theis et al. (2008), Drea et al. (2002), Jezierski
intruders but is less likely to do so when it is (Castor fiber); Ethiopian wolves (Canis et al. (2015), Roberts and Dunbar (2000),
an intruder simensis); rabbits; Alpine marmots (Marmota Rosell et al. (2000), Sillero-Zubiri and
marmota); aardwolves (Proteles cristata); Macdonald (1998), and Sliwa and
konik stallions (Konik polski); klipspringers Richardson (1998)
(Oreotragus oreotragus)
4. Over-marking is a form of mate attraction Meadow voles; zebras; hyenas; wolf; ring-tailed Ferkin et al. (2004a,b), Heymann (1998), Hurst
lemurs; moustached tamarins; house mice (1990c), Jezierski et al. (2015), Kappeler
(1998), Mertl-Millhollen et al. (1986),
Penzhorn (1984), and Tinnesand et al.
(2015)
5. Over-marking is an indication of a social or Hyena; prairie voles; wolf; klipspringers; Brashares and Arcese 1999, Drea et al. (2002),
sexual bond between the top- and bottom- antelope; moustached tamarins; ring-tailed Jordan et al. (2014), Heymann, 1998, Mertl-
scent donors lemurs Millhollen et al. (1986), Woodward et al.
(1999, 2000), and Palagi et al. (2004)
6. Over-marking allows scent marks to mix, Sifaka Greene and Drea (2014)
thereby providing group members with
a unique group scent
7. Over-marking the scent marks of Honey badgers (Mellivora capensis); otters Begg et al. (2003) and Kruuk (1995)
conspecifics increases foraging efficiency
by the individual and/or family members for
locating food sources
8. Over-marking allows aids in navigating Not tested directly Peterson (1988)
between familiar and unfamiliar areas
9. Over-marking serves no function Prairie voles Thomas and Wolff (2002) and Wolff (2004b)
10. Over-marking serves multiple functions Not tested directly Ferkin (2011) and Ferkin and Pierce (2007)
310 Communication by Chemical Signals

The fourth hypothesis is that over-marking is a form of mate condition, social status, age, and health (Ferkin and Pierce,
attraction. Thus, the top-scent mark may serve as a greeting or 2007).
invitation to the donor whose mark was over-marked, and
facilitate interactions between the two scent donors. The mate
1.10.4.3 An Over-mark versus a Single Scent Mark
attraction hypothesis has gained some support from studies
on ungulates, carnivores, rodents, and primates (Ferkin et al., It had been suggested that over-marks provide individuals with
2004a,b; Heymann, 1998; Hurst, 1990c; Jezierski et al., 2015; the opportunity to directly assess features of two same-sex
Kappeler, 1998; Mertl-Millhollen et al., 1986; Penzhorn, conspecifics that may not be available if individuals encoun-
1984; Tinnesand et al., 2015). tered the scent marks of these two conspecifics separately
The fifth hypothesis is that over-marking may be an (Hurst and Beynon, 2004; Johnston, 2003). A study by Ferkin
indication of a social or sexual bond between the top- and et al. (2011a) tested the hypothesis that after meadow voles
bottom-scent donors. If the over-mark is between opposite- enter areas containing different proportions of over-marks
sex conspecifics it may be a form of mate guarding (Hurst and single scent marks of two same-sex scent donors, voles
and Beynon, 2004; Woodward et al., 1999). For example, Jor- will differ in the amount of time they later spend investigating
dan et al. (2014) found that dominant African wild dogs the marks of these two donors. Males and females were allowed
(Lycaon pictus) over-mark the scent marks of their mates more to enter an arena that contained 10 over-marks and 0 single
often than their own marks and more likely to be part of marks of the conspecific that provided the bottom-scent mark
tandem marks with those of their siblings than those of a non- of the over-mark, nine over-marks and one single scent mark
siblings. Presumably, matched scents and tandem scent of the bottom-scent donor, eight over-marks, and two single
marking may be a way to combine territory defenses or to scent mark of the bottom-scent donor, and so on. Ferkin
advertise the pair’s relationship status to their group. et al. (2011a) discovered that after male voles entered an arena
The sixth hypothesis is that over-marking allows scent containing at least six over-marks and four single scent marks of
marks to mix, thereby providing group members with a unique the bottom-scent female donor of the over-mark, they spent
group scent. This group scent may provide cohesion among more time investigating the mark of the top-scent female donor
group members and allow individuals the means to distinguish than that of the bottom-scent female donor. When the ratio of
between group and nongroup members. Greene and Drea over-marks to single scent marks was less than 6:4, the males
(2014) discovered that mating pairs of sifakas copied each did not discriminate between the top- and bottom-scent female
other’s scent-marking behavior and produced similar odors donors. After female voles entered an arena containing at least
after they reproduced, whereas unmated pairs failed to do so. four over-marks and six single marks, they spent more time
The seventh hypothesis states that over-marking the scent investigating the mark of the top-scent male donor than that
marks of conspecifics increases foraging efficiency by the indi- of the bottom-scent male donor. When the ratio of over-
vidual and/or family members for locating food sources. marks to single scent marks was less than 4:6, the females did
Thus, individuals will over-mark the scent marks of conspe- not discriminate between the top- and bottom-scent male
cifics and their own scent marks to create a path that allows donors. Female voles also performed better than male voles
them to locate a food source, and may be akin to trail marking did in others tests involving discriminating between larger
in ants (Ferkin and Pierce, 2007). The foraging efficiency and smaller sets of scent marks (Ferkin and Hobbs, 2014; Fer-
hypothesis has gained indirect support from studies on honey kin et al., 2005; Vaughn and Ferkin, 2011). It is also worth
badgers (Mellivora capensis) and otters (Begg et al., 2003; Kruuk, pointing out that the proportions of over-marks that male
1995). and female voles needed to encounter to display a preference
The eighth hypothesis is that over-marking allows aids in for the mark of the second scent donor over that of the second
navigating between familiar and unfamiliar areas (Peterson, scent donor were similar to the proportion of scent marks
1988). This hypothesis has not been tested directly. males and females use to over-mark the scent marks of
The ninth hypothesis states that over-marking serves no a same-sex conspecific (Ferkin et al., 2004b).
function. This is essentially a null hypothesis, which is based Collectively, these results support and extend the view that
on reports of some mammals that rarely over-mark or over- over-marks provide individuals with the opportunity to
mark a very small proportion of the scent marks of conspecifics directly assess features of two same-sex conspecifics that may
(Thomas and Wolff, 2002; Wolff, 2004b). Although this not be available if individuals encountered the scent marks
hypothesis raises some interesting issues with regard to how of these two conspecifics separately (Ferkin et al., 2011a).
and where some over-marking studies are conducted, there is In this way, over-marks may also provide information about
little empirical evidence that shows that over-marking serves a possible association between the top- and bottom-scent
no function. donors (Ferkin and Pierce, 2007; Hurst and Beynon, 2004;
The tenth hypothesis states that over-marking serves Johnston, 2009). For example, over-marks provide a temporal
multiple functions. While it is tempting to assign all such tasks component in which the position of a donor’s scent mark as
to over-marking, it may be premature to suggest over-marking being either the top-scent donor or bottom-scent donor
is an all-purpose type of odor communication. The manner in conveys information to a receiver about the relative time frame
which individuals respond to scent marks and whether they in which both donors deposited their marks (Ferkin et al.,
over-mark the scent marks of conspecifics may be context- 2011b). An over-mark may also contain information about
dependent (Ferkin, 2011). Differences in over-marking the two scent donors that led to the establishment of the
behavior may be concomitant with changes in their life history over-mark itself. Thus, it may represent an olfactory snap
patterns, season, space use and social tolerance, reproductive shot of a past ‘olfactory-based’ encounter between two scent
Communication by Chemical Signals 311

donors (Hurst and Beynon, 2004). However, single marks may to levels comparable to intact male voles by testosterone
be easier for individuals to learn about features of the pheno- therapy (Ferkin and Leonard, 2010; Leonard et al., 2001,
type and genotype of individual scent donors (Ferkin, 2011, 2005). Additionally, food-deprived voles spent less time than
2015; Green et al., 2015; Kaur et al., 2014) and recall them those that were not food deprived self-grooming in response
later (Roberts et al., 2014). to the odors of a sexually receptive, opposite-sex conspecific
(Hobbs et al., 2008). Lastly, meadow voles and prairie voles
also spent more time self-grooming in response to odors of
1.10.4.4 Self-Grooming as a Form of Scent Marking
unfamiliar and unrelated females than in response to those
Thiessen (1977) suggested that self-grooming may spread of their reproductively active sisters (Paz-y-Miño et al., 2002).
odoriferous substances found in the integument, which may
affect the responses of the groomer and the receiver. For 1.10.4.4.1.2 Seasonal Differences
example, prairie voles, meadow voles (Ferkin et al., 1996), Seasonal differences also exist in self-grooming in meadow
and male Mongolian gerbils (Harriman and Thiessen, 1986; voles. Short-photoperiod voles spent similar amounts of time
Thiessen and Harriman, 1986) that recently self-groomed self-grooming in response to the odors of both opposite and
produced odors than those of males that did not self-groom same-sex long photoperiod and short-photoperiod scent
to females. These results suggest that self-grooming may be donors (Leonard and Ferkin, 2005). In contrast, long-
a form of olfactory communication, akin to scent marking, photoperiod voles spent more time self-grooming in response
which allows groomers to transmit information about features to the odors of long-photoperiod, opposite-sex conspecifics
of their identity and sex to nearby conspecifics (Ferkin et al., compared to that in response to the odors of short-
1996; Thiessen, 1977; Wiepkema, 1979). Self-grooming may photoperiod, opposite-sex conspecifics (Ferkin et al., 1996;
enhance features of this information, which may make nearby Leonard and Ferkin, 2005).
conspecifics that encounter these odors more responsive to the These observations and findings indicate a communicative
groomer (Ferkin and Leonard, 2010; Oliveira et al., 2013; role for self-grooming (Ferkin and Leonard, 2010; Ferkin
Thiessen and Harriman, 1986; Vaughn et al., 2010). et al., 1996). During the breeding season, self-grooming
behavior may be an indicator of detection and discrimination
1.10.4.4.1 Mate Attraction of sexually receptive females by males and vice-versa and serve
Several studies have also provided indirect and direct support to attract opposite-sex conspecifics to the groomer. In male
for the role of self-grooming in mate attraction. Ground squir- rodents, this response is mediated by testosterone (Achiraman
rels, deer (Odocoileus virginianus), Mongolian gerbils, hedge- et al., 2014; Ferkin and Leonard, 2010). Thus, seasonal differ-
hogs, blind mole rats (Spalax spp.), and voles self-groom ences in self-grooming in voles may be attributed to photoperi-
more when they are exposed to odors of opposite-sex conspe- odically induced changes in gonadal steroids, prolactin, and
cifics than when they are exposed to those of same-sex conspe- melatonin titers (Ferkin et al., 2007; Leonard et al., 2005),
cifics (Brockie, 1976; Ferkin and Leonard, 2010; Ferkin et al., which are needed to support reproduction (Ferkin and Leonard,
1996; Shanas and Terkel, 1995; Steiner, 1973, 1974; Thiessen 2010; Leonard and Ferkin, 2005). In contrast to its role during
and Harriman, 1986; Witt et al., 1988, 1990). Next, male and the breeding season, self-grooming during the nonbreeding
female voles spent more time self-grooming in response to may have a function outside of mate attraction. Perhaps, during
odors of reproductively active opposite-sex conspecifics than the nonbreeding seasons self-grooming is a communicative
those that are reproductively quiescent (Ferkin and Leonard, behavior directed at both male and female conspecifics, serving
2010). Male voles also spend more time self-grooming in to announce the groomer’s presence in an area (Ferkin and
response to odors of a female vole in postpartum estrus than Leonard, 2010; Leonard and Ferkin, 2005) or to reduce agonism
they did in response to odors of females not in postpartum between conspecifics (Simeonovska-Nikolova and Dekov,
estrus (Ferkin, 2006; Ferkin and delBarco-Trillo, 2014). Male 2013). These latter two hypotheses remain untested.
house mice spent more time self-grooming in response to urine
of estrus female mice compared to that of female mice not in
1.10.4.5 Scent Marks Provide Public Information
estrus (Achiraman et al., 2014). However, male root voles,
Microtus oeconomus, spent more time self-grooming in response Because scent marks and over-marks provide individually
to urinary odors of lactating females than to urinary odors of distinct information about senders (Halpin, 1986; Johnston
nonlactating females (Yu et al., 2010). Thus, the sexual condi- and delBarco-Trillo, 2009; Kaur et al., 2014), receivers gain
tion of the scent donor affects the amount of time that rodents specific information about the scent marks of individuals that
self-groom. they encounter (Charlton, 2015; Ferkin, 2015; Roberts, 2007;
Sharpe, 2015). Indeed, this information may be viewed as
1.10.4.4.1.1 Condition and Familiarity of the Scent Donor public information. Public information as defined by Valone
The condition and familiarity of the scent donor affects the (2007) is the “indirect information used by individuals to esti-
amount of time that rodents self-groom. Gonadectomized mate the quality of environmental parameters.” Public infor-
male voles, house mice, and rats spent less time self- mation may be gleaned by individuals noting the behavior of
grooming in response to odors of reproductively active others or by the overt decisions made by them (Danchin
opposite-sex females than did their intact counterparts et al., 2004). This social information provided may not be
(Achiraman et al., 2014; Ferkin, 2006; Moore, 1986). In house intentional. For example, scent marks may contain inadvertent
mice, self-grooming behavior was partially restored by testos- social information. To examine this possibility Ferkin and Fer-
terone treatment, whereas self-grooming rates were restored kin (unpublished data) first exposed male voles to the scented
312 Communication by Chemical Signals

bedding of a sexually receptive female and the bedding or appendage or rubbing the anogenital area against the surface
a varying number of male conspecifics that were placed next of a substrate. These behaviors may have been initially involved
to the bedding of that female. After the exposure, male voles in thermoregulation, maintenance of the pelage, or removal of
were tested to assess if they spend different amounts of time ectoparasites that may also have concomitantly released
investigating the fresh scent mark of the female from their substances in the environment. These secretions likely con-
exposure phase and the scent mark of a novel female. The tests tained odiferous substances that affected the behavior of
revealed that male voles preferred the scent mark of a female receivers (releasing effect) or modified the behavior of the
whose bedding was associated with the bedding of three or sender (priming effect) (Bossert and Wilson, 1963). If the net
five males compared to that of a novel female. Male voles result of these behaviors increased an individual’s survival
showed no preferences for the scent mark of a novel female and fitness and if variation existed among individuals, selection
or that of the female who was associated with the bedding of pressures may have led to formalization and stereotyping of
zero, one, or two males. These preliminary findings suggest these behaviors, such that over time senders would be able to
that male voles may use social information provided by the make this signal more easily detected (Rozhnov, 2004). Such
scent marks of both rival males and that of a female conspecific a process would lead to the ubiquity of scent marking in terres-
to influence their odor preferences for a particular female. Such trial mammals and its role in odor communication (Brown and
preferences for the ‘popular’ female may facilitate mate Macdonald, 1985; Ferkin. 2015; Johnston and delBarco-Trillo,
copying. 2009). For the sender, the specific behaviors involved in odor
Mate copying by males has been reported among species of communication would involve behaviors like scent marking,
fish. Male guppies (Poecilia reticulata), sail fin mollies (Poecilia over-marking, and self-grooming, which have been reported
latipinna), and mosquito fish (Gambusia affinis) preferred to in nearly all terrestrial mammals (Ferkin, 2011), as well as
associate and copulate with female conspecifics that they the production of odiferous substances that have primer and
have observed to have had more suitors compared to females releasing-like properties (Bossert and Wilson, 1963; Müller-
that had no suitors (Wagner and Danchin, 2003; Witte and Schwarze, 1984) and may be species-specific (delBarco-Trillo
Ryan, 2002). It appears that male meadow voles, akin to and Drea, 2014; Setchell et al., 2011). For the receiver, they
male guppies and mosquito fish (Bierbach and Somer- would need to have the functional morphology to detect chem-
Trembo, 2015; Callender et al., 2012; Mautz and Jennions, ical signals that allows them to attend to those of conspecifics
2011), make facultative adjustments in their choice of a female more than those of heterospecifics. For odor communication to
if male conspecifics are present and the number of male evolve, the sender must gain a benefit that outweighs the
competitors increases. This speculation is intriguing in that survival or fitness cost of signaling this to occur. Likewise, the
male meadow voles are promiscuous (Berteaux et al., 1999; receiver that encounters the scent mark must not incur in costs
Boonstra et al., 1993), and mating with a female that may that are greater than what is gained in terms of survival and
have mated with three other males would lower that male’s fitness by attending to and responding to the message (Roberts,
paternity assurance (Birkhead, 2000), and result in an increase 2007; Roberts et al., 2014). In addition, because male and
in the risk and intensity of sperm competition (Parker and female terrestrial mammals differ in some aspects of the
Pizzari, 2010), which result in him increasing the amount of frequency, duration, and timing of displaying odor-related
sperm in his ejaculate and lowering the amount of sperm in behavior suggests that odor communication may also have
future ejaculations (delBarco-Trillo and Ferkin, 2004; Vaughn been influenced by sexual selection (Ferkin, 2011, 2015; Petty
et al., 2008). Thus, it is likely that individuals may have to and Drea, 2015; Stockley et al., 2013).
balance the costs of sending and receiving potentially public
information, which may reduce their survival or attract compet-
1.10.5.2 Comparative Approaches to the Study of Olfactory
itors, with the benefits of attempting to mate with a ‘popular’
Communication
female or that of the female who was associated with the
bedding of zero, one, or two males (Morand-Ferron et al., As pointed out in a recent paper by delBarco-Trillo and Drea
2010). (2014), there is a paucity of data on the evolution of odor
communication in terrestrial mammals. This is partly due to
the difficulty of combining chemical data from different studies
1.10.5 Evolution to perform comparative studies, the only solution to this
problem being the chemical characterization of odors in the
1.10.5.1 Basic Concepts
several species to be compared, which is quite a complex and
Odor-related behaviors may have evolved from behaviors asso- time-consuming task (delBarco-Trillo et al., 2011a). This type
ciated with the excretion of by-products of digestion (Steiger of de novo approach has been used in strepsirrhine primates
et al., 2011). Scent marking likely evolved from the excretion (delBarco-Trillo et al., 2011a, 2012; delBarco-Trillo and Drea,
of metabolic wastes. Because the digestive exudates provide 2014). These studies have shown, for example, that (1) species
information about the phenotype and genotype of the sender that rely on urine as a signal for chemical communication have
it is likely to provide accurate information to receivers. Another more chemical compounds in their urine than species that rely
route that may have been involved in the evolution of odor more on glandular secretions; (2) such differences between
communication may be attributed to the specialization and urine markers and nonurine markers suggest urine marking being
elaboration of scent-producing tissues in the integument. an ancestral behavior in solitary and nocturnal species,
Perhaps, early terrestrial mammals were able to draw secretions whereas glandular marking would have evolved in diurnal
from the integument by scratching themselves with an and social species; (3) chemical richness is higher in
Communication by Chemical Signals 313

multimalemultifemale species (in which more interindi- Olfactory-receptor genes vary greatly among individuals, and
vidual interactions and thus transfer of information occur) this genetic variation may result in individualized receptor
than in pair-bonded species; (4) male odors are more chemi- repertoires that provide different individuals with a differential
cally complex in codominant species than in female- perception of the environment (Logan, 2014).
dominant species; (5) the chemical composition of odors is
very species-specific, even in species that belong to the same
genus, providing reliable species scent signatures; and (6) chem- 1.10.5.4 Odors and Sexual Selection
ical signals in strepsirrhines follow a gradual mode of evolution
(i.e., there is a linear and positive relationship between chem- Sexual selection is divided in four subprocesses, two of them
ical differences and phylogenetic distances), rather than occurring before copulation (male–male competition and
a saltational mode of evolution, both when we consider strep- female choice) and the other two occurring after copulation
sirrhines as a whole or a series of species from the same genus (sperm competition and cryptic female choice) (Kuijper et al.,
(Figure 8). 2012). The role of odors in female choice has already been
described earlier in this chapter, whereas the relationship
between olfactory communication and cryptic female choice
remains to be investigated. Consequently, in this section we
1.10.5.3 Genetic Variation, Natural Selection, and Odors
will address how odors can mediate behaviors involved in
Olfactory communication can be a very important component male–male competition and sperm competition.
during speciation processes (Smadja and Butlin, 2009). In the Some degree of male–male competition can be found in all
Skyros wall lizard, Podarcis gaigeae, islet and mainland popula- vertebrate species, and it includes behaviors in which males
tions differ in the chemical composition of their odors, and compete for sexual access to females and/or prevent other
there is a relationship between pheromonal and genetic diver- males from gaining access to their mates. Scent marks are often
gence among populations (Runemark et al., 2011). In two used in such competitions. For example, ring-tailed lemurs
subspecies of mice, Mus musculus musculus and Mus musculus spread odors on their tails and direct the odors toward
domesticus, hybrids in the sympatric area produce an odor competing individuals by waving their tails in the direction
phenotype that is distinct from the two parental species, which of their opponent, a behavioral interaction called a ‘stink fight’
facilitates the avoidance of intraspecific mating (Ganem et al., (Jolly, 1966). Many ungulates mark or urinate as part of terri-
2014). The genetic basis for odor divergence between these torial and/or aggressive contests for dominance. Some species,
two Mus subspecies involves autosomal genes and genes on such as the hartebeest, Alcelaphus buselaphus, perform ‘scent-
the X chromosome (Ganem et al., 2014). Particular proteina- marking fights’ at territorial boundaries, in which two males
ceous pheromone families, like androgen-binding proteins, scent mark alternately and also attempt to remove the scent
are also shaped by selection in mice (Karn and Laukaitis, marks of their opponents (Gosling, 1982). In addition to scent
2014). In several mammalian species a direct relationship has marking in his territory, a male may also rub his scent gland
been reported between chemical odor profiles and MHC geno- secretions, urine, or other scents onto his own body; other
types (Drea, 2015; Setchell et al., 2011). There is also a genetic males can then match the scent marks deposited in the terri-
underpinning for olfactory reception (Logan, 2014): tory with the male that deposited them. For example,

Figure 8 Types of information obtained by comparing the chemical composition of urine in several strepsirrhine primates. (a) Mean  SEM number
of volatile compounds (including either all compounds detected or only those with a purported semiochemical function) in the urine of six species of
urine markers and six species of nonurine markers. (b) Relationship between the urinary chemical distance between all possible pairs of species and
their phylogenetic distance. ***p < 0.0005. Reproduced from delBarco-Trillo, J., Burkert, B.A., Goodwin, T.E., Drea, C.M., 2011. Night and day: the
comparative study of strepsirrhine primates reveals socioecological and phylogenetic patterns in olfactory signals. J. Evol. Biol. 24, 82–98, with
permission from Wiley.
314 Communication by Chemical Signals

territorial male hartebeest rub their antorbital gland onto their When a male vole mates while being exposed to odors of
side, and also rub their body in their own feces (Gosling and many other males, he decreases the number of sperm ejacu-
Roberts, 2001). Many other ungulate species produce wallows lated in comparison to when he is exposed to odors of only
in which they urinate and then roll around in the wallow one competing male (delBarco-Trillo and Ferkin, 2006). This
(Grau, 1976), e.g., the American bison, Bison bison (Coppedge result is in agreement with sperm competition theory. That is,
and Shaw, 2000). Capuchin monkeys (Cebus spp.) ‘urine as the number of competing males increase, the advantage of
wash’ their hands and then rub their hands up their flanks increasing the number of sperm decreases.
(Zeller, 1987). The response of a male may depend on the status or condi-
An advantage of assessing rivals via their odors is that tion of competing males. If the odors of surrounding males
dangerous physical interactions may be reduced or avoided. come primarily from subordinate males that are not likely to
Males may recognize each other’s scent, and either withdraw mate, a subject male may respond differently than if those
or continue competing depending on the information ob- odors belong to a highly competitive male. Similarly, when
tained. For example, after male snow voles investigated an a male meadow vole mates with a female in the presence of
arena containing the odors of another male, they behaved odors from an ad libitum fed male, he increases the number
less aggressively toward that male (i.e., the resident male) of sperm ejaculated, but he will not do this in the presence of
than toward an unfamiliar male (Luque-Larena et al., 2001). odors from a food-deprived male, indicating that subject males
The level of response to the odors of other males may also assign a lower competitive threat to odors of food-deprived
differ between monogamous and promiscuous species. In the males than to odors of ad libitum fed males (Vaughn et al.,
monogamous California mouse, Peromyscus californicus, males 2008).
scent mark and over-mark more in response to the odors of Exposure during sexual development to conspecific odors
a conspecific male that is the case in the promiscuous white- that convey a context with a high level of sperm competition
footed mouse, Peromyscus leucopus (Becker et al., 2012). can have developmental effects. For example, in bank voles,
Sperm competition occurs when two or more males copu- Myodes glareolus, males raised in a context in which odors of
late with the same female and their sperm ‘compete’ within adult males resembled a high level of sperm competition devel-
the female’s reproductive tract for the fertilization of her ova oped larger seminal vesicles (Lemaître et al., 2011).
(Parker, 1970). Males may change their sexual behavior The increase in sperm allocation outline above in response
(Stockley and Preston, 2004) or the number of sperm ejacu- to contexts of sperm competition can be used for conservation
lated (delBarco-Trillo and Ferkin, 2006) depending on the purposes. In the giant pandas, males exposed to the odors of
risk of sperm competition (the probability that a female will a conspecific male became more interested in investigating
mate with other males) or the intensity of sperm competition females (Bian et al., 2013). In this and other species that repro-
(number of males mating with a female). Some studies have duce poorly in captivity, such enhancement in sexual motiva-
shown that males assess the risk and intensity of sperm compe- tion could lead to important increases in sexual performance.
tition by paying attention to the odors of other males on Given that the risk of sperm competition is lower in
females or in the immediate environment of a female. For unmated females than in recently mated females, males are
example, a ram will copulate faster when semen of another generally expected to show a preference for unmated females.
male is spread on the female’s vulva (Lezama et al., 2001). Indeed, in collared lemmings, sexually experienced males
The ram’s semen does not trigger the same response, suggesting prefer the odors of an estrous unmated female than the odors
that a male is able to discriminate the odor of his own semen of a female recently mated with another male, whereas sexually
from the semen of other males. inexperienced males do not (Huck et al., 1984). In some
A high risk of sperm competition can also be signaled by the species, however, as in Syrian hamsters, males mate indiscrim-
presence of the odors of a conspecific male in the environment. inately and do not show a preference when exposed to
A series of experiments investigated this possibility in meadow unmated and mated estrous females (Johnston and
voles (delBarco-Trillo and Ferkin, 2004; delBarco-Trillo and Rasmussen, 1984).
Ferkin, 2006). Male voles mated either in a context with high In promiscuous species, males that mate with several females
risk of sperm competition (conveyed solely by the presence may become sperm-depleted, and females mating with such
of odors of another male) or in a context with low risk of sperm males may incur a reproductive cost (Wedell et al., 2002). If
competition (no odors of conspecifics present during the trial). females are able to discriminate between recently mated and
When another male’s odors were present, male voles increased unmated males, they may reduce the cost of mating with
the number of sperm that they delivered to their mate a sperm-depleted male by avoiding mating with recently mated
(delBarco-Trillo and Ferkin, 2004). This increase was not due males. Female rats, for example, prefer the odors of males that
to any change in copulatory behavior (delBarco-Trillo and have not copulated than those of males that have recently mated
Ferkin, 2004), which suggests that the odors of another male with another female (Krames and Mastromatteo, 1973).
trigger a physiological response that ultimately produces an
increase in the contractility of the cauda epididymis and vas
deferens, and thus a larger number of sperm being ejaculated 1.10.6 Cognition and the Assessment
(delBarco-Trillo and Ferkin, 2007a). In contrast to what has of Scent Marks
been found in voles, male house mice do not increase the
number of sperm in their ejaculates in response to a high level Chemical signals from scent marks and odor-producing tissues
of sperm competition conveyed by odors of other males provide a great deal of information about the donor to receivers
(Ramm and Stockley, 2009). (Ferkin, 2011; Roberts, 2007; Wyatt, 2014). The hypotheses
Communication by Chemical Signals 315

that best explain the functions of scent marking are likely to be Episodic
several and dependent on the species and the variety of envi- memory
ronmental and biological challenges it must face. Nevertheless,
the manner in which an animal uses this information to make Recall Encode
decisions will depend on the context and manner in which it Percept
encounters these scent marks (Franklin and Ferkin, 2006, Perception Attention
Perceptual Reinforcement
2008; Roberts et al., 2014). It is unlikely that a learned a rule
learning
of thumb that will allow the animal to respond appropriately
when they encounter different scent marks of different donors. Action Procedural
To select the appropriate response, an individual will need to selection memory
identify the phenotype, genotype of the donor, recall any inter-
actions with that donor, be aware of its own condition and
respond in a manner that will presumably increase the individ-
Environment
ual’s survival and fitness (Ferkin, 2015; Green et al., 2015; Senses Acts
Tinnesand et al., 2015).
Figure 9 The cognitive processes that underlie or mediate the inter-
Receivers can use the information found in the scent marks,
pretation of the scent mark by the individual. Reproduced from
over-marks, and scent plumes from self-grooming to locate the
Franklin, S., Ferkin, M.H., 2006. An ontology for comparative cognition:
donor, learn its identity, and determine the donor’s phenotype a functional approach. Comp. Cogn. Behav. Rev. 1, 36–52, with
or genotype. This will involve the receivers learning spatial and permission from the authors.
temporal associations among scent donors and their marks
(Ferkin and Pierce, 2007; Roberts et al., 2014), storing them,
and later recalling these associations (Franklin and Ferkin, information about the sender posing some type of threat or
2006, 2008). Receivers will also need to learn other features a mating opportunity (Franklin and Ferkin, 2006, 2008). If the
associated with the scent donor such as when the scent mark scent mark is from a potential mate, the response would likely
was deposited, how long it has been since the receiver encoun- be to self-groom, scent mark, or over-mark and seek out the
tered the scent donor, and where that encounter occurred donor (Ferkin and Leonard, 2010; Ferkin and Pierce, 2007;
(Franklin and Ferkin, 2006, 2008). Higher level cognitive pro- Gosling and Roberts, 2001). If the scent mark is from a familiar
cessing involving procedural memory, episodic memory, auto- same-sex conspecific, it may be ignored (Hurst et al., 1993). If,
biographical memory, and making judgments of numerical however, the scent mark is from an unfamiliar same-sex conspe-
discrimination would be required for receivers to make poten- cific, it may be investigated (Gosling, 1982; Roberts, 2007). If
tially informed decisions, when they encounter the scent marks the scent mark is from a heterospecific, the response would
of particular conspecifics (Ferkin, 2011; Ferkin and Hobbs, depend on whether or not the donor was perceived as a threat
2014; Ferkin et al., 2005, 2008). (Eads et al., 2016; Vlautin et al., 2010). If the threat is such that
it is too costly for the individual to be detected, it would avoid
scent marking or seeking out interactions with the particular
1.10.6.1 Processing Scent Marks donor (Apfelbach et al., 1991), or it may shift its nest or area
Gosling and Roberts (2001) and Roberts (2007) provided us of activity away from that donor (Je˛ drzejewski et al., 1993).
with an excellent flowchart for the processing of scent marks in If a receiver chooses, it may deposit its own scent mark and
house mice. Ferkin (2015) extended this flow chart to incorpo- become a sender (Ferkin, 2015; Johnston, 2003, 2009). This
rate key cognitive elements that would be used by voles and action would return us to the first step of the schema (Figure 9).
potentially other mammals when they encounter scent marks,
which was developed by Franklin and Ferkin (2006, 2008),
1.10.6.2 Cognitive Ability, Numerosity, and Scent Marks
and illustrated in Figure 9. Briefly, the schema is as follows.
The first step in the schema involves the detection of the scent A variety of nonhuman animals in different taxa can discrimi-
mark, which involves sensory processes (environment, Figure 9). nate between a group and entity containing more objects from
After the scent mark is detected, it must be attended to (percep- one containing fewer of the same objects (Beran, 2001; Pahl
tion, Figure 9). This involves cognitive processes that underlie et al., 2013; Perdue et al., 2012; Reznikova and, Ryabko,
or mediate the interpretation of the scent mark by the individual. 2011). One way to assess if animals display numerousness is
Interpreting the scent mark will involve discrimination, percep- to expose them simultaneously to a set with relatively large
tual, procedural, spatial memory, and episodic memory, catego- numbers of stimuli for one response and a set with a relatively
rization, and the weighting or assessment of valence that may be small number of stimuli for another response, and then give
attached to the sender (donor) of the scent mark by the receiver them a test to determine if they respond differently to either
(Franklin and Ferkin, 2006, 2008; Roberts et al., 2014; Figure 9). set or if they respond selectively to the larger set (Boysen and
The number of scent donors, their identity, age, and sex and Capaldi, 1993; Hauser, 2000). If animals respond selectively to
eavesdroppers may affect the value of the scent mark to the the larger set over the smaller set, usually by attending to it or
receiver (Ferkin and Hobbs, 2014; Kaur et al., 2014; Roberts investigating it (Boysen, 1997; Hauser et al., 2003), that would
et al., 2014). At this point, the receiver may choose whether to be an indication that they may have the ability to spontaneously
respond to the scent mark (action selection, Figure 9). If the scent discriminate between two different numerosities and have
mark induces a response, the simplest response may depend on a concept of relative numerousness (Beran, 2001; Beran et al.,
the receiver’s determination of whether the scent mark provides 2012). This capacity for making judgments of numerousness
316 Communication by Chemical Signals

may allow individuals to discriminate between potential mates. The information found in a scent mark, for example, should
Carazo et al. (2009) found that male mealworm beetles (Tene- therefore be individually distinct (Halpin, 1986; Johnston, 1993;
brio molitor) discriminated between the odors of females if the Kaur et al., 2014). The response when they encounter scent
ratio between the female scent donors exceeded 1:2. By keeping marks from different donors or under different contexts should
track of the number of females and their odors, males can maxi- be flexible, allowing receivers to adjust their responses accord-
mize their mating opportunities. Likewise, mammals may keep ingly, depending on the identity of the sender, past associations,
track of the odors and scent marks produced by conspecifics to and context (Ferkin, 2011). Thus, the response chosen will be
find mates. In that scent marks are cheat-resistant signals (Ferkin, modulated by the receiver’s ability and capacity for discrimina-
2011; Roberts, 2007) that provide accurate and current informa- tion, categorization, judgments of relative numerousness, and
tion about the genotype and phenotype of a donor (Green et al., recollection of the emotional valence attached to the event
2015; Kaur et al., 2014) differences in the number of scent marks (Ferkin, 2011; Franklin and Ferkin, 2006, 2008; Johnston and
or over-marks deposited by donors may be used by conspecifics delBarco-Trillo, 2009; Roberts et al., 2014). The particular
to select potential mates (Ferkin et al., 2005; Ferkin and Hobbs, response should represent a balance of the costs and benefits
2014; Vaughn and Ferkin, 2011). For example, female voles were associated with that choice (Ferkin, 2015) and the likelihood
offered the scent mark of both male donors; they spent more of eavesdropping or copying by conspecifics (Danchin et al.,
time investigating the scent mark of the male donor that earlier 2004; Valone, 2007). The response in question would have
provided more scent marks to that of the male that had provided been modified over time through evolutionary pressures placed
fewer scent marks (Ferkin and Hobbs, 2014). In that study, on individuals in that species (Roberts, 2007). In this way,
female meadow voles discriminated between the scent marks receivers in species that face similar ecological and social chal-
of two males when the difference in scent marks was 2 and lenges may display responses to scent marks that are more
the total number of scent marks was 6 (Ferkin and Hobbs, similar compared to those facing different challenges (Wyatt,
2014). Spending more time investigating the scent marks of 2014), indicating some degree of convergence and stability
a particular conspecific compared to that of another conspecific over time (Ferkin, 2015).
has been interpreted as the subject anticipating an amicable
social interaction with the preferred individual if it was
Acknowledgments
a same-sex conspecific and a mating attempt with that individual
if it was an opposite-sex conspecific (Ferkin and Seamon, 1987).
We dedicate this chapter to Dr Robert E. Johnston. Bob was our mentor,
Similarly, mealworm beetles were trained by using a sponta- colleague, and friend. He will be missed.
neous two-choice procedure in which males were exposed to
substrates bearing odors from different numbers of females in
increasing numerosity ratios (1:2, 1:3 and 1:4). Males discrimi- References
nated sources of odors reflecting 1 versus 4 and 1 versus 3
females, but not 2 versus 4 or 1 versus 2 (Carazo et al., 2009). Achiraman, S., Ganesh, D.S., Kannan, S., Kamalakkannan, S., Nimala, N.,
So, the beetles demonstrated the ability to discriminate between Archunan, G., 2014. Response of male mice to odours of female mice in different
stages of oestrous cycle: self-grooming behaviour and the effect of castration. Ind.
‘more or less smells’ within the limit of four with an operational
J. Exp. Biol. 52, 30–35.
signature ratio of 1:2. Al Aïn, S., Chraïti, A., Schall, B., Patris, B., 2013. Orientation of newborn mice to
lactating females: Identify biological substrates of semiochemical interest. Dev.
Psychol. 55, 113–124.
1.10.7 Concluding Thoughts Albers, H.E., 2015. Species, sex and individual differences in the vasotocin/vaso-
pressin system: relationship to neurochemical signaling in the social behavior
neural network. Front. Neuroendocrinol. 36, 49–71.
The manner in which many terrestrial mammals behave Albers, H.E., Prishkolnik, J., 1992. Sex differences in odor-stimulated flank marking in
depends on the information they receive from their environ- the golden hamster (Mesocricetus auratus). Horm. Behav. 26, 229–239.
ment (Ferkin, 2011). This information should serve some Albers, H.E., Rowland, C.M., 1989. Ovarian hormones influence odor stimulated flank
marking behavior in the hamster (Mesocricetus auratus). Physiol. Behav. 45,
function or functions involved in intraspecific and interspe-
113–117.
cific communication (Johnston, 2009). For many terrestrial Albers, H.E., Karom, M., Smith, D., 2002. Serotonin and vasopressin interact in
mammals, scent marks convey information about the individ- the hypothalamus to control communicative behavior. NeuroReport 13,
ual’s biotic and abiotic environment to nearby conspecifics 931–933.
and heterospecifics (Hurst and Beynon, 2004). These scent Albers, H.E., Karom, M., Whitman, D.C., 1996. Ovarian hormones alter the behavioral
response of the medial preoptic anterior hypothalamus to arginine-vasopressin.
marks are typically deposited on prominent objects or along Peptides 17, 1359–1363.
paths that are shared with conspecifics (Brown and Albers, H.E., Liou, S.Y., Ferris, C.F., 1988. Testosterone alters the behavioral response
Macdonald, 1985). Thus a variety of individuals will enter of the medial preoptic-anterior hypothalamus to microinjection of arginine vaso-
areas that contain scent marks and overlapping scent marks, pressin in the hamster. Brain Res. 456, 382–386.
Albers, H.E., Pollock, J., Simmons, W.H., Ferris, C.F., 1986. A V1-like receptor
over-marks of a variety of donors, including heterospecifics
mediates vasopressin-induced flank marking behavior in hamster hypothalamus.
and conspecifics. First, these signals must be detected. And, J. Neurosci. 6, 2085–2089.
as we have described in this chapter, terrestrial mammals Albone, E.S., 1984. Mammalian Semiochemistry: The Investigation of Chemical
have exquisite sensory apparatus to detect these signals and Signals between Mammals. Wiley and Sons, New York.
specialized areas in the brain to decode, integrate, store, recol- Alderson, J., Johnston, R.E., 1975. Responses of male golden hamsters (Mesocricetus
auratus) to clean and male scented areas. Behav. Biol. 15, 505–510.
lect this information (Petrulis, 2013). The response should be Allen, M.L., Wittmer, H.U., Wilmers, C.C., 2014. Puma communication behaviors:
tailored to maximize the fitness of the participants (Ferkin, understanding functional use and variation among sex and age classes. Behaviour
2015; Wyatt, 2014). 151, 819–840.
Communication by Chemical Signals 317

Apfelbach, R., Russ, D., Slotnick, B.M., 1991. Ontogenic changes in odor sensitivity, Bean, N.Y., Nunez, A.A., Conner, R., 1981. Effects of medial preoptic lesions on male
olfactory receptor area and olfactory receptor density in the rat. Chem. Senses 16, mouse ultrasonic vocalizations and copulatory behavior. Brain Res. Bull. 6,
209–218. 109–112.
Apps, P.J., 2013. Are olfactory signals hiding right under our noses? Naurwissen- Becker, E.A., Petruno, S., Marler, C.A., 2012. A comparison of scent marking between
schaften 100, 487–508. a monogamous and promiscuous species of Peromyscus: pair bonded males do
Arakawa, H., Blanchard, D.C., Blanchard, R.J., 2015. Central oxytocin regulates social not advertise to novel females. PLoS One 7, e32002.
familiarity and scent marking behavior that involves amicable odor signals between Been, L.E., Petrulis, A., 2008. The neurobiology of sexual solicitation: vaginal marking
male mice. Physiol. Behav. 146, 36–46. in female Syrian hamsters (Mesocricetus auratus). In: Chemical Signals in Verte-
Arakawa, H., Arakawa, K., Blanchard, D.C., Blanchard, R.J., 2008a. A new test brates, 11. Springer, New York, pp. 231–239.
paradigm for social recognition evidenced by urinary scent marking behavior in Been, L.E., Bauman, J.M., Petrulis, A., Chang, Y.H., 2012. X-ray kinematics analysis
C57BL/6J mice. Behav. Brain Res. 190, 97–104. of vaginal scent marking in female Syrian hamsters (Mesocricetus auratus). Physiol.
Arakawa, H., Blanchard, D.C., Arakawa, K., Dunlap, C., Blanchard, R.J., 2008b. Scent Behav. 105, 1021–1027.
marking behavior as an odorant communication in mice. Neurosci. Biobehav. Rev. Been, L.E., Petrulis, A., 2011. Chemosensory and hormone information are relayed
32, 1236–1248. directly between the medial amygdala, posterior bed nucleus of the stria terminalis,
Bamshad, M., Cooper, T.T., Karom, M., Albers, H.E., 1996. Glutamate and vaso- and medial preoptic area in male Syrian hamsters. Horm. Behav. 59, 536–548.
pressin interact to control scent marking in Syrian hamsters (Mesocricetus aur- Begg, C.M., Begg, K.S., Du Toit, J.T., Mills, M.G.L., 2003. Scent-marking behaviour of
atus). Brain Res. 731, 213–216. the honey badger, Mellivora capensis (Mustelidae), in the southern Kalahari. Anim.
delBarco-Trillo, J., Drea, C.M., 2014. Socioecological and phylogenetic patterns in the Behav. 66, 917–929.
chemical signals of strepsirrhine primates. Anim. Behav. 97, 249–253. Beran, M.J., 2001. Summation and numerousness judgments of sequentially presented
delBarco-Trillo, J., Ferkin, M.H., 2004. Male mammals respond to a risk of sperm sets of items by chimpanzees (Pan troglodytes). J. Comp. Psychol. 115, 181–191.
competition conveyed by odours of conspecific males. Nature 431, 446–449. Beran, M.J., Perdue, B.M., Parrish, A.E., Evans, T.A., 2012. Do social conditions affect
delBarco-Trillo, J., Ferkin, M.H., 2005. Two modes of input processing in relation to capuchin monkeys’ (Cebus apella) choices in a quantity judgment task? Front.
sperm competition in mammals. Acta Zool. Sin. 51, 1122–1129. Psychol. 3, 492.
delBarco-Trillo, J., Ferkin, M.H., 2006. Male meadow voles respond differently to risk Berteaux, D., Bety, J., Rengifo, E., Bergeron, J., 1999. Multiple paternity in meadow
and intensity of sperm competition. Behav. Ecol. 17, 581–585. voles (Microtus pennsylvanicus): investigating the role of the female. Behav. Ecol.
delBarco-Trillo, J., Ferkin, M.H., 2007a. Increased sperm numbers in the vas deferens Sociobiol. 45, 283–291.
of meadow voles, Microtus pennsylvanicus, in response to odors of conspecific Bian, X., Liu, D., Zeng, H., Zhang, G., Wei, R., Hou, R., 2013. Exposure to odors of
males. Behav. Ecol. Sociobiol. 61, 1759–1764. rivals enhances sexual motivation in male giant pandas. PLoS One 8, e69889.
delBarco-Trillo, J., Ferkin, M.H., 2007b. Risk of sperm competition does not influence Bills, K.L., Samuelson, D.A., Larkin, I.L.V., 2013. Anal glands of the Florida manatee,
copulatory behavior in the promiscuous meadow vole (Microtus pennsylvanicus). Trichechus manatus latirostris: a potential source of chemsensory signal expres-
J. Ethol. 25, 139–145. sion. Mar. Mammal Sci. 29, 280–292.
delBarco-Trillo, J., Johnston, R.E., 2011. Adult female hamsters require long and Biben, M., 1980. Over-marking of alien conspecific odors by Mongolian gerbils. Biol.
sustained exposures to heterospecific males to avoid interspecific mating. Evol. Behav. 5, 139–145.
Ecol. 25, 391–401. Bierbach, D., Somer-Trembo, C., 2015. Personality affects mate choice: bolder males
delBarco-Trillo, J., Gulewicz, K., Johnston, R.E., 2009a. Medial amygdala involvement show stronger effects under high competition. Behav. Ecol. http://dx.doi.org/
in discrimination of same-species and closely-related-species male stimuli in 10.1093/beheco/arv079.
estrous female Mesocricetus hamsters. Behav. Neurosci. 123, 758–763. Birke, L.I., 1984. Effects of estradiol and progesterone on scent-marking behavior of
delBarco-Trillo, J., Gulewicz, K., Segal, A., McPhee, M.E., Johnston, R.E., 2009b. Can female rats. Horm. Behav. 18, 95–98.
captivity lead to inter-species mating in two Mesocricetus hamster species? Birkhead, T.R., 2000. Promiscuity: An Evolutionary History of Sperm Competition.
J. Zool. 278, 308–312. Harvard University Press, Cambridge, MA.
delBarco-Trillo, J., LaVenture, A.B., Johnston, R.E., 2009c. Male hamsters discrimi- Block, M.L., Volpe, L.C., Hayes, M.J., 1981. Saliva as a chemical cue in the devel-
nate estrous state from vaginal secretions and individuals from flank marks. Behav. opment of social behavior. Science 211, 1062–1064.
Process. 82, 18–24. Blocker, T., Ophir, A., 2013. Cryptic confounding compounds: a brief consideration of
delBarco-Trillo, J., McPhee, M.E., Johnston, R.E., 2009d. Nonagonistic familiarity the influences of anthropogenic contaminants on courtship and mating behavior.
decreases aggression in male Turkish hamsters, Mesocricetus brandti. Anim. Acta Ethol. 16, 105–125.
Behav. 77, 389–393. Boonstra, R., 1994. Population cycles in microtines: the senescence hypothesis. Evol.
delBarco-Trillo, J., McPhee, M.E., Johnston, R.E., 2010. Adult female hamsters avoid Ecol. 8, 196–219.
interspecific mating after exposure to heterospecific males. Behav. Ecol. Sociobiol. Boonstra, R., Xia, X., Pavone, L., 1993. Mating system of the meadow vole, Microtus
64, 1247–1253. pennsylvanicus. Behav. Ecol. 4, 83–89.
delBarco-Trillo, J., Burkert, B.A., Goodwin, T.E., Drea, C.M., 2011a. Night and day: the Bossert, W.H., Wilson, E.O., 1963. The analysis of olfactory communication among
comparative study of strepsirrhine primates reveals socioecological and phyloge- animals. J. Theor. Biol. 5, 443–469.
netic patterns in olfactory signals. J. Evol. Biol. 24, 82–98. Boulet, M., Charpentier, M.J.E., Drea, C.M., 2009. Decoding an olfactory mechanism
delBarco-Trillo, J., McPhee, M.E., Johnston, R.E., 2011b. Syrian hamster males below of kin recognition and inbreeding avoidance in a primate. BMC Evol. Biol. 9, 281.
an age threshold do not elicit aggression from unfamiliar adult males. Aggr. Behav. Boysen, S.T., 1997. Representation of quantities by apes. Adv. Study Behav. 26,
37, 91–97. 435–462.
delBarco-Trillo, J., Sacha, C.R., Dubay, G.R., Drea, C.M., 2012. Eulemur, me lemur: Boysen, S.T., Capaldi, E.J., 1993. The Development of Numerical Competence: Animal
the evolution of scent-signal complexity in a primate clade. Phil. Trans. R. Soc. and Human Models. Erlbaum, Hillsdale, NJ.
Lond. B Biol. Sci. 367, 1909–1922. Bowers, J.M., Alexander, B.K., 1967. Mice: individual recognition by olfactory cues.
Baudoin, C., Haim, A., Durand, J.-L., 2013. Effect of conspecific and heterospecific Science 158, 1208–1210.
urine odors on the foraging behavior of the golden spiny mouse. Integr. Zool. 8, 1–8. Brashares, J.S., Arcese, P., 1999. Scent marking in territorial African antelope: II the
Baum, M.J., Kelliher, K.R., 2009. Complementary roles of the main and accessory economics of marking with faeces. Anim. Behav. 57, 11–17.
olfactory systems in mammalian mate recognition. Annu. Rev. Physiol. 71, Brockie, R., 1976. Self-anointing by wild hedgehogs, Erinaceous europaeus. Anim.
141–160. Behav. 24, 68–71.
Batzli, G.O., 1985. Nutrition. In: Tamarin, R.H. (Ed.), Biology of New World Microtus. Bronson, F.H., 1989. Mammalian Reproductive Biology. University of Chicago Press,
The American Society of Mammalogists, Special Publication, Lawrence, KS, Chicago.
pp. 779–811. Brown, D.S., Johnston, R.E., 1983. Individual discrimination on the basis of urine in
Beach, F.A., Gilmore, R.W., 1949. Response of male dogs to urine from females in dogs and wolves. In: Müller-Schwarze, D., Silverstein, R.M. (Eds.), Chemical
heat. J. Mammal. 30, 391–392. Signals in Vertebrates, vol. 3. Plenum, New York, pp. 343–346.
Bean, N.J., 1982. Olfactory and vomeronasal mediation of ultrasonic vocalizations in Brown, J.L., Eklund, A., 1994. Kin recognition and the major histocompatibility
male mice. Physiol. Behav. 28, 31–37. complex: an integrative review. Am. Nat. 143, 435–461.
Bean, N.J., Nyby, J., Kerchner, M., Dahinden, Z., 1986. Hormonal regulation of Brown, R.E., 1977. Odor preference and urine-marking scales in male and female rats:
chemosignal-stimulated precopulatory behaviors in male housemice (Mus mus- effects of gonadectomy and sexual experience on responses to conspecific odors.
culus). Horm. Behav. 20, 390–404. J. Comp. Physiol. Psychol. 91, 1190–1206.
318 Communication by Chemical Signals

Brown, R.E., 1985. The rodents II: suborder Myomorpha. In: Brown, R.E., Charlton, B.D., 2015. Chemosensory discrimination of identity and familiarity in koalas.
Macdonald, D.W. (Eds.), Social Odours in Mammals, pp. 345–457. Behav. Process. 119, 38–43.
Brown, R.E., Macdonald, D.W., 1985. Social Odours in Mammals. Clarendon Press, Charpentier, M.J.E., Crawford, J.C., Boulet, M., Drea, C.M., 2010. Message ‘scent’:
Oxford. lemurs detect the genetic relatedness and quality of conspecifics via olfactory cues.
Brown, R.E., Roser, B., Singh, P.B., 1989. Class I and class II regions of the major Anim. Behav. 80, 101–108.
histocompatibility complex both contribute to individual odors in congenic inbred Cheetham, S.A., Thom, M.D., Jury, F., Ollier, W.E.R., Beynon, R.J., Hurst, J.L., 2007.
strains of rats. Behav. Genet. 19, 659–674. The genetic basis of individual-recognition signals in the mouse. Curr. Biol. 17,
Brown, R.E., Roser, B., Singh, P.B., 1990. The MHC and individual odors in rats. In: 1771–1777.
Macdonald, D.W., Müller-Schwarze, D., Natynczuk, S.E. (Eds.), Chemical Signals in Chen, R., Irwin, D.M., Zhang, Y.P., 2012. Differences in selection drive olfactory
Vertebrates, vol. 5. Oxford University Press, New York, pp. 228–243. receptor genes in different directions in dogs and wolf. Mol. Biol. Evol. 29,
Brown, R.E., Singh, P.B., Roser, B., 1987. The major histocompatibility complex and 3475–3484.
the chemosensory recognition of individuality in rats. Physiol. Behav. 40, 65–73. Clancy, A.N., Coquelin, A., Macrides, F., Gorski, R.A., Noble, E.P., 1984. Sexual
Buchanan-Smith, H.M., Anderson, D.A., Ryan, C.W., 1993. Responses of cotton-top behavior and aggression in male mice: involvement of the vomeronasal system.
tamarins (Saguinus oedipus) to faecal scents of predators and non-predators. J. Neurosci. 4, 2222–2229.
Anim. Welf. 2, 17–32. Clapham, M., Nevin, O.T., Ramsey, A.D., Rosell, F., 2013. The function of strategic
Buesching, C.D., Stopka, P., MacDonald, D.W., 2003. The social function of allo- tree selectivity in the chemical signalling of brown bears. Anim. Behav. 85,
marking in the European badger (Meles meles). Behaviour 140, 965–980. 1351–1357.
Bunnell, T., Hanisch, K., Hardege, J., Breithaupt, T., 2011. The fecal odor of sick Commins, D., Yahr, P., 1984. Lesions of the sexually dimorphic area disrupt mating
hedgehogs (Erinaceus europaeus) mediates olfactory attraction of the tick Ixodes and marking in male gerbils. Brain Res. Bull. 13, 185–193.
hexagonus. J. Chem. Ecol. 37, 340–347. Converse, L.J., Carlson, A.A., Ziegler, T.E., Snowdon, C.T., 1995. Communication of
Burgdorf, J., Wood, P.L., Kroes, R.A., Moskal, J.R., Panksepp, J., 2007. Neurobiology ovulatory state to mates by female pygmy marmosets, Cebuella pygmaea. Anim.
of 50-kHz ultrasonic vocalizations in rats: electrode mapping, lesion, and phar- Behav. 49, 615–621.
macology studies. Behav. Brain Res. 182, 274–283. Cooke, B.M., 2006. Steroid-dependent plasticity in the medial amygdala. Neuroscience
Burgener, N., Dehnhard, M., Hofer, H., East, M.L., 2009. Does anal gland scent signal 138, 997–1005.
identity in the spotted hyena? Anim. Behav. 77, 707–715. Coppedge, B.R., Shaw, J.H., 2000. American bison Bison bison wallowing behavior
Byrne, P.G., Keogh, J.S., 2007. Terrestrial toadlets use chemosignals to recognize and wallow formation on tallgrass prairie. Acta Theriol. 45, 103–110.
conspecifics, locate mates and strategically adjust calling behaviour. Anim. Behav. Coquelin, A., 1992. Urine-marking by female mice throughout their reproductive cycle.
74, 1155–1162. Horm. Behav. 26, 255–271.
Caldwell, R.L., 1985. A test of individual recognition in the stomatopod Gonodactylus Corona, R., Lévy, F., 2015. Chemical olfactory signals and parenthood in mammals.
festae. Anim. Behav. 33, 101–106. Horm. Behav. 68, 77–90.
Caldwell, H.K., Smith, D.A., Albers, H.E., 2008. Photoperiodic mechanisms controlling Coureaud, G., Moncomble, A.-S., Montigny, D., Dewas, M., Perrier, G., Schaal, B.,
scent marking: interactions of vasopressin and gonadal steroids. Eur. J. Neurosci. 2006. A pheromone that rapidly promotes learning in the newborn. Curr. Biol. 16,
27, 1189–1196. 1956–1961.
Callender, S., Blackwell, P.R.Y., Jennions, M.D., 2012. Context-dependent male Crawford, J.C., Boulet, M., Drea, C.M., 2011. Smelling wrong: hormonal contraception
choice: the effects of competitor presence and competitor size. Behav. Ecol. 23, in lemurs alters critical female odour cues. Proc. R. Soc. Lond. B Biol. Sci. 278,
355–360. 122–130.
Camacho-Arroyo, I., Cerbón, M., Gamboa-Domínguez, A., González-Agüero, G., Cross, H.B., Zedrosser, A., Nevin, O., Rosell, F., 2014. Sex discrimination via anal
González-Mariscal, G., 1999. Immunocytochemical detection of estrogen and gland secretion in a territorial monogamous mammal. Ethology 120, 1044–1052.
progesterone receptors in the rabbit submandibular gland. Comp. Biochem. Da Costa, A.P.C., Broad, K.D., Kendrick, K.M., 1997. Olfactory memory and maternal
Physiol. 123, 179–186. behaviour-induced changes in c-fos and zif/268 mRNA expression in the sheep
Carazo, P., Font, E., Forteza-Behrendt, E., Desfilis, E., 2009. Quantity discrimination in brain. Mol. Brain Res. 46, 63–76.
Tenebrio molitor: evidence of numerosity discrimination in an invertebrate? Anim. Danchin, E., Giraldeau, L.-A., Valone, T.J., Wagner, R.H., 2004. Public information:
Cogn. 12, 463–470. from nosy neighbors to cultural evolution. Science 305, 487–491.
Carr, W.J., Hirsch, J.T., Balazs, J.M., 1980. Responses of male rats to odors from Delville, Y., De Vries, G.J., Ferris, C.F., 2000. Neural connections of the anterior
familiar vs novel females. Behav. Neural Biol. 29, 331–332. hypothalamus and agonistic behavior in golden hamsters. Brain Behav. Evol. 55,
Carr, W.J., Krames, L., Costanzo, D.J., 1970. Previous sexual experience and olfactory 53–76.
preference for novel versus original sex partners in rat. J. Comp. Physiol. Psychol. Delville, Y., De Vries, G.J., Schwartz, W.J., Ferris, C.F., 1998. Flank-marking behavior
71, 216–222. and the neural distribution of vasopressin innervation in golden hamsters with
Carr, W.J., Loeb, L.S., Dissinger, M.L., 1965. Responses of rats to sex odors. suprachiasmatic lesions. Behav. Neurosci. 112, 1486–1501.
J. Comp. Physiol. Psychol. 59, 370–377. Desjardins, C., Maruniak, J.A., Bronson, F.H., 1973. Social rank in house mice:
Caspers, B., Schroeder, F., Franke, S., Streich, W., Voigt, C., 2009. Odour-based differentiation revealed by ultraviolet visualization of urinary marking patterns.
species recognition in two sympatric species of sac-winged bats (Saccopteryx Science 182, 939–941.
bilineata, S. leptura): combining chemical analyses, behavioural observations and Dewsbury, D.A., Ferguson, B., Hodges, A.W., Taylor, S.A., 1986. Tests of preferences
odour preference tests. Behav. Ecol. Sociobiol. 63, 741–749. of deer mice (Peromyscus maniculatus bairdi) for individuals and their odors as
Ceacero, F., Pluhácek, J., Zábransky, M., 2015. Pre-orbital gland opening during a function of sex and estrous condition. J. Comp. Psychol. 100, 117–127.
aggressive interactions in rusa deer (Rusa timorensis). Behav. Process. 111, Doty, R.L., 1972. Odor preferences of female Peromyscus maniculatus bairdi for male
51–54. mouse odors of P. m. bairdi and P. leucopus noveboracensis as a function of
Cerbón, M.A., Camacho-Arroyo, I., Gamboa-Domínguez, A., González-Mariscal, G., estrous state. J. Comp. Physiol. Psychol. 81, 191–197.
1996. The rabbit submandibular gland: sexual dimorphism, effects of gonadec- Doty, R.L., 1973. Reactions of deermice (Peromyscus maniculatus) and whitefooted
tomy, and variations across the female reproductive cycle. J. Comp. Physiol. A mice (Peromyscus leucopus) to homospecific and heterospecific urine odors.
178, 351–357. J. Comp. Physiol. Psychol. 84, 296–303.
Chabout, J., Sarkar, A., Dunson, D.B., Jarvis, E.D., 2015. Male mice song syntax Doty, R.L., 1986. Odor-guided behavior in mammals. Experientia 42, 257–271.
depends on social contexts and influences female preferences. Front. Behav. Doty, R.L., Kamath, V., 2014. The influences of age on olfaction: a review. Front.
Neurosci. 9. Psychol. 5, 20.
Chamero, P., Leinders-Zufall, T., Zufall, F., 2012. From genes to social communi- Drea, C.M., 2015. D’scent of man: a comparative survey of primate chemosignals in
cation: molecular sensing by the vomeronasal organ. Trends Neurosci. 35, relation to sex. Horm. Behav. 68, 117–133.
597–606. Drea, C.M., Vignieri, S.N., Kim, H.S., Weldele, M.L., Glickman, S.E., 2002. Responses
Chang, Y.M., Kelliher, K.R., Baum, M.J., 2000. Steroidal modulation of scent inves- to olfactory stimuli in spotted hyenas (Crocuta crocuta): II. Discrimination of
tigation and marking behaviors in male and female ferrets (Mustela putorius furo). conspecific scent. J. Comp. Psychol. 116, 342–349.
J. Comp. Psychol. 114, 401–407. Drickamer, L.C., 1995. Rates of urine excretion by house mouse (Mus domesticus):
Charlton, B.D., 2014. Discrimination of sex and reproductive state in koalas, Phas- differences by age, sex, social status, and reproductive condition. J. Chem. Ecol.
colarctos cinerus, using chemical cues in urine. Anim. Behav. 91, 1118–1124. 21, 1481–1493.
Communication by Chemical Signals 319

Drickamer, L.C., 2001. Urine marking and social dominance in male house mice (Mus Ferkin, M.H., Dunsavage, J., Johnston, R.E., 1999. Meadow voles can discriminate
musculus domesticus). Behav. Process. 53, 113–120. between the top and bottom scent of an over-mark and prefer the top scent.
Dröscher, I., Kappeler, P.M., 2014. Maintenance of familiarity and social bonding via J. Comp. Psychol. 113, 43–51.
communal latrine use in a solitary primate (Lepilemur leucopus). Behav. Ecol. Ferkin, M.H., Lee, D.N., Leonard, S.T., 2004a. The reproductive state of female voles
Sociobiol. 68, 2043–2058. affects their scent marking behavior and the responses of male conspecifics to
Duarte, M., da Luz Mathias, M., Bastos-Silveira, C., 2015. Pair-bonding behaviour of such marks. Ethology 110, 257–272.
the sister species Microtus lusitanicus and M. duodecimcostatus. J. Ethol. 33, Ferkin, M.H., Leonard, S.T., Gilless, J.P., 2007. Exogeneous melatonin administration
213–223. affects self-grooming and conspecific odor preferences in long-photoperiod
Dunbar, I.F., 1977. Olfactory preferences in dogs: the response of male and female meadow voles (Microtus pennsylvanicus). Physiol. Behav. 91, 255–263.
beagles to conspecific odors. Behav. Biol. 20, 471–481. Ferkin, M.H., Li, H.Z., Leonard, S.T., 2004b. Meadow voles and prairie voles differ in
Eads, D.A., Browser, J., Poonamallee, M., Molina, S., Neill, J., White, L., 2016. Black- the percentage of conspecific marks that they over-mark. Acta Ethol. 7, 1–7.
tailed prairie dogs selectively urinate near rabbit urine: the scent of competition Ferkin, M.H., Sorokin, E.S., Johnston, R.E., 1996. Self-grooming as a sexually
between a rodent and a lagomorph? Ethol. Ecol. Evol. 28 (1), 102–109. dimorphic communicative behaviour in meadow voles, Microtus pennsylvanicus.
Egid, K., Brown, J.L., 1989. The major histocompatibility complex and female mating Anim. Behav. 51, 801–810.
preferences in mice. Anim. Behav. 38, 548–549. Ferkin, M.H., Sorokin, E.S., Johnston, R.E., 1997a. Effect of prolactin on the attrac-
Eisenberg, J.F., Kleiman, D.G., 1972. Olfactory communication in mammals. Annu. tiveness of male odors to females in meadow voles: independent and additive
Rev. Ecol. Syst. 3, 1–32. effects with testosterone. Horm. Behav. 31, 55–63.
Engel, C.R., 1990. Scent marking, residency, and female choice in the European rabbit Ferkin, M.H., Sorokin, E.S., Johnston, R.E., Lee, C.J., 1997b. Attractiveness of scents
(Oryctolagus cuniculus). In: Macdonald, D.W., Müller-Schwarze, D., varies with protein content of the diet in meadow voles. Anim. Behav. 53,
Natynczuk, S.E. (Eds.), Chemical Signals in Vertebrates, vol. 5. Oxford University 133–141.
Press, New York, pp. 329–335. Ferkin, M.H., Vlautin, C.T., Pierson, L.M., 2016. Age-related effects on individual
Erlinge, S., Sandell, M., Brinck, C., 1982. Scent-marking and its territorial significance discrimination among meadow voles, Microtus pennsylvanicus. In: Schulte, B.,
in stoats, Mustela erminea. Anim. Behav. 30, 811–818. Goodwin, T., Ferkin, M.H. (Eds.), Chemical Signals in Vertebrates, vol. 13. Springer
Fan, S., Luo, M., 2009. The organization of feedback projections in a pathway Press, New York, pp. 121–136.
important for processing pheromonal signals. Neuroscience 161, 489–500. Ferkin, M.H., Ferkin, D.A., Ferkin, B.D., Vlautin, C.T., 2010. Olfactory experience
Feldhoff, R.C., Rollmann, S.M., Houck, L.D., 1999. Chemical analysis of courtship affects the response of meadow voles to the opposite-sex scent donor of mixed-sex
pheromones in a plethodontid salamander. In: Johnston, R.E., Müller-Schwarze, D., over-marks. Ethology 116, 821–831.
Sorensen, P.W. (Eds.), Advances in Chemical Signals in Vertebrates. Plenum Press, Ferkin, M.H., Pierce, A.A., Sealand, R.O., delBarco-Trillo, J., 2005. Meadow voles,
New York, pp. 117–125. Microtus pennsylvanicus, can distinguish more over-marks from fewer over-marks.
Fell, R.J., Buesching, C.D., Macdonald, D.W., 2006. The social integration of Anim. Cogn. 8, 82–89.
European badger (Meles meles) cubs into their natal group. Behaviour 143, Ferkin, M.H., Briley, D., Ferkin, B.D., Hardaway, A., Applebury, T., 2011a. Responses
683–700. of meadow voles, Microtus pennsylvanicus, to areas containing over-marks and
Ferdenzi, C., Schaal, B., Roberts, S.C., 2010. Family scents: developmental changes in single scent marks of two opposite-sex conspecifics. Behaviour 148, 117–130.
the perception of kin body odor? J. Chem. Ecol. 36, 847–854. Ferkin, M.H., Combs, A., delBarco-Trillo, J., Pierce, A.A., Franklin, S.P., 2008.
Ferkin, M.H., 1988. The effect of familiarity on social interactions in meadow voles, Meadow voles display a capacity for what, where, and when. Anim. Cogn. 11,
Microtus pennsylvanicus: a laboratory and field study. Anim. Behav. 36, 147–159.
1816–1822. Ferkin, M.H., Hobbs, N.J., Ferkin, B.D., Ferkin, A.C., Ferkin, D.A., 2011b. Male and
Ferkin, M.H., 1999. Attractiveness of opposite-sex odor and responses to it vary with female meadow voles, Microtus pennsylvanicus, respond differently to scent
age and sex in meadow voles (Microtus pennsylvanicus). J. Chem. Ecol. 25, marks from the top- middle-, and bottom-scent donors of an over-mark. Curr. Zool.
757–769. 57, 441–447.
Ferkin, M.H., 2006. The amount of time that a meadow vole, Microtus pennsylvanicus, Ferris, C.F., Axelson, J.F., Shinto, L.H., Albers, H.E., 1987. Scent marking and the
self-grooms is affected by its reproductive state and that of the odor donor. Behav. maintenance of dominant/subordinate status in male golden hamsters. Physiol.
Process. 73, 266–271. Behav. 40, 661–664.
Ferkin, M.H., 2007. Effects of previous interactions and sex on over-marking in Ferris, C.F., Axelson, J.F., Martin, A.M., Roberge, L.F., 1989. Vasopressin immuno-
meadow voles. Behaviour 144, 1297–1313. reactivity in the anterior hypothalamus is altered during the establishment of
Ferkin, M.H., 2011. Odor-related behavior and cognition in meadow voles, Microtus dominant/subordinate relationships between hamsters. Neuroscience 29,
pennsylvanicus (Arvicolidae, Rodentia). Folia Zool. 60, 262–276. 675–683.
Ferkin, M.H., 2015. The response of rodents to scent marks: four broad hypotheses. Ferris, C.F., Delville, Y., Irvin, R.W., Potegal, M., 1994. Septo-hypothalamic organi-
Horm. Behav. 68, 43–52. zation of a stereotyped behavior controlled by vasopressin in golden hamsters.
Ferkin, M.H., delBarco-Trillo, J., 2014. The behavior of female meadow voles, Physiol. Behav. 55, 755–759.
Microtus pennsylvanicus, during postpartum estrus and the responses of males to Ferris, C.F., Gold, L., De Vries, G.J., Potegal, M., 1990a. Evidence for a functional and
them. Mammal. Biol. 79, 81–89. anatomical relationship between the lateral septum and the hypothalamus in the
Ferkin, M.H., Hobbs, N.J., 2014. Female meadow voles, Microtus pennsylvanicus, control of flank marking behavior in golden hamsters. J. Comp. Neurol. 293,
respond differently to the scent marks of multiple male conspecifics. Anim. Cogn. 476–485.
17, 715–722. Ferris, C.F., Irvin, R.W., Potegal, M., Axelson, J.F., 1990b. Kainic acid lesion of
Ferkin, M.H., Johnston, R.E., 1993. Roles of gonadal hormones in control of five vasopressinergic neurons in the hypothalamus disrupts flank marking behavior in
sexually attractive odors of meadow voles (Microtus pennsylvanicus). Horm. Behav. golden hamsters. J. Neuroendocrinol. 2, 123–129.
27, 523–538. Ferris, C.F., Meenan, D.M., Albers, H.E., 1986a. Microinjection of kainic acid into the
Ferkin, M.H., Johnston, R.E., 1995a. Effects of pregnancy, lactation and postpartum hypothalamus of golden hamsters prevents vasopressin-dependent flank-marking
oestrus on odour signals and the attraction to odours in female meadow voles, behavior. Neuroendocrinology 44, 112–116.
Microtus pennsylvanicus. Anim. Behav. 49, 1211–1217. Ferris, C.F., Meenan, D.M., Axelson, J.F., Albers, H.E., 1986b. A vasopressin
Ferkin, M.H., Johnston, R.E., 1995b. Meadow voles, Microtus pennsylvanicus, use antagonist can reverse dominant/subordinate behavior in hamsters. Physiol. Behav.
multiple sources of scent for sex recognition. Anim. Behav. 49, 37–44. 38, 135–138.
Ferkin, M.H., Leonard, S.T., 2010. Self-grooming as a form of olfactory communication Ferris, C.F., Albers, H.E., Wesolowski, S.M., Goldman, B.D., Luman, S.E., 1984.
in meadow voles and prairie voles (Microtus spp.). In: Kalueff, A.V., LaPorte, J.L., Vasopressin injected into the hypothalamus triggers a stereotypic behavior in
Bergner, C. (Eds.), Neurobiology of Grooming Behavior. Cambridge University golden hamsters. Science 224, 521–523.
Press, UK, pp. 19–47. Ferris, C.F., Pilapil, C.G., Hayden-Hixson, D., Wiley, R.G., Koh, E.T., 1992. Functionally
Ferkin, M.H., Pierce, A.A., 2007. Perspectives on over-marking: is it good to be on and anatomically distinct populations of vasopressinergic magnocellular neurons in
top? J. Ethol. 25, 107–116. the female golden hamster. J. Neuroendocrinol. 4, 193–205.
Ferkin, M.H., Seamon, J.O., 1987. Odor preferences and social behavior in meadow Fischer, R.B., McQuiston, J., 1991. A possible role for Syrian hamster, Mesocricetus
voles, Microtus pennsylvanicus: seasonal differences. Can. J. Zool. 65, auratus, vaginal secretion in inter-female competition. Anim. Behav. 42,
2931–2937. 949–954.
320 Communication by Chemical Signals

Fisher, H.S., Rosenthal, G.G., 2006. Female swordtail fish use chemical cues to select González-Mariscal, G., Caba, M., Martínez-Gómez, M., Bautista, A., Hudson, R., 2016.
well-fed mates. Anim. Behav. 72, 721–725. Mothers and offspring: the rabbit as a model system in the study of mammalian
Fisher, H.S., Swaisgood, R.R., Fitch-Snyder, H., 2003a. Countermarking by male maternal behavior and sibling interactions. Horm. Behav. 77, 30–41.
pygmy lorises (Nycticebus pygmaeus): do females use odour cues to select mates González-Mariscal, G., Melo, A.I., Zavala, A., Chirino, R., Beyer, C., 1993. Sex steroid
with high competitive ability? Behav. Ecol. Sociobiol. 53, 123–130. regulation of chin-marking behavior in male New Zealand rabbits. Physiol. Behav.
Fisher, H.S., Swaisgood, R.R., Fitch-Snyder, H., 2003b. Odor familiarity and female 54, 1035–1040.
preferences for males in a threatened primate, the pygmy loris Nycticebus pyg- Gorman, M.L., 1984. The response of prey to stoat (Mustela erminea) scent. J. Zool.
maeus: applications for genetic management of small populations. Natur- 202, 419–423.
wissenschaften 90, 509–512. Gosling, L.M., 1982. A reassessment of the function of scent marking in territories.
Fisher, H.S., Wong, B.B.M., Rosenthal, G.G., 2006. Alteration of the chemical envi- Z. Tierpsychol. 60, 89–118.
ronment disrupts communication in a freshwater fish. Proc. R. Soc. Lond. B Biol. Gosling, L.M., 1990. Scent-marking by resource holders: alternative mechanisms for
Sci. 273, 1187–1193. advertising the costs of competition. In: Macdonald, D.W., Müller-Schwarze, D.,
Fleming, A.S., Chee, P., Vaccarino, F., 1981. Sexual behaviour and its olfactory control Natynczuk, S.E. (Eds.), Chemical Signals in Vertebrates, vol. 5. Oxford University
in the desert woodrat (Neotoma lepida lepida). Anim. Behav. 29, 727–745. Press, Oxford, pp. 315–328.
Floody, O.R., 1989. Dissociation of hypothalamic effects on ultrasound production and Gosling, L.M., McKay, H.V., 1990. Competitor assessment by scent matching: an
copulation. Physiol. Behav. 46, 299–307. experimental test. Behav. Ecol. Sociobiol. 26, 415–420.
Floody, O.R., 2002. Time course of VMN lesion effects on lordosis and proceptive Gosling, L.M., Roberts, S.C., 2001. Scent-marking by male mammals: cheat-proof
behavior in female hamsters. Horm. Behav. 41, 366–376. signals to competitors and mates. Adv. Study Behav. 30, 169–217.
Floody, O.R., Comerci, J.T., Lisk, R.D., 1987. Hormonal control of sex differences in Gosling, L.M., Roberts, S.C., Thornton, E.A., Andrew, M.J., 2000. Life history costs of
ultrasound production by hamsters. Horm. Behav. 21, 17–35. olfactory status signalling in mice. Behav. Ecol. Sociobiol. 48, 328–332.
Floody, O.R., DeBold, J.F., 2004. Effects of midbrain lesions on lordosis and ultra- Grau, G.A., 1976. Olfaction and reproduction in ungulates. In: Doty, R.L. (Ed.),
sound production. Physiol. Behav. 82, 791–804. Mammalian Olfaction, Reproductive Processes and Behavior. Academic Press, New
Floody, O.R., O’Donohue, T.L., 1980. Lesions of the mesencephalic central gray York, pp. 219–242.
depress ultrasound production and lordosis by female hamsters. Physiol. Behav. Green, J.P., Holmes, A.M., Davidson, A.J., Paterson, S., Stockley, P., Beynon, R.J.,
24, 79–85. Hurst, J.L., 2015. The genetic basis of kin recognition in a cooperatively breeding
Floody, O.R., Petropoulos, A.C., 1987. Aromatase inhibition depresses ultrasound mammal. Curr. Biol. 25, 2631–2641.
production and copulation in male hamsters. Horm. Behav. 21, 100–104. Greene, L.K., Drea, C.M., 2014. Love is in the air: sociality and pair bondedness
Floody, O.R., Lisk, R.D., Vomachka, A.J., 1986. Facilitation of lordosis by estradiol in influence sifaka reproductive signalling. Anim. Behav. 88, 147–156.
the mesencephalic central gray. Physiol. Behav. 37, 587–595. Griffin, A.S., Blumstein, D.T., Evans, C.S., 2000. Training captive-based or trans-
Floody, O.R., Pfaff, D.W., Lewis, C.D., 1977. Communication among hamsters by located animals to avoid predators. Conserv. Biol. 14, 1317–1326.
high-frequency acoustic signals: II. Determinants of calling by females and males. Grimm, D., 2014. Animal research: male scent may compromise biomedical studies.
J. Comp. Physiol. Psychol. 91, 807. Science 344, 461.
Floody, O.R., Walsh, C., Flanagan, M.T., 1979b. Testosterone stimulates ultrasound Gromov, V.S., 2015. Scent marking in gerbils and its possible functions. Russ. J.
production by male hamsters. Horm. Behav. 12, 164–171. Theriol. 14, 113–128.
Floody, O.R., Merkle, D.A., Cahill, T.J., Shopp Jr., G.M., 1979a. Gonadal hormones Guo, Z., Holy, T.E., 2007. Sex selectivity of mouse ultrasonic songs. Chem. Senses 32,
stimulate ultrasound production by female hamsters. Horm. Behav. 12, 172–184. 463–473.
Franklin, S., Ferkin, M.H., 2006. An ontology for comparative cognition: a functional Halpern, M., Martinez-Marcos, A., 2003. Structure and function of the vomeronasal
approach. Comp. Cogn.Behav. Rev. 1, 36–52. system: an update. Progr. Neurobiol. 70, 245–318.
Franklin, S., Ferkin, M.H., 2008. Using broad cognitive models to apply computational Halpin, Z.T., 1986. Individual odors among mammals: origins and functions. Adv.
intelligence to animal cognition. In: Smolinski, T.G., Milanova, M.M., Hassanien, A.- Study Behav. 16, 39–70.
E. (Eds.), Applications of Computational Intelligence in Biology: Current Trends and Harding, S.M., McGinnis, M.Y., 2003. Effects of testosterone in the VMN on copu-
Open Problems. Springer-Verlag, Berlin, pp. 363–394. lation, partner preference, and vocalizations in male rats. Horm. Behav. 43,
Fu, X.W., Brudzynski, S.M., 1994. High-frequency ultrasonic vocalization induced by 327–335.
intracerebral glutamate in rats. Pharmacol.Biochem. Behav. 49, 835–841. Harding, S.M., McGinnis, M.Y., 2004. Androgen receptor blockade in the MPOA or
Gadbois, S., Reeve, C., 2014. Canine olfaction: scent, sign, and situation. In: VMN: effects on male sociosexual behaviors. Physiol. Behav. 81, 671–680.
Horowitz, A. (Ed.), Domestic Dog Cognition and Behavior. Springer-Verlag, Berlin Harding, S.M., McGinnis, M.Y., 2005. Microlesions of the ventromedial nucleus of the
Heidelberg, pp. 3–29. hypothalamus: effects on sociosexual behaviors in male rats. Behav. Neurosci.
Ganem, G., Rueff, C., Perriat-Sanguinet, M., 2014. The genetic architecture of che- 119, 1227–1234.
mosensory cues involved in species recognition: a behavioral approach in the Hart, B.L., 1974. Medial preoptic-anterior hypothalamic area and sociosexual behavior
house mouse. Behav. Genet. 44, 56–67. of male dogs: a comparative neuropsychological analysis. J. Comp. Physiol.
Garratt, M., Stockley, P., Armstrong, S.D., Beynon, R.J., Hurst, J.L., 2011. The scent Psychol. 86, 328–349.
of senescence: sexual signalling and female preference in house mice. J. Evol. Hart, B.L., Voith, V.L., 1978. Changes in urine spraying, feeding and sleep behavior of
Biol. 24, 2398–2409. cats following medial preoptic-anterior hypothalamic lesions. Brain Res. 145,
Getz, L.L., 1985. Habitats. In: Tamarin, R.H. (Ed.), Biology of New World Microtus. 406–409.
American Society of Mammalogists, Lawrence, Kansas, pp. 286–309. Harriman, A.E., Thiessen, D.D., 1986. Harderian letdown in male Mongolian gerbils
Geyer, L.A., Barfield, R.J., 1978. Influence of gonadal hormones and sexual behavior (Meriones unguiculatus) contributes to proceptive behavior. Horm. Behav. 19,
on ultrasonic vocalization in rats: I. Treatment of females. J. Comp. Physiol. 213–219.
Psychol. 92, 438–446. Harris, M.A., Murie, J.O., 1984. Discrimination of oestrous status by scent in
Giller, K., Huebbe, P., Doering, F., Pallauf, K., Rimbach, G., 2013. Major urinary Columbian ground squirrels. Anim. Behav. 32, 939–940.
protein 5, a scent communication protein, is regulated by dietary restriction and Hauser, M.D., 2000. What do animals think about numbers? Am. Sci. 88,
subsequent re-feeding in mice. Proc. R. Soc. Lond. B Biol. Sci. 280 (1757), 144–151.
20130101. Hauser, M.D., Tsao, F., Garcia, P., Spelke, E.S., 2003. Evolutionary foundations of
Gomes, L.A.P., Salgado, P.M.P., Barata, E.N., Mira, A.P.P., 2013. Alarm scent- number: spontaneous representation of numerical magnitudes by cotton-top
marking during predatory attempts in the Cabrera vole (Microtus cabrerae tamarins. Proc. R. Soc. Lond. B Biol. Sci. 270, 1441–1446.
Thomas, 1906). Ecol. Res. 28, 335–343. Hayashi, S., Kimura, T., 1974. Sex-attractant emitted by female mice. Physiol. Behav.
Gómez-Baena, G., Armstrong, S.D., Phelan, M.M., Hurst, J.L., Beynon, R.J., 2014. 13, 563–567.
The major urinary protein system in the rat. Biochem. Soc. Trans. 42, 886–892. Heeb, M.M., Yahr, P., 2000. Cell-body lesions of the posterodorsal preoptic
González-Mariscal, G., Chirino, R., Hudson, R., 1994. Prolactin stimulates emission of nucleus or posterodorsal medial amygdala, but not the parvicellular sub-
nipple pheromone in ovariectomized New Zealand white rabbits. Biol. Reprod. 50, parafascicular thalamus, disrupt mating in male gerbils. Physiol. Behav. 68,
373–376. 317–331.
González-Mariscal, G., Melo, A.I., Zavala, A., Beyer, C., 1990. Variations in chin- Hennessey, A.C., Whitman, D.C., Albers, H.E., 1992. Microinjection of arginine-
marking behavior of New Zealand female rabbits throughout the whole repro- vasopressin into the periaqueductal gray stimulates flank marking in Syrian
ductive cycle. Physiol. Behav. 48, 361–365. hamsters (Mesocricetus auratus). Brain Res. 569, 136–140.
Communication by Chemical Signals 321

Heth, G., Todrank, J., 2000. Individual odour similarities across species parallel Hurst, J.L., 1990a. Urine marking in populations of wild house mice Mus domesticus
phylogenetic relationships in the S. ehrenbergi superspecies of mole-rats. Anim. rutty. I. Communication between males. Anim. Behav. 40, 209–222.
Behav. 60, 789–795. Hurst, J.L., 1990b. Urine marking in populations of wild house mice Mus domesticus
Heth, G., Todrank, J., Johnston, R.E., 1998. Kin recognition in golden hamsters: rutty. II. Communication between females. Anim. Behav. 40, 223–232.
evidence for phenotype matching. Anim. Behav. 56, 409–417. Hurst, J.L., 1990c. Urine marking in populations of wild house mice Mus domesticus
Heth, G., Todrank, J., Johnston, R.E., 1999. Similarity in the qualities of individual Rutty. III. Communication between the sexes. Anim. Behav. 40, 233–243.
odors among kin and species in Turkish (Mesocricetus brandti) and golden Hurst, J.L., Beynon, R.J., 2004. Scent wars: the chemobiology of competitive sig-
(Mesocricetus auratus) hamsters. J. Comp. Psychol. 113, 321–326. nalling in mice. Bioessays 26, 1288–1298.
Heth, G., Todrank, J., Busquet, N., Baudoin, C., 2001. Odour-genes covariance and Hurst, J.L., Rich, T.J., 1999. Scent marks as competitive signals of mate quality. In:
differential investigation of individual odours in the Mus species complex. Biol. J. Johnston, R.E., Müller-Schwarze, D., Sorensen, P.W. (Eds.), Advances in Chemical
Linn. Soc. 73, 213–220. Signals in Vertebrates. Plenum Press, New York, pp. 209–225.
Heymann, E., 1998. Sex differences in olfactory communication in a primate, the Hurst, J.L., Fang, J., Barnard, C.J., 1993. The role of substrate odours in maintaining
moustached tamarin, Saginus mystax (Callitrichinae). Behav. Ecol. Sociobiol. 43, social tolerance between male house mice, Mus musculus domesticus. Anim.
37–45. Behav. 45, 997–1006.
Higham, J., Hebets, E., 2013. An introduction to multimodal communication. Behav. Hurst, J.L., Payne, C.E., Nevison, C.M., Marie, A.D., Humphries, R.E.,
Ecol. Sociobiol. 67, 1381–1388. Robertson, D.H.L., Cavaggioni, A., Beynon, R.J., 2001. Individual recognition in
Hobbs, N.J., Ferkin, M.H., 2011a. Effect of protein content of the diet on scent mice mediated by major urinary proteins. Nature 414, 631–634.
marking and over-marking behavior in meadow voles, Microtus pennsylvanicus. Hurst, J.L., Robertson, D.H.L., Tolladay, U., Beynon, R.J., 1998. Proteins in urine scent
Behaviour 148, 1027–1044. marks of male house mice extend the longevity of olfactory signals. Anim. Behav.
Hobbs, N.J., Ferkin, M.H., 2011b. Dietary protein content affects the responses of 55, 1289–1297.
meadow voles, Microtus pennsylvanicus, to over-marks. Acta Ethol. 14, 57–64. Hurst, J.L., Thom, M.D., Nevison, C.M., Humphries, R.E., Beynon, R.J., 2005. MHC
Hobbs, N.J., Ferkin, M.H., 2012a. The response of male meadow voles, Microtus odours are not required or sufficient for recognition of individual scent owners.
pennsylvanicus, to same- and mixed-sex over-marks depends on the reproductive Proc. R. Soc. Lond. B Biol. Sci. 272, 715–724.
state of the top- and bottom-female scent donors. Behaviour 149, 705–722. Irvin, R.W., Szot, P., Dorsa, D.M., Potegal, M., Ferris, C.F., 1990. Vasopressin in the
Hobbs, N.J., Ferkin, M.H., 2012b. Effects of food availability on proceptivity: a test of septal area of the golden hamster controls scent marking and grooming. Physiol.
the reproduction at all costs and metabolic fuels hypotheses. Behav. Process. 91, Behav. 48, 693–699.
192–197. Jaeger, R.G., Wise, S.E., 1991. A reexamination of the male salamander “sexy faeces
Hobbs, N.L., Aven, A.M., Ferkin, M.H., 2008. Self-grooming responses of meadow hypothesis”. J. Herpetol. 25, 370–373.
voles to the odors of opposite-sex conspecifics in relation to the dietary protein Je˛ drzejewski, W., Rychlik, L., Je˛ drzejewska, B., 1993. Responses of bank voles to
content of both sexes. Ethology 114, 1210–1217. odours of seven species of predators: experimental data and their relevance to
Huck, U.W., Banks, E.M., 1979. Behavioral components of individual recognition in the natural predator-vole relationships. Oikos 68, 251–257.
collared lemming (Dicrostonyx groenlandicus). Behav. Ecol. Sociobiol. 6, 85–90. Jemiolo, B., Xie, T.M., Novotny, M., 1991. Socio-sexual olfactory preference in female
Huck, U.W., Banks, E.M., 1980. The effects of cross-fostering on the behavior of two mice: attractiveness of synthetic chemosignals. Physiol. Behav. 50, 1119–1122.
species of North American lemmings, Dicrostonyx groenlandicus and Lemmus Jezierski, T., Jaworski, Z., Sobcyznska, M., Kaminska, B., Gorecka-Bruzda, A.,
trimucronatus: I. Olfactory preferences. Anim. Behav. 28, 1046–1052. Walczak, M., 2015. Excreta-mediated olfactory communication in konik stallions:
Huck, U.W., Banks, E.M., 1982. Differential attraction of females to dominant males: a preliminary study. J. Vet. Behav. Clin. Appl. Res. 10, 353–364.
olfactory discrimination in the brown lemming (Lemmus trimucronatus). Behav. Jia, H., Pustovyy, O.M., Waggoner, P., Beyers, R.J., Schumacher, J., Wildey, C.,
Ecol. Sociobiol. 11, 217–222. Barrett, J., Morrison, E., Salibi, N., Denney, T.S., Vodyanoy, V.J., Deshpande, G.,
Huck, U.W., Banks, E.M., 1984. Social olfaction in male brown lemmings (Lemmus 2014. Functional MRI of the olfactory system in conscious dogs. PLoS One. http://
sibiricus trimucronatus) and collared lemmings (Dicrostonyx groenlandicus): I. dx.doi.org/10.1371/journal.pone.0086362.
Discrimination of species, sex, and estrous condition. J. Comp. Psychol. 98, Johnson, E.K., 1977. Seasonal changes in the skin of mammals. Symp. Zool. Soc.
54–59. Lond. 39, 373–404.
Huck, U.W., Banks, E.M., Coopersmith, C.B., 1984. Social olfaction in male brown Johnson, R.P., 1973. Scent marking in mammals. Anim. Behav. 21, 521–535.
lemmings (Lemmus sibiricus ¼ trimucronatus) and collared lemmings (Dicrosto- Johnston, R.E., 1975a. Scent marking by male golden hamsters (Mesocricetus aur-
nyx groenlandicus): II. Discrimination of mated and unmated females. J. Comp. atus). I. Effects of odors and social encounters. Z. Tierpsychol. 37, 75–98.
Physiol. Psychol. 98, 60–65. Johnston, R.E., 1975b. Scent marking by male golden hamsters (Mesocricetus aur-
Huck, U.W., Banks, E.M., Wang, S.-C., 1981. Olfactory discrimination of social status atus). III. Behavior in a seminatural environment. Z. Tierpsychol. 37, 213–221.
in the brown lemming. Behav. Neural Biol. 33, 364–371. Johnston, R.E., 1977. The causation of two scent-marking behaviour patterns in
Huck, U.W., Lisk, R.D., Gore, A.C., 1985a. Scent marking and mate choice in the female hamsters (Mesocricetus auratus). Anim. Behav. 25, 317–327.
golden hamster. Physiol. Behav. 35, 389–393. Johnston, R.E., 1979. Olfactory preferences, scent marking, and ‘proceptivity’ in
Huck, U.W., Quinn, R.P., Lisk, R.D., 1985b. Determinants of mating success in the female hamsters. Horm. Behav. 13, 21–39.
golden hamster (Mesocricetus auratus) IV. Sperm competition. Behav. Ecol. Johnston, R.E., 1980. Responses of male golden hamsters to odors of females in
Sociobiol. 17, 239–252. different reproductive states. J. Comp. Physiol. Psychol. 94, 894–904.
Hudson, R., Distel, H., 1983. Nipple location by newborn rabbits: behavioural evidence Johnston, R.E., 1981. Attraction to odors in hamsters: an evaluation of methods.
for pheromonal guidance. Behaviour 85, 260–275. J. Comp. Physiol. Psychol. 95, 951–960.
Hudson, R., Distel, H., 1986. Pheromonal release of suckling in rabbits does not Johnston, R.E., 1983. Chemical signals and reproductive behavior. In:
depend on the vomeronasal organ. Physiol. Behav. 37, 123–128. Vandenbergh, J.G. (Ed.), Pheromones and Reproduction in Mammals. Academic
Hudson, R., González-Mariscal, G., Beyer, C., 1990. Chin marking behavior, sexual Press, New York, pp. 3–37.
receptivity, and pheromone emission in steroid-treated, ovariectomized rabbits. Johnston, R.E., 1986. Effects of female odors on the sexual behavior of male
Horm. Behav. 24, 1–13. hamsters. Behav. Neural Biol. 46, 168–188.
Hughes, N.K., Banks, P.B., 2010. Interacting effects of predation risk and signal Johnston, R.E., 1992. Vomeronasal and/or olfactory mediation of ultrasonic calling and
patchiness on activity and communication in house mice. J. Anim. Ecol. 79, scent marking by female golden hamsters. Physiol. Behav. 51, 437–448.
88–97. Johnston, R.E., 1993. Memory for individual scent in hamsters (Mesocricetus auratus)
Hughes, N.K., Korpimäki, E., Banks, P.B., 2010. The predation risks of interspecific as assessed by habituation methods. J. Comp. Psychol. 107, 201–207.
eavesdropping: weasel–vole interactions. Oikos 119, 1210–1216. Johnston, R.E., 2003. Chemical communication in rodents: from pheromones to
Huhman, K.L., Albers, H.E., 1993. Estradiol increases the behavioral response to individual recognition. J. Mammal. 84, 1141–1162.
arginine vasopressin (AVP) in the medial preoptic-anterior hypothalamus. Peptides Johnston, R.E., 2009. Individual odors and social communication, individual recogni-
14, 1049–1054. tion, kin recognition, and scent over-marking. Adv. Study Behav. 30, 439–505.
Humphries, R.E., Robertson, D.H.L., Beynon, R.J., Hurst, J.L., 1999. Unravelling the Johnston, R.E., Brenner, D., 1982. Species-specificity of scent marking in hamsters.
chemical basis of competitive scent marking in house mice. Anim. Behav. 58, Behav. Neural Biol. 35, 46–55.
1177–1190. Johnston, R.E., Bullock, T.A., 2001. Individual recognition by use of odours in
Hurst, J.L., 1987. The functions of urine marking in a free-living population of house golden hamsters: the nature of individual representations. Anim. Behav. 61,
mice, Mus domesticus Rutty. Anim. Behav. 35, 1433–1442. 545–557.
322 Communication by Chemical Signals

Johnston, R.E., delBarco-Trillo, 2009. Communication by chemical signals: behavior, Kean, E.F., Chadwick, E.A., Müller, C.T., 2015. Scent signals individual identity and
social recognition, hormones and the role of the vomeronasal and olfactory country of origin in otters. Mammal. Biol. 80, 99–105.
systems. In: Pffaf, D., Arnold, A.P., Fahrbach, S.E., Etgen, A.M., Rubin, R.T. (Eds.), Kent, L., Tang-Martínez, Z., 2014. Evidence of individual odors and individual
Hormones, Brain, and Behavior, vol. 2. Elsevier Science, New York, pp. 797–870. discrimination in the raccoon, Procyon lotor. J. Mammal. 95, 1254–1262.
Johnston, R.E., Jernigan, P., 1994. Golden hamsters recognize individuals, not just Kikuyama, S., Toyoda, F., Ohmiya, Y., Matsuda, K., Tanaka, S., Hayashi, H., 1995.
individual scents. Anim. Behav. 48, 129–136. Sodefrin: a female-attracting peptide pheromone in newt cloacal glands. Science
Johnston, R.E., Kwan, M., 1984. Vaginal scent marking: effects on ultrasonic calling 267, 1643–1645.
and attraction of male golden hamsters. Behav. Neural Biol. 42, 158–168. Kimchi, T., Xu, J., Dulac, C., 2007. A functional circuit underlying male sexual
Johnston, R.E., Mueller, U.G., 1990. Olfactory but not vomeronasal mediation of scent behaviour in the female mouse brain. Nature 448, 1009–1014.
marking by male golden hamsters. Physiol. Behav. 48, 701–706. Kimura, T., Hagiwara, Y., 1985. Regulation of urine marking in male and female mice:
Johnston, R.E., Peng, A., 2008. Memory for individuals: hamsters (Mesocricetus effects of sex steroids. Horm. Behav. 19, 64–70.
auratus) require contact to develop multicomponent representations (concepts) of Kirn, J., Floody, O.R., 1985. Differential effects of lesions in three limbic areas on
others. J. Comp. Psychol. 122, 121–131. ultrasound production and lordosis by female hamsters. Behav. Neurosci. 99,
Johnston, R.E., Rasmussen, K., 1984. Individual recognition of female hamsters by 1142–1152.
males: role of chemical cues and of the olfactory and vomeronasal systems. Klailova, M., Lee, P.C., 2014. Wild western lowland gorilla signal selectively using
Physiol. Behav. 33, 95–104. odor. PLoS One e99554.
Johnston, R.E., Chiang, G., Tung, C., 1994. The information in scent over-marks of Krames, L., Mastromatteo, L.A., 1973. Role of olfactory stimuli during copulation in
golden hamsters. Anim. Behav. 48, 323–330. male and female rats. J. Comp. Physiol. Psychol. 85, 528–535.
Johnston, R.E., Munver, R., Tung, C., 1995. Scent counter marks: selective memory Kruuk, H., 1995. Wild Otters: Predation and Populations. Oxford University Press,
for the top scent by golden hamsters. Anim. Behav. 49, 1435–1442. Oxford.
Johnston, R.E., Sorokin, E.S., Ferkin, M.H., 1997a. Female voles discriminate males’ Kuijper, B., Pen, I., Weissing, F.J., 2012. A guide to sexual selection theory. Annu.
over-marks and prefer top-scent males. Anim. Behav. 54, 679–690. Rev. Ecol. Evol. Syst. 43, 287–311.
Johnston, R.E., Sorokin, E.S., Ferkin, M.H., 1997b. Scent counter-marking by male Kumari, S., Prakash, I., 1984. Perception of oestrous odours by male Meriones
meadow voles: females prefer the top-scent male. Ethology 103, 443–453. hurrianae. Anim. Behav. 32, 927–929.
Johnston, R.E., Derzie, A., Chiang, G., Jernigan, P., Lee, H.-C., 1991. Sources of Kwan, M., Johnston, R.E., 1980. The role of vaginal secretion in hamsters sexual
individual odors in golden hamsters. Chem. Senses 16, 540–541. behavior: male’s responses to normal and vaginectomized females and their odors.
Jolly, A., 1966. Lemur Behaviour: A Madagascar Field Study. University of Chicago J. Comp. Physiol. Psychol. 94, 905–913.
Press, Chicago. Kyuhou, S., Gemba, H., 1998. Two vocalization-related subregions in the midbrain
Jones, R.B., Nowell, N.W., 1973. Aversive and aggression-promoting properties of periaqueductal gray of the guinea pig. Neuroreport 9, 1607–1610.
urine from dominant and subordinate male mice. Anim. Learn. Behav. 1, Labov, J.B., Wysocki, C.J., 1989. Vomeronasal organ and social factors affect urine
207–210. marking by male mice. Physiol. Behav. 45, 443–447.
Jordan, N.R., Golabek, K.A., Apps, P.J., Glifilian, G.D., NcNutt, J.W., 2013. Scent- Lai, S.-C., Johnston, R.E., 1994. Individual odors in Djungarian hamsters (Phodopus
mark identification and scent-marking behaviour in African wild dogs (Lycaon campbelli). Ethology 96, 117–126.
pictus). Ethology 119, 644–652. Lai, S.-C., Vasilieva, N.Y., Johnston, R.E., 1996. Odors providing sexual information in
Jordan, N.R., Mwanguhya, F., Kyabulima, S., Rüedi, P., Cant, M.A., 2010. Scent Djungarian hamsters: evidence for an across-odor code. Horm. Behav. 30, 26–36.
marking within and between groups of wild banded mongooses. J. Zool. 280, Lai, W.-S., Chen, A., Johnston, R.E., 2004. Patterns of neural activation associated
72–83. with exposure to odors from a familiar winner in male golden hamsters. Horm.
Jordan, N.R., Apps, P.J., Golabek, K.A., McNull, J.W., 2014. Top marks from top Behav. 46, 319–329.
dogs: tandem marking and pair bond advertisement in African wild dogs. Anim. Lai, W.-S., Johnston, R.E., 2002. Individual recognition after fighting by golden
Behav. 88, 211–217. hamsters: a new method. Physiol. Behav. 76, 225–239.
Juntti, S.A., Tollkuhn, J., Wu, M.V., Fraser, E.J., Soderborg, T., Tan, S., Honda, S.-I., Lai, W.-S., Ramiro, L.-L.R., Yu, H.A., Johnston, R.E., 2005. Recognition of familiar
Harada, N., Shah, N.M., 2010. The androgen receptor governs the execution, but individuals in golden hamsters: a new method and functional neuroanatomy.
not programming of male sexual and territorial behaviors. Neuron 66, 260–272. J. Neurosci. 25, 11239–11247.
Jürgens, U., 2009. The neural control of vocalization in mammals: a Review. J. Voice Landauer, M.R., Banks, E.M., Carter, C.S., 1978. Sexual and olfactory preferences of
23, 1–10. naive and experienced male hamsters. Anim. Behav. 26, 611–621.
Kairys, D.J., Magalhaes, H., Floody, O.R., 1980. Olfactory bulbectomy depresses Latour, Y., Perriat-Sanguinet, M., Caminade, P., Boursot, P., Smadja, C.M.,
ultrasound production and scent marking by female hamsters. Physiol. Behav. 25, Ganem, G., 2014. Sexual selection against natural hybrids may contribute to
143–146. reinforcement in a house mouse hybrid zone. Proc. R. Soc. Lond. B Biol. Sci. 281,
Kang, N., Baum, M.J., Cherry, J.A., 2009. A direct main olfactory bulb projection to 20132733.
the ‘vomeronasal’ amygdala in female mice selectively responds to volatile pher- Lazaro-Perea, C., Snowdon, C.T., de Fatima Arruda, M., 1999. Scent marking behavior
omones from males. Eur. J. Neurosci. 29, 624–634. in wild groups of common marmosets (Callithrix jacchus). Behav. Ecol. Sociobiol.
Kappeler, P.M., 1990. Social status and scent-marking behaviour in Lemur catta. 46, 313–324.
Anim. Behav. 40, 774–776. Leclaire, S., Nielsen, J.F., Thavarajah, N.K., Manser, M., Clutton-Brock, T.H., 2013.
Kappeler, P.M., 1998. To whom it may concern: the transmission and function of Odour-based kin discrimination in the cooperatively breeding meerkat. Biol. Lett. 9.
chemical signals in Lemur catta. Behav. Ecol. Sociobiol. 42, 411–421. Lefcort, H., Meguire, R.A., Wilson, L.H., Ettinger, W.F., 1998. Heavy metals alter the
Karn, R.C., Laukaitis, C.M., 2014. Selection shaped the evolution of mouse androgen- survival, growth, metamorphosis, and antipredatory behavior of Columbia spotted
binding protein (ABP) function and promoted the duplication of Abp genes. Bio- frog (Rana luteiventris) tadpoles. Arch. Environ. Contamin. Toxicol. 35, 447–456.
chem. Soc. Trans. 42, 851–860. Lemaître, J.-F., Ramm, S.A., Hurst, J.L., Stockley, P., 2011. Social cues of sperm
Kaur, Angeldeep W., Ackels, T., Kuo, T.-H., Cichy, A., Dey, S., Hays, C., Kateri, M., competition influence accessory reproductive gland size in a promiscuous
Logan, Darren W., Marton, Tobias F., Spehr, M., Stowers, L., 2014. Murine mammal. Proc. R. Soc. Lond. B Biol. Sci. 278, 1171–1176.
pheromone proteins constitute a context-dependent combinatorial code governing Leonard, S.T., Ferkin, M.H., 2005. Seasonal differences in self-grooming in meadow
multiple social behaviors. Cell 157, 676–688. voles, Microtus pennsylvanicus. Acta Ethol. 8, 86–91.
Kavaliers, M., Choleris, E., Pfaff, D.W., 2005. Genes, odours and the recognition of Leonard, S.T., Ferkin, M.H., Johnson, M.M., 2001. The response of meadow voles to
parasitized individuals by rodents. Trends Parasitol. 21, 423–429. an over-mark in which the two donors differ in gonadal hormone status. Anim.
Kavaliers, M., Colwell, D.D., Choleris, E., 1998. Parasitized female mice display Behav. 62, 1171–1177.
reduced aversive responses to the odours of infected males. Proc. R. Soc. Lond. B Leonard, S.T., Alizadeh-Naderi, R., Stokes, K., Ferkin, M.H., 2005. The role of pro-
Biol. Sci. 265, 1111–1118. laction and testosterone in mediating seasonal differences in the self-grooming
Kavaliers, M., Wiebe, J.P., Galea, L.A., 1994. Male preference for the odors of estrous behavior of male meadow voles, Microtus pennsylvanicus. Physiol. Behav. 85,
female mice is enhanced by the neurosteroid 3 alpha-hydroxy-4-pregnen-20-one 461–468.
(3-alpha HP). Brain Res. 646, 140–144. Lévy, F., Keller, M., Poindron, P., 2004. Olfactory regulation of maternal behavior in
Kavaliers, M., Fudge, M.A., Colwell, D.D., Choleris, E., 2003. Aversive and avoidance mammals. Horm. Behav. 46, 284–302.
responses of female mice to the odors of males infected with an ectoparasite and Lewis, R.J., 2005. Sex differences in scent-marking in sifaka: mating conflict or male
the effects of prior familiarity. Behav. Ecol. Sociobiol. 54, 423–430. services? Am. J. Phys. Anthropol. 128, 389–398.
Communication by Chemical Signals 323

Lezama, V., Orihuela, A., Angulo, R., 2001. Sexual behavior and semen characteristics Martinez, L.A., Petrulis, A., 2013. The medial preoptic area is necessary for sexual
of rams exposed to their own semen or semen from a different ram on the vulva of odor preference, but not sexual solicitation, in female Syrian hamsters. Horm.
the ewe. Appl. Anim. Behav. Sci. 75, 55–60. Behav. 63, 606–614.
Liang, H., Zhang, J., Zhang, Z., 2004. Food restriction in pregnant rat-like hamsters Martinez, L.A., Albers, H.E., Petrulis, A., 2010. Blocking oxytocin receptors inhibits
(Cricetulus triton) affects endocrine, immune function and odor attractiveness of vaginal marking to male odors in female Syrian hamsters. Physiol. Behav. 101,
male offspring. Physiol. Behav. 82, 453–458. 685–692.
Lin, D.Y., Zhang, S.Z., Block, E., Katz, L.C., 2005. Encoding social signals in the Martinez, L.A., Levy, M.J., Petrulis, A., 2013. Endogenous oxytocin is necessary for
mouse main olfactory bulb. Nature 434, 470–477. preferential Fos expression to male odors in the bed nucleus of the stria terminalis
Lindsay, D.R., 1965. The importance of olfactory stimuli in the mating behaviour of the in female Syrian hamsters. Horm. Behav. 64, 653–664.
ram. Anim. Behav. 13, 75–78. Martinez-Marcos, A., 2009. On the organization of olfactory and vomeronasal cortices.
Lisberg, A.E., Snowdon, C.T., 2009. The effects of sex, gonadectomy and status on Progr. Neurobiol. 87, 21–30.
investigation patterns of unfamiliar conspecific urine in domestic dogs, Canis Maruniak, J.A., Bronson, F.H., 1976. Gonadotropic responses of male mice to female
familiaris. Anim. Behav. 77, 1147–1154. urine. Endocrinology 99, 963–969.
Lischka, F.W., Gomez, G., Yee, K.K., Dankulich-Nagrudny, L., Lo, L., Haskins, M.E., Maruniak, J.A., Wysocki, C.J., Taylor, J.A., 1986. Mediation of male mouse urine
Rawson, N.E., 2008. Altered olfactory epithelial structure and function in feline marking and aggression by the vomeronasal organ. Physiol. Behav. 37, 655–657.
models of mucopolysaccharidoses I and VI. J. Comp. Neurol. 511, 360–372. Mateo, J.M., 2009. The causal role of odours in the development of recognition
Lisk, R.D., Nachtigall, M.J., 1988. Estrogen regulation of agonistic and proceptive templates and social preferences. Anim. Behav. 77, 115–121.
responses in the golden hamster. Horm. Behav. 22, 35–48. Mateo, J.M., Johnston, R.E., 2000a. Kin recognition and the ‘armpit effect’: evidence
Llaneza, L., Garcia, E.J., López-Bao, J.V., 2014. Intensity of territorial marking predicts of self-referent phenotype matching. Proc. R. Soc. Lond. B Biol. Sci. 267,
wolf reproduction: implications for wolf monitoring. PLoS One e93015. 695–700.
Logan, D.W., 2014. Do you smell what I smell? Genetic variation in olfactory Mateo, J.M., Johnston, R.E., 2000b. Retention of social recognition after hibernation in
perception. Biochem. Soc. Trans. 42, 861–865. Belding’s ground squirrels. Anim. Behav. 59, 491–499.
Lonstein, J.S., Lévy, F., Fleming, A.S., 2015. Common and divergent psychobiological Matochik, J.A., Barfield, R.J., 1991. Hormonal control of precopulatory sebaceous
mechanisms underlying maternal behaviors in non-human and human mammals. scent marking and ultrasonic mating vocalizations in male rats. Horm. Behav. 25,
Horm. Behav. 73, 156–185. 445–460.
Lumley, L.A., Sipos, M.L., Charles, R.C., Charles, R.F., Meyerhoff, J.L., 1999. Social Matochik, J.A., Sipos, M.L., Nyby, J.G., Barfield, R.J., 1994. Intracranial androgenic
stress effects on territorial marking and ultrasonic vocalizations in mice. Physiol. activation of male-typical behaviors in house mice: motivation versus performance.
Behav. 67, 769–775. Behav. Brain Res. 60, 141–149.
Luque-Larena, J.J., López, P., Gosálbez, J., 2001. Scent matching modulates space Mautz, B.S., Jennions, M.D., 2011. The effect of competitor presence and relative
use and agonistic behaviour between male snow voles, Chionomys nivalis. Anim. competitive ability on male mate choice. Behav. Ecol. 22, 769–775.
Behav. 62, 1089–1095. Mayeaux, D.J., Johnston, R.E., 2002. Discrimination of individual odors by hamsters
Luque-Larena, J.J., López, P., Gosálbez, J., 2002. Relative dominance affects use of (Mesocricetus auratus) varies with the location of those odors. Anim. Behav. 64,
scent-marked areas in male snow voles Chionomys nivalis. Ethology 108, 269–281.
273–285. McLean, S., 2014. Scent glands of the common bushtail possum (Trichosurus vul-
Lürling, M., Scheffer, M., 2007. Info-disruption: pollution and the transfer of chemical pecula). NZ J. Zool. 41, 193–202.
information between organisms. Trends Ecol. Evol. 22, 374–379. McClintock, M.K., 2002. Pheromones, odors, and vasanas: the neuroendocrinology of
Lydell, K., Doty, R.L., 1972. Male rat odor preferences for female urine as a function of social chemosignals in humans and animals. In: Pffaf, D., Arnold, A.P.,
sexual experience, urine age, and urine source. Horm. Behav. 3, 205–212. Fahrbach, S.E., Etgen, A.M., Rubin, R.T. (Eds.), Hormones, Brain, and Behavior.
Macdonald, D.W., 1980. Patterns of scent marking with urine and feces among Elsevier Science, pp. 797–870.
carnivore communities. Symp. Zool. Soc. Lond. 45, 107–139. Meikle, D.B., Kruper, J.H., Browning, C.R., 1995. Adult male house mice born to
Macrides, F., Clancy, A.N., Singer, A.G., Agosta, W.C., 1986. Male hamster undernourished mothers are unattractive to oestrous females. Anim. Behav. 50,
investigatory and copulatory responses to vaginal discharge: an attempt to 753–758.
impart sexual significance to an arbitrary chemosensory stimulus. Physiol. Melo, A.I., Chirino, R., Jiménez, A., Cuamatzi, E., Beyer, C., González-Mariscal, G.,
Behav. 33, 627–632. 2008. Effect of forebrain implants of testosterone or estradiol on scent-marking
Maggio, J.C., Whitney, G., 1985. Ultrasonic vocalizing by adult female mice (Mus and sexual behavior in male and female rabbits. Horm. Behav. 54, 676–683.
musculus). J. Comp. Psychol. 99, 420–436. Meredith, M., 1994. Chronic recording of vomeronasal pump activation in awake
Mahoney, P.D., Koh, E.T., Irvin, R.W., Ferris, C.F., 1990. Computer-aided mapping of behaving hamsters. Physiol. Behav. 56, 345–354.
vasopressin neurons in the hypothalamus of the male golden hamster: evidence of Meredith, M., 1998. Vomeronasal function. Chem. Senses 23, 463–466.
magnocellular neurons that do not project to the neurohypophysis. Mertl-Millhollen, A.S., Goodmann, P.A., Klinghammer, E., 1986. Wolf scent-marking
J. Neuroendocrinol. 2, 113–122. with raised-leg urination. Zoo Biol. 5, 7–20.
Malsbury, C.W., Kow, L.M., Pfaff, D.W., 1977. Effects of medial hypothalamic lesions Miceli, M.O., Malsbury, C.W., 1982. Sagittal knife cuts in the near and far lateral
on the lordosis response and other behaviors in female golden hamsters. Physiol. preoptic area-hypothalamus reduce ultrasonic vocalizations in female hamsters.
Behav. 19, 223–237. Physiol. Behav. 29, 953–956.
Maras, P.M., Petrulis, A., 2010a. Anatomical connections between the anterior and Mobley, A.S., Rodriguez-Gil, D.J., Imamura, F., Greer, C.A., 2014. Aging in the
posterodorsal sub-regions of the medial amygdala: integration of odor and hormone olfactory system. Trends Neurosci. 37, 77–84.
signals. Neuroscience 170, 610–622. Mohedano-Moriano, A., De La Rosa-Prieto, C., Saiz-Sanchez, D., Ubeda-Banon, I.,
Maras, P.M., Petrulis, A., 2010b. The anterior medial amygdala transmits sexual odor Pro-Sistiaga, P., De Moya-Pinilla, M., Martinez-Marcos, A., 2012. Centrifugal
information to the posterior medial amygdala and related forebrain nuclei. Eur. J. telencephalic afferent connections to the main and accessory olfactory bulbs.
Neurosci. 32, 469–482. Front. Neuroanat. 6, 19.
Mares, R., Young, A.J., Levesque, D.L., Harrison, N., Clutton-Brock, T.H., 2011. Moles, A., Costantini, F., Garbugino, L., Zanettini, C., D’Amato, F.R., 2007. Ultrasonic
Responses to intruder scents in the cooperatively breeding meerkat: sex and social vocalizations emitted during dyadic interactions in female mice: a possible index of
status differences and temporal variation. Behav. Ecol. 22, 594–600. sociability? Behav. Brain Res. 182, 223–230.
Martel, K.L., Baum, M.J., 2009. A centrifugal pathway to the mouse accessory Morand-Ferron, J., Doligez, B., Dall, S.R.X., Reader, S.M., 2010. Social information
olfactory bulb from the medial amygdala conveys gender-specific volatile phero- use. Encycl. Anim. Behav. 3, 242–250.
monal signals. Eur. J. Neurosci. 29, 368–376. Monclús, R., Rödel, H.G., Von Holst, D., De Miguel, J., 2005. Behavioural and
Martin, I.G., Beauchamp, G.K., 1982. Olfactory recognition of individuals by male physiological responses of naïve European rabbits to predator odour. Anim. Behav.
cavies (Cavia aperea). J. Chem. Ecol. 8, 1241–1249. 70, 753–761.
Martin, J., Carranza, J., López, P., Alarcoa, S., Pedro-González, J., 2014. A new Moore, C.L., 1986. A hormonal basis for sex differences in the self-grooming of rats.
sexual signal in rutting male red deer: age related chemical scent constituents in Horm. Behav. 20, 155–165.
the belly black spot. Mammal. Biol. 79, 362–368. Moore, R.E., 1965. Olfactory discrimination as an isolating mechanism between
Martinez, L.A., Petrulis, A., 2011. The bed nucleus of the stria terminalis is critical for Peromyscus maniculatus and Peromyscus polionotus. Am. Midl. Nat. 73, 85–100.
sexual solicitation, but not for opposite-sex odor preference, in female Syrian Müller-Schwarze, D., 1971. Pheromones in black-tailed deer (Odocoileus hemionus
hamsters. Horm. Behav. 60, 651–659. columbianus). Anim. Behav. 19, 141–152.
324 Communication by Chemical Signals

Müller-Schwarze, D., 1984. Chemical communication in mammals: ecological context Perdue, B.M., Talbot, C.F., Stone, A., Beran, M.J., 2012. Putting the elephant back in
and future research. Acta Zool. Fenn. 171, 55–56. the herd: elephant relative quantity judgments match those of other species. Anim.
Murthy, V.N., 2011. Olfactory maps in the brain. Annu. Rev. Neurosci. 34, Cogn. 15, 955–961.
233–258. Pearl, C.A., Cervantes, M., Chan, M., Ho, U., Shoji, R., Thomas, E.O., 2000. Evidence
Musolf, K., Hoffmann, F., Penn, D.J., 2010. Ultrasonic courtship vocalizations in wild for a mate-attracting chemosignal in the dwarf African clawed frog Hymenochirus.
house mice, Mus musculus musculus. Anim. Behav. 79, 757–764. Horm. Behav. 38, 67–74.
Mykytowycz, R., 1965. Further observations on the territorial function and histology of Penn, D., Potts, W.K., 1998a. Chemical signals and parasite-mediated sexual
the submandibular cutaneous (chin) glands in the rabbit, Oryctolagus cuniculus selection. Trends Ecol. Evol. 13, 391–396.
(L.). Anim. Behav. 13, 400–412. Penn, D., Potts, W.K., 1998b. Untrained mice discriminate MHC-determined odors.
Mykytowycz, R., 1968. Territorial marking by rabbits. Sci. Am. 218, 116–126. Physiol. Behav. 64, 235–243.
Mykytowycz, R., 1970. The role of skin glands in mammalian communication. In: Penn, D., Schneider, G., White, K., Slev, P., Potts, W., 1998. Influenza infection
Johnston, J.W., Moulton, D.G., Turk, A. (Eds.), Advances in Chemoreception, neutralizes the attractiveness of male odour to female mice (Mus musculus).
Communication by Chemical Signals, vol. 1. Appleton-Century-Crofts, New York, Ethology 104, 685–694.
pp. 327–360. Penn, D.J., Potts, W.K., 1999. The evolution of mating preferences and major
Mykytowycz, R., 1973. Reproduction of mammals in relation to environment odours. histocompatibility complex genes. Am. Nat. 153, 145–164.
J. Reprod. Fertil. (Suppl. 19), 433–446. Penzhorn, B.J., 1984. A long-term study of social organization and behaviour of Cape
Nevison, C.M., Armstrong, S., Beynon, R.J., Humphries, R.E., Hurst, J.L., 2003. The Mountain zebras (Equs zebra zebra). Z. Tierspsychol. 64, 97–146.
ownership signature in mouse scent marks is involatile. Proc. R. Soc. Lond. B Biol. Peterson, S.C., 1988. Chemical trail following by caterpillars of Malacosoma neustria.
Sci. 270, 1957–1963. J. Chem. Ecol. 14, 815–824.
Nevo, E., Bodmer, M., Heth, G., 1976. Olfactory discrimination as an isolating Petrulis, A., 2013a. Chemosignals, hormones and mammalian reproduction. Horm.
mechanism in speciating mole rats. Experientia 32, 1511–1512. Behav. 63, 723–741.
Newman, S.W., 1999. The medial extended amygdala in male reproductive behavior. A Petrulis, A., 2013b. Chemosignals and hormones in the neural control of mammalian
node in the mammalian social behavior network. Ann. NY Acad. Sci. 877, sexual behavior. Front. Neuroendocrinol. 34, 255–267.
242–257. Petrulis, A., 2015. Introduction to the special issue on chemical signals and repro-
Norrdahl, K., Korpimäki, E., 1998. Does mobility or sex of voles affect risk of predation duction. Horm. Behav. 68, 1–2.
by mammalian predators? Ecology 79, 226–232. Petrulis, A., Desouza, I., Schiller, M., Johnston, R.E., 1998. Role of frontal cortex in
Nunes, S., 2007. Dispersal and philopatry. In: Wolff, J.O., Sherman, P.W. (Eds.), social odor discrimination and scent-marking in female golden hamsters (Meso-
Rodent Societies: An Ecological and Evolutionary Perspective. The University of cricetus auratus). Behav. Neurosci. 112, 199–212.
Chicago Press, Chicago, pp. 150–162. Petrulis, A., Johnston, R.E., 1997. Causes of scent marking in female golden hamsters
Nunez, A.A., Nyby, J., Whitney, G., 1978. The effects of testosterone, estradiol, and (Mesocricetus auratus): specific signals or classes of information? J. Comp. Psy-
dihydrotestosterone on male mouse (Mus musculus) ultrasonic vocalizations. chol. 111, 25–36.
Horm. Behav. 11, 264–272. Petrulis, A., Johnston, R.E., 1999. Lesions centered on the medial amygdala impair
Nyby, J., 1983a. Volatile and nonvolatile chemosignals of female rodents: differences scent-marking and sex-odor recognition but spare discrimination of individual odors
in hormonal regulation. In: Müller-Schwarze, D., Silverstein, R.M. (Eds.), Chemical in female golden hamsters. Behav. Neurosci. 113, 345–357.
Signals in Vertebrates, vol. 3. Plenum, New York, pp. 179–193. Petrulis, A., Peng, M., Johnston, R.E., 1999. Effects of vomeronasal organ removal on
Nyby, J., 1983b. Ultrasonic vocalizations during sex behavior of male house mice (Mus individual odor discrimination, sex-odor preference, and scent marking by female
musculus): a description. Behav. Neural Biol. 39, 128–134. hamsters. Physiol. Behav. 66, 73–83.
Nyby, J., Dizinno, G., Whitney, G., 1977. Sexual dimorphism in ultrasonic vocalizations Petrulis, A., Peng, M., Johnston, R.E., 2000. The role of the hippocampal system in
of mice (Mus musculus): gonadal hormone regulation. J. Comp. Physiol. Psychol. social odor discrimination and scent-marking in female golden hamsters (Meso-
91, 1424–1431. cricetus auratus). Behav. Neurosci. 114, 184–195.
Nyby, J., Matochik, J.A., Barfield, R.J., 1992. Intracranial androgenic and estrogenic Petrulis, A., Weidner, M., Johnston, R.E., 2004. Recognition of competitors by male
stimulation of male-typical behaviors in house mice (Mus domesticus). Horm. golden hamsters. Physiol. Behav. 81, 629–638.
Behav. 26, 24–45. Petty, J.M.A., Drea, C.M., 2015. Female rule in lemurs is ancestral and hormonally
O’Connell, L.A., Hofmann, H.A., 2012. Evolution of a vertebrate social decision-making mediated. Sci. Rep. 5, e9631.
network. Science 336, 1154–1157. Pfaff, D.W., Kow, L.M., Loose, M.D., Flanagan-Cato, L.M., 2008. Reverse engineering
Oliveira, E.S., Japyassú, H.F., Melange, J., Alberts, C.C., 2013. The evolution of the lordosis behavior circuit. Horm. Behav. 54, 347–354.
behavioural systems: a study of grooming in rodents. Behaviour 150, Pierce, A.A., Ferkin, M.H., Williams, T.K., 2005. Food-deprivation-induced changes in
1295–1324. sexual behaviour of meadow voles, Microtus pennsylvanicus. Anim. Behav. 70,
Orrock, J.L., Danielson, B.J., Brinkerhoff, R.J., 2004. Rodent foraging is affected by 339–348.
indirect, but not by direct, cues of predation risk. Behav. Ecol. 15, 433–437. Pierce, A.A., Iwueke, I., Ferkin, M.H., 2007. Food deprivation and the role of estradiol
Osada, K., Kashiwayanagi, M., Izumi, H., 2009. Profiles of volatiles in male rat urine: in mediating sexual behaviors in meadow voles. Physiol. Behav. 90, 353–361.
the effect of puberty on the female attraction. Chem. Senses 34, 713–721. Pierce Jr., J.D., Sawrey, D.K., Dewsbury, D.A., 1989. A comparative study of rodent
Owen, K., Thiessen, D.D., 1974. Estrogen and progesterone interaction in the regu- ultrasonic vocalizations during copulation. Behav. Neural Biol. 51, 211–221.
lation of scent marking in the female Mongolian gerbil (Meriones unguiculatus). Pierman, S., Sica, M., Allieri, F., Viglietti-Panzica, C., Panzica, G.C., Bakker, J., 2008.
Physiol. Behav. 12, 351–355. Activational effects of estradiol and dihydrotestosterone on social recognition and
Owen, M.A., Swasigood, R.R., Slocomb, A., Amstrup, S.C., Dumer, G.M., Simac, K., the arginine-vasopressin immunoreactive system in male mice lacking a functional
Pessier, A.P., 2015. An experimental investigation of chemical communication in aromatase gene. Horm. Behav. 54, 98–106.
the polar bear. J. Zool. 295, 36–43. Pierson, L.M., Ferkin, M.H., 2015. The impact of phytoestrogens on sexual behavior
Pahl, M., Si, A., Zhang, S., 2013. Numerical cognition in bees and other insects. Front. and cognition in rodents. Mammal. Biol. 80, 148–154.
Comp. Psychol. http://dx.doi.org/10.3389/fpsyg.2013.00162. Pillay, N., 2000. Female mate preference and reproductive isolation in populations of
Palagi, E., Telara, S., Borgognini-Tarli, S.M., 2004. Reproductive strategies in Lemur the striped mouse Rhabdomys pumilio. Behaviour 137, 1431–1441.
catta: balance among sending, receiving, and counter-marking scent signals. Int. J. Piñeiro, A., Barja, I., 2015. Evaluating the function of wildcat faecal marks in relation to
Primatol. 25, 1019–1031. the defence of favourable hunting areas. Ethol. Ecol. Evol. 27, 161–172.
Pankevich, D.E., Baum, M.J., Cherry, J.A., 2004. Olfactory sex discrimination persists, Pitcher, B.J., Harcourt, R.G., Schaal, B., Charrier, I., 2011. Social olfaction in marine
whereas the preference for urinary odorants from estrous females disappears in mammals: wild female Australian sea lions can identify their pup’s scent. Biol. Lett.
male mice after vomeronasal organ removal. J. Neurosci. 24, 9451–9457. 7, 60–62.
Parker, G.A., 1970. Sperm competition and its evolutionary consequences in insects. Pluhácek, J., Ceacero, F., Lupták, P., 2015. First records of preorbital gland opening in
Biol. Rev. 45, 525–567. rare wild barasingha (Rucervus duvaucelii). Behav. Process. 119, 28–31.
Parker, G.A., Pizzari, T., 2010. Sperm competition and ejaculate economics. Biol. Rev. Pochron, S.T., Morelli, T.L., Scirbona, J., Wright, P.C., 2005. Sex differences in scent
85, 897–934. marking in Propithecus edwardsi of Ranomafana National Park, Madagascar. Am.
Paz-y-Miño, G.C., Leonard, C.T., Ferkin, M.H., Trimble, J.F., 2002. Self-grooming and J. Primatol. 66, 97–110.
sibling recognition in meadow voles, Microtus pennsylvanicus, and prairie voles M, Pomerantz, S.M., Nunez, A.A., Bean, N.J., 1983. Female behavior is affected by male
ochrogaster. Anim. Behav. 63, 331–338. ultrasonic vocalizations in house mice. Physiol. Behav. 31, 91–96.
Communication by Chemical Signals 325

Porter, R.H., 1983. Communication in rodents: adults to infants. In: Elwood, R.W. (Ed.), Sabau, R.M., Pierson, L., Ferkin, M.H., 2014. Body mass and attractivity of
Parental Behaviour of Rodents. Wiley and Sons, New York, pp. 95–125. female offspring are negatively affected by food restriction of meadow vole
Porter, R.H., Moore, J.D., 1981. Human kin recognition by olfactory cues. Physiol. (Microtus pennsylvanicus) dams during lactation. Behaviour 151,
Behav. 27, 493–495. 1847–1868.
Potts, W.K., Wakeland, E.K., 1993. Evolution of MHC genetic diversity: a tale of incest, Sachs, B.D., 1999. Airborne aphrodisiac odor from estrous rats: implication for
pestilence and sexual preference. Trends Genetics 9, 408–412. pheromonal classification. In: Johnston, R.E., Müller-Schwarze, D., Sorensen, P.W.
Potts, J.R., Harris, S., Giuggoli, L., 2012. Territorial dynamics and stable home range (Eds.), Advances in Chemical Signals in Vertebrates. Kluwer Academic/Plenum
formation for central place foragers. PLoS One 7, e34033. Publishers, New York, pp. 333–342.
Preston-Taylor, A., Gunther, M.S., Allen, M.L., 2015. Black bear marking behaviour at Sayag, N., Hoffman, N.W., Yahr, P., 1994. Telencephalic connections of the sexually
rub trees during the breeding season in northern California. Behaviour 152, dimorphic area of the gerbil hypothalamus that influence male sexual behavior.
1097–1111. Behav. Neurosci. 108, 743–757.
Ralls, K., 1971. Mammalian scent marking. Science 171, 443–449. Scalia, F., Winans, S.S., 1975. The differential projections of the olfactory bulb and
Ramm, S.A., Stockley, P., 2009. Male house mice do not adjust sperm allocation accessory olfactory bulb in mammals. J. Comp. Neurol. 161, 31–55.
in response to odours from related or unrelated rivals. Anim. Behav. 78, Schaal, B., Coureaud, G., Langlois, D., Giniès, C., Sémon, E., Perrier, G., 2003.
685–690. Chemical and behavioural characterization of the rabbit mammary pheromone.
Randall, J.A., 1986. Preference for estrous female urine by male kangaroo rats Nature 424, 68–72.
(Dipodomys spectabilis). J. Mammal. 67, 736–739. Schellinck, H.M., Slotnick, B.M., Brown, R.E., 1997. Odors of individuality originating
Rasa, O.A., 1973. Marking behaviour and its social significance in the African dwarf from the major histocompatibility complex are masked by diet cues in the urine of
mongoose, Helogale undulate rufula. Z. Tierpsychol. 32, 293–318. rats. Anim. Learn. Behav. 25, 193–199.
Reznikova, Z., Ryabko, B., 2011. Numerical competence in animals, with an insight Schulte, B.A., Bagley, K.R., Castelda, S., Loziz, H., Nasseri, N., Vyas, D.K.,
from ants. Behaviour 148, 405–434. Goodwin, T.E., 2013. From exploration to selective information gathering: the
Rich, T.J., Hurst, J.L., 1999. The competing countermarks hypothesis: reliable development of chemosensory investigation in male African elephants (Loxodonta
assessment of competitive ability by potential mates. Anim. Behav. 58, afriana). In: East, M., Denhard, M. (Eds.), Chemical Signals in Vertebrates, vol. 12.
1027–1037. Springer Press, New York, pp. 135–145.
Roberts, S.A., Davidson, S.J., Beynon, R.J., Hurst, J.L., 2014. Female attraction to Schultze-Westrum, T.G., 1969. Social communication by chemical signals in flying
mate scent and associative learning: the house mouse as a mammalian model. phalangers (Petaurus breviceps papuanus). In: Pfaffmann, C. (Ed.), Olfaction and
Anim. Behav. 97, 313–321. Taste, Proceedings of the Third International Symposium. Rockefeller University
Roberts, S.A., Simpson, D.M., Armstrong, S.D., Davidson, A.J., Robertson, D.H., Press, New York, pp. 268–277.
McLean, L., Beynon, R.J., Hurst, J.L., 2010. Darcin: a male pheromone that Secondi, J., Haerty, W., Lodé, T., 2005. Female attraction to conspecific chemical
stimulates female memory and sexual attraction to an individual male’s odour. BMC cues in the palmate newt Triturus helveticus. Ethology 111, 726–735.
Biol. 8, 75. Secondi, J., Hinot, E., Djalout, Z., Sourice, S., Jadas-Hécart, A., 2009. Realistic nitrate
Roberts, S.C., 2007. Scent marking. In: Wolff, J.O., Sherman, P.W. (Eds.), Rodent concentration alters the expression of sexual traits and olfactory male attractive-
Societies: An Ecological and Evolutionary Perspective. The University of Chicago ness in newts. Funct. Ecol. 23, 800–808.
Press, Chicago, pp. 255–266. Segovia, S., Guillamon, A., 1993. Sexual dimorphism in the vomeronasal pathway and
Roberts, S.C., Dunbar, R.I.M., 2000. Female territoriality and the function of scent sex differences in reproductive behaviors. Brain Res. Rev. 18, 51–74.
marking in a monogamous antelope (Oreotagus oreotagus). Behav. Ecol. Sociobiol. Setchell, J.M., Vaglio, S., Abbott, K.M., Moggi-Cecchi, J., Boscaro, F., Pieraccini, G.,
47, 417–423. Knapp, L.A., 2011. Odour signals major histocompatibility complex genotype in an
Roberts, S.C., Gosling, L.M., Thornton, E.A., McClung, J., 2001. Scent-marking by Old World monkey. Proc. R. Soc. Lond. B Biol. Sci. 278, 274–280.
male mice under the risk of predation. Behav. Ecol. 12, 698–705. Shanas, U., Terkel, J., 1995. Grooming expresses Harderian gland materials in the
Rodgers, T.W., Giacalone, J., Heske, E.J., Pawlikowski, N.C., Schooley, R.L., 2015. blind mole rat. Aggr. Behav. 21, 137–146.
Communal latrines act as potentially important communication centers in ocelots, Sharpe, L.L., 2015. Handstand scent marking: height matters to dwarf mongooses.
Leopardus pardalis. Mammal. Biol. 80, 380–384. Anim. Behav. 105, 173–179.
Romeyer, A., Poindron, P., Orgeur, P., 1994. Olfaction mediates the establishment of Sherborne, A.L., Thom, M.D., Paterson, S., Jury, F., Ollier, W.E.R., Stockley, P.,
selective bonding in goats. Physiol. Behav. 56, 693–700. Beynon, R.J., Hurst, J.L., 2007. The genetic basis of inbreeding avoidance in
Ronkainen, H., Ylönen, H., 1994. Behaviour of cyclic bank voles under risk of mustelid house mice. Curr. Biol. 17, 2061–2066.
predation: do females avoid copulations? Oecologia 97, 377–381. Signoret, J.P., 1976. Chemical communication and reproduction in domestic
Rose, E., Drickamer, L.C., 1975. Castration, sexual experience, and female urine odor mammals. In: Doty, R.L. (Ed.), Mammalian Olfaction, Reproductive Processes and
preferences in adult BDF 1 male mice. Bull. Psych. Soc. 5, 84–86. Behavior. Academic Press, New York, pp. 243–256.
Rosell, F., Bjorkoyli, T., 2002. A test of the dear enemy phenomenon in the Eurasian Sillero-Zubiri, C., Macdonald, D.W., 1998. Scent-marking and territorial behaviour of
beaver. Anim. Behav. 63, 1073–1078. Ethiopian wolves, Canis simensis. J. Zool. 245, 351–361.
Rosell, F., Sanda, J., 2006. Potential risks of olfactory signaling: the effect of predators Simeonovska-Nikolova, D., Dekov, O., 2013. Some aspects of the behavior and
on scent marking by beavers. Behav. Ecol. 17, 897–904. defensive vocalization of the Romanian hamster, Mesocricetus newtoni. Acta Zool.
Rosell, F., Johnson, G., Parker, H., 2000. Eurasian beavers (Castor fiber) behavioral Bulg. 65, 462–468.
response to simulated territorial intruders. Can. J. Zool. 78, 931–935. Simerly, R.B., 1990. Hormonal control of neuropeptide gene expression in sexually
Rozenfeld, F.M., LeBoulenge, E., Rasmont, R., 1987. Urine marking by male bank dimorphic olfactory pathways. Trends Neurosci. 13, 104–110.
voles Clethrionomys glareolus Schreber, 1780; Microtidae, Rodentia) in relation to Singer, A., Macrides, F., Clancy, A.N., Agosta, W.C., 1986. Purification and analysis of
social rank. Can. J. Zool. 65, 2549–2601. a proteinaceous aphrodisiac pheromone from hamster vaginal discharge. J. Biol.
Rozhnov, V.V., 2004. Mediated communication by scent mark in mammals: on Chem. 261, 13323–13326.
changes in the paradigm and a new approach to studies of marking behavior. Zool. Sipos, M.L., Nyby, J.G., 1996. Concurrent androgenic stimulation of the ventral
Z. 83, 132–158. tegmental area and medial preoptic area: synergistic effects on male-typical
Runemark, A., Gabirot, M., Svensson, E.I., 2011. Population divergence in chemical reproductive behaviors in house mice. Brain Res. 729, 29–44.
signals and the potential for premating isolation between islet- and mainland Sipos, M.L., Nyby, J.G., 1998. Intracranial androgenic activation of male-typical
populations of the Skyros wall lizard (Podarcis gaigeae). J. Evol. Biol. 24, behaviours in house mice: concurrent stimulation of the medial preoptic area
795–809. and medial nucleus of the amygdala. J. Neuroendocrinol. 10, 577–586.
Sabau, R.M., Ferkin, M.H., 2013a. Food deprivation and restriction during late Sipos, M.L., Wysocki, C.J., Nyby, J.G., Wysocki, L., Nemura, T.A., 1995. An
gestation affect the sexual behavior of postpartum female meadow voles, Microtus ephemeral pheromone of female house mice: perception via the main and
pennsylvanicus. Ethology 119, 29–38. accessory olfactory systems. Physiol. Behav. 58, 529–534.
Sabau, R.M., Ferkin, M.H., 2013b. Food restriction affects the maternal behavior Sliwa, A., Richardson, P.R.K., 1998. Responses of aardwolves, Proteles cristatus,
provided by female meadow voles (Microtus pennsylvanicus). J. Mammal. 94, Sparrman 1783, to translocated scent marks. Anim. Behav. 56, 137–146.
1068–1076. Smadja, C., Butlin, R.K., 2009. On the scent of speciation: the chemosensory system
Sabau, R.M., Ferkin, M.H., 2014. Maternal food restriction during lactation affects and its role in premating isolation. Heredity 102, 77–97.
body weight and sexual behavior of male offspring in meadow voles (Microtus Smith, T., 2006. Individual olfactory signatures in common marmosets (Callithrix
pennsylvanicus). Ethology 120, 793–803. jacchus). Am. J. Primatol. 68, 585–604.
326 Communication by Chemical Signals

Smith, T.E., Gordon, S.J., 2002. Sex differences in olfactory communication in Tierney, K.B., Baldwin, D.H., Hara, T.J., Ross, P.S., Scholz, N.L., Kennedy, C.J., 2010.
Saguinus labiatus. Int. J. Primatol. 23, 429–441. Olfactory toxicity in fishes. Aquat. Toxicol. 96, 2–26.
Soulsbury, C.D., Fawcett, J.K., 2015. Ontogenic patterns of scent marking in red Tinnesand, H.V., Jojola, S., Zedrosser, A., Rosell, F., 2013. The smell of desperadoes?
foxes, Vulpes vulpes (Carnivora: Canidae). Folia Zool. 94, 40–44. Beavers distinguish between dominant and subordinate intruders. Behav. Ecol.
Steiger, S., Schmitt, T., Schaefer, H.M., 2011. The origin and dynamic evolution of Sociobiol. 67, 895–904.
chemical information transfer. Proc. R. Soc. Lond. B Biol. Sci. 278, 970–979. Tinnesand, H.V., Buesching, C.D., Noonan, M.J., Newman, C., Zedrosser, A.,
Steiner, A.L., 1973. Self- and allo-grooming behavior in some ground squirrels Rosell, F., Macdonald, D.W., 2015. Will trespassers be prosecuted or
(Sciuridae), a descriptive study. Can. J. Zool. 51, 151–161. assessed according to their merits? A consilient interpretation of territoriality
Steiner, A.L., 1974. Body-rubbing, marking, and other scent-related behavior in some in a group-living carnivore, the European badger (Meles meles). PLoS One
ground squirrels (Sciuridae). Can. J. Zool. 52, 889–906. e0132432.
Stensmyr, M., Maderspacher, F., 2013. Olfactory evolution: mice rethink stink. Curr. Todrank, J., Heth, G., 2003. Odor-genes covariance and genetic relatedness
Biol. 23, R59–R61. assessments: rethinking odor-based “recognition” mechanisms in rodents. Adv.
Stockley, P., Bottell, L., Hurst, J.L., 2013. Wake up and smell the conflict: odour signals Study Behav. 32, 77–130.
in female competition. Philos. Trans. R. Soc. Lond. B Biol. Sci. 368, 20130082. Todrank, J., Heth, G., Johnston, R.E., 1998. Kin recognition in golden hamsters:
Stockley, P., Preston, B.T., 2004. Sperm competition and diversity in rodent copulatory evidence for kinship odors. Anim. Behav. 55, 377–386.
behaviour. J. Evol. Biol. 17, 1048–1057. Tuomainen, U., Candolin, U., 2011. Behavioural responses to human-induced envi-
Stopková, R., Zdráhal, Z., Ryba, S., Sedo, O., et al., 2010. Novel OBP genes similar to ronmental change. Biol. Rev. 86, 640–657.
hamster aphrodisin in the bank vole, Myodes glareolus. BMC Genomics 11, 45. Ulibarri, C., Yahr, P., 1988. Role of neonatal androgens in sexual differentiation of brain
Stowers, L., Holy, T.E., Meister, M., Dulac, C., Koentges, G., 2002. Loss of sex structure, scent marking, and gonadotropin secretion in gerbils. Behav. Neural Biol.
discrimination and male-male aggression in mice deficient for TRP2. Science 295, 49, 27–44.
1493–1500. Ulibarri, C., Yahr, P., 1996. Effects of androgens and estrogens on sexual differen-
Sugimoto, H., Okabe, S., Kato, M., Koshida, N., Shiroishi, T., Mogi, K., Kikusui, T., tiation of sex behavior, scent marking, and the sexually dimorphic area of the gerbil
Koide, T., 2011. A role for strain differences in waveforms of ultrasonic vocali- hypothalamus. Horm. Behav. 30, 107–130.
zations during male–female interaction. PLoS One 6, e22093. Unger, E.K., Burke, J., Kenneth, J., Yang, C.F., Bender, K.J., Fuller, P.M., Shah, N.M.,
Takahashi, L.K., Gladstone, C.D., 1988. Medial amygdaloid lesions and the regulation 2015. Medial amygdalar aromatase neurons regulate aggression in both sexes.
of sociosexual behavioral patterns across the estrous cycle in female golden Cell Rep. 10, 453–462.
hamsters. Behav. Neurosci. 102, 268–275. Vagell, M.E., McGinnis, M.Y., 1998. The role of gonadal steroid receptor activation in
Takahashi, L.K., Lisk, R.D., Burnett, A.L., 1985. Dual estradiol action in diencephalon the restoration of sociosexual behavior in adult male rats. Horm. Behav. 33,
and the regulation of sociosexual behavior in female golden hamsters. Brain Res. 163–179.
359, 194–207. Valone, T.J., 2007. From eavesdropping on performance to copying the behavior of
Taylor, S.A., Dewsbury, D.A., 1990. Male preferences for females of different others: a review of public information use. Behav. Ecol. Sociobiol. 62, 1–14.
reproductive conditions: a critical review. In: Macdonald, D.W., Müller- Vaughn, A.A., Ferkin, M.H., 2011. The presence and number of male competitor’s
Schwarze, D., Natynczuk, S.E. (Eds.), Chemical Signals in Vertebrates, vol. 5. scent marks and female reproductive state affects the response of male meadow
Oxford University Press, Oxford, pp. 184–198. voles to female conspecifics’ odours. Behaviour 148, 927–943.
Theis, K.R., Heckla, A.L., Verge, J.R., Holekamp, K.E., 2008. The ontogeny of pasting Vaughn, A.A., delBarco-Trillo, J., Ferkin, M.H., 2008. Sperm investment in male
behavior in free-living spotted hyenas, Crocuta crocuta. In: Hurst, J.L., meadow voles is affected by the condition of the nearby male conspecifics. Behav.
Beynon, R.J., Roberts, S.C., Wyatt, T.D. (Eds.), Chemical Signals in Vertebrates, Ecol. 19, 1159–1164.
vol. 11. Springer Press, New York, pp. 179–187. Vaughn, A.A., delBarco-Trillo, J., Ferkin, M.H., 2010. Self-grooming by male meadow
Thiessen, D.D., 1977. Thermoenergetics and the evolution of pheromone communi- voles differs across copulation but is not affected by the risk and intensity of sperm
cation. In: Sprague, J.M., Epstein, A.N. (Eds.), Progress in Psychobiology and competition. Behaviour 147, 259–274.
Physiological Psychology, vol. 7. Academic Press, New York, pp. 91–191. Viviers, M.Z., Burger, B.V., le Roux, N.J., Morris, J., le Roux, M., 2014. Temporal
Thiessen, D.D., Harriman, A.E., 1986. Harderian gland exudates in the male Meriones changes in the neonatal recognition cue of Dohne Merino lambs (Ovis aries).
unguiculatus regulate female proceptive behavior, aggression, and investigation. Chem. Senses 39, 249–262.
J. Comp. Psychol. 100, 85–87. Vlautin, C.T., Ferkin, M.H., 2012. The influence of predator and conspecific odor on
Thiessen, D.D., Rice, M., 1976. Mammalian scent gland marking and social behavior. sex differences in path choice in meadow voles. Behaviour 149, 133–152.
Psychol. Bull. 83, 505–539. Vlautin, C.T., Ferkin, M.H., 2014. Female meadow voles, Microtus pennsylvanicus, do
Thiessen, D.D., Yahr, P., 1970. Central control of territorial marking in the Mongolian not later their over-marking in response to female conspecifics that were food
gerbil. Physiol. Behav. 5, 275–278. deprived. Acta Ethol. 17, 69–75.
Thiessen, D.D., Owen, K., Lindzey, G., 1971. Mechanisms of terrritorial marking in the Vlautin, C.T., Hobbs, N.J., Ferkin, M.H., 2010. Male and female meadow voles,
male and female Mongolian gerbil (Meriones unguiculatus). J. Comp. Physiol. Microtus pennsylvanicus, differ in their responses to heterospecific/conspecific
Psychol. 77, 38–47. over-marks. Ethology 116, 797–895.
Thiessen, D.D., Blum, S.L., Lindzey, G., 1970. A scent marking response associated Vogt, K., Zimmermann, F., Kolliker, M., Breitenmoser, U., 2014. Scent-marking
with the ventral sebaceous gland of the Mongolian gerbil (Meriones unguiculatus). behaviour and social dynamics in a wild population of Eurasian lynx. Behav.
Anim. Behav. 18, 26–30. Process. 106, 98–106.
Thiessen, D.D., Friend, H.C., Lindzey, G., 1968. Androgen control of territorial marking Vyas, A., Kim, S.-K., Giacomini, N., Boothroyd, J.C., Sapolsky, R.M., 2007. Behavioral
in the Mongolian gerbil. Science 160, 432–434. changes induced by Toxoplasma infection of rodents are highly specific to aversion
Thiessen, D.D., Yahr, P., Owen, K., 1973. Regulatory mechanisms of territorial of cat odors. Proc. Natl Acad. Sci. USA 104, 6442–6447.
marking in the Mongolian gerbil. J. Comp. Physiol. Psychol. 82, 382–393. Wachowiak, M., 2011. All in a sniff: olfaction as a model for active sensing. Neuron
Tinbergen, N., 1963. On aims and methods of ethology. Z. Tierpsychol. 20, 410–433. 71, 962–973.
Thom, M.D., Hurst, J.L., 2004. Individual recognition by scent. Ann. Zool. Fenn. 41, Wade, G.N., Schneider, J.E., Li, H.-Y., 1996. Control of fertility by metabolic cues. Am.
765–787. J. Physiol. 270, E1–E19.
Thomas, M.L., 2011. Detection of female mating status using chemical signals and Wagner, R.H., Danchin, E., 2003. Conspecific copying: a general mechanism of social
cues. Biol. Rev. 86, 1–13. aggregation. Anim. Behav. 65, 405–408.
Thomas, S.A., Wolff, J.O., 2002. Scent marking in voles: a reassessment of over Wallace, P., Owen, K., Thiessen, D.D., 1973. The control and function of maternal
marking, counter marking, and self-advertisement. Ethology 108, 51–62. scent marking in the Mongolian gerbil. Physiol. Behav. 10, 463–466.
Thonhauser, K.E., Raveh, S., Hettyey, A., Beissmann, H., Penn, D.J., 2013a. Why do Walls, S.C., Mathis, A., Jaeger, R.G., Gergits, W.F., 1989. Male salamanders with
male mice mate with multiple mates? Behav. Ecol. Sociobiol. 67, 1961–1970. high-quality diets have faeces attractive to females. Anim. Behav. 38, 546–548.
Thonhauser, K.E., Raveh, S., Hettyey, A., Beissmann, H., Penn, D.J., 2013b. Scent Wang, H., Liang, S., Burgdorf, J., Wess, J., Yeomans, J., 2008. Ultrasonic vocali-
marking increases male reproductive success in wild house mice. Anim. Behav. zations induced by sex and amphetamine in M2, M4, M5 muscarinic and D2
86, 1013–1021. dopamine receptor knockout mice. PLoS One 3, e1893.
Thoß, M., Luzynski, K., Ante, M., Miller, I., Penn, D.J., 2015. Major urinary protein Warburton, V.L., Sales, G.D., Milligan, S.R., 1989. The emission and elicitation of
(MUP) profiles show dynamic changes rather than individual “barcode” signatures. mouse ultrasonic vocalizations: the effects of age, sex and gonadal status. Physiol.
Front. Ecol. Evol. 3, 71. Behav. 45, 41–47.
Communication by Chemical Signals 327

Wedekind, C., Seebeck, T., Bettens, F., Paepke, A.J., 1995. MHC-dependent mate Wysocki, C.J., Nyby, J., Whitney, G., Beauchamp, G.K., Katz, Y., 1982. The vomer-
preferences in humans. Proc. R. Soc. Lond. B Biol. Sci. 260, 245–249. onasal organ: primary role in mouse chemosensory gender recognition. Physiol.
Wedell, N., Gage, M.J.G., Parker, G.A., 2002. Sperm competition, male prudence and Behav. 29, 315–327.
sperm-limited females. Trends Ecol. Evol. 17, 313–320. Yamazaki, K., Beauchamp, G.K., Curran, M., Boyse, E.A., 2000. Parent-progeny
White, N.R., Colona, L.C., Barfield, R.J., 1991. Sensory cues that elicit ultrasonic recognition as a function of MHC odor type identity. Proc.Natl Acad. Sci. USA
vocalizations in female rats (Rattus norvegicus). Behav. Neural Biol. 55, 154–165. 97, 10500–10502.
White, N.R., Prasad, M., Barfield, R.J., Nyby, J.G., 1998. 40- and 70-kHz vocalizations Yamazaki, K., Yamaguchi, M., Boyse, E.A., Thomas, L., 1980. The major histocom-
of mice (Mus musculus) during copulation. Physiol. Behav. 63, 467–473. patibility complex as a source of odors imparting individuality among mice. In:
Wiepkema, P.R., 1979. The social significance of self-grooming in rats. Netherlands J. Müller-Schwarze, D., Silverstein, R.M. (Eds.), Chemical Signals. Vertebrates and
Zool. 29, 622–623. Aquatic Invertebrates. Plenum, New York, pp. 267–273.
Willis, C., Poulin, R., 2000. Preference of female rats for the odours of non-parasitised Yahr, P., 1977. Social subordination and scent-marking in male Mongolian gerbils
males: the smell of good genes? Folia Parasitol. 47, 6–10. (Meriones unguiculatus). Anim. Behav. 25, 292–297.
Witt, D.M., Carter, C.S., Carlstead, K., Read, L.D., 1988. Sexual and social interaction Yahr, P., Gregory, J.E., 1993. The medial and lateral cell groups of the sexually
preceding and during male-induced oestrous in prairie voles, Microtus ochrogaster. dimorphic area of the gerbil hypothalamus are essential for male sex behavior and
Anim. Behav. 36, 1465–1471. act via separate pathways. Brain Res. 631, 287–296.
Witt, D.M., Carter, C.S., Chayer, R., Adams, K., 1990. Patterns of behavior during Yahr, P., Newman, A., Stephens, D.R., 1979. Sexual behavior and scent marking in
postpartum oestrus in prairie voles, Microtus ochrogaster. Anim. Behav. 39, male gerbils: comparison of changes after castration and testosterone replace-
528–534. ment. Horm. Behav. 13, 175–184.
Witte, K., Ryan, M.J., 2002. Mate-choice copying in the sailfin molly (Poecilia lat- Yahr, P., Stephens, D.R., 1987. Hormonal control of sexual and scent marking
ipinna). Anim. Behav. 63, 943–949. behaviors of male gerbils in relation to the sexually dimorphic area of the hypo-
Whitsett, J.M., Thiessen, D.D., 1972. Sex difference in the control of scent-marking thalamus. Horm. Behav. 21, 331–346.
behavior in the Mongolian gerbil (Meriones unguiculatus). J. Comp. Physiol. Yahr, P., Thiessen, D.D., 1975. Estrogen control of scent marking female Mongolian
Psychol. 78, 381–385. gerbils (Meriones unguiculatus). Behav. Biol. 13, 95–101.
Wintink, A.J., Brudzynski, S.M., 2001. The related roles of dopamine and glutamate in Yahr, P., Commins, D., Jackson, J.C., Newman, A., 1982. Independent control of
the initiation of 50-kHz ultrasonic calls in adult rats. Pharmacol. Biochem. Behav. sexual and scent marking behaviors of male gerbils by cells in or near the medial
70, 317–323. preoptic area. Horm. Behav. 16, 304–322.
Wolff, J.O., 2004a. Scent marking by voles in response to predation risk: a field- Young, L.J., Wang, Z., Cooper, T.T., Albers, H.E., 2000. Vasopressin (V1a) receptor
laboratory validation. Behav. Ecol. 15, 286–289. binding, mRNA expression and transcriptional regulation by androgen in the Syrian
Wolff, J.O., 2004b. Laboratory studies with rodents: facts of artifacts. Bioscience 53, hamster brain. J. Neuroendocrinol. 12, 1179–1185.
421–427. Yu, H., Zhao, X., Yue, P., Sun, P., 2010. Chemical communication in mammal pop-
Wood, R.I., 1997. Thinking about networks in the control of male hamster sexual ulations: urinary olfactory chemosignals in lactating female root voles (Microtus
behavior. Horm. Behav. 32, 40–45. oeconomus pallas). Pol. J. Ecol. 58, 153–165.
Woodward, R.L., Schmick, M.K., Ferkin, M.H., 1999. Response of prairie voles, Zala, S.M., Potts, W.K., Penn, D.J., 2004. Scent-marking displays provide honest
Microtus ochrogaster (Rodentia, Arvicolidae), to scent over-marks of two same- signals of health and infection. Behav. Ecol. 15, 338–344.
sex conspecifics: a test of the scent masking hypothesis. Ethology 105, Zeigler, T.E., Epple, G., Snowdon, C.T., Porter, T.A., Belcher, A.M., Kuderling, I., 1993.
1009–1017. Detection of the chemical signals of ovulation in the cotton-top tamarin, Saginus
Woodward, R.L., Bartos, K., Ferkin, M.H., 2000. Meadow voles (Microtus pennsyl- oedipus. Anim. Behav. 45, 313–322.
vanicus) and prairie voles (M. ochrogaster) differ in their responses to over-marks Zeller, A.C., 1987. Communication by sight and smell. In: Smuts, B.B., Cheney, D.L.,
from opposite- and same-sex conspecifics. Ethology 106, 979–992. Seyfarth, R.M., Wrangham, R.W., Struhsaker, T.T. (Eds.), Primate Societies. The
Wu, M.V., Manoli, D.S., Fraser, E.J., Coats, J.K., Tollkuhn, J., Honda, S.-I., Harada, N., University of Chicago Press, Chicago, pp. 433–439.
Shah, N.M., 2009. Estrogen masculinizes neural pathways and sex-specific Zhou, Y., Chen, W., Buesching, C.D., Newman, C., Kaneko, Y., Xiang, M., Nie, C.,
behaviors. Cell 139, 61–72. Macdonald, D.W., Xie, Z., 2015. Hog badger (Arctonyx collaris) latrine use in
Wuensch, K.L., 1979. Adrenal hypertrophy in mice following exposure to crowded relation to food abundance: evidence of the scarce factor paradox. Ecosphere
males’ odors. Behav. Neural Biol. 27, 222–226. 6, e19.
Wyatt, T.D., 2014. Pheromones and Animal Behavior: Chemical Signals and Signa- Zufall, F., Leinders-Zufall, T., 2007. Mammalian pheromone sensing. Curr. Opin.
tures. Cambridge University Press. Neurobiol. 17, 483–489.
This page intentionally left blank
1.11 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network,
and Mating Behavior
Aras Petrulis, Georgia State University, Atlanta, GA, USA
Jeannie M Fiber, Contract Scientist, Cambridge, MA, USA
Jennifer M Swann, Lehigh University, Bethlehem, PA, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by J. Swann, C. Fabre-Nys, R. Barton, volume 1, pp. 441–474, Ó 2009, Elsevier Inc.

1.11.1 Introduction 329


1.11.2 Pheromones in Mammalian Reproduction: The Rodent Model 330
1.11.2.1 Impact of Mating-Related Pheromones on Males 330
1.11.2.2 Impact of Mating-Related Pheromones on Females 330
1.11.3 The Impact of Mating-Related Hormones on Social Behaviors 331
1.11.3.1 The Impact of Gonadal Hormones on Mating in Males 331
1.11.3.2 The Impact of Gonadal Hormones on Mating in Females 331
1.11.4 The Social Behavior Network 331
1.11.5 MA Subdivisions 333
1.11.6 Comparative Anatomy 334
1.11.7 Sex Differences 334
1.11.8 Role of the MA and Connected Structures in Social Odor Investigation and Sexual Behavior in Males 334
1.11.8.1 MA Connections with BST 335
1.11.8.2 MA Connections with MPOA 336
1.11.8.3 MA Connections with VMH 336
1.11.9 The Role of the MA in Females 336
1.11.9.1 MA Connections with BST and MPOA 336
1.11.9.2 MA Connections with VMH 337
1.11.10 Steroids May Act Centrally and Peripherally to Impact Responses to Pheromones 337
1.11.11 Future Directions 337
References 338

Abbreviations
AH Anterior hypothalamus MApv Posteroventral subdivision of the medial nucleus of
AOB Accessory olfactory bulb the amygdala
AR Androgen receptor MOB Main olfactory bulb
AVP Arginine-vasopressin MPOA Medial preoptic area
BST Bed nucleus of the stria terminalis OBX Olfactory bulbectomy
DHT Dihydrotestosterone OT Oxytocin
E Estrogen SBN Social behavior network
ER Estrogen receptor USV Ultrasonic vocalizations
IEG Immediate-early gene T Testosterone
MA Medial nucleus of the amygdala VMH Ventromedial hypothalamus
MAa Anterior subdivision of the medial nucleus of the VMHdm Dorsomedial subdivision of the ventromedial
amygdala hypothalamus
MApd Posterodorsal subdivision of the medial nucleus of VMHvl Ventrolateral subdivision of the ventromedial
the amygdala hypothalamus

1.11.1 Introduction pheromones coordinate between conspecifics by signaling their


location and internal state. In many vertebrates, these signals
Sexual reproduction is a complicated business requiring coor- come together to act on a network of interconnected neural
dination within and between individuals. Hormones and pher- groups to affect appropriate social behavior. At the heart of
omones play key roles: hormones coordinate within the this circuit is the medial amygdala (MA), an almond-shaped
organism by aligning the activity of organs and cells, while nucleus in the temporal lobe that serves as a gatekeeper,

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00011-0 329


330 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior

integrating hormonal and pheromonal signals and facilitating aspects of odor cues, to orient toward mating partners (Ferkin
social behavior. The MA is a bellwether for the individual’s et al., 2008; Johnston, 2008; Pankevich et al., 2006), and will
internal state as its morphology and function are modified readily approach and investigate female odors as part of their
and supported by gender and hormonal milieu. The MA is mate-location strategy (Beauchamp and Beruter, 1973; Brown,
but a few synapses away from the external environment, 1978; Johnston, 1974). Upon detecting female odors, males of
receiving direct and indirect projections from the main and several rodent species, including rats (Geyer and Barfield,
accessory olfactory bulbs. Hormonal sensitivity and sex differ- 1978), mice (Nyby et al., 1977), hamsters (Johnston and
ences in structure, neurochemistry and perhaps connectivity, Kwan, 1984), and guinea pigs (Eisthen et al., 1987), will
impact the perception of pheromonal cues that regulate emit ultrasonic vocalizations (USVs), which appear to facilitate
behavior. While this ensures appropriate reproductive behav- the location of the female (Pfaff et al., 2008). These responses
iors, it also has larger implications for how (sometimes are eliminated by olfactory bulbectomy (OBX) (Bean, 1982;
competing) social cues are filtered and perceived by males Eisthen et al., 1987; Wysocki et al., 1982).
and females and by individuals, to achieve specific, appropriate Once a male has located a female, he will usually engage in
behavioral outcomes. Thus, the MA, as a hub in several neural substantial amounts of chemoinvestigation targeting the ano-
networks involved in social behaviors, plays a critical role in genital region of the female, prior to mounting (Dewsbury,
a wide variety of social interactions including aggression, kin 1975). OBX eliminates AGI and subsequent mounting,
recognition, parenting, and copulation. Our review explores and therefore copulation, in Syrian hamsters (Murphy and
the complexity of that role in mating. It concludes that what Schneider, 1970) and mice (Rowe and Edwards, 1972), and
we have learned over the past 40 years lays a solid foundation it impairs AGI/copulation in rats and guinea pigs (Beauchamp
for new questions to be addressed with new technologies et al., 1977; Edwards et al., 1990, 1996). In some species, such
that further our understanding of pheromones and mating as rats, robust investigation of female odors depends on socio-
behavior and more broadly, neural networks and behavioral sexual experience with the opposite sex, and exposure to vola-
outcomes. This chapter represents a new version of a previously tile chemosignals from female urine/feces alone can elicit
published review on this topic (Swann et al., 2009). noncontact erections (NCEs) (Kondo et al., 1999; Sachs et al.,
1994). In contrast, both sexually naïve and sexually experi-
enced male Syrian hamsters are strongly attracted to female
1.11.2 Pheromones in Mammalian Reproduction: odors (reviewed in Petrulis, 2013), and nonvolatile compo-
The Rodent Model nents of female vaginal secretion (O’Connell and Meredith,
1984; Singer et al., 1984) are sufficient, but not necessary
Although chemical communication is involved in all social (Johnston, 1986), for eliciting mounting behavior (Johnston,
contexts, it is most evident in reproduction. Chemosignals 1975; Murphy, 1973).
impact reproductive development and physiology and, impor-
tantly, drive the appetitive components of reproductive behav-
1.11.2.2 Impact of Mating-Related Pheromones on Females
iors such as investigation and approach toward opposite-sex
conspecifics, precopulatory behaviors that advertise the pres- Female Syrian hamsters (Johnston, 1979), rats (Carr et al.,
ence/absence of appropriate partners, as well as behaviors 1965), mice (Hurst, 1990), meadow voles (Ferkin and Zucker,
such as anogenital investigation (AGI) that initiate copulation 1991), and ferrets (Kelliher and Baum, 2002) are preferentially
(Ball and Balthazart, 2008; Petrulis, 2013). However, phero- attracted to chemical cues of adult male conspecifics. Moreover,
mones are generally less important for the maintenance of females scent-mark to advertise their current or impending
sexual behavior once copulation has commenced. For example, receptivity in mice (Rich and Hurst, 1999), Syrian hamsters
the display of lordosis in rats and hamsters is primarily stimu- (Johnston, 1977), rabbits (Gonzalez-Mariscal et al., 1990),
lated by somatosensory input (reviewed by Pfaff, 1980), and ferrets (Chang et al., 2000), and rats (Birke, 1984). For
the role of chemosignals is limited to reinitiation of interest example, female Syrian hamsters engage in vaginal marking,
in the male (Rajendren et al., 1990). Removal of the olfactory a stereotyped behavior that places a vaginal fluid on the
bulbs of female rats or hamsters does not inhibit and may substrate (Been and Petrulis, 2008, 2012; Been et al., 2012)
even enhance (Williams et al., 1992) the expression of lordosis and is specifically targeted toward the location of male odors
(Carter, 1973; Kelche and Aron, 1984). This, however, does not (Johnston and Brenner, 1982; Johnston, 1977; Petrulis and
occur in mice (Keller et al., 2006a,b). In addition, in male Johnston, 1997). Vaginal marking increases substantially on
hamsters (a species that requires chemosensory input for the night before behavioral receptivity but is absent during
expression of male-typical copulatory behavior (Murphy and sexual receptivity, consistent with its role in attracting dispersed
Schneider, 1970)), blocking chemosignal processing during males for mating (Gattermann et al., 2001; Johnston, 1977).
copulation has no effect on sexual behavior, whereas blocking Similarly, estrous female rats (White et al., 1991) and hamsters
chemosensory input before copulation or during long intervals (Floody et al., 1977) also produce USVs in response to males or
between copulatory bouts eliminates male sexual behavior their odors, a behavior that aids in short-range localization of
(Devor and Murphy, 1973). mates (Pfaff et al., 2008). Both types of precopulatory behavior,
marking and USVs, critically require chemosensory input as
OBX female hamsters strongly reduce both their USVs and
1.11.2.1 Impact of Mating-Related Pheromones on Males
vaginal marking toward males (Kairys et al., 1980).
Males may use mnemonic (anticipated location of females), Even though chemosignals are not required for the expres-
temporal (freshness), and spatial (location, arrangement) sion of the sexually receptive posture (lordosis) in some
The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 331

mammalian species (Pfaff, 1980), chemosignals can facilitate formed in the brain and peripheral tissues (Degtiar et al.,
lordosis, e.g., in pigs (Melrose et al., 1971; Reed et al., 1974), 1981). Moreover T has been shown to upregulate the produc-
rats (Rajendren and Moss, 1994), musk shrews (Rissman, tion of both enzymes in the mammalian brain (Wagner
1989), and prairie voles (Curtis et al., 2001). In contrast, recep- and Morrell, 1997; Aste et al., 1998), suggesting a role for
tivity is much more dependent of chemosignals in female mice DHT and E in the regulation of mating by T.
(Haga et al., 2010; Keller et al., 2006a,b) although it is not clear Neither hormone alone is sufficient for the expression of
if this chemosensory requirement is limited to the initiation mating; simultaneous treatment with both steroids restores the
of sex behavior, or the maintenance of the receptive posture. full expression of male sexual behavior in castrated rats (Baum
and Vreeburg, 1973; Larsson et al., 1976, 1973b,a; Beyer et al.,
1973) and rabbits (Beyer et al., 1975; Beyer and Rivaud, 1973).
1.11.3 The Impact of Mating-Related Hormones on
Social Behaviors
1.11.3.2 The Impact of Gonadal Hormones on Mating in
Females
Hormones exert a powerful impact on social behaviors. For
example, the presence or absence of gonadal steroids dictates Females also rely on gonadal steroids for the expression of
whether mating takes place or not. Information about the mating behavior. In spontaneous (Erb et al., 1971) – though
internal steroid milieu and external chemosensory signals are not in induced (Kauffman and Rissman, 2006) – ovulators,
integrated within brain regions involved in social behaviors. gonadal hormones are released in a cyclical manner with
This section provides a brief overview. Extensive reviews of estrogen rising before progesterone rises. Estrogen recruits
this topic are available in other chapters within this volume. progesterone receptors (Blaustein et al., 1980) which must be
activated for mating to occur (Mani and Oyola, 2012). Ovarec-
tomized females can be made sexually receptive if treated with
1.11.3.1 The Impact of Gonadal Hormones on Mating in
estrogen and progesterone in a pattern that mimics their
Males
normal release. For example, most rodents have 4-day estrous
Gonadectomy, which removes the greatest source of gonadal cycles. Treatment with estradiol for 2 days followed by proges-
steroids, gradually and permanently eliminates male mating terone is sufficient to induce motivational and consumatory
behavior (Clark et al., 1995; Ballard and Wood, 2007). aspects of female mating behavior. Both steroids appear to
Systemic treatment with testosterone (T) via injections or have central sites of action (Maggi and Perez, 1985), as estrogen
timed-release capsules restores mating (Hull and Dominguez, and progesterone placed in the hypothalamus restore mating
2007). T competes with dihydrotestosterone (DHT) to bind (Barfield et al., 1984). Ovarian steroids act peripherally as
to androgen receptors (ARs) (Pihlajamaa et al., 2015) and to well as centrally to coordinate responses to somatosensory
exert its actions. While ARs are found throughout the central stimuli and act within the hypothalamus to optimize and coor-
nervous system (Sarkey et al., 2008; Simerly et al., 1990) they dinate hypothalamic neurotransmitter (noradrenergic and
are densely concentrated in the MA and bed nucleus of the stria others) release, cellular responses to neurotransmitter release,
terminalis (BST) (Wood et al., 1992). An interesting experi- and second messenger systems (i.e., cAMP, cGMP) to facilitate
ment by Sheridan suggests the possibility of more than one expression of lordosis at the appropriate time (Etgen et al.,
AR: one that binds unmetabolized T, and another one that 1999; González-Flores et al., 2010).
binds DHT (with greater affinity for DHT than T). The study
utilized radioactive T that would lose its label if metabolized
or aromatized (Sheridan, 1979). The study found the labeled, 1.11.4 The Social Behavior Network
unmetabolized T in only two brain areas – the BST and the
MA suggesting that the MA and BST alone may be directly influ- Early research outlined a series of pathways, collectively
enced by T in addition to its metabolites (Sheridan, 1983, described as the limbic system, connecting the amygdala with
1991). Therefore, the BST and MA are unique in that they are the hippocampus for learning and memory and the cortex for
responsive to actions by unmetabolized T in addition to its subsequent decision-making (MacLean, 1990; Papez, 1937).
metabolites. More recent work with more sophisticated techniques has ques-
Circulating androgen is essential for male sex behavior, and tioned the designation of these groups as a single system and
treatment with T restores mating in castrate male hamsters suggested, instead, that it operates as two or three circuits with
over several weeks (Ballard and Wood, 2007). Gonadal different, specific roles (Rolls, 2015; Catani et al., 2013).
hormones in males are released in pulses or bursts that reflect Research using rodent models has identified a subset of limbic
the pulsatile nature of the pituitary hormones that regulate system nuclei that play major roles in the regulation of social
steroid hormone release (Gan and Quinton, 2010). However, behaviors (Newman, 1999). Described as the ‘social behavior
pulsatile release of pituitary hormone is not critical for T produc- network’ (SBN), it is comprised of six reciprocally connected
tion (Chase et al., 1988) or steroidal regulation of male regions: the extended MA (central-medial amygdala and BST),
behavior, as copulation is restored in male hamsters, rats, and anterior hypothalamus, midbrain, lateral septum, medial pre-
mice given injections (Piekarski et al., 2009) or timed-release optic area (MPOA), and the ventromedial hypothalamus
capsules (Matochik et al., 1994; Antonio-Cabrera and Paredes, (VMH) (Newman, 1999; O’Connell and Hofmann, 2011;
2012). Figure 1). Together, these regions are nodes within a network
T is metabolized to DHT and estradiol (E) by 5-alpha that regulates sexual, aggressive, and parental behavior in males
reductase and aromatase, respectively. Both metabolites are and females. Newman’s model broke new ground in suggesting
332 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior

Mesolimbic
reward system
Social behavior
network Str

POA NAcc

AH LS VP

VMH BNST/ blAMY


meAMY

PAG/CG HIP

VTA

Figure 1 The social behavior network and social decision-making network: the left panel shows the six reciprocally interconnected regions of the
social behavior network as described by Newman (1999). The lateral septum and BST/MA are also involved in the social decision-making network
(O’Connell and Hofmann, 2011). Arrows indicate projection neurons from one region to the other, and double-headed arrows indicate that the
connection is reciprocal. AH, anterior hypothalamus; blAMY, basolateral amygdala; BNST/meAMY, bed nucleus of the stria terminalis/medial amyg-
dala; HIP, hippocampus; LS, lateral septum; NAcc, nucleus accumbens; PAG/CG, periaqueductal gray/central gray; POA, preoptic area; Str, striatum;
VMH, ventromedial hypothalamus; VP, ventral pallidum; VTA, ventral tegmental area. Reprinted with permission from O’Connell, L.A., Hofmann, H.A.,
2011. The vertebrate mesolimbic reward system and social behavior network: a comparative synthesis. J. Comp. Neurol. 519, 3599–3639.

that it is the collective, rather than the individual, elements of the cues related to arousal, reinforcement, motivation, and
group that drives behavior. Exactly which behavior is expressed emotion surrounding survival are processed (LeDoux, 2012;
is determined by the degree to which relevant environmental Sternson, 2013). For example, as part of this circuit, chemosen-
and internal parameters activate a given area. sory cues related to responses to unconditioned threats are
The critical node of this network for processing social che- mediated via the MA (LeDoux, 2012). In addition to inte-
mosensory cues related to male mating behavior is the medial grating chemosensory and hormonal signals to allow mating
nucleus of the amygdala (MA). The nucleus forms a small behavior or other behaviors to occur, the MA appears to serve
almond-shaped collection of cells in the ventromedial extent as a filter or sorting center to inhibit certain behaviors or choose
of the temporal lobe in vertebrates (Medina et al., 2011; between competing chemosensory-driven behaviors (e.g.,
Martinez-Garcia et al., 2002). It is in the MA that chemosensory defensive response versus mating behavior if both cues are
signals from both the accessory and the main olfactory bulbs present).
are integrated with hormonal signals to initiate and regulate There is evidence for this type of MA-initiated inhibitory
reproductive behavior. The MA also processes chemosensory chemosensory circuit regulating maternal behavior. Lesions of
cues salient to other social behaviors, including parental the MA (especially the posteroventral subdivision), anterior
behavior, aggression, and conditioned defeat (Newman, hypothalamus, or VMN decrease the latency for nonmaternal
1999; Markham and Huhman, 2008). The MA has also been rats to exhibit maternal behavior, suggesting that these regions
implicated as an extension of the mesolimbic dopamine- normally act to inhibit the behavior (Sheehan et al., 2001).
based reward system, in a ‘social decision-making network’ In addition, pup-induced Fos stimulation in the anterior
(O’Connell and Hofmann, 2011; Figure 1). In the expanded hypothalamus and VMN as well as other regions of the SBN
group or social decision-making network the rewarding proper- are reduced with ipsilateral lesions of the MA (Sheehan et al.,
ties of dopamine impart salience to stimuli and associated 2001). Taken together, these data suggest that the MA, through
behaviors (O’Connell and Hofmann, 2011). However, it its projection to the hypothalamus, and acting together with
appears that the reward component of sexual pheromones is other parts of the SBN, is involved in both facilitating and
mediated via the vomeronasal amygdala (medial and cortical inhibiting chemosensory-induced behaviors depending on
posteromedial nuclei) projections to the ventral striatum and the internal hormonal milieu and external signals received.
not via the dopaminergic tegmento-striatal pathway (Lanuza It should be noted that more recent models describe a more
et al., 2008). Two nuclei, the septum and the BST (the exten- complex brain circuitry involved in chemosensory-driven
sion of the MA), belong to both the SBN and social decision- expression of the initiation of maternal behavior (Olazabal
making network, supporting the role of the MA as a central et al., 2013). For example, stimulation from pup chemosensory
organizing hub for social behaviors. cues can differentially impact specific subdivisions of the lateral
In addition, the MA, MPOA, and parts of the hypothalamus septum or MPOA to inhibit or facilitate maternal behavior
involved in the SBN overlap as part of a ‘survival circuit’ where (reviewed by Olazabal et al., 2013). The nucleus accumbens
The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 333

and ventral pallidum are also involved in processing the reward (pv) subdivision – is also connected to nuclei that mediate
salience of pup olfactory cues; and the central and basolateral defensive behaviors (Canteras, 2002; Figure 2). Using axon
amygdala are involved in processing the emotional salience of tracers genetically encoded for different LIM homeodomain
pup cues to facilitate or inhibit behavior (see Olazabal et al., transcription factors, Choi et al. (2005) identified GABAergic
2013 for review). While the MA is one of the first regions to cells within the MApd of the male rat that respond to mating-
integrate external chemosensory signals and internal hormonal related olfactory cues (female urine). These cells project to the
milieu to help guide appropriate social behavior, other regions ventrolateral (vl) VMH, and the authors suggest that this allows
are involved in an extraordinarily complex circuitry of decision- reproductive behavior to occur, likely by disinhibiting cells
making and expression of social behaviors, concomitantly within the VMHvl, or indirectly through the BST. These
processing different aspects of chemosensory salience for the GABAergic cells in MApd that respond to mating-related chemo-
individual. sensory cues and project to regions that facilitate mating
behavior also contain the transcription factor Lhx6 (Choi et al.,
2005). They are distinct from cells within the MApv that contain
1.11.5 MA Subdivisions other Lhx transcription factors, respond to olfactory defensive
cues (cat odor), and are glutamatergic. Choi et al. (2005) have
The MA can be divided into three main subdivisions based on proposed that the MApv glutamatergic cells that project to the
their connections within the SBN. Two of these – the anterior dorsomedial (dm) part of the VMH are allowing or facilitating
(MAa) and the posterodorsal (MApd) – maintain connections defensive behaviors in response to odor cues and may serve to
with nuclei involved in reproduction (Coolen and Wood, inhibit mating behavior when a concomitant threatening cue is
1998; Maras and Petrulis, 2010a; Gomez and Newman, 1992; present via projection to the VMHvl. This elegant study suggests
Canteras et al., 1995) while the third – the posterior ventral that the subdivisions of the MA respond to different types of

MA posterodorsal (pd)
Receives input from AOB and MOB
Projects to VMH(vl) and posteromedial BST
Contains androgen receptors (++++) and
estrogen receptors (+++(+))
Regulates mul ple aspects of reproduc ve
MApd
behavior; regulates female odor preference
via androgen receptors in pd
Contains GABAergic cells that respond to
ma ng-related chemosignals

MAa

MA anterior (a)
Receives input from AOB and MOB
Projects strongly to to VMH(vl), posterointermediate
BST and laterodorsal BST MApv
Contains androgen receptors (+++) and estrogen MA posteroventral (pv)
receptors (+) Receives input from AOB and MOB
Regulates interest in male and female odors (males) Projects to VMH(dm) (and vl to inhibit ma ng
and responses to odor in BST/MPOA behavior when threat is present), and the
May act as social filter for salience of cues via posteromedial BST
interac on with MApd Contains androgen receptors (+++) and
Oxytocin and arginine-vasopressin implicated in social estrogen receptors (+)
chemoinves ga on in MAa Regulates defensive behavior via glutaminergic
cells that respond to threat/defense-related
chemosignals

Figure 2 Subdivisions of the medial nucleus of the amygdala (MA) and their roles in chemosensory-driven mating behaviors. The double arrows in
this schematic illustrate the reciprocally connected subdivisions of the MA. All three subdivisions, anterior (a), posteroventral (pv), and posterodorsal
(pd), receive input from the AOB, and pd and anterior receive input from the MOB (Choi et al., 2005; Kang et al., 2009; Cadiz-Moretti et al., 2016).
The AOB projects to deeper layer Ia laminae of the amygdala versus MOB (Kang et al., 2009). All three subdivisions contain steroid receptors, to
different degrees as indicated by number of þ symbols (Simerly et al., 1990), and play differential roles in regulating aspects of chemosignal-driven
mating behaviors. All MA subdivisions project to the medial preoptic area, with the strongest projection from pd to the medial preoptic nucleus
(based on a mouse study) (Pardo-Bellver et al., 2012). Differential projections to the BST are described in the schematic. Despite some differences,
the three main subdivisions appear to interact to cooperatively regulate expression of (sometimes competing) chemosensory-driven behaviors. The
drawings of MA subdivisions are adapted from “A Stereotaxic Atlas of the Golden Hamster Brain” (Morin and Wood, 2001). This figure and legend
are also based on the work of Wood et al. (1992), Pardo-Bellver et al. (2012), Portillo et al. (2006), Greco et al. (1996), Ferguson et al. (2001),
Arakawa et al. (2010), Maras and Petrulis (2010a), and Keshavarzi et al. (2014).
334 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior

cues, but there is some prioritization and filtering occurring at the nucleus is greater in males and controlled by circulating T
this hub of the SBN to ensure that responding to threats takes (Morris et al., 2008; Rasia-Filho et al., 1991). Male rats have
precedence over mating behavior. In social mammals, this mech- a greater number of cells – neuronal and supporting (Johnson
anism may be akin to social judgment. Details of the pathways et al., 2008) – than females in the MApd and MApv (Rasia
and mechanisms of this type of filtering or gating require further et al., 2002). The morphology of the cells is also dimorphic.
study. A cell fate mapping study showed a differential pattern of Both glia (Johnson et al., 2008) and neurons (Rasia et al.,
expression of Sonic hedgehog (Shh)- and Nkx2-1-derived inhib- 1999) are more complex in males than females. Moreover,
itory output cells in the MApv and MApd, and that these cell neuronal morphology is sex specific so that dendrites are differ-
groups possess different neurotransmitter profiles (Carney et entially oriented in both the MApd and MApv (Dall’Oglio
al., 2010). These findings support the idea that the two subdivi- et al., 2008) of male and female rats. While most of these differ-
sions of MA, which have differing cell lineages, play different ences are in the steroid-responsive MApd, sex differences in
integral roles within the SBN. The findings suggest that the MA dendritic spines are also found in the anterior versus posterior
subdivisions are programmed by cell migration early in develop- divisions of MA (Rasia et al., 1999). Male rats have greater
ment to manage chemosensory input and regulate appropriate concentrations of opioid receptors (Wilson et al., 2002),
behavioral responses. The MApd is anatomically sculpted by substance P (Malsbury and McKay, 1989), and CCK (Micevych
internal processes during puberty that determine axon place- et al., 1994) in the MApd than females. There are species-
ment and pruning of dendrites and spines (Zehr et al., 2006). specific neurochemistry differences as well. For example, while
In addition, social signals and experiences after weaning impact the MApd of male prairie voles has more tyrosine hydroxylase
MApd volume and neuronal soma size, as well as male mating than that of females (Cavanaugh and Lonstein, 2010), this sex
behavior (Cooke et al., 2000). Thus, both pubertal anatomical difference is not found in rats or hamsters (Northcutt et al.,
developments and social experiences wire the MA for processing 2007). Taken together the data suggest that the MA of males
the array of social olfactory cues that allow appropriate behav- and females are differentially organized, and this may play
ioral responses to those cues via the SBN. a role in differential responses to chemosignals. This hypoth-
esis is further explored below.

1.11.6 Comparative Anatomy


1.11.8 Role of the MA and Connected Structures in
Beyond mammals, key elements of the SBN appear to be Social Odor Investigation and Sexual Behavior in
similar in the nonmammalian vertebrate brain (Goodson, Males
2005; O’Connell and Hofmann, 2011, 2012) suggesting that
the SBN is an evolutionarily conserved, key feature of the verte- The MA plays a critical role in the attraction of males to the
brate brain (Goodson, 2005) and that diversity in social odors of females. Bilateral ablation of either the MAa or
behavior is likely due to variation in an otherwise conserved, MApd eliminates preferences for female odors in Syrian
homologous neural network and the array of gene expression hamsters and rats, albeit in different ways. Specifically, in
within that network (O’Connell and Hofmann, 2011, 2012). hamsters, MAa damage causes unusually high levels of interest
It should be noted that, while not listed as part of the SBN, in male odors but unaltered (high) levels of interest in female
the olfactory bulbs also play an obvious and critical role in the odors, whereas males with damage to MApd reduce their inves-
organization and expression of olfactory-mediated social behav- tigation of female odors (Dhungel et al., 2011a,b; Maras and
iors. The pioneering work of Keverne (Lloyd-Thomas and Petrulis, 2006). This may suggest that the rostral aspect of the
Keverne, 1982; Kaba et al., 1989) showed that memory of nucleus, with its strong chemosensory input, provides a ‘social
a mate may not be encoded in the amygdala or the SBN. Rather, filter’ that evaluates the significance of a social odor. This infor-
the site of the phenomenon known as the Bruce effect (preg- mation then interacts with a steroid-dependent mechanism in
nancy termination after exposure to chemosignals from an unfa- the caudal aspect of MA that adjusts the ‘gain’ driving increased
miliar male) lies in the accessory olfactory bulb (Kaba et al., investigation. Further evidence for this distinction between
1989, 1994; Brennan et al., 1990). Moreover, research suggests MAa and MApd function comes from (1) the ability of MAa,
that the main olfactory system may play a larger role in process- but not MApd, lesions to eliminate immediate-early gene
ing reproductively relevant chemosignals than initially consid- (IEG) responses throughout MA, BST, and MPOA following
ered (Baum and Cherry, 2015; Woodley and Baum, 2004; exposure to social odors (Maras and Petrulis, 2010b,c; Wang
Dhungel et al., 2011a; Swann et al., 2001; Kang et al., 2009) et al., 2013; Maras and Petrulis, 2010b), (2) highly specific
and that the piriform cortex may play a significant role in the IEG and electrophysiological responses to social odors in
processing of socially relevant cues (Veyrac et al., 2011; Petrulis, MApd, but not MAa (Bergan et al., 2014; Meredith and
2009). Thus, the basic networks and circuitry do not function Westberry, 2004; Samuelsen and Meredith, 2009), (3) finding
as solitary units, but as part of an interconnected system of that olfactory bulb stimulation only increases IEG expression
networks that also includes afferent and efferent brain regions. in MAa, but not MApd (Blake and Meredith, 2010; Meredith
and Westberry, 2004; Nolte and Meredith, 2005), and (4) the
ability of AR blockade to impair female odor preference in
1.11.7 Sex Differences MApd, but not MAa (Hosokawa and Chiba, 2010). Indeed,
the MAa and MApd must interact to drive attraction to female
The MA is sexually dimorphic in a number of species for odors, either through reciprocal MAa–MApd connections
a variety of parameters. The volume of both the neurons and (Maras and Petrulis, 2010a,b,c) or through downstream
The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 335

structures, as functional disconnection of MAa and MApd elim- (ER) blockade here reduces mounting in hormonally intact
inates odor preference (Maras and Petrulis, 2010c; Figure 2). males (Wood and Williams, 2001). Although these results
Although the MA is critical for male attraction and interest suggest that estrogen action in MApd is critical for sexual
in female odors, the particular neurochemical mechanisms behavior, systemic aromatase blockade has no effect on female
within MA that regulate this process are not well understood. investigation or mounting behavior in hamsters (Cooper et al.,
There is evidence, however, that the oxytocin (OT) and 2000). In rats, implants of estradiol within MApd restore
arginine-vasopressin (AVP) neuropeptide systems within the mounting behavior in castrates (Huddleston et al., 2003;
MA are implicated in social investigation. This relationship Rasia-Filho et al., 1991), and blocking aromatase action here
appears bidirectional because, in mice, OT receptor blockade (Huddleston et al., 2006) reduces mounting in intact male
impairs IEG responses in MA (and connected structures) to rats. However, blocking ER-alpha in MApd does not impair
females (Ferguson et al., 2001) or their odors (Samuelsen mounting (Paisley et al., 2012) nor is mounting maintained
and Meredith, 2011). Additionally, MA lesions reduce social by specific ER-alpha or ER-beta agonist infusions in MApd
IEG responses in hypothalamic OT/AVP cell groups (Wang (Russell et al., 2012). This suggests that combined ER-alpha
et al., 2013) and central/peripheral OT release (Wang et al., and ER-beta activity is required in the MApd for sexual
2015). initiation and/or AR activation is also needed. Indeed,
The typical reductions in male chemoinvestigation of blocking AR within MApd reduces mounting in rats
familiar females (‘social memory’) appear to require OT action (McGinnis et al., 1996) and is necessary and sufficient for
in the MA: OT receptor antagonist injected into MApd of male sexual arousal to distal female cues in rats (Bialy and Sachs,
mice and rats blocks social memory whereas OT injected into 2002; Bialy et al., 2011). Similarly, the initiation of sexual
MApd rescues social memory in OT knockout mice (Ferguson behavior in mice survives deletion of virtually all aromatase
et al., 2001; Lukas et al., 2013). This effect may be due to OT neurons in MApd (Unger et al., 2015).
facilitation of long-term depression of AOB, but not MOB, Perhaps the strongest argument against DHT acting through
inputs to MA (Gur et al., 2014). However, OT effects on MA the MAa is in studies of MA morphology. Systemic T, E, and
are not limited to opposite-sex investigation as OT receptor DHT þ E affect changes in neuronal morphology of the MAa,
antagonist infusions in MA increase approach toward predator but DHT alone has no effect in either the hamster (Gomez
odors (Samuelsen and Meredith, 2011) and decrease approach and Newman, 1991), or the rat (Cooke et al., 2003), suggesting
toward odors of healthy same-sex conspecifics (Arakawa et al., that DHT acts on other sites to affect behavior or affects
2010). This pattern of results would suggest that OT in MA nor- behavior without affecting morphology of the MA.
mally promotes interest in complex odors except that avoid- The neurochemical mechanisms by which the MA initiates
ance of odors from sick males is unaffected by OT receptor investigation and sexual behavior are becoming somewhat
blockade (Arakawa et al., 2010); this discrepancy remains to clearer. Even though there is little overlap between the MApd
be explained. A similar discrepancy is apparent with AVP action cells that respond during copulation or during aggression
in MA: blockade of AVP receptors (V1a type) here inhibits (Lin et al., 2011), the initiation of copulation and aggressive
avoidance of sick conspecifics (Arakawa et al., 2010), but upre- behavior both require GABA neuron activation in MApd
gulation of AVP within MA by Toxoplasma gondii infection leads (Hong et al., 2014). A low-intensity increase in activity of
to an abnormal attraction to predator (cat) odors in rats (Hari GABAergic neurons in MApd stimulates mounting behavior
Dass and Vyas, 2014). This difference in results may reflect or social grooming whereas attack behavior is initiated at
different loci of AVP action. That is, local AVP release and higher levels of GABA cell activity/recruitment. In contrast,
action within MA promotes avoidance of unhealthy or MApd glutamatergic neurons appear to normally inhibit
dangerous odors whereas increased AVP produced in MA ongoing social behavior. Perhaps glutamatergic MApd cells
may be released at distal targets to drive attraction. produce these effects by coreleasing beta-endorphin as injec-
The MA initiates male sexual behavior through its integra- tions of this peptide into MApd decrease investigation
tion of chemosensory and sex steroid signals (Wood and of females and subsequent transition to mounting in rats
Coolen, 1997). MA ablation in hamsters, like olfactory bulb (McGregor and Herbert, 1992b,c). There is also evidence that
removal (Murphy and Schneider, 1970), eliminates sexual AVP cells within MA play a role in decreasing hippocampal
behavior and strongly inhibits AGI of females; this deficit is activity during initial investigation and mounting in male
much more evident following MAa lesions than after MApd rats, but the precise functional role of this connection has not
damage (Lehman et al., 1980). In contrast, in male rats it is been established (Smock et al., 1998).
damage to MApd, and to a lesser extent MAa, which impairs
initiation of sexual behavior (McGregor and Herbert, 1992a)
1.11.8.1 MA Connections with BST
and sexual arousal (NCEs) toward female odors (Kondo
et al., 1998; Kondo and Sachs, 2002). The massive interconnections between MA and BST are a key
Sex steroids acting within MApd appears to be necessary and neural substrate for attraction to, and interest in, opposite-sex
sufficient for initiating mounting and female investigation. odors. In male hamsters, damage to just the BST eliminates the
However, the precise nature of this relationship may vary across high levels of interest in volatile female odors and strongly
species, and the evidence is, in some cases, contradictory. For decreases interest in contacting female scents (Been and Petrulis,
example, implanting T or estradiol, but not dihydrotestoster- 2010a; Powers et al., 1987). Similarly, male rats with damage to
one, into the MApd restores mounting and female investiga- BST also show a lack of preferential interest in sexually active
tion in castrated hamsters (Coolen and Wood, 1999; Wood females (Edwards et al., 1996). The preference to investigate
and Newman, 1995c; Wood, 1996), and estrogen receptor volatile (but not contacted) components of female odors
336 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior

requires functional interactions between BST and the MA (Been Wood and Newman, 1995a,b), rats (Lisk, 1967), and mice
and Petrulis, 2012). This pattern of results suggests that MA-BST (Nyby et al., 1992) (also see Sipos and Nyby, 1996, 1998).
interactions are critical for attraction to distal female cues, Conversely, blocking aromatase or ER-alpha action in MPOA
but that preference for odors when contact is allowed is reduces mounting in rats (Clancy et al., 2000; Huddleston
mediated by the limited, but direct, AOB projections to BST. et al., 2007; Paisley et al., 2012; Russell et al., 2012; Sano
Although the BST, or its connections with MA, is not critical et al., 2013). However, AR action in the MPOA also appears
for copulatory behavior (Been and Petrulis, 2010a, 2012), BST important for initiating mounting as blocking AR in MPOA
lesions do reduce the AGI that precedes copulation (Claro reduces mount behavior in rats (Harding and McGinnis,
et al., 1995; Powers et al., 1987) as well as penile erections in 2004; McGinnis et al., 1996, 2002).
response to female odors (Liu et al., 1997). T or E implants
in BST/MPOA can restore female investigation and mounting
1.11.8.3 MA Connections with VMH
in hamsters (Wood and Newman, 1995b) and mice (Matochik
et al., 1994; Nyby et al., 1992), but no studies have investigated In the context of sexual behavior, there has been a strong focus
how hormonal manipulations restricted to BST influence on the VMH as a key brain substrate for hormonal control of
sexual behavior. female, rather than male, sexual behavior (Pfaff, 1980).
However, several lines of evidence now indicate that the VMH
actually plays a very important role in initiating male investiga-
1.11.8.2 MA Connections with MPOA
tion and mounting of females. First, lesions limited to the MAa
The MPOA may be part of the circuit that links interest in projection zone of the VMH impair precopulatory behavior in
opposite-sex cues to motivated approach behavior (Everitt, male rats (Harding and McGinnis, 2005). Second, within the
1990), but this relationship is complicated by the oft- VMH, T implants restore, and AR blockade impairs, preference
observed concomitant damage to BST following MPOA lesions for investigating sexually active females (McGinnis et al., 1996;
and interactions with the sexual experience of the subject. Harding and McGinnis, 2003, 2004). Third, deletion of proges-
Damage that includes the MPOA reverses the preference for terone receptor-bearing neurons in VMH decreases male
female odors in sexually naïve ferrets (Alekseyenko et al., mounting behavior as well as female sexual behavior (Yang
2007; Hurtazo and Paredes, 2005) and eliminates preference et al., 2013). Lastly, low-level stimulation of ER-alpha neurons
for estrous female odors in sex-experienced rats (Dhungel within VMH, much like MApd stimulation (Hong et al.,
et al., 2011b; Hurtazo et al., 2008; Hurtazo and Paredes, 2014), induces prolonged investigation of females and
2005). However, the ventral/caudalmost part of the BST was subsequent mounting (Lee et al., 2014), suggesting that MA-
also lesioned in these studies and so any deficits in investiga- VMH connections are critical for managing and filtering
tion could be due to ventral or caudal BST damage and not conflicting social states (i.e., aggression vs copulation).
to MPOA damage itself. Indeed, in hamsters, only damage to
both the MPOA and the ventral/caudal BST was sufficient to
eliminate interest in volatile female odors by sexually experi- 1.11.9 The Role of the MA in Females
enced male hamsters (Been and Petrulis, 2010b). Specific
MPOA lesions only reduce volatile odor preference in sexually The MA plays a critical role in chemosensory-driven sexual
naïve males, not in sex-experienced animals (Been and Petrulis, behavior in females. Damage to MA eliminates differential
2010b). In fact, this caudal/ventral aspect of BST is already investigation by females of male odors in Syrian hamsters
known to be critical for sexual behavior in both rats (Finn (Petrulis and Johnston, 1999), mice (DiBenedictis et al.,
and Yahr, 2005) and gerbils (Yahr and Gregory, 1993). Simi- 2012), and rats (Kondo and Sakuma, 2005) as well as the
larly, within MPOA, AR blockade only impairs opposite-sex heightened receptivity following repeated mating in rats (Rajen-
preference when targeted to caudal and dorsal MPOA, an dren and Moss, 1993). However, ER-alpha knockdown within
area contiguous with caudal/ventral BST (Harding and MApd has no effect on preference and proceptive behavior in
McGinnis, 2004; McGinnis et al., 2002). female rats (Spiteri et al., 2012) and so the behavioral role
The MPOA has long been known to be critical for male that gonadal steroids play within MA of females is currently
sexual behavior (Hull and Dominguez, 2007). Indeed, lesions unclear.
limited to the MPOA eliminate copulation in Syrian hamsters
(Been and Petrulis, 2010b; Powers et al., 1987), gerbils
1.11.9.1 MA Connections with BST and MPOA
(Heeb and Yahr, 2000; Yahr and Gregory, 1993; Yahr et al.,
1994), mice (Bean et al., 1981), and rats (Dorries et al., The BST is well known as a brain region that is sexually differ-
1997). Connections from MA to MPOA are critical for the initi- entiated in size and cell phenotype (de Vries and Sodersten,
ation of copulation in hamsters (Been and Petrulis, 2012), rats 2009) which may explain the relative lack of data on the role
(Kondo and Arai, 1995), and gerbils (Heeb and Yahr, 2000), of BST in female sexual attraction or behavior. What evidence
indicating that this pathway is the likely circuit for chemosen- exists suggests that the female BST is involved in appropriate
sory control of male copulatory behavior (Triemstra et al., targeting of sexual solicitation, via an OT mechanism, and is
2005). Downstream of MA, the MPOA is also a site of conver- not involved in directing interest toward male odors (Martinez
gence of steroid and chemosensory signals for regulating copu- et al., 2010; Martinez and Petrulis, 2011). Although the MPOA
lation (Greco et al., 1998; Wood and Newman, 1995b) as is more typically associated with male copulatory behavior,
implanting T or E into MPOA restores mounting and female this brain area does play a role in regulating both interest in
investigation in castrated hamsters (Lisk and Bezier, 1980; males and initiation of female copulatory behavior. For
The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 337

example, when females are allowed to pace mate, MPOA are modified by steroids and sex. How can one area be respon-
lesions impair female copulatory behavior (Yang and Clem- sible for multiple behaviors? To answer that question, one
ents, 2000; Whitney, 1986) and eliminate preferences for need only look to the vast research on the VMH. Decades of
male scents in rats (Xiao et al., 2005) and hamsters (Martinez research have implicated this tiny group of cells in feeding,
and Petrulis, 2013) but not in ferrets (Robarts and Baum, copulation, and lordosis. Clues to the mechanisms that
2007). Moreover, knockdown of ER-alpha within rat MPOA underlie its complex roles have been addressed using optoge-
impairs preferential investigation of reproductively active netics coupled with genetic cell markers and other genetic
males (Spiteri et al., 2012). approaches. Stimulation and ablation of specific cells within
the VMH reveal that the functional subdivisions of the VMH
defy conventional approaches. Neurons participating in
1.11.9.2 MA Connections with VMH
specific behaviors are not segregated to different regions –
The VMH has long been implicated in the control of female they are distributed throughout (Lin et al., 2011). Moreover,
sexual receptivity (Pfaff et al., 2008), but it also regulates the study supports earlier findings, described above, that the
female precopulatory behavior. For example, lesions of the VMH is also involved in male sexual behavior (Lin et al.,
VMH in ferrets eliminate preference for investigating male 2011). Similar work in the MA could begin with optogenetic
cues (Robarts and Baum, 2007) whereas blocking ER-alpha approaches, building on previous LIM homeodomain data
in VMH reduces preference for reproductively active males in (Choi et al., 2005).
rats (Spiteri et al., 2010) as well as proceptive behaviors in More comparative work also needs to be done. The large
mice (Musatov et al., 2006). Similarly, estrogenic stimulation body of work presented here is focused on mammals primarily
of VMH reinstates sexual solicitation in ovariectomized because work in birds and reptiles is in short supply. Homolog-
hamsters (Takahashi et al., 1985). ical studies identifying the amygdala in birds and fishes suggest
functional differences in the rostral and caudal regions of the
tenia (homologous to the MA) (Absil et al., 2002; Thompson
1.11.10 Steroids May Act Centrally and Peripherally et al., 1998). Additional work would broaden our
to Impact Responses to Pheromones understanding of the relationship between the development of
the amygdala and the evolution of the behaviors it subserves.
Both sex and circulating hormones impact neural responses to Similarly, studies of the primate MA are few and far
chemosensory cues (Fiber and Swann, 1996; Brock et al., 2012; between. With recognition of the powerful work of LeDoux
Kang et al., 2011; Bodo and Rissman, 2007; Swann and Fiber, (2007), researchers have focused on fear as mediated by the
1997). Circulating steroids not only influence behavioral lateral amygdaloid nucleus, virtually ignoring the contribu-
responses to pheromones, but may also impact the sensitivity tions of the MA. This may be an oversight in the search for
to reception or perception of mating-related chemosignals. neural correlates of cognition. MA mediates aggression which
Steroids likely act on both central regions mediating behavioral is an important hallmark of some cognitive diseases (Van den
responses as well as peripheral structures that process incoming Stock et al., 2015). Olfaction has been implicated in cognitive
sensory information (Gandelman, 1983). Studies using Fos as diseases such as Parkinson’s, schizophrenia, and Alzheimer’s
a marker for neural stimulation show that T increases phero- (Good and Sullivan, 2015; Yildirim and Derksen, 2012;
mone (female vaginal secretion)-stimulated Fos in medial pre- Markham, 2012). Plaques and Lewy bodies’ characteristic of
optic nucleus magnocellular (MPNmag) of male hamsters these diseases are present in the olfactory bulbs of patients
(Fiber and Swann, 1996; Kollack-Walker and Newman, with disease (Attems et al., 2014; Wilson et al., 2011). Olfac-
1997) and fails to affect pheromone-stimulated Fos in the post- tory dysfunction is also associated with combat-related trau-
eromedial BST and the posterior MA of either sex (Fiber and matic brain injury (Xydakis et al., 2015). Anosmia
Swann, 1996). In a recent study using calcium imaging of vom- determined by smell tests has become a useful part of diag-
eronasal sensory neurons to directly visualize cellular response nostic confirmation and to inform medical decisions (Otta-
to male pheromone ligands, it was demonstrated that the viano et al., 2015; Xydakis et al., 2015). Steroids have also
pheromone-responsive sensory neurons respond to the phero- been implicated in the prognosis for cognitive diseases (Yil-
mone ligands during estrus, but not during diestrus when dirim and Derksen, 2012; Markham, 2012). Facial expression
progesterone is present (Dey et al., 2015). Taken together, these or emotion recognition are common deficits among certain
studies appear to confirm that circulating steroids act at both cognitive disorders (Torres et al., 2015; Yoshimura et al.,
peripheral sites and central sites to govern the reception or 2005; Rangel et al., 2015), and one study found the amygdala
perception of sensory cues and the behavioral responses to che- function was compromised in individuals with Parkinson’s
mosensory cues. disease (Yoshimura et al., 2005). The combination of olfac-
tory dysfunction and impairments of social cue perception
in individuals with cognitive disorders suggests the possible
1.11.11 Future Directions involvement of the MA.
As we seek to know more about these devastating cognitive
What next for this complex nucleus? While the posterior dorsal diseases, we ignore this area at our own peril. As a central hub
and posterior ventral subdivisions seem committed to specific of several important social networks within the connectome,
behavioral roles, the anterior subdivision does not, and may the amygdala with its ability for internal and external refer-
play a role in filtering social information. The MA also medi- ence may ultimately be where we find the answer to Pee
ates aggression and parenting, behaviors that, such as mating, Wee Herman’s question “I know you are, but what am I?”
338 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior

References Bialy, M., Sachs, B.D., 2002. Androgen implants in medial amygdala briefly maintain
noncontact erection in castrated male rats. Horm. Behav. 42, 345–355.
Absil, P., Braquenier, J.B., Balthazart, J., Ball, G.F., 2002. Effects of lesions of nucleus Birke, L.I., 1984. Effects of estradiol and progesterone on scent-marking behavior of
taeniae on appetitive and consummatory aspects of male sexual behavior in female rats. Horm. Behav. 18, 95–98.
japanese quail. Brain Behav. Evol. 60, 13–35. Blake, C.B., Meredith, M., 2010. Selective enhancement of main olfactory input to the
Alekseyenko, O.V., Waters, P., Zhou, H., Baum, M.J., 2007. Bilateral damage to the medial amygdala by GnRH. Brain Res. 1317, 46–59.
sexually dimorphic medial preoptic area/anterior hypothalamus of male ferrets Blaustein, J.D., Ryer, H.I., Feder, H.H., 1980. A sex difference in the progestin receptor
causes a female-typical preference for and a hypothalamic fos response to male system of guinea pig brain. Neuroendocrinology 31, 403–409.
body odors. Physiol. Behav. 90, 438–449. Bodo, C., Rissman, E.F., 2007. Androgen receptor is essential for sexual differentiation
Antonio-Cabrera, E., Paredes, R.G., 2012. Effects of chronic estradiol or testosterone of responses to olfactory cues in mice. Eur. J. Neurosci. 25, 2182–2190.
treatment upon sexual behavior in sexually sluggish male rats. Pharmacol. Bio- Brennan, P., Kaba, H., Keverne, E.B., 1990. Olfactory recognition: a simple memory
chem. Behav. 101, 336–341. system. Science 250, 1223–1226.
Arakawa, H., Arakawa, K., Deak, T., 2010. Oxytocin and vasopressin in the medial Brock, O., Keller, M., Douhard, Q., Bakker, J., 2012. Female mice deficient in alpha-
amygdala differentially modulate approach and avoidance behavior toward illness- fetoprotein show female-typical neural responses to conspecific-derived phero-
related social odor. Neuroscience 171, 1141–1151. mones. PLoS One 7, e39204.
Aste, N., Panzica, G.C., Viglietti-Panzica, C., Harada, N., Balthazart, J., 1998. Brown, R.E., 1978. Hormonal control of odor preferences and urine-marking in male
Distribution and effects of testosterone on aromatase mRNA in the quail and female rats. Physiol. Behav. 20, 21–24.
forebrain: a non-radioactive in situ hybridization study. J. Chem. Neuroanat. Cadiz-Moretti, B., Otero-Garcia, M., Martinez-Garcia, F., Lanuza, E., 2016. Afferent
14, 103–115. projections to the different medial amygdala subdivisions: a retrograde tracing
Attems, J., Walker, L., Jellinger, K.A., 2014. Olfactory bulb involvement in neurode- study in the mouse. Brain Struct. Funct 221, 1033–1065.
generative diseases. Acta Neuropathol. 127, 459–475. Canteras, N.S., 2002. The medial hypothalamic defensive system: hodological
Ball, G.F., Balthazart, J., 2008. How useful is the appetitive and consummatory organization and functional implications. Pharmacol. Biochem. Behav. 71,
distinction for our understanding of the neuroendocrine control of sexual behavior? 481–491.
Horm. Behav. 53, 307–311, author reply 315–318. Canteras, N.S., Simerly, R.B., Swanson, L.W., 1995. Organization of projections from
Ballard, C.L., Wood, R.I., 2007. Partner preference in male hamsters: steroids, sexual the medial nucleus of the amygdala: a PHAL study in the rat. J. Comp. Neurol.
experience and chemosensory cues. Physiol. Behav. 91, 1–8. 360, 213–245.
Barfield, R.J., Glaser, J.H., Rubin, B.S., Etgen, A.M., 1984. Behavioral effects of Carney, R.S., Mangin, J.M., Hayes, L., Mansfield, K., Sousa, V.H., Fishell, G.,
progestin in the brain. Psychoneuroendocrinology 9, 217–231. Machold, R.P., Ahn, S., Gallo, V., Corbin, J.G., 2010. Sonic hedgehog expressing
Baum, M.J., Cherry, J.A., 2015. Processing by the main olfactory system of che- and responding cells generate neuronal diversity in the medial amygdala. Neural
mosignals that facilitate mammalian reproduction. Horm. Behav. 68, 53–64. Dev. 5, 14.
Baum, M.J., Vreeburg, J.T., 1973. Copulation in castrated male rats following Carr, W.J., Loeb, L.S., Dissinger, M.L., 1965. Responses of rats to sex odors.
combined treatment with estradiol and dihydrotestosterone. Science 182, J. Comp. Physiol. Psychol. 59, 370–377.
283–285. Carter, C.S., 1973. Olfaction and sexual receptivity in the female golden hamster.
Bean, N.J., 1982. Olfactory and vomeronasal mediation of ultrasonic vocalizations in Physiol. Behav. 10, 47–51.
male mice. Physiol. Behav. 28, 31–37. Catani, M., Dell’acqua, F., Thiebaut de Schotten, M., 2013. A revised limbic system
Bean, N.Y., Nunez, A.A., Conner, R., 1981. Effects of medial preoptic lesions on male model for memory, emotion and behaviour. Neurosci. Biobehav. Rev. 37,
mouse ultrasonic vocalizations and copulatory behavior. Brain Res. Bull. 6, 1724–1737.
109–112. Cavanaugh, B.L., Lonstein, J.S., 2010. Androgenic and oestrogenic influences on
Beauchamp, G.K., Beruter, J., 1973. Source and stability of attractive components in tyrosine hydroxylase-immunoreactive cells of the prairie vole medial amygdala and
guinea pig (Cavia porcellus) urine. Behav. Biol. 9, 43–47. bed nucleus of the stria terminalis. J. Neuroendocrinol. 22, 217–225.
Beauchamp, G.K., Magnus, J.G., Shmunes, N.T., Durham, T., 1977. Effects of Chang, Y.M., Kelliher, K.R., Baum, M.J., 2000. Steroidal modulation of scent inves-
olfactory bulbectomy on social behavior of male guinea pigs (Cavia procellus). tigation and marking behaviors in male and female ferrets (Mustela putorius furo).
J. Comp. Physiol. Psychol. 91, 336–346. J. Comp. Psychol. 114, 401–407.
Been, L.E., Bauman, J.M., Petrulis, A., Chang, Y.H., 2012. X-ray kinematics analysis Chase, D.J., Schanbacher, B.D., Lunstra, D.D., 1988. Effects of pulsatile and
of vaginal scent marking in female syrian hamsters (Mesocricetus auratus). Physiol. continuous luteinizing hormone (lh) infusions on testosterone responses to lh in
Behav. 105, 1021–1027. rams actively immunized against gonadotropin-releasing hormone. Endocrinology
Been, L.E., Petrulis, A., 2008. The neurobiology of sexual solicitation: vaginal marking 123, 816–826.
in female syrian hamsters (Mesocricetus auratus. In: Hurst, J.L. (Ed.), Chemical Choi, G.B., Dong, H.W., Murphy, A.J., Valenzuela, D.M., Yancopoulos, G.D.,
Signals in Vertebrates, vol. 11. Springer, New York. Swanson, L.W., Anderson, D.J., 2005. Lhx6 delineates a pathway mediating innate
Been, L.E., Petrulis, A., 2010a. Lesions of the posterior bed nucleus of the stria reproductive behaviors from the amygdala to the hypothalamus. Neuron 46,
terminalis eliminate opposite-sex odor preference and delay copulation in male 647–660.
syrian hamsters: role of odor volatility and sexual experience. Eur. J. Neurosci. 32, Clancy, A.N., Zumpe, D., Michael, R.P., 2000. Estrogen in the medial preoptic area of
483–493. male rats facilitates copulatory behavior. Horm. Behav. 38, 86–93.
Been, L.E., Petrulis, A., 2010b. The role of the medial preoptic area in appetitive and Clark, J.T., Micevych, P.E., Panossian, V., Keaton, A.K., 1995. Testosterone-induced
consummatory reproductive behaviors depends on sexual experience and odor copulatory behavior is affected by the postcastration interval. Neurosci. Biobehav.
volatility in male syrian hamsters. Neuroscience 170, 1120–1132. Rev. 19, 369–376.
Been, L.E., Petrulis, A., 2012. Dissociated functional pathways for appetitive and Claro, F., Segovia, S., Guilamon, A., Del Abril, A., 1995. Lesions in the medial posterior
consummatory reproductive behaviors in male syrian hamsters. Horm. Behav. 61, region of the BST impair sexual behavior in sexually experienced and inexperienced
204–211. male rats. Brain Res. Bull. 36, 1–10.
Bergan, J.F., Ben-Shaul, Y., Dulac, C., 2014. Sex-specific processing of social cues in Cooke, B.M., Breedlove, S.M., Jordan, C.L., 2003. Both estrogen receptors and
the medial amygdala. eLife 3, e02743. androgen receptors contribute to testosterone-induced changes in the morphology
Beyer, C., de la Torre, L., Larsson, K., Perez-Palacios, G., 1975. Synergistic actions of of the medial amygdala and sexual arousal in male rats. Horm. Behav. 43,
estrogen and androgen on the sexual behavior of the castrated male rabbit. Horm. 336–346.
Behav. 6, 301–306. Cooke, B.M., Chowanadisai, W., Breedlove, S.M., 2000. Post-weaning social isolation
Beyer, C., Larsson, K., Pérez-Palacios, G., Morali, G., 1973. Androgen structure and of male rats reduces the volume of the medial amygdala and leads to deficits in
male sexual behavior in the castrated rat. Horm. Behav. 4, 99–108. adult sexual behavior. Behav. Brain Res. 117, 107–113.
Beyer, C., Rivaud, N., 1973. Differential effect of testosterone and dihydrotestosterone Coolen, L.M., Wood, R.I., 1998. Bidirectional connections of the medial amygdaloid
on the sexual behavior of prepuberally castrated male rabbits. Horm. Behav. 4, nucleus in the syrian hamster brain: simultaneous anterograde and retrograde tract
175–180. tracing. J. Comp. Neurol. 399, 189–209.
Bialy, M., Nikolaev-Diak, A., Kalata, U., Nikolaev, E., 2011. Blockade of androgen Coolen, L.M., Wood, R.I., 1999. Testosterone stimulation of the medial preoptic area
receptor in the medial amygdala inhibits noncontact erections in male rats. Physiol. and medial amygdala in the control of male hamster sexual behavior: redundancy
Behav. 103, 295–301. without amplification. Behav. Brain Res. 98, 143–153.
The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 339

Cooper, T.T., Clancy, A.N., Karom, M., Moore, T.O., Albers, H.E., 2000. Conversion of Gattermann, R., Fritzsche, P., Neumann, K., Al-Hussein, I., Kayser, A., Abiad, M.,
testosterone to estradiol may not be necessary for the expression of mating behavior Yakti, R., 2001. Notes on the current distribution and the ecology of wild golden
in male syrian hamsters (Mesocricetus auratus). Horm. Behav. 37, 237–245. hamsters (Mesocricetus auratus). J. Zool. 254, 359–365.
Curtis, J.T., Liu, Y., Wang, Z., 2001. Lesions of the vomeronasal organ disrupt mating- Geyer, L.A., Barfield, R.J., 1978. Influence of gonadal hormones and sexual behavior
induced pair bonding in female prairie voles (Microtus ochrogaster). Brain Res. on ultrasonic vocalization in rats: I. Treatment of females. J. Comp. Physiol.
901, 167–174. Psychol. 92, 438–446.
Dall’Oglio, A., Gehlen, G., Achaval, M., Rasia-Filho, A.A., 2008. Dendritic branching Gomez, D.M., Newman, S.W., 1991. Medial nucleus of the amygdala in the adult
features of golgi-impregnated neurons from the “ventral” medial amygdala sub- syrian hamster: a quantitative golgi analysis of gonadal hormonal regulation of
nuclei of adult male and female rats. Neurosci. Lett. 439, 287–292. neuronal morphology. Anat. Rec. 231, 498–509.
de Vries, G.J., Sodersten, P., 2009. Sex differences in the brain: the relation between Gomez, D.M., Newman, S.W., 1992. Differential projections of the anterior and
structure and function. Horm. Behav. 55, 589–596. posterior regions of the medial amygdaloid nucleus in the syrian hamster. J. Comp.
Degtiar, V.G., Loseva, L.A., Isatchenkov, B.A., 1981. In vitro metabolism of androgens Neurol. 317, 195–218.
in hypothalamus and pituitary from infantile and adolescent rats of both sexes. Gonzalez-Mariscal, G., Melo, A.I., Zavala, A., Beyer, C., 1990. Variations in chin-
Endocrinol. Exp. 15, 181–190. marking behavior of New Zealand female rabbits throughout the whole repro-
Devor, M., Murphy, M.R., 1973. The effect of peripheral olfactory blockade on the ductive cycle. Physiol. Behav. 48, 361–365.
social behavior of the male golden hamster. Behav. Biol. 9, 31–42. González-Flores, O., Beyer, C., Gómora-Arrati, P., García-Juárez, M., Lima-
Dewsbury, D.A., 1975. Diversity and adaptation in rodent copulatory behavior. Science Hernández, F.J., Soto-Sánchez, A., Etgen, A.M., 2010. A role for Src kinase in
190, 947–954. progestin facilitation of estrous behavior in estradiol-primed female rats. Horm.
Dey, S., Chamero, P., Pru, J.K., Chien, M.S., Ibarra-Soria, X., Spencer, K.R., Behav. 58, 223–229.
Logan, D.W., Matsunami, H., Peluso, J.J., Stowers, L., 2015. Cyclic regulation of Good, K.P., Sullivan, R.L., 2015. Olfactory function in psychotic disorders: insights
sensory perception by a female hormone alters behavior. Cell 161, 1334–1344. from neuroimaging studies. World J. Psychiatry 5, 210–221.
Dhungel, S., Masaoka, M., Rai, D., Kondo, Y., Sakuma, Y., 2011a. Both olfactory Goodson, J.L., 2005. The vertebrate social behavior network: evolutionary themes and
epithelial and vomeronasal inputs are essential for activation of the medial variations. Horm. Behav. 48, 11–22.
amygdala and preoptic neurons of male rats. Neuroscience 199, 225–234. Greco, B., Edwards, D.A., Michael, R.P., Clancy, A.N., 1996. Androgen receptor
Dhungel, S., Urakawa, S., Kondo, Y., Sakuma, Y., 2011b. Olfactory preference in the immunoreactivity and mating-induced fos expression in forebrain and midbrain
male rat depends on multiple chemosensory inputs converging on the preoptic structures in the male rat. Neuroscience 75, 161–171.
area. Horm. Behav. 59, 193–199. Greco, B., Edwards, D.A., Zumpe, D., Michael, R.P., Clancy, A.N., 1998. Fos induced
DiBenedictis, B.T., Ingraham, K.L., Baum, M.J., Cherry, J.A., 2012. Disruption of by mating or noncontact sociosexual interaction is colocalized with androgen
urinary odor preference and lordosis behavior in female mice given lesions of the receptors in neurons within the forebrain, midbrain, and lumbosacral spinal cord of
medial amygdala. Physiol. Behav. 105, 554–559. male rats. Horm. Behav. 33, 125–138.
Dorries, K.M., Adkins-Regan, E., Halpern, B.P., 1997. Sensitivity and behavioral Gur, R., Tendler, A., Wagner, S., 2014. Long-term social recognition memory is
responses to the pheromone androstenone are not mediated by the vomeronasal mediated by oxytocin-dependent synaptic plasticity in the medial amygdala. Biol.
organ in domestic pigs. Brain, Behav. Evol. 49, 53–62. Psychiatry 76, 377–386.
Edwards, D.A., Griffis, K.T., Tardivel, C., 1990. Olfactory bulb removal: effects on Haga, S., Hattori, T., Sato, T., Sato, K., Matsuda, S., Kobayakawa, R., Sakano, H.,
sexual behavior and partner-preference in male rats. Physiol. Behav. 48, Yoshihara, Y., Kikusui, T., Touhara, K., 2010. The male mouse pheromone esp1
447–450. enhances female sexual receptive behaviour through a specific vomeronasal
Edwards, D.A., Walter, B., Liang, P., 1996. Hypothalamic and olfactory control of receptor. Nature 466, 118–122.
sexual behavior and partner preference in male rats. Physiol. Behav. 60, Harding, S.M., McGinnis, M.Y., 2003. Effects of testosterone in the VMN on copu-
1347–1354. lation, partner preference, and vocalizations in male rats. Horm. Behav. 43,
Eisthen, H.L., Wysocki, C.J., Beauchamp, G.K., 1987. Behavioral responses of male 327–335.
guinea pigs to conspecific chemical signals following neonatal vomeronasal organ Harding, S.M., McGinnis, M.Y., 2004. Androgen receptor blockade in the MPOA or
removal. Physiol. Behav. 41, 445–449. VMN: effects on male sociosexual behaviors. Physiol. Behav. 81, 671–680.
Erb, R.E., Randel, R.D., Callahan, C.J., 1971. Female sex steroid changes during the Harding, S.M., McGinnis, M.Y., 2005. Microlesions of the ventromedial nucleus of the
reproductive cycle. J. Animal Sci. 32 (Suppl. 1), 80–106. hypothalamus: effects on sociosexual behaviors in male rats. Behav. Neurosci.
Etgen, A.M., Chu, H.P., Fiber, J.M., Karkanias, G.B., Morales, J.M., 1999. Hormonal 119, 1227–1234.
integration of neurochemical and sensory signals governing female reproductive Hari Dass, S.A., Vyas, A., 2014. Toxoplasma gondii infection reduces predator
behavior. Behav. Brain Res. 105, 93–103. aversion in rats through epigenetic modulation in the host medial amygdala. Mol.
Everitt, B.J., 1990. Sexual motivation: a neural and behavioural analysis of the Ecol. 23, 6114–6122.
mechanisms underlying appetitive and copulatory responses of male rats. Neu- Heeb, M.M., Yahr, P., 2000. Cell-body lesions of the posterodorsal preoptic nucleus or
rosci. Biobehav. Rev. 14, 217–232. posterodorsal medial amygdala, but not the parvicellular subparafascicular thal-
Ferguson, J.N., Aldag, J.M., Insel, T.R., Young, L.J., 2001. Oxytocin in the medial amus, disrupt mating in male gerbils. Physiol. Behav. 68, 317–331.
amygdala is essential for social recognition in the mouse. J. Neurosci. 21, Hong, W., Kim, D.W., Anderson, D.J., 2014. Antagonistic control of social versus
8278–8285. repetitive self-grooming behaviors by separable amygdala neuronal subsets. Cell
Ferkin, M.H., Combs, A., delBarco-Trillo, J., Pierce, A.A., Franklin, S., 2008. Meadow 158, 1348–1361.
voles, Microtus pennsylvanicus, have the capacity to recall the “what”, “where”, Hosokawa, N., Chiba, A., 2010. Androgen receptor blockade in the posterodorsal
and “when” of a single past event. Anim. Cogn. 11, 147–159. medial amygdala impairs sexual odor preference in male rats. Horm. Behav. 58,
Ferkin, M.H., Zucker, I., 1991. Seasonal control of odour preferences of meadow voles 493–500.
(Microtus pennsylvanicus) by photoperiod and ovarian hormones. J. Reprod. Fertil. Huddleston, G.G., Michael, R.P., Zumpe, D., Clancy, A.N., 2003. Estradiol in the male
92, 433–441. rat amygdala facilitates mounting but not ejaculation. Physiol. Behav. 79,
Fiber, J.M., Swann, J.M., 1996. Testosterone differentially influences sex-specific 239–246.
pheromone-stimulated fos expression in limbic regions of syrian hamsters. Huddleston, G.G., Paisley, J.C., Clancy, A.N., 2006. Effects of estrogen in the male rat
Horm. Behav. 30, 455–473. medial amygdala: infusion of an aromatase inhibitor lowers mating and bovine
Finn, P.D., Yahr, P., 2005. Projection from the ventral bed nucleus of the stria ter- serum albumin-conjugated estradiol implants do not promote mating. Neuroen-
minalis to the retrorubral field in rats and the effects of cells in these areas on docrinology 83, 106–116.
mating in male rats versus gerbils. Horm. Behav. 47, 123–138. Huddleston, G.G., Paisley, J.C., Graham, S., Grober, M.S., Clancy, A.N., 2007.
Floody, O.R., Pfaff, D.W., Lewis, C.D., 1977. Communication among hamsters by Implants of estradiol conjugated to bovine serum albumin in the male rat medial
high-frequency acoustic signals: ii. Determinants of calling by females and males. preoptic area promote copulatory behavior. Neuroendocrinology 86, 249–259.
J. Comp. Physiol. Psychol. 91, 807. Hull, E.M., Dominguez, J.M., 2007. Sexual behavior in male rodents. Horm. Behav. 52,
Gan, E.H., Quinton, R., 2010. Physiological significance of the rhythmic secretion of 45–55.
hypothalamic and pituitary hormones. Prog. Brain Res. 181, 111–126. Hurst, J.L., 1990. Urine marking in populations of wild house mice Mus
Gandelman, R., 1983. Gonadal hormones and sensory function. Neurosci. Biobehav. domesticus rutty. II. Communication between the sexes. Anim. Behav. 40,
Rev. 7, 1–17. 233–243.
340 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior

Hurtazo, H.A., Paredes, R.G., 2005. Olfactory preference and fos expression in the Kondo, Y., Tomihara, K., Sakuma, Y., 1999. Sensory requirements for noncontact
accessory olfactory system of male rats with bilateral lesions of the medial preoptic penile erection in the rat. Behav. Neurosci. 113, 1062–1070.
area/anterior hypothalamus. Neuroscience 135, 1035–1044. Lanuza, E., Novejarque, A., Martinez-Ricos, J., Martinez-Hernandez, J., Agustin-
Hurtazo, H.A., Paredes, R.G., Agmo, A., 2008. Inactivation of the medial preoptic area/ Pavon, C., Martinez-Garcia, F., 2008. Sexual pheromones and the evolution of
anterior hypothalamus by lidocaine reduces male sexual behavior and sexual the reward system of the brain: the chemosensory function of the amygdala. Brain
incentive motivation in male rats. Neuroscience 152, 331–337. Res. Bull. 75, 460–466.
Johnson, R.T., Breedlove, S.M., Jordan, C.L., 2008. Sex differences and laterality in Larsson, K., Sodersten, P., Beyer, C., 1973a. Induction of male sexual behaviour by
astrocyte number and complexity in the adult rat medial amygdala. J. Comp. oestradiol benzoate in combination with dihydrotestosterone. J. Endocrinol. 57,
Neurol. 511, 599–609. 563–564.
Johnston, R.E., 1974. Sexual attraction function of golden hamster vaginal secretion. Larsson, K., Sodersten, P., Beyer, C., 1973b. Sexual behavior in male rats treated with
Behav. Biol. 12, 111–117. estrogen in combination with dihydrotestosterone. Horm. Behav. 4, 289–299.
Johnston, R.E., 1975. Sexual excitation function of hamster vaginal secretion. Anim. Larsson, K., Sodersten, P., Beyer, C., Morali, G., Perez-Palacios, G., 1976. Effects of
Learn. Behav. 3, 161–166. estrone, estradiol and estriol combined with dihydrotestosterone on mounting and
Johnston, R.E., 1977. The causation of two scent-marking behaviour patterns in lordosis behavior in castrated male rats. Horm. Behav. 7, 379–390.
female hamsters (Mesocricetus auratus). Anim. Behav. 25, 317–327. LeDoux, J., 2007. The amygdala. Curr. Biol. 17, R868–R874.
Johnston, R.E., 1979. Olfactory preferences, scent marking, and “proceptivity” in LeDoux, J., 2012. Rethinking the emotional brain. Neuron 73, 653–676.
female hamsters. Horm. Behav. 13, 21–39. Lee, H., Kim, D.W., Remedios, R., Anthony, T.E., Chang, A., Madisen, L., Zeng, H.,
Johnston, R.E., 1986. Effects of female odors on the sexual behavior of male Anderson, D.J., 2014. Scalable control of mounting and attack by esr1þ neurons
hamsters. Behav. Neural Biol. 46, 168–188. in the ventromedial hypothalamus. Nature 509, 627–632.
Johnston, R.E., 2008. Individual odors and social communication: individual recogni- Lehman, M.N., Winans, S.S., Powers, J.B., 1980. Medial nucleus of the amygdala
tion, kin recognition, and scent over-marking. In: Brockmann, H.J., Roper, T.J., mediates chemosensory control of male hamster sexual behavior. Science 210,
Naguib, M., Wynne-Edwards, K.E., Barnard, C., John, C.M. (Eds.), Advances in the 557–560.
Study of Behavior. Academic Press. Lin, D., Boyle, M.P., Dollar, P., Lee, H., Lein, E.S., Perona, P., Anderson, D.J., 2011.
Johnston, R.E., Brenner, D., 1982. Species-specificity of scent marking in hamsters. Functional identification of an aggression locus in the mouse hypothalamus. Nature
Behav. Neural Biol. 35, 46–55. 470, 221–226.
Johnston, R.E., Kwan, M., 1984. Vaginal scent marking: effects on ultrasonic calling Lisk, R.D., 1967. Neural localization for androgen activation of copulatory behavior in
and attraction of male golden hamsters. Behav. Neural Biol. 42, 158–168. the male rat. Endocrinology 80, 754–761.
Kaba, H., Hayashi, Y., Higuchi, T., Nakanishi, S., 1994. Induction of an olfactory Lisk, R.D., Bezier, J.L., 1980. Intrahypothalamic hormone implantation and activation
memory by the activation of a metabotropic glutamate receptor. Science 265, of sexual behavior in the male hamster. Neuroendocrinology 30, 220–227.
262–264. Liu, Y.C., Salamone, J.D., Sachs, B.D., 1997. Lesions in medial preoptic area and bed
Kaba, H., Rosser, A., Keverne, B., 1989. Neural basis of olfactory memory in the nucleus of stria terminalis: differential effects on copulatory behavior and
context of pregnancy block. Neuroscience 32, 657–662. noncontact erection in male rats. J. Neurosci. 17, 5245–5253.
Kairys, D.J., Magalhaes, H., Floody, O.R., 1980. Olfactory bulbectomy depresses Lloyd-Thomas, A., Keverne, E.B., 1982. Role of the brain and accessory olfactory
ultrasound production and scent marking by female hamsters. Physiol. Behav. 25, system in the block to pregnancy in mice. Neuroscience 7, 907–913.
143–146. Lukas, M., Toth, I., Veenema, A.H., Neumann, I.D., 2013. Oxytocin mediates rodent
Kang, N., Baum, M.J., Cherry, J.A., 2009. A direct main olfactory bulb projection to social memory within the lateral septum and the medial amygdala depending on
the ‘vomeronasal’ amygdala in female mice selectively responds to volatile pher- the relevance of the social stimulus: male juvenile versus female adult conspecifics.
omones from males. Eur. J. Neurosci. 29, 624–634. Psychoneuroendocrinology 38, 916–926.
Kang, N., McCarthy, E.A., Cherry, J.A., Baum, M.J., 2011. A sex comparison of the MacLean, P.D., 1990. The Triune Brain in Evolution : Role in Paleocerebral Functions.
anatomy and function of the main olfactory bulb-medial amygdala projection in Plenum Press, New York.
mice. Neuroscience 172, 196–204. Maggi, A., Perez, J., 1985. Role of female gonadal hormones in the cns: clinical and
Kauffman, A., Rissman, E., 2006. Neuroendocrine control of mating-induced ovulation. experimental aspects. Life Sci. 37, 893–906.
In: Neill, J.D., Plant, T.M., Pfaff, D.W., Challis, J.R.G., de Kretser, D.M., Malsbury, C.W., McKay, K., 1989. Sex difference in the substance p-immunoreactive
Richards, J.S., Wassarman, P.M. (Eds.), Knobil and Neill’s Physiology of Repro- innervation of the medial nucleus of the amygdala. Brain Res. Bull. 23, 561–567.
duction, 3rd ed. Elsevier. Mani, S.K., Oyola, M.G., 2012. Progesterone signaling mechanisms in brain and
Kelche, C., Aron, C., 1984. Olfactory cues and accessory olfactory bulb lesion: effects behavior. Front. Endocrinol. (Lausanne) 3, 7.
on sexual behavior in the cyclic female rat. Physiol. Behav. 33, 45–48. Maras, P.M., Petrulis, A., 2006. Chemosensory and steroid-responsive regions of the
Keller, M., Douhard, Q., Baum, M.J., Bakker, J., 2006a. Destruction of the main medial amygdala regulate distinct aspects of opposite-sex odor preference in male
olfactory epithelium reduces female sexual behavior and olfactory investigation in syrian hamsters. Eur. J. Neurosci. 24, 3541–3552.
female mice. Chem. Senses 31, 315–323. Maras, P.M., Petrulis, A., 2010a. Anatomical connections between the anterior and
Keller, M., Pierman, S., Douhard, Q., Baum, M.J., Bakker, J., 2006b. The vomeronasal posterodorsal sub-regions of the medial amygdala: integration of odor and hormone
organ is required for the expression of lordosis behaviour, but not sex discrimi- signals. Neuroscience 170, 610–622.
nation in female mice. Eur. J. Neurosci. 23, 521–530. Maras, P.M., Petrulis, A., 2010b. The anterior medial amygdala transmits sexual odor
Kelliher, K., Baum, M., 2002. Effect of sex steroids and coital experience on information to the posterior medial amygdala and related forebrain nuclei. Eur. J.
ferrets’ preference for the smell, sight and sound of conspecifics. Physiol. Neurosci. 32, 469–482.
Behav. 76, 1–7. Maras, P.M., Petrulis, A., 2010c. Lesions that functionally disconnect the anterior and
Keshavarzi, S., Sullivan, R.K., Ianno, D.J., Sah, P., 2014. Functional properties and posterodorsal sub-regions of the medial amygdala eliminate opposite-sex odor
projections of neurons in the medial amygdala. J. Neurosci 34, 8699–8715. preference in male syrian hamsters (Mesocricetus auratus). Neuroscience 165,
Kollack-Walker, S., Newman, S.W., 1997. Mating-induced expression of c-fos in the 1052–1062.
male syrian hamster brain: role of experience, pheromones, and ejaculations. Markham, C.M., Huhman, K.L., 2008. Is the medial amygdala part of the neural circuit
J. Neurobiol. 32, 481–501. modulating conditioned defeat in syrian hamsters? Learn. Mem. 15, 6–12.
Kondo, Y., Arai, Y., 1995. Functional association between the medial amygdala and the Markham, J.A., 2012. Sex steroids and schizophrenia. Rev. Endocr. Metab. Disord.
medial preoptic area in regulation of mating behavior in the male rat. Physiol. 13, 187–207.
Behav. 57, 69–73. Martinez, L.A., Albers, H.E., Petrulis, A., 2010. Blocking oxytocin receptors inhibits
Kondo, Y., Sachs, B.D., 2002. Disparate effects of small medial amygdala lesions on vaginal marking to male odors in female syrian hamsters. Physiol. Behav. 101,
noncontact erection, copulation, and partner preference. Physiol. Behav. 76, 443–447. 685–692.
Kondo, Y., Sachs, B.D., Sakuma, Y., 1998. Importance of the medial amygdala in rat Martinez, L.A., Petrulis, A., 2011. The bed nucleus of the stria terminalis is critical for
penile erection evoked by remote stimuli from estrous females. Behav. Brain Res. sexual solicitation, but not for opposite-sex odor preference, in female syrian
91, 215–222. hamsters. Horm. Behav. 60, 651–659.
Kondo, Y., Sakuma, Y., 2005. The medial amygdala controls the coital access of Martinez, L.A., Petrulis, A., 2013. The medial preoptic area is necessary for sexual
female rats: a possible involvement of emotional responsiveness. Jpn. J. Physiol. odor preference, but not sexual solicitation, in female syrian hamsters. Horm.
55, 345–353. Behav. 63, 606–614.
The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 341

Martinez-Garcia, F., Martinez-Marcos, A., Lanuza, E., 2002. The pallial amygdala of Pardo-Bellver, C., Cadiz-Moretti, B., Novejarque, A., Martinez-Garcia, F., Lanuza, E.,
amniote vertebrates: evolution of the concept, evolution of the structure. Brain Res. 2012. Differential efferent projections of the anterior, posteroventral, and poster-
Bull. 57, 463–469. odorsal subdivisions of the medial amygdala in mice. Front Neuroanat 6, 33.
Matochik, J.A., Sipos, M.L., Nyby, J.G., Barfield, R.J., 1994. Intracranial androgenic Paisley, J.C., Huddleston, G.G., Carruth, L.L., Petrulis, A., Grober, M.S., Clancy, A.N.,
activation of male-typical behaviors in house mice: motivation versus perfor- 2012. Sexual responses of the male rat medial preoptic area and medial amygdala
mance. Behav. Brain Res. 60, 141–149. to estrogen i: site specific suppression of estrogen receptor alpha. Horm. Behav.
McGinnis, M.Y., Montana, R.C., Lumia, A.R., 2002. Effects of hydroxyflutamide in the 62, 50–57.
medial preoptic area or lateral septum on reproductive behaviors in male rats. Pankevich, D.E., Cherry, J.A., Baum, M.J., 2006. Accessory olfactory neural fos
Brain Res. Bull. 59, 227–234. responses to a conditioned environment are blocked in male mice by vomeronasal
McGinnis, M.Y., Williams, G.W., Lumia, A.R., 1996. Inhibition of male sex behavior by organ removal. Physiol. Behav. 87, 781–788.
androgen receptor blockade in preoptic area or hypothalamus, but not amygdala Papez, J.W., 1937. A proposed mechanism of emotion. Archiv. Neurol. Psychiatry 38,
or septum. Physiol. Behav. 60, 783–789. 25–743.
McGregor, A., Herbert, J., 1992a. Differential effects of excitotoxic basolateral and Petrulis, A., 2009. Neural mechanisms of individual and sexual recognition in syrian
corticomedial lesions of the amygdala on the behavioural and endocrine hamsters (Mesocricetus auratus). Behav. Brain Res. 200, 260–267.
responses to either sexual or aggression-promoting stimuli in the male rat. Petrulis, A., 2013. Chemosignals, hormones and mammalian reproduction. Horm.
Brain Res. 574, 9–20. Behav. 63, 723–741.
McGregor, A., Herbert, J., 1992b. The effects of beta-endorphin infusions into the Petrulis, A., Johnston, R.E., 1997. Causes of scent marking in female golden hamsters
amygdala on visual and olfactory sensory processing during sexual behaviour in the (Mesocricetus auratus): specific signals or classes of information? J. Comp. Psy-
male rat. Neuroscience 46, 173–179. chol. 111, 25–36.
McGregor, A., Herbert, J., 1992c. Specific effects of beta-endorphin infused into the Petrulis, A., Johnston, R.E., 1999. Lesions centered on the medial amygdala impair
amygdala on sexual behaviour in the male rat. Neuroscience 46, 165–172. scent-marking and sex-odor recognition but spare discrimination of individual odors
Medina, L., Bupesh, M., Abellan, A., 2011. Contribution of genoarchitecture to in female golden hamsters. Behav. Neurosci. 113, 345–357.
understanding forebrain evolution and development, with particular emphasis on Pfaff, D.W., 1980. Estrogens and Brain Function : Neural Analysis of a Hormone-
the amygdala. Brain Behav. Evol. 78, 216–236. controlled Mammalian Reporductive Behavior. Springer-Verlag, New York.
Melrose, D.R., Reed, H.C., Patterson, R.L., 1971. Androgen steroids associated with Pfaff, D.W., Kow, L.M., Loose, M.D., Flanagan-Cato, L.M., 2008. Reverse engineering
boar odour as an aid to the detection of oestrus in pig artificial insemination. Br. the lordosis behavior circuit. Horm. Behav. 54, 347–354.
Vet. J. 127, 497–502. Piekarski, D.J., Routman, D.M., Schoomer, E.E., Driscoll, J.R., Park, J.H., Butler, M.P.,
Meredith, M., Westberry, J.M., 2004. Distinctive responses in the medial amygdala to Zucker, I., 2009. Infrequent low dose testosterone treatment maintains male sexual
same-species and different-species pheromones. J. Neurosci. 24, 5719–5725. behavior in syrian hamsters. Horm. Behav. 55, 182–189.
Micevych, P.E., Abelson, L., Fok, H., Ulibarri, C., Priest, C.A., 1994. Gonadal steroid Pihlajamaa, P., Sahu, B., Janne, O.A., 2015. Determinants of receptor- and tissue-
control of preprocholecystokinin mRNA expression in the limbic-hypothalamic specific actions in androgen signaling. Endocr. Rev. 36, 357–384.
circuit: comparison of adult with neonatal steroid treatments. J. Neurosci. Res. Portillo, W., Diaz, N.F., Cabrera, E.A., Fernandez-Guasti, A., Paredes, R.G., 2006.
38, 386–398. Comparative analysis of immunoreactive cells for androgen receptors and oes-
Morin, L.P., Wood, R.I., 2001. A Stereotaxic Atlas of the Golden Hamster Brain. trogen receptor alpha in copulating and non-copulating male rats. J. Neuro-
Academic Press, San Diego, CA; London. endocrinol 18, 168–176.
Morris, J.A., Jordan, C.L., Breedlove, S.M., 2008. Sexual dimorphism in neuronal Powers, J.B., Newman, S.W., Bergondy, M.L., 1987. MPOA and BNST lesions in male
number of the posterodorsal medial amygdala is independent of circulating syrian hamsters: differential effects on copulatory and chemoinvestigatory behav-
androgens and regional volume in adult rats. J. Comp. Neurol. 506, 851–859. iors. Behav. Brain Res. 23, 181–195.
Murphy, M.R., 1973. Effects of female hamster vaginal discharge on the behavior of Rajendren, G., Dudley, C.A., Moss, R.L., 1990. Role of the vomeronasal organ in the
male hamsters. Behav. Biol. 9, 367–375. male-induced enhancement of sexual receptivity in female rats. Neuroendocri-
Murphy, M.R., Schneider, G.E., 1970. Olfactory bulb removal eliminates mating nology 52, 368–372.
behavior in the male golden hamster. Science 167, 302–304. Rajendren, G., Moss, R.L., 1993. The role of the medial nucleus of amygdala in the
Musatov, S., Chen, W., Pfaff, D.W., Kaplitt, M.G., Ogawa, S., 2006. RNAi-mediated mating-induced enhancement of lordosis in female rats: the interaction with
silencing of estrogen receptor {alpha} in the ventromedial nucleus of hypothal- luteinizing hormone-releasing hormone neuronal system. Brain Res. 617, 81–86.
amus abolishes female sexual behaviors. Proc. Natl. Acad. Sci. U.S.A. 103, Rajendren, G.V., Moss, R.L., 1994. Vomeronasal organ-mediated induction of fos in
10456–10460. the central accessory olfactory pathways in repetitively mated female rats. Brain
Newman, S.W., 1999. The medial extended amygdala in male reproductive behavior. A Res. Bull. 34, 53–59.
node in the mammalian social behavior network. Ann. N.Y. Acad. Sci. 877, Rangel, A., Munoz, C., Ocampo, M.V., Quintero, C., Escobar, M., Botero, S., Marin, C.,
242–257. Jaramillo, L.E., Sanchez, R., Rodriguez-Losada, J., Ospina-Duque, J., Palacio, C.,
Nolte, C.M., Meredith, M., 2005. mGluR2 activation of medial amygdala input impairs CA, J., Valencia, A.V., Aguirre-Acevedo, D.C., Garcia, J., 2015. Neurocognitive
vomeronasal organ-mediated behavior. Physiol. Behav. 86, 314–323. subtypes of schizophrenia. Actas Esp. Psiquiatr. 43, 80–90.
Northcutt, K.V., Wang, Z.X., Lonstein, J.S., 2007. Sex and species differences in Rasia, A.A., Londero, R.G., Achaval, M., 1999. Effects of gonadal hormones on the
tyrosine hydroxylase-synthesizing cells of the rodent olfactory extended amygdala. morphology of neurons from the medial amygdaloid nucleus of rats. Brain Res.
J. Comp. Neurol. 500, 103–115. Bull. 48, 173–183.
Nyby, J., Matochik, J.A., Barfield, R.J., 1992. Intracranial androgenic and estrogenic Rasia, A.A., Xavier, L.L., dos Santos, P., Gehlen, G., Achaval, M., 2002. Glial fibrillary
stimulation of male-typical behaviors in house mice (Mus domesticus). Horm. acidic protein immunodetection and immunoreactivity in the anterior and posterior
Behav. 26, 24–45. medial amygdala of male and female rats. Brain Res. Bull. 58, 67–75.
Nyby, J., Wysocki, C.J., Whitney, G., Dizinno, G., 1977. Pheromonal regulation of Rasia-Filho, A.A., Peres, T.M., Cubilla-Gutierrez, F.H., Lucion, A.B., 1991. Effect of
male mouse ultrasonic courtship (Mus musculus). Anim. Behav. 25, 333–341. estradiol implanted in the corticomedial amygdala on the sexual behavior of cas-
O’Connell, L.A., Hofmann, H.A., 2011. The vertebrate mesolimbic reward system and social trated male rats. Braz. J. Med. Biol. Res. 24, 1041–1049.
behavior network: a comparative synthesis. J. Comp. Neurol. 519, 3599–3639. Reed, H.C., Melrose, D.R., Patterson, R.L., 1974. Androgen steroids as an aid to the
O’Connell, L.A., Hofmann, H.A., 2012. Evolution of a vertebrate social decision-making detection of oestrus in pig artificial insemination. Br. Vet. J. 130, 61–67.
network. Science 336, 1154–1157. Rich, T.J., Hurst, J.L., 1999. The competing countermarks hypothesis: reliable
O’Connell, R.J., Meredith, M., 1984. Effects of volatile and nonvolatile chemical signals assessment of competitive ability by potential mates. Anim. Behav. 58,
on male sex behaviors mediated by the main and accessory olfactory systems. 1027–1037.
Behav. Neurosci. 98, 1083–1093. Rissman, E.F., 1989. Male-related chemical cues promote sexual receptivity in the
Olazabal, D.E., Pereira, M., Agrati, D., Ferreira, A., Fleming, A.S., Gonzalez- female musk shrew. Behav. Neural Biol. 51, 114–120.
Mariscal, G., Levy, F., Lucion, A.B., Morrell, J.I., Numan, M., Uriarte, N., 2013. Robarts, D.W., Baum, M.J., 2007. Ventromedial hypothalamic nucleus lesions disrupt
Flexibility and adaptation of the neural substrate that supports maternal behavior in olfactory mate recognition and receptivity in female ferrets. Horm. Behav. 51,
mammals. Neurosci. Biobehav. Rev. 37, 1875–1892. 104–113.
Ottaviano, G., Frasson, G., Nardello, E., Martini, A., 2015. Olfaction deterioration in Rolls, E.T., 2015. Limbic systems for emotion and for memory, but no single limbic
cognitive disorders in the elderly. Aging Clin. Exp. Res. 28 (1). system. Cortex 62, 119–157.
342 The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior

Rowe, F.A., Edwards, D.A., 1972. Olfactory bulb removal: influences on the mating Torres, B., Santos, R.L., Sousa, M.F., Simoes Neto, J.P., Nogueira, M.M., Belfort, T.T.,
behavior of male mice. Physiol. Behav. 8, 37–41. Dias, R., Dourado, M.C., 2015. Facial expression recognition in Alzheimer’s
Russell, N.V., Ogaga-Mgbonyebi, E.V., Habteab, B., Dunigan, A.I., Tesfay, M.A., disease: a longitudinal study. Arq. Neuro Psiquiatr. 73, 383–389.
Clancy, A.N., 2012. Sexual responses of the male rat medial preoptic area and Triemstra, J.L., Nagatani, S., Wood, R.I., 2005. Chemosensory cues are essential for
medial amygdala to estrogen ii: site specific effects of selective estrogenic drugs. mating-induced dopamine release in MPOA of male syrian hamsters. Neuro-
Horm. Behav. 62, 58–66. psychopharmacology 30, 1436–1442.
Sachs, B.D., Akasofu, K., Citron, J.H., Daniels, S.B., Natoli, J.H., 1994. Noncontact Unger, E.K., Burke Jr., K.J., Yang, C.F., Bender, K.J., Fuller, P.M., Shah, N.M., 2015.
stimulation from estrous females evokes penile erection in rats. Physiol. Behav. 55, Medial amygdalar aromatase neurons regulate aggression in both sexes. Cell Rep.
1073–1079. 10, 453–462.
Samuelsen, C.L., Meredith, M., 2009. Categorization of biologically relevant chemical Van den Stock, J., De Winter, F.L., Ahmad, R., Sunaert, S., Van Laere, K.,
signals in the medial amygdala. Brain Res. 1263, 33–42. Vandenberghe, W., Vandenbulcke, M., 2015. Functional brain changes under-
Samuelsen, C.L., Meredith, M., 2011. Oxytocin antagonist disrupts male mouse medial lying irritability in premanifest Huntington’s disease. Hum. Brain Mapp. 36,
amygdala response to chemical-communication signals. Neuroscience 180, 2681–2690.
96–104. Veyrac, A., Wang, G., Baum, M.J., Bakker, J., 2011. The main and accessory olfactory
Sano, K., Tsuda, M.C., Musatov, S., Sakamoto, T., Ogawa, S., 2013. Differential systems of female mice are activated differentially by dominant versus subordinate
effects of site-specific knockdown of estrogen receptor alpha in the medial male urinary odors. Brain Res. 1402, 20–29.
amygdala, medial pre-optic area, and ventromedial nucleus of the hypothalamus on Wagner, C.K., Morrell, J.I., 1997. Neuroanatomical distribution of aromatase mRNA in
sexual and aggressive behavior of male mice. Eur. J. Neurosci. 37, 1308–1319. the rat brain: indications of regional regulation. J. Steroid Biochem. Mol. Biol. 61,
Sarkey, S., Azcoitia, I., Garcia-Segura, L.M., Garcia-Ovejero, D., DonCarlos, L.L., 307–314.
2008. Classical androgen receptors in non-classical sites in the brain. Horm. Wang, Y., He, Z., Zhao, C., Li, L., 2013. Medial amygdala lesions modify aggressive
Behav. 53, 753–764. behavior and immediate early gene expression in oxytocin and vasopressin
Sheehan, T., Paul, M., Amaral, E., Numan, M.J., Numan, M., 2001. Evidence neurons during intermale exposure. Behav. Brain Res. 245, 42–49.
that the medial amygdala projects to the anterior/ventromedial hypothalamic Wang, Y., Zhao, S., Wu, Z., Feng, Y., Zhao, C., Zhang, C., 2015. Oxytocin in the
nuclei to inhibit maternal behavior in rats. Neuroscience 106, 341–356. regulation of social behaviours in medial amygdala-lesioned mice via the inhibition
Sheridan, P.J., 1979. The nucleus interstitialis striae terminalis and the nucleus of the extracellular signal-regulated kinase signalling pathway. Clin. Exp. Phar-
amygdaloideus medialis: prime targets for androgen in the rat forebrain. Endo- macol. Physiol. 42, 465–474.
crinology 104, 130–136. White, N.R., Colona, L.C., Barfield, R.J., 1991. Sensory cues that elicit ultrasonic
Sheridan, P.J., 1983. Androgen receptors in the brain: what are we measuring? vocalizations in female rats (Rattus norvegicus). Behav. Neural Biol. 55, 154–165.
Endocr. Rev. 4, 171–178. Whitney, J.F., 1986. Effect of medial preoptic lesions on sexual behavior of female rats
Sheridan, P.J., 1991. Can a single androgen receptor fill the bill? Mol. Cell. Endocrinol. is determined by test situation. Behav. Neurosci. 100, 230–235.
76, C39–C45. Williams, G.W., McGinnis, M.Y., Lumia, A.R., 1992. The effects of olfactory bulbec-
Simerly, R.B., Chang, C., Muramatsu, M., Swanson, L.W., 1990. Distribution of tomy and chronic psychosocial stress on serum glucocorticoids and sexual
androgen and estrogen receptor mRNA-containing cells in the rat brain: an in situ behavior in female rats. Physiol. Behav. 52, 755–760.
hybridization study. J. Comp. Neurol. 294, 76–95. Wilson, M.A., Mascagni, F., McDonald, A.J., 2002. Sex differences in delta opioid
Singer, A.G., Clancy, A.N., Macrides, F., Agosta, W.C., 1984. Chemical studies of receptor immunoreactivity in rat medial amygdala. Neurosci. Lett. 328, 160–164.
hamster vaginal discharge: male behavioral responses to a high molecular weight Wilson, R.S., Yu, L., Schneider, J.A., Arnold, S.E., Buchman, A.S., Bennett, D.A.,
fraction require physical contact. Physiol. Behav. 33, 645–651. 2011. Lewy bodies and olfactory dysfunction in old age. Chem. Senses 36,
Sipos, M.L., Nyby, J.G., 1996. Concurrent androgenic stimulation of the ventral 367–373.
tegmental area and medial preoptic area: synergistic effects on male-typical Wood, R.I., 1996. Estradiol, but not dihydrotestosterone, in the medial amygdala
reproductive behaviors in house mice. Brain Res. 729, 29–44. facilitates male hamster sex behavior. Physiol. Behav. 59, 833–841.
Sipos, M.L., Nyby, J.G., 1998. Intracranial androgenic activation of male-typical Wood, R.I., Brabec, R.K., Swann, J.M., Newman, S.W., 1992. Androgen and estrogen
behaviours in house mice: concurrent stimulation of the medial preoptic area concentrating neurons in chemosensory pathways of the male syrian hamster
and medial nucleus of the amygdala. J. Neuroendocrinol. 10, 577–586. brain. Brain Res. 596, 89–98.
Smock, T., Albeck, D., Stark, P., 1998. A peptidergic basis for sexual behavior in Wood, R.I., Coolen, L.M., 1997. Integration of chemosensory and hormonal cues is
mammals. Prog. Brain Res. 119, 467–481. essential for sexual behaviour in the male syrian hamster: role of the medial
Spiteri, T., Musatov, S., Ogawa, S., Ribeiro, A., Pfaff, D.W., Agmo, A., 2010. Estrogen- amygdaloid nucleus. Neuroscience 78, 1027–1035.
induced sexual incentive motivation, proceptivity and receptivity depend on Wood, R.I., Newman, S.W., 1995a. Hormonal influence on neurons of the mating
a functional estrogen receptor alpha in the ventromedial nucleus of the hypo- behavior pathway in male hamsters. In: Neurobiological Effects of Sex Steroid
thalamus but not in the amygdala. Neuroendocrinology 91, 142–154. Hormones.
Spiteri, T., Ogawa, S., Musatov, S., Pfaff, D.W., Agmo, A., 2012. The role of the Wood, R.I., Newman, S.W., 1995b. Integration of chemosensory and hormonal cues is
estrogen receptor alpha in the medial preoptic area in sexual incentive motivation, essential for mating in the male syrian hamster. J. Neurosci. 15, 7261–7269.
proceptivity and receptivity, anxiety, and wheel running in female rats. Behav. Brain Wood, R.I., Newman, S.W., 1995c. The medial amygdaloid nucleus and medial
Res. 230, 11–20. preoptic area mediate steroidal control of sexual behavior in the male syrian
Sternson, S.M., 2013. Hypothalamic survival circuits: blueprints for purposive hamster. Horm. Behav. 29, 338–353.
behaviors. Neuron 77, 810–824. Wood, R.I., Williams, S.J., 2001. Steroidal control of male hamster sexual behavior in
Swann, J., Fabre-Nys, C., Barton, R., 2009. Hormonal and pheromonal modu- me and MPOA: effects of androgen dose and tamoxifen. Physiol. Behav. 72,
lation of the extended amygdala: implications for social behaviour. In: 727–733.
Pfaff, D., Arnold, A., Fahrbach, S., Etgen, A., Rubin, R. (Eds.), Hormones, Woodley, S.K., Baum, M.J., 2004. Differential activation of glomeruli in the ferret’s
Brain and Behavior. main olfactory bulb by anal scent gland odours from males and females: an early
Swann, J., Fiber, J.M., 1997. Sex differences in function of a pheromonally stimulated step in mate identification. Eur. J. Neurosci. 20, 1025–1032.
pathway: role of steroids and the main olfactory system. Brain Res. Bull. 44, Wysocki, C.J., Nyby, J., Whitney, G., Beauchamp, G.K., Katz, Y., 1982. The vomer-
409–413. onasal organ: primary role in mouse chemosensory gender recognition. Physiol.
Swann, J., Rahaman, F., Bijak, T., Fiber, J., 2001. The main olfactory system Behav. 29, 315–327.
mediates pheromone-induced fos expression in the extended amygdala and pre- Xiao, K., Kondo, Y., Sakuma, Y., 2005. Differential regulation of female rat olfactory
optic area of the male syrian hamster. Neuroscience 105, 695–706. preference and copulatory pacing by the lateral septum and medial preoptic area.
Takahashi, L.K., Lisk, R.D., Burnett 2nd, A.L., 1985. Dual estradiol action in dien- Neuroendocrinology 81, 56–62.
cephalon and the regulation of sociosexual behavior in female golden hamsters. Xydakis, M.S., Mulligan, L.P., Smith, A.B., Olsen, C.H., Lyon, D.M., Belluscio, L.,
Brain Res. 359, 194–207. 2015. Olfactory impairment and traumatic brain injury in blast-injured combat
Thompson, R.R., Goodson, J.L., Ruscio, M.G., Adkins-Regan, E., 1998. Role of the troops: a cohort study. Neurology 84, 1559–1567.
archistriatal nucleus taeniae in the sexual behavior of male japanese quail Yahr, P., Finn, P.D., Hoffman, N.W., Sayag, N., 1994. Sexually dimorphic cell groups in
(Coturnix japonica): a comparison of function with the medial nucleus of the the medial preoptic area that are essential for male sex behavior and the neural
amygdala in mammals. Brain Behav. Evol. 51, 215–229. pathways needed for their effects. Psychoneuroendocrinology 19, 463–470.
The Medial Amygdala, Hormones, Pheromones, Social Behavior Network, and Mating Behavior 343

Yahr, P., Gregory, J.E., 1993. The medial and lateral cell groups of the sexually Yildirim, B.O., Derksen, J.J., 2012. A review on the relationship between testosterone
dimorphic area of the gerbil hypothalamus are essential for male sex behavior and and the interpersonal/affective facet of psychopathy. Psychiatry Res. 197, 181–198.
act via separate pathways. Brain Res. 631, 287–296. Yoshimura, N., Kawamura, M., Masaoka, Y., Homma, I., 2005. The amygdala of
Yang, C.F., Chiang, M.C., Gray, D.C., Prabhakaran, M., Alvarado, M., Juntti, S.A., patients with parkinson’s disease is silent in response to fearful facial expressions.
Unger, E.K., Wells, J.A., Shah, N.M., 2013. Sexually dimorphic neurons in the Neuroscience 131, 523–534.
ventromedial hypothalamus govern mating in both sexes and aggression in males. Zehr, J.L., Todd, B.J., Schulz, K.M., McCarthy, M.M., Sisk, C.L., 2006. Dendritic
Cell 153, 896–909. pruning of the medial amygdala during pubertal development of the male syrian
Yang, L.Y., Clements, L.G., 2000. MPOA lesions affect female pacing of copulation in hamster. J. Neurobiol. 66, 578–590.
rats. Behav. Neurosci. 114, 1191–1202.
This page intentionally left blank
1.12 Circadian Regulation of Endocrine Functions
Matthew P Butler, Oregon Health & Science University, Portland, OR, USA
Ilia Karatsoreos, Washington State University, Pullman, WA, USA
Lance J Kriegsfeld, University of California, Berkeley, CA, USA
Rae Silver, Columbia University, New York, NY, USA; Barnard College, New York, NY, USA; and College of Physicians and
Surgeons, New York, NY, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by M.P. Butler, L.J. Kriegsfeld, R. Silver, volume 1, pp. 473–507, Ó 2009, Elsevier Inc.

1.12.1 Introduction to Circadian Aspects of Endocrine Function 345


1.12.2 The Circadian Timing System 346
1.12.2.1 Entrainment through a Hierarchy of Clocks 350
1.12.3 Circadian Regulation of the Endocrine System 353
1.12.4 Circadian Regulation of Hypothalamo–Pituitary–Adrenal Axis 353
1.12.5 Circadian Rhythmicity in the Hypothalamic–Pituitary–Gonadal Axis 355
1.12.6 Circadian Regulation of Melatonin 359
1.12.7 Circadian Dysfunction and Chronotherapy 360
Acknowledgments 361
References 362

1.12.1 Introduction to Circadian Aspects of While this signal occurs every day, it is effective at stimulating
Endocrine Function gonadotropin-releasing hormone (GnRH) neurons to produce
the LH surge only when estrogen levels reach a critical
Overview of circadian timing. The significance of circadian threshold. The occurrence of this daily stimulatory suprachias-
rhythms to hormones and behavior is twofold. First, time of matic nucleus (SCN) signal can be demonstrated by implant-
day must be taken into account in considering ‘normal’ ing female rodents with capsules that produce high
hormone levels as most hormones are secreted with circadian concentrations of circulating estrogens, in which case an LH
periodicity. These rhythms are determined by the interaction surge occurs daily (Legan et al., 1975; Christian et al., 2005).
between the brain’s master clock which is typically synchro- Clearly, this work points to a role for neural mechanisms in
nized to the external light–dark cycle and hormone secreting timing daily surges in hormone secretion. The principle that
tissues that are also rhythmic. Second, the physiological effects time of day of exposure to a hormone has marked effects on
of a hormone depend on the timing of its secretion and sensi- the physiological response has also been recognized (Figure 1).
tivity of the target tissue to its effects. Cellular actions are deter- For example, glucocorticoid secretion by the adrenal gland
mined by the concentration of a hormone relative to affinity for in vitro responds differentially to a fixed dose of ACTH depend-
its specific receptor, receptor availability and/or abundance, ing on time of day (Ungar and Halberg, 1962).
and postreceptor signaling factors. Time of day, acting through Since the time of these initial studies, the phenomenology
the circadian timing system, modulates each of these processes. of circadian rhythmicity has exploded, with ubiquitous exam-
The evidence of time of day effects has long been available. Our ples of circadian regulation. We now understand more fully
understanding of the causal mechanisms continues to emerge. how the circadian clocks gate and modulate endocrine
The importance of time of day in the regulation of endo- rhythms. Developments in molecular biology have led to the
crine processes was known well before the biological basis of identification of genes that are gated or modulated by the
the circadian timing system was understood. As an interesting circadian system and canonical circadian ‘clock’ genes that
example, Everett and Sawyer’s (1950) pioneering work on the are part of the cellular timing mechanism. While questions
timing of ovulation was motivated by the question of whether of how the circadian system regulates endocrine secretions
there was any role for the central nervous system in the regula- remain, the nature of the unknowns and the kind of questions
tion of hormone release. Everett and Sawyer knew that the asked and answered has changed dramatically. This is due, in
luteinizing hormone (LH) (Grundschober et al., 2001) surge part, to the ability to more easily and accurately measure circa-
in rats occurs at a particular time of day – termed the critical dian variables over long time intervals making the work of col-
period – on the afternoon of proestrus. They demonstrated lecting time series data less grueling. Technical breakthroughs
that when hypothalamic activity is blocked in proestrus now allow real-time measurement of clock function in vivo and
females by transient barbiturate anesthesia, the LH surge is in vitro making high-throughput screening of gene expression
delayed for a full 24 h rather than for the 2 h of anesthetic seda- possible.
tion, pointing to the participation of a circadian timing system Functions of rhythms: entrainment, internal synchronization,
in generating the LH surge. It is now known that a daily signal and anticipation. The fundamental functions of the circadian
from the brain’s master clock times the preovulatory LH surge. timing system are to ensure the correct temporal staging of

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00012-2 345


346 Circadian Regulation of Endocrine Functions

12 such as stress, jet lag, shift work, and aging. The goal of this
Direct extraction
Incubation without ACTH chapter is to delineate the basic phenomena important in
Incubation with 0.04 I.U.ACTH the field of circadian rhythmicity and to highlight ways in
Incubation with 0.40 I.U.ACTH
Incubation with 4.00 I.U.ACTH which circadian timing impacts the understanding of
10
hormone effects on behavior and physiology. Terminological
Mean of 2 pools notes and definitions esoteric to circadian studies are
Corticosterone (μg/IO adrenals/2 h)

Individual mean provided in Box 1.


8

1.12.2 The Circadian Timing System


6
Overview. The trajectory of chronobiological research has been
‘top-down’ from macroscopic studies of rhythms in animals
and humans to more reductionist levels. An excellent review
4
of the history of research is available for work up to the initial
discovery of clock genes in mammals (Weaver, 1998). Biolog-
ically relevant mathematical models have guided research
2 from early days and play increasingly important roles in
revealing how oscillators are coupled from the neuron to
the tissue level (Bechtel, 2016; Pauls et al., 2016). The enigma
of how isolated and interacting cells measure the passage of
0
24 08
time has been fruitfully addressed at the biochemical,
08 16
Time (clock hours) network, and whole organism levels. The role of transcrip-
tion–translation feedback loops is very well documented
Figure 1 Time of hormone administration determines the amplitude (Albrecht, 2012). More recently, the contribution of
of the rhythmic response. In phenomenological studies done nontranscriptional mechanisms has drawn attention to the
throughout the world, Franz Halberg and his associates monitored multitude of oscillatory mechanisms (Hoyle and O’Neill,
physiological rhythms in numerous variables as part of the ‘neuro- 2015). Importantly, it is now well established that virtually
endocrinologic armamentarium.’ In experimental laboratory work, he
all tissues in the body exhibit circadian rhythms that are
showed a circadian influence on the response of mouse adrenals to
based on a cellular clock and a robustly cycling transcriptome
fixed doses of adrenocorticotropic hormone (ACTH) administered
in vitro. Corticosterone was extracted either directly from adrenal (Zhang et al., 2014). This literature lays the groundwork for
glands or after incubation with several doses of ACTH. Figure from understanding circadian rhythms in endocrine and neuroen-
Ungar, F., Halberg, F., 1962. Circadian rhythm in the in vitro response docrine systems. In the following section, we highlight key
of mouse adrenal to adrenocorticotropic hormone. Science 137, aspects of circadian timing in a ‘bottom-up’ manner, starting
1058–1060. from the cellular basis to the hierarchical control of clocks
throughout the body.
Core clock genes and transcription/translation feedback loops.
numerous processes internal to the body and to enable the Cell-based circadian oscillators had initially been construed
entrainment of these processes to the local environment. as composed of a small number of gene families linked in tran-
This coordination is necessary for optimal regulation of phys- scription–translation feedback loops that complete one cycle in
iology and behavior. Coordination is achieved by a master approximately 24 h (Figure 2). In the primary loop, two basic
clock, located in the SCN, which is synchronized each day to helix-loop-helix transcription factors, CLOCK and BMAL1,
the local photoperiod. The SCN, in turn, sets the phase of dimerize and drive transcription of the Period (Per1, Per2, and
the cellular clocks throughout the body. The output of some Per3) and the Cryptochrome (Cry1 and Cry2) genes by binding
of these downstream systems, such as hormones produced to the E-box motif (CACGTG) in the promoter regions of
by the major glands of the body, in turn, feeds back to provide these genes. The resultant PER and CRY proteins form hetero-
temporal information to the brain clock. Together the brain and homodimers in the cytoplasm of the cell and are then
and bodywide circadian clocks enable the anticipation of regu- translocated back to the nucleus where they inhibit their own
larly recurring daily events and the optimal temporal phasing transcription via direct interactions with the CLOCK:BMAL1
of key events within the body, relative to each other. Notably, protein complex (Zhang and Kay, 2010; Partch et al., 2014;
conditions such as shift work can disrupt the delicate balance Dibner and Schibler, 2015).
of oscillators across the body and impair this coordination In addition to rhythmic negative feedback via PER and CRY,
leading to pathology. there is a rhythm in CLOCK:BMAL1 transactivation based on
Intense interest in circadian chronobiology derives from cycling abundance of BMAL1. These rhythms are generated by
a convergence of practical and medical needs with technolog- a second loop: CLOCK:BMAL1 acts as a positive regulator, driving
ical and theoretical developments. In the practical domain, transcription of two Rev-erb genes and four Ror genes. The protein
the range of medical phenomena that are modulated by the products compete to bind retinoic acid–related orphan receptor
circadian system has expanded to include every organ of the response elements (RORE) in the promoter region of Bmal1,
body, numerous disrupted states such as alcoholism, cardio- with REV-ERBa-inhibiting and RORa-stimulating transcription,
vascular crises, cancer and cell division, and phenomena ultimately leading to rhythms in BMAL1 abundance
Circadian Regulation of Endocrine Functions 347

Box 1 Terminological notes


Amplitude: The strength or magnitude of an oscillation.
Arrhythmia: When a system that is normally rhythmic loses rhythmicity. For example, lesions of the SCN lead to arrhythmia in mice.
Circadian time (CT): Unit of time based on an organism’s free-running period. Typically CT0 is the onset of activity in diurnal organisms and CT12 is the onset of
activity for nocturnal organisms.
Circadian: A cycle or oscillation with a period close to 24h, from circa ¼ about and diem ¼ day. This term is generally used to describe processes that are
endogenously driven and that will persist in the absence of any external cues, such as light.
Clock gene: A gene involved in the basic genetic negative feedback loops underlying circadian rhythms.
Desynchronization: When a rhythm becomes uncoupled from a synchronizing cue or when two or more internal rhythms are no longer coupled.
Diurnal: Used to describe rhythmic phenomena that are not endogenous but rather driven by the external light–dark cycle. Also used to refer to day-active animals.
Endogenous rhythm: Self-sustained rhythm that persists in constant conditions, such as constant darkness.
Entrainment: Synchronization of the circadian system to external cues. The light–dark cycle is the dominant entraining signal for most organisms, but other cues, such
as food, can also entrain organisms.
Free-running: The state of an oscillator running at its own endogenous period, without external perturbations, such as entrainment to a light–dark cycle. The free-
running period of an animal is usually measured in constant darkness.
Nocturnal: Night active.
Pacemaker: Anything that can generate robust, endogenous rhythms and can synchronize other rhythmic entities to its own rhythm. The SCN is the master
pacemaker, responsible for synchronizing all of the body’s rhythms to its own.
Period: The duration of a cycle or oscillation, typically close to 24 h for circadian processes.
Phase: The time at which a cyclic event occurs as measured against a reference time frame, such as time of day. A phase advance causes the event to occur earlier in
the day, while a phase delay causes it to occur later.
Subjective day/night: Under free-running conditions subjective day is the period from CT0 to CT12, when diurnal organisms would typically be active. Subjective night
is the period from CT12 to CT0 when nocturnal organisms would typically be active.
Zeitgeber: Any cue or signal that can synchronize or entrain a rhythm. For example light is a very powerful zeitgeber capable of entraining the body clock to
environmental light/dark cycles. Zeitgeber time (ZT) is a unit of time measured from the onset of a zeitgeber. For example, ZT0 is the moment when the cue appears
and ZT12 is 12 hours after the cue is presented.

Figure 2 The cartoon depicts the fundamental core clock elements in mammalian cells. At the genetic level, circadian rhythms are generated in
single cells by interlocked positive and negative feedback loops. BMAL1 and CLOCK (or its homolog NPAS2 depending on the tissue) dimerize and
transactivate several clock genes, including those of the Period (Per) and Cryptochrome (Cry) families and Rev-erba and other clock-controlled genes
(Ccg). The negative regulators, Per and Cry, are translated and then translocated as a single complex to the nucleus where they inhibit CLOCK:BMAL1
transactivation. Casein Kinase 1-epsilon (CK1ε) and other kinases then alter the phosphorylation states of core clock components, thereby regulating
their retention in the cytoplasm or degradation, which determines the period and amplitude of the cellular rhythms. Outputs of the clock set the
phase of expression of numerous additional genes, regulating myriad cellular functions.
348 Circadian Regulation of Endocrine Functions

(Guillaumond et al., 2005). The Bmal1 gene is necessary for protein family. On a circadian timescale, they display circa-
circadian function – indeed it is the only single gene whose dian rhythms in oxidation, detectable in red blood cells,
knockout produces arrhythmic behavior. Numerous other which lack a transcriptional mechanism. The interconnec-
integrated feedback loops, often with elements that are tions between the circadian transcription/translation feed-
themselves regulated by the core clock loop, mediate clock back loop and redox pathways point to a substantial
output to clock-controlled genes (Mirsky et al., 2009; Albrecht, contribution of metabolism to circadian timekeeping (Asher
2012; Takahashi, 2015). and Sassone-Corsi, 2015; Perelis et al., 2015; Brown, 2016).
Posttranslational and metabolic modulation of oscillation: Post- This is important in the present context as, to date, the endo-
translational effects have a strong influence on clock speed, espe- crine systems have been considered primarily as outputs of
cially regulatory kinases that determine phosphorylation state of the circadian timing system. The discovery of circadian meta-
clock proteins (Albrecht, 2012). For example, the circadian bolic clocks points to new models of circadian rhythmicity
mutation named tau encodes a dominant negative allele of and new roles for endocrine factors, as these have major
casein kinase I epsilon (CK1ε) that shortens the period of loco- effects on metabolism (Figure 3).
motor activity rhythms (Ralph and Menaker, 1988; Lowrey et al., Clock-controlled genes. The core clock genes are the same across
2000). The wild-type forms of CK1ε and CK1d shape endoge- tissues, but the eventual set of rhythmic genes is large and tissue
nous period by determining the phosphorylation state of core specific. There is a small subset of canonical clock-controlled
clock genes (Etchegaray et al., 2009; Isojima et al., 2009; Mong genes, defined as those that exhibit E or D boxes and are rhythmi-
et al., 2011). cally transactivated by CLOCK:BMAL1, exemplified by albumin
Phosphorylation can modulate rhythm speed, but may D-element-binding protein (Dbp) (Ripperger and Schibler,
also represent a timekeeping mechanism. Phosphorylation 2006). Nevertheless, the circadian clock may have a much
state drives the protein-based circadian timer in cyanobacte- longer reach via its transcriptional and epigenetic effects.
ria (Nakajima et al., 2005), and kinase–phosphatase Across the genome, the core clock genes bind to thousands of
coupling alone can generate endogenous rhythms (Jolley sites suggesting that the clock genes have large spheres of
et al., 2012). Circadian timekeeping by the transcriptional/ influence (from 4600 CLOCK sites to 16 000 CRY1 sites)
translational feedback loop is also linked to nontranscrip- (Koike et al., 2012). Microarray studies have identified
tional metabolic and redox oscillations (Rey and Reddy, many cycling transcripts in several tissues, including the
2015). Peroxiredoxins are members of an antioxidant SCN, liver, heart, skeletal muscle, adrenals, and fibroblasts

Figure 3 Within a cell, rhythms depend on an intact molecular clock based on transcription and translation of core clock proteins (molecular
clock). This clock is intimately associated with nontranscriptional oscillations, including self-sustaining redox rhythms that depend on the redox state
of peroxiredoxins (nontranscriptional clocks) as well as metabolic flux rhythms in ATP and NADH (metabolic rhythms). There are several key interme-
diate molecules that link the molecular and metabolic clocks, including the sirtuins, PPAR, and PARP-1 (Brown, 2016).
Circadian Regulation of Endocrine Functions 349

# circadian genes detected


0 1,000 2,000 3,000

Liver 16%
Kidney 13%
Lung 12%
Brown fat 8%
Heart 6%
Adrenal gland 5%
Aorta 4%
White fat 4%
Skeletal muscle 4% 1+ spliceforms present
Cerebellum 4% 2+ spliceforms present
Brainstem 4%
Hypothalamus 3%

Figure 4 The graph depicts the number of protein-coding genes that express circadian rhythms in various organs (Zhang et al., 2014).

(Figure 4; Grundschober et al., 2001; Akhtar et al., 2002; Duffield associated with specific tissue functions. Interestingly, in most
et al., 2002; Panda et al., 2002a; Storch et al., 2002; Ueda et al., organs, the expression of many oscillating genes peak just
2002; Oster et al., 2006a; McCarthy et al., 2007). Zhang et al. before dawn and/or after dusk (Zhang et al., 2014). Finally, the
(2014) used RNA-seq and DNA arrays to quantify the majority of the top-selling drugs target genes that exhibit
transcriptomes of 12 mouse organs (adrenal gland, aorta, brain circadian rhythms (Zhang et al., 2014).
stem, brown fat, cerebellum, heart, hypothalamus, kidney, Rhythmic transcription within a tissue is intimately related
liver, lung, skeletal muscle, and white fat) and produced an to rhythms in RNA polymerase function and epigenetic regu-
atlas of changes over time in these tissues. What is most lation of the chromatin (Koike et al., 2012; Masri et al., 2015).
surprising is that 43% of all protein-coding genes showed Indeed, the gene CLOCK is a histone acetyltransferase (Doi
circadian rhythms in transcription somewhere in the body, et al., 2006), and numerous bodily tissues exhibit daily
largely in an organ-specific manner with little overlap between rhythm in patterns of chromatin modifications (Koike et al.,
organs. Only 10 genes, including the core clock genes, Per1, 2012). There is a strong dawn peak in genome binding by
Per2, and Per3, oscillated in all organs. This maps well to prior BMAL1 and CLOCK coincident with increased histone acetyla-
data indicating that there is only 5% overlap between the tion (H3K9ac) and methylation (H3K4me1), and there is
rhythmic genes in the SCN and in the liver (Panda et al., a strong evening peak in PER and CRY binding, also with
2002a), or between the heart and liver (Storch et al., 2002). distinct acetylation and methylation changes (H3K27ac and
This finding immediately suggests that cycling transcripts are K3K4me3) (Figure 5). This suggests that systems biology

Figure 5 Circadian chromatin rhythms in the liver. Profiles show the timing and degree of genome binding by core clock genes, the times of major
histone modifications including acetylation and methylation, and the time of peak binding by RNA polymerase. Reprinted with permission from AAAS,
Koike, N., Yoo, S.H., Huang, H.C., Kumar, V., Lee C., Kim, T.K., Takahashi, J.S., 2012. Transcriptional architecture and chromatin landscape of the
core circadian clock in mammals. Science 338 (6105), 349–354.
350 Circadian Regulation of Endocrine Functions

approaches will be valuable in the field of circadian rhythms This non-image-forming pathway is necessary and sufficient
(Hogenesch and Ueda, 2011). for photic entrainment. If the primary visual pathway is trans-
ected at the level of the optic tract beyond the optic chiasm (i.e.,
caudal to the SCN), then the animal is visually blind, but the
1.12.2.1 Entrainment through a Hierarchy of Clocks
circadian system continues to respond to photic cues and the
Hierarchy of circadian oscillators. The many clocks within the animal remains entrained (Klein and Moore, 1979; Johnson
body are synchronized in a hierarchical system in which et al., 1988).
a master circadian clock in the SCN entrains downstream The threshold of entrainment and the relative contributions
rhythms via an array of signaling mechanisms (Figure 6). The of the retinal photoreceptors-rods, cones, and intrinsically
SCN receives environmental time information via direct retinal photosensitive retinal ganglion cells (ipRGCs) have been exam-
projections and also receives information as to the internal ined (Butler and Silver, 2011). The entrainment threshold is
state of the body, from hormonal signals that act on receptors determined by rods and is more sensitive by 1–2 log units
in the SCN, or indirectly via afferent input from other sites. than other measures of the non-image-forming visual system.
SCN outputs entrain rhythms in target sites in the CNS and That is, dim light that entrained mice did not elicit either mask-
periphery. The oscillatory behavior of these extra-SCN sites ing or the pupillary light response.
can communicate phase information back to the SCN, creating Although the dim threshold of entrainment is determined
a closed-loop feedback system. The stability of this hierarchical by rod function, all three major photoreceptor classes (rods,
arrangement is necessary for normal body functioning. Disrup- cones, and melanopsin-expressing intrinsically sensitive retinal
tion of coordinated timing is a hallmark of numerous subop- ganglion cells) are capable of driving entrainment (Panda et al.,
timal and disease states. We now characterize the hierarchical 2002b, 2003; Ruby et al., 2002; Hattar et al., 2003; Lucas et al.,
organization of the circadian timing system, from input to 2003). All three photoreceptors must be knocked out to
output. abolish entrainment (Hattar et al., 2003). Anatomically,
Input to the brain clock. Circadian rhythms are entrained (or ipRGCs make up 2% of the total RGC population and serve
synchronized) to local environmental time primarily via light as the principal conduit to the SCN: ablation of these cells
information conveyed to the SCN by the retinohypothalamic alone prevents entrainment even while preserving pattern
tract (Moore and Klein, 1974; Klein and Moore, 1979). vision (Guler et al., 2008; Figure 7). The axon collaterals of

SCN

Pineal gland Melatonin

Pituitary gland

Thyroid
Prolactin

Adrenal
gland
Liver

Cortisol

Ovary
Testis

Testosterone

Figure 6 The circadian system is hierarchically organized. The master clock of the suprachiasmatic nucleus (SCN) provides timing cues to periph-
eral clocks in the rest of the body, including those in endocrine organs, via both direct (e.g., neural control) and indirect (e.g., humoral outputs,
feeding rhythms, body temperature) signals. These peripheral clocks in turn participate in coordinate local endocrine function and feed back at many
sites (not shown). Each hormone rhythm cycles with its own characteristic waveform and phase, shown here for a typical diurnal mammal. On the
right panel, the shaded area indicates night.
Circadian Regulation of Endocrine Functions 351

(a) Retinal input (b) SCN (c) Output

hν hν Actogram Endocrinogram

Neural Intact
and DD
diffusible
Rod Cone output
Shell SCN-x
hν Core

SCN
RGC ipRGC
Optic chiasm transplant
Anterior 0 24 0 24
Posterior Hours Hours

Figure 7 Inputs and outputs from the suprachiasmatic nucleus (SCN) master clock. (a) A small fraction of retinal ganglion cells contain the photo-
pigment melanopsin and are photosensitive (termed intrinsically photosensitive retinal ganglion cells: ipRGCs). The ipRGCs also receive photic infor-
mation from rods and cones, and they project via the retinohypothalamic tract (RHT) to the SCN. Elimination of ipRGCs eliminates circadian
responses to light (entrainment and phase shifting) but does not impact vision (Guler et al., 2008). (b) The core region of the SCN receives a dense
retinal input. Both core and shell send efferents to target sites in the brain. The SCN communicates timing information to the body by means of both
neural and diffusible signals. (c) The schematic shows locomotor activity in constant conditions in an animal before and after an SCN lesion. Each
consecutive day is plotted on lines from the top, and the black bars indicate the periods of activity. The animal becomes arrhythmic following the
lesion. In support of an SCN diffusible output signal, transplantation of fetal SCN tissue sealed in a semipermeable membrane into the lesioned host
restores locomotor rhythmicity. In contrast, the graft does not restore endocrine rhythms, indicating that neural outputs are required for these
responses.

these ipRGCs are extensive (Fernandez et al., 2016) and are in but dissociated individual SCN neurons exhibit different free-
a position to directly or indirectly regulate reproductive func- running periods (Welsh et al., 1995). Neurons communicate
tion and a variety of other endocrine-mediated behaviors and among themselves, and there is a functionally important
physiological processes (Fernandez et al., 2016). Note that underlying circuitry. The SCN has a core (also termed the
there is no evidence for extraretinal photoreception in rodents ventral or ventromedial subregion) and shell (dorsal or dorso-
(Nelson and Zucker, 1981). Extraocular photoreception also medial subregion), based on anatomical, neurochemical, and
appears to be absent in humans (Lockley et al., 1998; Barinaga, functional studies (reviewed in Antle and Silver, 2005). In
2002; Wright and Czeisler, 2002), despite one report to the the core, a light pulse induces the expression of clock genes,
contrary (Campbell and Murphy, 1998). including Per1 and Per2, but these genes do not oscillate detect-
The SCN and its oscillator neurons. Many lines of evidence ably. In the shell compartment, Per1 and Per2 oscillate with
confirm that the SCN is a light-entrainable master pacemaker. a circadian rhythm, but are not induced by light (Hamada
It receives direct projections from the retina (Moore and Lenn, et al., 2001). The question of how light-induced cells and oscil-
1972), ablation of this nucleus abolishes circadian rhythms lating cells of the SCN interact to communicate their phasic and
(Moore and Lenn, 1972; Stephan and Zucker, 1972), the rhythmic information to the rest of the brain/body has been
SCN exhibits rhythms of electrical activity and gene expres- examined both in terms of effects on molecular components
sion both in vivo and in vitro (Inouye and Kawamura, 1979; and its overall circuitry, evident in the spatial and temporal
Green and Gillette, 1982; Groos and Hendriks, 1982; Shibata patterns of clock gene expression (Hamada et al., 2001; Herzog,
et al., 1982; Yamazaki et al., 2000; Ono et al., 2015), and 2007; Yan et al., 2007; Pauls et al., 2014). The network exhibits
transplants of SCN tissue restore rhythmicity in arrhythmic a spatiotemporal pattern of oscillation that adopts different
hosts (Lehman et al., 1987; Ralph et al., 1990). In the last, conformations in response to the environment (e.g., long or
the donor SCN, rather than the host, determines period, short day lengths) (Evans et al., 2013). While multiple mecha-
showing that the transplanted tissue does not act by restoring nisms have been proposed for coupling among SCN neurons
host-brain function but that the ‘clock’ is contained in the (Antle et al., 2003, 2007), to date, relatively little is known of
transplanted tissue. The fact that SCN rhythms persist for the precise nature of its anatomical circuitry (Drouyer et al.,
many cycles in vitro (longest reported is 478 days!) (Yamazaki 2010). Furthermore, while the SCN oscillates independent of
and Takahashi, 2005) shows that input from other brain areas photic input from the retina, some cells within the SCN act
is not necessary. as ‘slave oscillators’ whose rhythm depends on the presence
The cells of the SCN are heterogeneous in neurotransmitter of connections from the retina (Lee et al., 2003).
and peptide content, in their dendritic and axonal morphol- SCN output is diffusible and neural. The SCN signals the body
ogies, and in their afferent and efferent connections. There by several mechanisms. Among them are both diffusible and
are distinct components of the clock and important emergent neural output signals. Some aspects of rhythmicity, including
properties of its interacting elements. In the intact animal, the locomotor behavior, are sustained in the presence of diffusible
SCN oscillation generates a coordinated circadian rhythm, signals from the master clock. On the other hand, the circadian
352 Circadian Regulation of Endocrine Functions

control of the endocrine system requires neural connections hypothalamus (DMH), and orexin neurons of the lateral
from the SCN to sustain oscillation. hypothalamus all exhibit 12-h rhythms. These data suggest
SCN tissue from a fetal donor, when implanted into the third each SCN (left and right) may signal each monosynaptic target
ventricle of an adult, SCN-lesioned host, restores circadian once daily, thereby producing 12-h days in the animal (Butler
patterns in activity-related behaviors such as locomotor, et al., 2012).
drinking, and gnawing rhythms (Lehman et al., 1987; Ralph Neural output from the SCN has been extensively investi-
et al., 1990; Silver et al., 1990). While some neural gated in mice, rats, and hamsters using tract-tracing techniques
connections are established between the donor tissue and the (Morin, 2013). Projections arise from both the core and the
host brain, the transplant also produces diffusible factors. That shell of the SCN. Importantly, many of these monosynaptic
a diffusible signal is sufficient to restore locomotor projections target brain regions containing neuroendocrine
rhythmicity in SCN-lesioned hosts was demonstrated cells producing hypothalamic releasing hormones. Direct
definitively by encapsulating donor SCN tissue in a membrane projections have been traced from the SCN to the medial pre-
that prevented neural outgrowth while allowing the diffusion optic area (MPOA), supraoptic nucleus, anteroventral periven-
of signals between graft and host (Silver et al., 1996). The tricular nucleus (AVPV), the PVN, DMH, lateral septum, and
efficacy of paracrine signaling in circadian timing was the arcuate nucleus (Arc). The SCN also projects to the pineal
demonstrated with an in vitro coculture technique using wild- through a multisynaptic pathway (Klein et al., 1983; Klein,
type SCN atop a membrane that communicated by shared 1985). There is abundant evidence for direct neural SCN
culture medium enabling it to drive oscillation in a target control of neuroendocrine cell populations (van der Beek
‘host’ SCN deficient either in elements of neuropeptidergic et al., 1993, 1997; Vrang et al., 1995; Kalsbeek et al., 1996,
signaling or in elements of the core feedback loop (Maywood 2000; Buijs et al., 1998, 2003; Horvath et al., 1998; Gerhold
et al., 2011). Paracrine signaling restored pacemaking in SCN et al., 2001; Kalsbeek and Buijs, 2002; Kriegsfeld et al., 2002;
circuits lacking vasoactive intestinal peptide (VIP) and Egli et al., 2004). Because these cell populations can regulate
maintained rhythmicity in arrhythmic Cry-null SCN. More neurochemicals that are secreted into the cerebrospinal fluid,
specifically, this work suggested a role for VIP, arginine pituitary portal system, and general circulation, SCN-derived
vasopressin (AVP), and gastrin-releasing peptide (GRP) in signals can control widespread systems in the brain and body
synchronizing SCN neurons. Thus, a hierarchy of paracrine (Skinner and Malpaux, 1999; Reiter and Tan, 2002; Skinner
neuropeptidergic signals determines cell- and circuit-level and Caraty, 2002; Tricoire et al., 2003).
circadian pacemaking in the SCN. Extra-SCN Oscillators. Unlike the long-lasting and self-
SCN output: Neural efferents. In contrast to behavioral sustained rhythms exhibited by the SCN in culture, most
rhythms, endocrine rhythms require neural projections from gene expression rhythms damp out after several cycles in vitro
the SCN to neurosecretory cells in the brain. Endocrine rhythms (Balsalobre et al., 1998; Yamazaki et al., 2000). This damping
are abolished after knife cuts severing SCN efferents (Nunez and of the rhythms reflects desynchronization of self-sustained
Stephan, 1977; Hakim et al., 1991) and are not restored in SCN- and cell-autonomous oscillators (Nagoshi et al., 2004; Welsh
lesioned animals with SCN transplants, presumably due to et al., 2004). In vivo, similar desynchronization among organs
inadequate neural innervation of the host brain by the graft is observed in the absence of the SCN (Yoo et al., 2004).
(Nunez and Stephan, 1977; Silver et al., 1996; Meyer-Bernstein The coordination of peripheral oscillations, and therefore
et al., 1999). Further evidence for a neural SCN output signal the expression of overt rhythms in physiology and behavior,
regulating hormone secretion is seen in studies of hamsters. depends primarily on the SCN, but may be modulated by other
When housed in constant light, the locomotor activity of mechanisms. The SCN projects via multisynaptic pathways to
hamsters ‘splits’ into two separate activity bouts within a 24-h peripheral endocrine organs, including the pineal gland, adre-
interval (as though they have 12-h days). Split females display nals, thyroid, and pancreas (Moore, 1996; Buijs et al., 1999;
two daily preovulatory LH surges, 12 h apart and each Kalsbeek et al., 2000; Bartness et al., 2001; Buijs and Kalsbeek,
approximately half the concentration of a single surge in 2001). The SCN can also transmit timing cues indirectly via
a nonsplit female (Swann and Turek, 1985). While both halves SCN-dependent behaviors, such as eating, that can alter
of the bilaterally symmetrical SCN are active in synchrony physiological signals. The presence of oscillators throughout
under normal conditions, in split hamsters, rhythms of SCN the body confers flexibility in entraining to cues. For
activity on each side of the brain are 12 h out of phase (de la example, when the feeding schedule and the light–dark
Iglesia et al., 2000). Remarkably, FOS expression in GnRH schedule are dissociated, rhythms in SCN gene expression
neurons is only observed ipsilaterally to the FOS-expressing remain entrained to the light–dark schedule, but rhythms in
side of the SCN (de la Iglesia et al., 2003). These findings the liver entrain to the feeding schedule (Damiola et al.,
support the conclusion that the precise timing of the LH surge 2000; Stokkan et al., 2001). By examining SCN-lesioned mice
is derived from a neural signal originating in the SCN and parabiotically paired with animals bearing an intact SCN
communicated to ipsilateral GnRH neurons (likely via (sharing 1–4% of their blood supply), it was revealed that
kisspeptin neurons, see below), as a diffusible output signal nonneural signals are adequate to maintain circadian
would reach both sides of the brain. Further analysis of the rhythms of clock gene expression of Per1, Per2, or Bmal1 in
split hamster indicates that within each nucleus, the SCN splits the liver and kidney (but not in the heart, spleen, or skeletal
into two parts, oscillating in antiphase (Nakajima et al., 2005; muscle) (Guo et al., 2005). This implicates a blood-borne
Tavakoli-Nezhad and Schwartz, 2005). Surprisingly, in most cue, perhaps endocrine or metabolic, that can provide phase
brain areas, SCN targets, including the preoptic area, information to some but not all peripheral oscillators.
paraventricular nucleus (PVN), and dorsomedial nucleus of the Alternatively, the intact animal may communicate timing
Circadian Regulation of Endocrine Functions 353

information to the SCN-lesioned mouse via activity and/or between nocturnal and diurnal species (Ramanathan et al.,
body temperature fluctuations. One interesting implication of 2010), the precise pattern of variation in plasma hormone
this work is that an SCN-derived cue may synchronize each levels does differ and results from interactions between regu-
individual liver hepatocyte every single day in order to larly recurring periodic cues (such as eating) and nonrhythmic
sustain phase coherence in peripheral tissues. A similar factors such as homeostatic mechanisms (e.g., rest/activity
principle may apply to the process of synchronizing cycles) (Kriegsfeld and Silver, 2006; Morin, 2013; Dibner and
individual cells of glandular organs. Schibler, 2015). This is shown elegantly in studies of humans
Interactions among central and peripheral oscillators: Peripheral where cycles of fasting–feeding, sleep–wake, and rest–activity
organs are entrained by the SCN, but there are many potential are easily dissociated (Morris et al., 2012).
intermediate pathways, and their relative importance is still not
well described. In addition to direct effects of SCN neural output
or SCN-derived diffusible cues, SCN-controlled behaviors and 1.12.4 Circadian Regulation of Hypothalamo–
secondary messengers are critical zeitgebers. One of the most Pituitary–Adrenal Axis
important of these mechanisms is the feeding–fasting rhythm.
Rhythmic presentation of food is sufficient to entrain the Rhythms of the hypothalamo–pituitary–adrenal (HPA) axis.
locomotor activity rhythms of animals (food anticipatory Rhythms in glucocorticoid secretion and serum concentration
activity) even in otherwise arrhythmic SCN-lesioned animals are well established (Dickmeis, 2009); indeed, elimination of
(Mistlberger, 2006). The endogenous nature of this rhythm is corticosterone rhythms after SCN lesions were among the first
revealed during subsequent total food deprivation. At these data implicating the SCN as a brain clock (Moore and Eichler,
times, the food-deprived animals continue to display free- 1972). Humans, nonhuman primates, and rodents all display
running anticipatory behavior, and this behavior demonstrates circadian changes in glucocorticoid secretion that continue in
the existence of a food-entrained oscillator (FEO). There is constant conditions (Weitzman et al., 1971; Moore and Eichler,
considerable interest in understanding whether this FEO is 1972; Gallagher et al., 1973; Dubey et al., 1983; Van Cauter and
localized in a specific site or constitutes a network – a topic of Refetoff, 1985; Czeisler and Klerman, 1999). The phase of the
heated debate (Gooley and Saper, 2007; Landry and rhythm differs between nocturnal and diurnal species, with
Mistlberger, 2007; Landry et al., 2007). The phenomenon of corticoid secretion beginning to rise before waking in both
food-entrained oscillation is important because food-derived but then peaking during the day in diurnal species and during
signals constitute cues that can influence rhythmicity of the night in nocturnal species (Wong et al., 1983; Albers et al.,
downstream oscillators in both the brain and in endocrine 1985; Ottenweller et al., 1987). In humans, cortisol levels have
organs (discussed below). In turn, changes in the phase a robust daily rhythm, peaking around the morning wake-up
hormone secretion may influence other timed physiological time. The adrenal gland itself manifests rhythms in clock
and behavioral responses. gene expression and these underlie rhythmic responsiveness
In addition to food-related entraining cues, peripheral to pituitary adrenocorticotropic hormone (Gallagher et al.,
tissues may also be entrained by glucocorticoids (Balsalobre 1973), and to physical and physiological stressors (Ungar
et al., 2000; Le Minh et al., 2001). Body temperature rhythms and Halberg, 1962; Dunn et al., 1972; Buijs et al., 1997;
as well, governed directly and indirectly through rest–activity Bittman et al., 2003; Kalsbeek et al., 2003; Oishi et al., 2003;
cycles, are also cues for peripheral oscillators (Buhr et al., Guo et al., 2006; Oster et al., 2006b; Fahrenkrug et al., 2008).
2010). Impressively, while the molecular makeup of circadian The robust circadian pattern of circulating corticoids is
oscillators is nearly identical in all cells, they may not be generated by changes in pulsatile secretion that occurs on an
equally sensitive or similarly responsive to entraining cues. ultradian basis, with a periodicity of about 1 h (Walker et al.,
Though tempting to consider a peripheral tissue clock phase 2012; Spiga et al., 2014; Russell et al., 2015). Both modeling
as the endpoint of entrainment, this is simplistic. Each cell and empirical data show that this ultradian pattern of secretion
exhibits a multilayered rhythm from epigenetic regulation of is driven by biological delays and feed-forward mechanisms at
the chromatin, to transcription, to cellular function. These the level of the pituitary and adrenal gland, without any
may differ in their sensitivity to the potential internal and requirement for a pulse generator in the hypothalamus (Walker
external entraining cues. For example, of 2000 genes that et al., 2010, 2012). These ultradian pulses are important for
are rhythmic in whole brain extracts, only 400 remain adrenal function as it has been demonstrated that the adrenal
rhythmic in sleep-deprived mice, indicating a strong entraining gland responds optimally to an oscillatory profile of ACTH,
role for sleep and its correlates (Maret et al., 2007). Similarly, of but that constant ACTH infusion results in a decrease in corti-
3000 genes that cycle in the liver during ad-lib feeding condi- coid output (Spiga et al., 2011).
tions, only 400 genes remain rhythmic during fasting condi- Role of SCN in HPA axis rhythmicity. Rhythms in circulating
tions (Vollmers et al., 2009). glucocorticoids are eliminated by SCN lesions and are not
restored by SCN transplants (Moore and Eichler, 1972;
Meyer-Bernstein et al., 1999). Similarly, rhythms in clock
1.12.3 Circadian Regulation of the Endocrine gene expression in the adrenal depend on the SCN (Guo
System et al., 2006). Two SCN efferent pathways are implicated in
the regulation of the adrenal axis (Figure 8). First, monosyn-
Circadian patterns of hormone secretion occur in both diurnal aptic projections to the corticotropin releasing hormone
and nocturnal species (Kriegsfeld et al., 2002a). Although the (CRH) neurons in the PVN are thought to directly regulate
timing of expression of clock genes in the SCN is not different ACTH release (Vrang et al., 1995; Kalsbeek et al., 1996;
354 Circadian Regulation of Endocrine Functions

PVN

CRH cells

IML
SCN

Eye Pituitary
H ion
ACT nervat
mic in
Circadian rhythm ono
Aut
of ACTH sensitivity

Rhythmic
glucocorticoid
Adrenal release

Figure 8 Schematic depiction of the hypothalamo–pituitary–adrenal (HPA) axis and its connections. The suprachiasmatic nucleus (SCN) projects to
the adrenal gland via synapses in the autonomic portion of the paraventricular nucleus (PVN) and the intermediolateral column (IML). In addition,
SCN neurons make contacts with corticotropin releasing hormone (CRH)-expressing cells of the PVN, which in turn promote the release of ACTH
from the pituitary. The combination of signals synchronizes the cycling transcriptome of the adrenal which includes genes associated with adrenal
function. The local cycling of gene expression within the adrenal gland may also contribute to gating the response to ACTH.

Buijs et al., 1998). In addition, the SCN may directly control results indicate that functioning peripheral clocks are necessary
adrenal rhythms via multisynaptic autonomic innervation of for optimal glucocorticoid production by the adrenal gland.
the adrenal cortex (Buijs et al., 1999). This second pathway Consequences of adrenal hormone rhythms in brain and
may be critical to establishing proper adrenal function, as periphery. As noted, glucocorticoid hormones produced by the
hypophysectomy does not alter adrenal clock gene expression adrenal gland cortex are rhythmically released under the
rhythms, indicating that rhythms in ACTH do not by them- control of the SCN. The SCN itself does not express glucocorti-
selves induce rhythms in the adrenal glands (Fahrenkrug coid receptors (GRs) (Rosenfeld et al., 1988; Balsalobre et al.,
et al., 2008). 2000). Glucocorticoids, nevertheless, may have indirect effects
Regulation of HPA axis by peripheral oscillators. Transcription on the SCN: glucocorticoid treatment in humans disrupts sleep
profiles of the mouse adrenal reveal a large number of cycling and in the SCN, reduces AVP (Liu et al., 2006). In rats, gluco-
genes (5% transcripts on an Affymetrix MG430v2.0 chip), corticoids upregulate glial fibrillary acidic protein in the SCN,
among which are those involved in such adrenal functions possibly through effects on serotonergic neurons (Maurel
as steroid biosynthesis and catecholamine metabolism (Oster et al., 2000). For the most part, the specific neural targets by
et al., 2006a). The majority of the cycling transcriptome peaks which corticoids influence SCN function remain to be
at the sleep to wake transition; notably, 10 of 14 rhythmic determined.
genes associated with steroid biosynthesis were synchronous, In extra-SCN brain regions, it is quite clear that glucocorti-
peaking in early subjective night in parallel with the coid rhythms can communicate phase information in a region-
increasing plasma corticoid levels at this time (Oster et al., specific manner. In rats, PER2 is rhythmic in several brain
2006a). The gating of adrenal ACTH sensitivity depends on areas, including the oval nucleus of the bed nucleus of the stria
this local cycling, as rhythms in sensitivity are eliminated in terminalis (BNST-OV), and the central (CEA) and basolateral
cultured adrenal slices from arrhythmic Per2/Cry1 knockout nuclei of the amygdala (BLA) (Amir et al., 2004; Lamont
mice (Oster et al., 2006b). Synchronization of the adrenal et al., 2005). The PER2 rhythm in the BLA is not affected by
oscillators by other cues allows for rhythmic function in the adrenalectomy. In contrast, rhythms in the BNST-OV and
absence of the SCN. For example, SCN lesions do not prevent CEA depend on peripheral glucocorticoid cues for
the corticosterone peak associated with restricted feeding entrainment, for in these two structures, the rhythm in PER2
(Krieger et al., 1977). is abolished by adrenalectomy. Rhythms are restored in
In an elegant study, Oster et al. (2006b) tested the role of animals provided with corticosterone in the drinking water
a functioning adrenal clock by cross-implanting adrenals (producing a rhythm in circulating corticosterone), but not
between wild-type and Per2/Cry1 mutant mice: the mutant by implanted constant-release corticosterone pellets (Segall
mice lack rhythmic ACTH, corticosterone, and adrenal clock et al., 2006). While these effects have been well characterized
gene expression. They found that the rhythm of ACTH respon- in subcortical brain areas, there are similar effects observed
siveness observed in vivo was also observed in vitro in wild-type in the cortex. A direct role for diurnal changes in corticoids
but not in Per2/Cry1 knockout adrenals. This implicated the has been shown, with diurnal changes in circulating
local, intra-adrenal clock in responses to ACTH. They then corticosterone important for spine turnover in motor cortex.
showed that corticoid excretion rhythms of wild-type mice This has implications for performance, showing that
with wild-type adrenal implants were nearly double the ampli- glucocorticoid rhythms promote learning of motor tasks
tude of wild-type mice that had mutant adrenal implants. These (Liston and Gan, 2011).
Circadian Regulation of Endocrine Functions 355

Glucocorticoids also communicate timing information to Miller et al., 2004; Chu et al., 2013) Two aspects of gonado-
peripheral organs (Balsalobre et al., 2000). Dexamethasone tropin regulation have been described. First, a circadian rhythm
(DEX), a potent agonist of the GR, is effective in synchronizing in gonadotropin secretion is seen around the time of puberty
circadian gene expression in cultured cells and in activating due to increases in nighttime secretion of LH and follicle-
transcription of Per1, a core clock gene. Intraperitoneal injec- stimulating hormone (FSH) (Dunkel et al., 1992; Apter et al.,
tion of DEX in mice during the day results in phase shifts in 1993). As peripubertal children enter adulthood, daytime secre-
the liver, suggesting that glucocorticoid signaling synchronizes tion of the gonadotropins increases; consequently, adults lack
liver clocks. Using gene chip microarrays, Oishi et al. (2005) circadian patterns of gonadotropin secretion (Krieger et al.,
described 169 genes that differed in expression level between 1972; Veldhuis et al., 1986). Second, as already noted, circadian
night and day in the livers of normal mice. The day–night rhythms are striking in the regulation of the preovulatory LH
difference in 100 of these genes was abolished by surge. The high estradiol (E) concentrations during proestrus
adrenalectomy. Reddy et al. (2007) further identified 366 are permissive in this regard, and constant E treatment in ovari-
transcripts whose rhythmic expression were abolished by ectomized (OVX) females reveals a circadian rhythm in daily LH
SCN lesions; of these, rhythms in 57% were reinstated by surges at the same time each day (Legan et al., 1975). In adult
DEX treatment. Multiple pathways drive rhythmicity across males, there is a robust circadian rhythm in circulating testos-
the whole cycling transcriptome. For example, only two- terone (T), with a trough in late evening and a peak in the early
thirds of the genes whose rhythms were induced by DEX had morning (Spratt et al., 1988). T rhythms persist in constant
glucocorticoid response elements (GRE) in their promoters conditions (Dubey et al., 1983). The disparity between a lack
(Reddy et al., 2007). Rhythms in some of the remaining genes of adult male gonadotropin rhythms and a robust rhythm in
may have been induced by clock genes, themselves with DEX- T suggests some regulation of the T rhythm downstream from
induced rhythms. Finally, other rhythmic genes must act as LH release, potentially through autonomous clock function in
intermediaries between the clock- or DEX-responsive genes the testes (see below). In adult women, plasma levels of ovarian
and remaining genes of the cycling transcriptome that lack hormones vary across the menstrual cycle, but their circadian
both glucocorticoid and clock responsive promoter elements. rhythmicity is less clear. Nonhuman primates show diurnal vari-
ation in estrogen and progesterone (P) during the luteal, but not
during the follicular, stage of the menstrual cycle (Spies et al.,
1.12.5 Circadian Rhythmicity in the Hypothalamic– 1974). In humans, results are mixed, with some studies finding
Pituitary–Gonadal Axis no diurnal variation in P or E, and others finding rhythms
during some phases of the menstrual cycle (Aedo et al., 1981;
Overview. As noted in the introduction to this chapter, the Carandente et al., 1989; Rossmanith et al., 1990).
importance of the brain in regulating the hypothalamic–pitui- SCN regulation of the HPG axis rhythms. Work in rodents indi-
tary–gonadal (HPG) axis was first established in the 1950s. It cates, at the whole-animal level, that the circadian system deter-
was in this era that the existence of neurosecretory hormones mines the duration of the female reproductive cycle. In Syrian
was first described in the classical studies of Harris where he hamsters entrained to a 24-h day, the duration of the estrous
showed that the hypothalamohypophysial portal vasculature cycle is normally 4 days. When the animals free-run, their
carries neurosecretions from hypothalamic nerve fibers to the estrous cycle is a quadruple-multiple of the free-running
anterior pituitary, unlike the direct neural control of the poste- rhythm period (Fitzgerald and Zucker, 1976). This is a general
rior pituitary. The first hypothalamic releasing hormone, pattern: in treatments that lengthen the circadian period (e.g.,
GnRH, was isolated in 1969, and its importance was acknowl- with deuterium oxide in the drinking water), or in non-24-h
edged in a Nobel Prize in 1977 for its seekers, Guillemin and days (e.g., 11 h light:11 h dark), the estrous cycle duration is
Schally (Initially GnRH was called luteinizing hormone reliably a fourfold function of the circadian day. Similarly,
releasing hormone as it was thought to regulate only LH the preovulatory LH surge occurs at a specific time of day on
(Grundschober et al., 2001)). Following that work, other hypo- proestrus (Colombo et al., 1974). Notably, surgical lesions of
thalamic releasing hormones were soon isolated and synthe- the SCN abolish both the LH surge and estrous cycling
sized, leaving one with the impression that the most (Brown-Grant and Raisman, 1977; Gray et al., 1978). Work
important neural elements of the HPG axis were known. with mutant mice confirms a role for circadian clocks at the
Thus, it came as a surprise to find major new neuroendocrine cellular level. Mice with mutated Clock or Bmal1 have an
elements in the brain in the twenty-first century, namely kiss- impaired LH surge and abnormal estrous cyclicity (van der
peptin and gonadotropin-inhibitory hormone (GnIH), funda- Horst et al., 1999; Miller et al., 2004; Chu et al., 2013). Women
mental stimulatory and inhibitory regulators of the HPG axis, bearing single-nucleotide polymorphisms in ARNTL (Bmal1)
respectively. The search for brain hormones has been dramatic have more miscarriages than those lacking this mutation
(Loriaux, 2016), and the tale of the more recent discoveries is (Kovanen et al., 2010).
an interesting (but perhaps not unusual) example of the jagged SCN efferents from core (VIP) and shell (AVP) cells. The finding
course of discovery in research. that the SCN underlies the unilateral activation of neurosecre-
Rhythms of the HPG axis. Circadian rhythms are evident at all tory cells in split hamsters (de la Iglesia et al., 2003; Gibson
levels of the HPG axis. The importance of the SCN is under- et al., 2008; Butler et al., 2012) provides strong evidence that
scored by the observation that disruptions of circadian rhythms, the SCN controls neuroendocrine secretion through neural
either by surgical lesions or by mutations in the underlying output signaling. Neuroanatomical and physiological studies
clock genes, interfere with or abolish female reproductive indicate two sets of neural SCN efferents that signal daily
rhythms and maintenance of pregnancy (Gray et al., 1978; temporal information to the reproductive axis: VIP in the
356 Circadian Regulation of Endocrine Functions

ventrolateral core part of the SCN and AVP containing neurons controlled in the SCN, Clock mutant mice (which have
in the dorsomedial shell part. The VIP peptide does not vary reduced AVP mRNA expression in the SCN) lack an LH surge,
rhythmically in constant darkness, but under light–dark condi- a response that is rescued by central injections of AVP, further
tions, VIP content decreases over the course of the light period linking this peptide to the circadian control of ovulation
and then gradually recovers during the dark period, and thus (Miller et al., 2006). How does AVP have such a pronounced
may transmit lighting information (Shinohara et al., 1993). impact on the LH surge without communication directly to
While VIP decreases, the content of GRP, another less well GnRH cells? As discussed further below, AVP appears to
studied peptide in the core, increases. mediate the LH surge via indirect projections to kisspeptin
VIP: Efferents of VIP SCN neurons contact GnRH neurons cells in the preoptic area that, in turn, project to GnRH cells.
which express the VIP receptor (VPAC2) (van der Beek et al., FMRFamide. The cardioexcitatory neuropeptide containing
1993, 1997; Smith et al., 2000). Interestingly, female rats the C-terminal Phe-Met-Arg-Phe-NH2 (FMRFamide) was first
have greater VIPergic innervation than males (Horvath et al., identified in the ganglia of the clam, Macrocallista nimbosa
1998) and VIP contacts increase following puberty (Kriegsfeld (Price and Greenberg, 1977). Following this discovery,
et al., 2002). Pharmacological studies indicate that blocking antibodies to FMRFamide peptides were applied as a tool for
the VIP receptor, VPAC2, attenuates GnRH cell firing on the labeling structurally similar peptides across taxa, although the
proestrous afternoon surge in female mice. In brain slices taken identity of labeled peptides remained unknown. Subsequently,
from OVX E-primed mice, acute VIP treatment increases firing a host of vertebrate peptides sharing the RF motif (Arg-Phe-
rates in GnRH neurons from mice sacrificed around the time NH2: RFamide) have emerged as prominent regulators of
of LH surge onset, but generally had little effect on neurons neuroendocrine activity. Two RFamide peptides, kisspeptin
before the surge (Christian and Moenter, 2008). Central and GnIH, have marked direct, but opposing, actions on the
administration of VIP antiserum reduces the LH surge in reproductive axis. Kisspeptin provides positive drive to the
female rats (van der Beek et al., 1999). By contrast, infusion GnRH system, whereas GnIH acts to suppress GnRH output.
of VIP into the third ventricle inhibits LH surges triggered by Given the location of these peptides in the rodent brain, both
progesterone injection in OVX E-primed rats (Weick and are in a position to relay circadian information to the
Stobie, 1995). The reasons for this discrepancy are unclear. reproductive axis (Figure 9).
Finally, VIP might contribute to the LH surge through Gonadotropin-inhibitory hormone. GnIH was first identified in
interactions with neuroglia that envelop GnRH cells. quail brain by probing the proteome using competitive
Astrocytes express VIP receptors, and their envelopment of enzyme-linked immunosorbent assays (ELISA) with antibodies
GnRH neurons varies with time of day (Gerhold and Wise,
2006). Suppression of the SCN VIP rhythm with antisense
nucleotides prevents these rhythms in astrocyte coverage.
Such glial–neuronal signaling is an important aspect of the
regulation of neuroendocrine secretion (Garcia-Segura and
McCarthy, 2004) and is a key element of SCN oscillation
(Marpegan et al., 2009).
AVP neurons of the SCN: Another important SCN peptide,
AVP, has been implicated in the LH surge. The observation
that vasopressin peaks coincident with the onset of the preovu-
latory LH surge provided the initial impetus for further investi-
gating the role of the SCN peptide in surge generation
(Schwartz et al., 1983). However, unlike VIP, there is no
evidence of direct SCN to GnRH connections by AVPergic
neurons, and GnRH cells very rarely express the 1a subtype of
AVP receptor (V1a) (Kalamatianos et al., 2004). Despite not
directly acting on the GnRH system, pharmacological work
established that AVP was critical for surge generation. For
example, administration of AVP into the MPOA induces an
LH surge in SCN-lesioned, OVX rats treated with E2, whereas
treatment with an AVP receptor, V1a, antagonist reduced the
E2-induced LH surge (Palm et al., 1999; Funabashi et al., Figure 9 Cartoon depicts the circadian control of two key RFamide
2000). Likewise, in cocultures of POA and SCN tissue, the mediators of the reproductive axis. Kisspeptin (Kp) and gonadotropin-
rhythm of GnRH release is in phase with the rhythm of AVP inhibitory hormone (GnIH) neurons project directly to the gonadotropin-
release, but not VIP (Funabashi et al., 2000). AVP gene releasing hormone (GnRH) system to positively and negatively regulate
GnRH secretion, respectively. GnIH cells also communicate directly with
transcription in the SCN is directly controlled by the CLOCK/
the anterior pituitary portal system to regulate gonadotropin secretion.
BMAL1 dimer (Jin et al., 1999; Silver et al., 1999). The SCN
The suprachiasmatic nucleus (SCN) controls the daily and seasonal
content of AVP and its release in the cerebrospinal fluid activity of the kisspeptin and GnIH systems. There are prominent direct
displays a daily rhythm that is maintained in constant projections from the SCN to anteroventral periventricular nucleus
darkness (Schwartz and Reppert, 1985; Gillette and Reppert, (AVPV). Whether these projections specifically make synapses on kiss-
1987; Takahashi et al., 1989; Kalsbeek et al., 1995; Krajnak peptin cells is not known. Finally, the SCN projects directly to GnIH and
et al., 1998). As one might expect given that AVP is clock- GnRH neurons (Williams and Kriegsfeld, 2012).
Circadian Regulation of Endocrine Functions 357

directed against the RF motif. Using this approach, a novel RFa- et al., 2009; Williams et al., 2011). These findings, combined
mide peptide was identified that rapidly and dose-dependently with the fact that the SCN projects extensively to the AVPV
inhibits gonadotropin release from cultured quail pituitaries (Leak and Moore, 2001; Kriegsfeld et al., 2004), suggested
(Tsutsui et al., 2000) and was named GnIH. In rodents, that the circadian system may stimulate the LH surge via
GnIH cells are confined to the dorsomedial hypothalamus projections to the kisspeptin cells in the AVPV. Indeed,
(DMH), with widespread projections to hypothalamic and AVPergic SCN cells project directly to GnRH cells in mice and
limbic structures (Kriegsfeld, 2006). GnIH cells project upon hamsters (Sarvari et al., 2010; Williams et al., 2011) and AVP
GnRH perikarya and to the median eminence. Injections of increases kisspeptin cellular activity as measured by FOS
GnIH lead to rapid suppression of LH in rats, mice, and Syrian (Williams et al., 2011) and intracellular calcium signaling (Piet
hamsters. Importantly, GnIH cells express estrogen receptors et al., 2015). A substantial amount of work has been done on
and subserve the steroid negative feedback on the reproductive the neuroanatomy and function of the kisspeptin signaling
axis (Kriegsfeld et al., 2006). The SCN sends pronounced system, its receptors (Kiss1r), and its projections across
projections to the DMH (Leak and Moore, 2001; Kriegsfeld mammalian species (Lehman et al., 2013). Across species, the
et al., 2004), and in Syrian hamsters, over 60% of GnIH cells majority of Arc kisspeptin cells coexpress the positive and
are contacted by SCN terminals (Gibson et al., 2008; Russo negative regulators of the reproductive axis, neurokinin B
et al., 2015). Important in the present context, the SCN acts (NKB) and dynorphin, respectively (Goodman et al., 2007).
to suppress GnIH cell activity at the time of the LH surge, These triple-phenotype neurons are now commonly
thereby allowing the coordinated release of the GnRH system referred to as KNDy (pronounced like ‘candy’) and
from estrogenic negative feedback at this time via VIPergic (K ¼ kisspeptin, N ¼ NKB, and Dy ¼ dynorphin) (Lehman
projections (Gibson et al., 2008; Russo et al., 2015). In et al., 2010). KNDy neurons are reciprocally connected and
summary, GnIH is regulated by the circadian timing system express receptors for NKB and dynorphin, providing the ability
and serves as a key player regulating reproduction in verte- to propagate stimulatory or inhibitory signaling (Lehman et al.,
brates, acting on the brain and pituitary to modulate reproduc- 2010). KNDy neurons also express progesterone as well as
tive physiology and behavior (Smith and Clarke, 2010; estrogen receptors, providing a mechanism for mediating
Kriegsfeld, 2013; Tsutsui et al., 2015). steroid negative feedback (Akhtar et al., 2002; Foradori et al.,
Kisspeptin. The Kiss1 gene was originally discovered in a screen 2002). This Arc population of kisspeptin neurons is thought
for tumor metastasis suppressors, and its protein was named to participate in the regulation of GnRH pulses and sex steroid
metastin for this property when it was identified in 2001 as negative feedback. Additional kisspeptin neurons, fibers, and
the endogenous ligand for the orphan G-protein-coupled Kiss1R can be detected in areas outside the hypothalamus,
receptor, GPR54 (Kriegsfeld, 2006; Kauffman et al., 2007). A but their physiological role(s) in these regions is not
role for this RFamide, now designated kisspeptin, in understood.
regulating reproductive function emerged in 2003 when Intrinsic oscillation in GnRH neurons. Circadian oscillators in
individuals with hypogonadism were shown to have GnRH neurons or rhythmic changes in the properties of these
a mutation in GPR54 (de Roux et al., 2003; Seminara et al., neurons may participate in regulating GnRH secretion. In hypo-
2003). The effects of kisspeptin are mediated through its thalamic slice cultures in the absence of the SCN, circadian
actions on the GnRH system; administration of kisspeptin rhythms in neural firing rate of GnRH neurons persist for several
leads to immediate early gene expression (i.e., FOS) in GnRH days in vitro when the slices are prepared from ovariectomized,
cells (Irwig et al., 2004; Matsui et al., 2004). Additionally, E-treated mice but not from those of ovariectomized, untreated
kisspeptin-induced gonadotropin secretion is blocked across mice (Christian et al., 2005). Clock gene and GnRH mRNA
species by the GnRH receptor antagonist, acyline (Gottsch expression are rhythmic in GnRH-secreting GT1-7 cell lines
et al., 2004; Irwig et al., 2004; Shahab et al., 2005). Finally, (Gillespie et al., 2003). Moreover, transfection of the
a majority of GnRH cells coexpress GPR54 mRNA across dominant negative mutant Clock-D19 gene into GT1-7 cells
species (Irwig et al., 2004; Han et al., 2005; Messager et al., decreases GnRH pulse frequency, indicating that the circadian
2005), and kisspeptin acts directly on GnRH cells to increase clock genes could participate in regulating pulsatility
their neural firing in brain slices (Liu et al., 2008). In rodents, (Chappell et al., 2003). The evidence for a functional clock in
kisspeptin cell bodies are concentrated in the AVPV and Arc GnRH cells in vivo is less clear, however. GnRH mRNA is
nuclei. As with the GnIH system in rodents, kisspeptin cells rhythmic in both OVX and intact female rats, but the GnRH
colocalize sex-steroid receptors, making them a direct target neurons in vivo do not themselves seem to exhibit rhythms in
for sex steroid actions (Smith, 2008). Several lines of evidence clock gene expression (Kriegsfeld et al., 2006; Schirman-
led researchers to suspect that kisspeptin might act as a critical Hildesheim et al., 2006). The GnRH system exhibits time-
node in the circadian network stimulating the LH surge. First, dependent sensitivity to stimulation directly by VIP and
the AVPV was known to be a critical brain region mediating indirectly by AVP (Zhao and Kriegsfeld, 2009; Williams et al.,
the positive feedback effects of estrogen, crucial to the 2011), suggesting that this timekeeping mechanism might
initiation of the preovulatory LH surge and ovulation subserve such daily changes. In GT1-7 cells, Kiss1R is
(Wintermantel et al., 2006). The AVPV expresses both expressed rhythmically when cells are exposed to estradiol
estrogen receptor subtypes (ERa and ERb) and V1a (Shughrue (Tonsfeldt et al., 2011) suggesting that autonomous GnRH
et al., 1997; Kalamatianos et al., 2004). In the AVPV, Kiss1 cell clocks might alter relevant receptor expression to mediate
mRNA expression is at a maximum at the time of the LH these daily changes in sensitivity to upstream neurochemicals.
surge, and these neurons show peak activity, as measured by Ovarian rhythms. The ovary exhibits clock gene rhythms
FOS expression, on the afternoon of proestrous (Robertson in vivo and these rhythms are self-sustained for several days in
358 Circadian Regulation of Endocrine Functions

culture (Nakamura et al., 2010). Both Per1 and Per2 message luciferase driven by the Per2 promoter (Per2-dluc) in rats (He
and protein are rhythmically expressed in follicles, corpora et al., 2007). Several 24 h cycles of Per2-dluc biolumines-
lutea, and the interstitium; furthermore, distinct rhythms of cence, although gradually decreasing in amplitude, could be
cytoplasmic and nuclear localization are observed (Fahrenkrug induced by medium changes or by DEX in nondifferentiating
et al., 2006). These circadian oscillations may be synchronized ovarian luteal cells, proliferative uterine stromal cells, and
directly by rhythmic release of LH and FSH, both of which testicular interstitial cells. In contrast, no rhythms were detected
induce Per1 and Per2 rhythms in cultured granulosa cells (He in decidualizing uterine stromal cells, differentiating Leydig
et al., 2007). Research in birds suggests a connection between cells, or thymocytes. Notably, ovarian granulosa cells displayed
ovarian rhythms and ovulation. Clock genes are rhythmic in a single circadian cycle of bioluminescence; loss of Per2-dluc
preovulatory follicles in Japanese quail, but not in smaller folli- rhythmicity was concomitant with increased LH receptor
cles (Nakao et al., 2007). This follicular clock in turn may expression, a marker of granulose cell differentiation (He
control other physiological processes: the steroidogenic acute et al., 2007). Whether circadian rhythms interfere with differen-
regulatory protein (StAR), a component of the progesterone tiation if not inactivated is an interesting possibility.
synthesis pathway, is expressed rhythmically, again in preovu- Estrogenic feedback to the SCN. Ovarian hormones have
latory but not in smaller follicles. The StAR promoter region profound effects on circadian locomotor activity rhythms. Nor-
includes several E-box elements: the authors used a luciferase mally cycling hamsters display a phase advance every fourth
assay to show that Clock and Bmal1 induce promoter activity day on the day of estrus when E levels are highest (Morin
and that mutation of the E-boxes abolishes this response. et al., 1977). Continuous administration of E shortens the
In rodents, granulosa cells cultured from mouse or rat free-running rhythm compared to OVX controls (Morin et al.,
ovaries of transgenic reporter strains show many cycles of clock 1977), and when such animals are held in constant light that
gene expression (Sellix, 2015). Circadian oscillations in the normally reduces consolidation of the activity bout and can
corpus luteum, which contains both thecal and granulosa cells, lead to splitting of activity, continuous E prevents these changes
appear to be stronger than in the follicle (Chen et al., 2013). (Morin, 1980). Finally, E may act either directly on the SCN,
These data support the notion that clock-driven events in the where ER are sparsely expressed (Shughrue et al., 1997;
gonad are necessary for critical local processes and the timing Hileman et al., 1999; Gundlah et al., 2000; Vida et al., 2008),
of hormone secretion. The clock/StAR relationship has been or indirectly (Figure 10). Cells that express ERa in the preoptic
explored in the mammalian ovary. Levels of StAR were reduced area, amygdala, BNST, and Arc provide input to the SCN (de la
in ovary, as in testis and adrenal, of Bmal1-deficient mice Iglesia et al., 1999). Estrogens may act by altering gap junction
(Alvarez et al., 2008). In female Bmal1 knockouts, suppression connectivity within the SCN. In female rats, estrogen increased
of gonadal steroidogenesis leads to prevention of implantation expression of connexin-36, a subunit that makes up interneu-
(Liu et al., 2014). ronal gap junction channels in the SCN (Shinohara et al.,
Testicular rhythms. Unlike data for the ovary, the evidence for 2001; Rash et al., 2007). Functional gap junctions are revealed
a functional role of clock genes in the testis is complex; period- in cultured SCN cells by dye and electrical coupling (Jiang et al.,
icity of testicular function has been described along multiple 1997; Colwell, 2000; Long et al., 2005), and gap junction
timescales, from ultradian to infradian to circadian and blockers reversibly alter rhythms in electrical activity and
seasonal (Bittman, 2016). Evidence for circadian rhythms in hormone release in vitro (Prosser et al., 1994; Shinohara
T secretion is well established (Van Reeth et al., 1994), but its et al., 2000). Locomotor activity rhythms are dampened in
origins are not well understood (Neill, 2005). There is some male connexin-36 knockout mice, suggesting that gap junc-
evidence of circadian variation in Per gene expression (Zylka tions may be important to normal behavioral rhythms in vivo
et al., 1998; Guo et al., 2005), but other studies have not (Long et al., 2005).
detected such rhythms (Miyamoto and Sancar, 1999; Bittman Interactions of clock genes with ERa and ERb. Per2, a core clock
et al., 2003; Morse et al., 2003; Yamamoto et al., 2004; Alvarez gene, is regulated both transcriptionally and posttranslationally
and Sehgal, 2005; Liu et al., 2007; Bittman, 2016). Additional by multiple cues (Albrecht, 2012). Relevant in the present
findings have led to further confusion: although Bmal1 message context is a feedback loop comprising ovarian hormones, the
is not expressed rhythmically, BMAL1 protein oscillates in Ley- ER signaling network, and Per2. ERb expression is rhythmic
dig cells, and knockout of Bmal1 reduces T production and in several peripheral tissues in wild-type but not in arrhythmic
fertility in mice (Alvarez and Sehgal, 2005; Alvarez et al., Bmal1 knockout mice. Its promoter region includes an E-box,
2008). Although much work appears contradictory, real-time a target for the CLOCK:BMAL1 complex, suggesting direct
reporting of bioluminescence of testis slices from Per2::luc regulation by the circadian clock (Cai et al., 2008). Per2
mice confirms that at least ex vivo, circadian rhythmicity occurs enhances ERa protein degradation, and Per2 suppression
in testes (Nishide et al., 2014). This method, however, does not leads to ERa stabilization. Estrogen can in turn feedback
clarify the cell type that produces the oscillation. The resolution induce Per2 expression (Gery et al., 2007). Such feedback
of apparently contradictory evidence likely lies in the heteroge- mechanisms point to a critical role for peripheral circadian
neity of testicular tissue, with different temporal patterns of regulation in tissue homeostasis. While this work was done
change unique to various cell types (Bittman, 2016). in a cancer cell model, it is likely to apply to other ERa
Circadian rhythms and differentiation of endocrine tissue. containing cells.
Alvarez et al. have suggested that circadian rhythms are sus- Androgenic feedback to the SCN. Castration of mice lengthens
pended during differentiation based on data from testis and the free-running period, decreases the precision of activity
thymus (Alvarez et al., 2003; Alvarez and Sehgal, 2005). This onset, and reduces consolidation of activity (Daan et al.,
contention is supported by in vitro data using a destabilized 1975; Karatsoreos et al., 2007). Both T and dihydrotestosterone
Circadian Regulation of Endocrine Functions 359

Figure 10 Schematic representation of differences between the sites of action of estrogenic and androgenic hormones on the suprachiasmatic
nucleus (SCN). Estrogen receptor (ER)-rich nuclei (pink) including the retina, the intergeniculate leaflet (IGL), and the dorsal raphe (DR), via the
median raphe project to the SCN. The DR also contains androgen receptors (ARs, blue) and projects to the SCN. ERs are largely constrained to the
shell SCN region. Unlike ERs, ARs are densely localized in the core SCN. The SCN regulates circadian timing in physiology and behavior by sending
outputs to the neuroendocrine systems, including the gonads.

(DHT: a nonaromatizable androgen) restore the gonadally length (Arendt, 2006; Johnston and Skene, 2015). Melatonin
intact phenotype, suggesting that the effects of T are mediated is also a chronotherapeutic and can phase shift circadian
by androgen receptors (ARs) rather than by ERs. While it is rhythms in both rodents and humans, although only at times
likely that androgens modulate locomotor behavior at multiple when melatonin would not be secreted endogenously
sites, the AR is concentrated in the core region of the SCN of (Armstrong, 1989; Lewy et al., 1998). Here we consider the
mice, and local administration of T to the SCN restores regulation of melatonin and its potential effects on clocks
intact-typical circadian periods, strongly indicating a direct within the body.
effect of androgens, mediated by the AR, in the SCN (Figure 8; SCN regulation of melatonin secretion. Melatonin is secreted at
Karatsoreos and Silver, 2007; Model et al., 2015). More diffuse night by the pineal gland; its synthesis is governed rhythmically
AR expression has also been detected in other species (see by cues from the SCN via a multisynaptic pathway with
Karatsoreos et al., 2007 for review). Androgenic effects on synapses in the PVN, the medial forebrain bundle, and the
period in males are mediated by an increased sensitivity of superior cervical ganglion of the spinal cord (Moore and Klein,
the SCN to light cues from the retina (Butler et al., 2012). 1974). From the superior cervical ganglion, b-adrenergic
Although the mechanism is not known, this may be associated neurons drive pineal melatonin secretion during the dark but
with an increased response to NMDA as observed in the hippo- not during the light when their activity is inhibited (Cassone
campus (Pouliot et al., 1996). et al., 1990; Larsen et al., 1998; Larsen, 1999; Teclemariam-
Comparison of AR expression indicates sex differences in Mesbah et al., 1999; Card, 2000). Both stimulatory and inhib-
the SCN in humans (Fernandez-Guasti et al., 2000) and in itory outputs of the SCN are implicated in controlling the final
rodents (Iwahana et al., 2008). Western blots and immuno- noradrenergic stimulation of the pineal gland at night. During
chemistry indicate that ARs are more highly expressed in the day, GABAergic projections to the PVN inhibit excitatory
male than in female mice; gonadectomy eliminates and output to the pineal and therefore suppress melatonin
androgen treatment restores these sex differences. At the behav- (Kalsbeek et al., 2000). During the night, stimulatory glutama-
ioral level, gonadectomy produces a dramatic loss of the tergic outputs from the SCN to the PVN stimulate melatonin
evening activity onset bout in males, but has no such effect synthesis (Perreau-Lenz et al., 2004). As with other hormonal
in females. Consistent with the implications of these findings, systems, lesions of the SCN abolish circadian rhythms in
treatment with T or DHT restores male locomotor activity and melatonin production and secretion (Scott et al., 1995;
eliminates sex differences in the behavioral response (Iwahana Tessonneaud et al., 1995). Given its high-amplitude rhythm,
et al., 2008). strong dependence on circadian phase, and rapid rise from
basal levels, melatonin secretion patterns are the gold standard
for measuring clock phase in humans, usually defined as the
1.12.6 Circadian Regulation of Melatonin time of melatonin secretion onset under dim light conditions
(Lewy and Sack, 1989).
Overview. The study of melatonin and the clock grew from Melatonin as an entraining agent (endogenous and exogenous).
a strong literature on the mechanisms of seasonality, and mela- Stimuli such as light and melatonin can shift the phase of the
tonin’s important role as an endocrine representation of day circadian clock, but only when they are administered at
360 Circadian Regulation of Endocrine Functions

1.12.7 Circadian Dysfunction and Chronotherapy

The universal experience of our sleep–wake cycles and feelings


associated with their disruption lend immediate salience to this
realm of basic research. It is easy to intuit how physiological
functioning suffers without an internal circadian clock synchro-
nized to the environment. Most of us have experienced this
state, manifested by general feelings of malaise after a long
flight to a new time zone. While we can recover from acute
jet lag within a few days, millions of frequent flyers, shift
workers, and those with sleep disorders are exposed repeatedly
to temporal disruptions. However, it has also been demon-
Figure 11 Melatonin is most effective in phase shifting the circadian strated that the nearly weekly changes in sleep patterns caused
clock at times when endogenous melatonin secretion is low. Here, the
by workdays and days off also cause significant temporal disor-
phase response curve to 3.0 mg of oral melatonin is shown, with
ganization, a phenomenon known as ‘social jet lag’ (Wittmann
maximum advances in the afternoon and delays in the morning. Endog-
enous melatonin secretion (gray), in contrast, is high during the night-
et al., 2006).
time hours. Modified from Burgess, H.J., Revell, V.L., Molina, T.A., To return to points made in the first paragraphs of this
Eastman, C.I., 2010. Human phase response curves to three days of chapter, the function of the SCN brain clock is to synchronize
daily melatonin: 0.5 mg versus 3.0 mg. J. Clin. Endocrinol. Metab. the many cellular clocks in the rest of the body. The function
95 (7), 3325–3331. of individual cellular clocks is to generate oscillations in the
production of clock-controlled output genes, which in turn
drive overt rhythms in physiology and behavior. This is
particular times of day. The relationship of time of treatment to achieved, in part, by oscillating hormones of the adrenal and
the magnitude of response is described by a phase response gonadal axes, which have been discussed here.
curve. Melatonin can cause phase advances, phase delays, or Under normal conditions, signals from the SCN to clocks in
have no effect depending on its dose and time of administra- peripheral tissues produce coherent and stable phase relation-
tion (Figure 11). It is noteworthy, however, that the most effec- ships among different organs. Of course, the precise phase of
tive phase advancing effect occurs in the evening, well before tissue clocks is specific to each organ due to tissue-specific func-
bedtime, at a time when melatonin suppression is minimal. tions. When perturbed, such as occurs following alterations in
Therefore, endogenous melatonin secretion is unlikely to play lighting cycle subsequent to long distance travel or shift work,
a role in phase resetting the central clock. the SCN shifts in phase much more quickly (within a day or
Whether endogenous melatonin secretion plays a role in two) than do peripheral clocks (which can take a week or
entraining peripheral clocks is not well studied, although this more), creating desynchrony (Lowrey et al., 2000). Similarly,
remains an intriguing mechanism for clock coordination (Pevet systematic manipulation of external cues such as unusual
and Challet, 2011). Melatonin receptors are described in feeding times can disturb the usual relationship between
several endocrine tissues, including the pituitary gland, adrenal peripheral clocks and the SCN clock (Damiola et al., 2000),
gland, pancreas, and ovaries (Slominski et al., 2012). In the and these effects of food are independent of the SCN and
pituitary gland, long and short duration endogenous mela- activity rhythms (Pitts et al., 2003).
tonin secretion differentially entrain clock genes in the pars How might mistimed feeding-related signals contribute to
tuberalis; the altered relationships among clock genes are circadian desynchrony? The HPA axis activity is modulated
thought to be involved in day-length encoding in seasonally by leptin, a hormone involved in regulating feeding behavior
breeding species (Lincoln et al., 2002). Until melatonin phase and energy homeostasis. Leptin binds to its receptors in the
response curves are defined for other specific peripheral tissues, hypothalamus and midbrain and signals the availability of
it will not be possible to determine the role that melatonin peripheral energy stores (Stieg et al., 2015). Leptin is released
plays in coordinating peripheral rhythms. The one area in rhythmically and in antiphase to the release of cortisol
which endogenous melatonin has a strong documented (Aschbacher et al., 2014). Elevated leptin inhibits glucocorti-
entraining effect is in maternal–fetal communication. Maternal coid activity in the medial hypothalamus (Grosbellet et al.,
melatonin synchronizes the clocks of rat pups (Bellavia et al., 2015) and thus suppresses appetite and stimulates energy
2006). expenditure. Reduction in leptin levels increases glucocorticoid
Exogenous melatonin, when administered at the proper release, activating circuits that stimulate eating (Ahima et al.,
times, can have strong phase resetting properties that are worth 1996). Thus the interaction between rhythms in leptin and
highlighting. Melatonin is an effective treatment to synchronize cortisol provide a pathway by which neuroendocrine stimuli
the free-running rhythms of blind subjects (Sack et al., 2000). and the brain circuits on which they act can influence circadian
In sighted individuals, evening melatonin can phase advance rhythmicity and the production of clock-controlled genes by
rhythms and its effect is additive to that of light (Burke et al., altering normal phase relationships among organs.
2013). Some affective disorders, such as seasonal affective Chronic circadian dysregulation has significant negative
disorder and major depression, are associated with altered consequences for health and has been associated with
clock phase (Lewy et al., 2006; Emens et al., 2009). In the increased cancer susceptibility, inflammation, and altered
former, melatonin timed to normalize clock phase improves metabolism (Karatsoreos et al., 2011; Golombek et al.,
mood (Lewy et al., 2006). 2013). Experimental evidence indicates that individuals with
Circadian Regulation of Endocrine Functions 361

chronic circadian disruptions often present with clinical


pathologies, pointing to the importance of synchrony between
brain and environmental rhythms. Elderly mice subjected to
temporal disruptions equivalent to a flight from Washington
to Paris, once a week for 8 weeks, suffered high mortality rates
due to these phase shifts (Davidson et al., 2006). Flight atten-
dants who frequently travel across time zones exhibit cognitive
deficits associated with reductions in temporal lobe structures,
have slower reaction times, and make more errors on cognitive
tasks (Cho et al., 2000; Cho, 2001). Shift workers have a higher
incidence of cancer (Hansen, 2006; Kubo et al., 2006; O’Leary
et al., 2006; Patel, 2006; Conlon et al., 2007), diabetes
(Sanborn et al., 1982; Robinson et al., 1990; Poole et al.,
1992; Karlsson et al., 2005; Morikawa et al., 2005), ulcers
(Kolmodin-Hedman and Swensson, 1975; Segawa et al.,
1987; Costa, 1996; Koda et al., 2000), hypertension and cardio-
vascular disease (Costa, 1996; Alstadhaug et al., 2005; Hwang
and Lee, 2005; Wolk et al., 2005; Kivimaki et al., 2006),
psychological disorders (Skipper et al., 1990; De Koninck,
1991; Leonard et al., 1998; Venuta et al., 1999; Munakata
et al., 2001; Bildt and Michelsen, 2002), and are a host of other
clinical issues. Social jet lag has also been associated with alter-
ations in metabolism and increases in obesity (Roenneberg
et al., 2012).
One important aspect of the circadian system is individual
differences – and these appear to be characteristic of individ-
uals. In one study, eight hormones were measured over two
different nights (Schulz et al., 1996). The temporal organiza- Figure 12 Circadian genes, genes associated with disease and drug
tion of hormone secretion into the blood was highly indi- targets. There is substantial overlap between circadian genes, known
vidual, and the intraindividual patterns were conserved. disease-associated genes, and drug targets (Zhang et al., 2014).
Colloquially, we have similar intuitive experiences and can
often categorize ourselves as larks or owls. A major break- Brown, 2008). These cell-based clock models are ideal for
through for circadian biologists interested in humans has target discovery and chemical biology and some of the
been the use of cells of peripheral tissues as proxies for the screening principles and measuring technologies may be
oscillators found in the SCN. Interindividual genetic differ- useful in the study of relationships between hormones, clock
ences appear to be manifest similarly in central and periph- genes, and clock-controlled genes at various levels of the neu-
eral oscillators (Yagita et al., 2001; Pando et al., 2002). raxis (Figure 12).
Several strategies for assessing individual differences in We have emphasized in this chapter the significance of the
timing, with the potential for optimizing medical treatment, circadian system in the brain and periphery in studies of
are promising. Molecular gene cycling is useful for deter- endocrine physiology and behavior. Circadian regulation
mining body time with important applications in personal- and dysregulation have profound consequences in health
ized medicine, including cardiovascular disease and cancer, and disease. It is clear that precise temporal organization
our leading causes of death (Liu et al., 2007). Detection of helps to promote optimal function of many endocrine and
an individual’s body time via a single-time-point assay can neuroendocrine systems. Disruption of this organization is
be achieved by a ‘molecular timetable’ composed of also linked to disease, even at the molecular level. Clock genes
hundreds of ‘time-indicating genes’ (Ueda et al., 2004). link estrogen receptors and breast cancer, and their dysregula-
Diurnal protein cycling in blood using high-throughput pro- tion has been implicated in cancers of other endocrine tissues
teomics is also an attractive possibility; blood draws are mini- (Gery et al., 2007; Suzuki et al., 2008). Understanding circa-
mally invasive, and the proteomic profile can indicate what is dian rhythms can increase experimental control in empirical
happening elsewhere in the body in health and disease studies and improved understanding of tissue-specific
(Martino et al., 2007). Rhythms in fibroblasts also vary rhythms should help to refine clinical diagnosis and hasten
among individuals, and the correlation between activity the development of individualized, timed courses of
patterns and fibroblast rhythms suggest that these cells treatment.
accurately indicate circadian properties (Brown et al.,
2005). Because peripheral cells oscillate and are easily acces-
sible, it has also been possible to introduce genetic materials Acknowledgments
into such cells by transfection or transduction; this holds
promise for the analysis of biochemical mechanisms of oscil- We thank Erica Mezias and Malini Riddle for invaluable assistance in
lation, quantitative trait mapping, and the search for indi- preparing this chapter and figures. Our work has been supported by
vidual chronotypes (Brown et al., 2008; Cuninkova and NIH grants MH075045, NS37919, and by NSF grant 125605.
362 Circadian Regulation of Endocrine Functions

References Brown, S.A., Fleury-Olela, F., Nagoshi, E., Hauser, C., Juge, C., Meier, C.A.,
Chicheportiche, R., Dayer, J.M., Albrecht, U., Schibler, U., 2005. The period length
Aedo, A.R., Landgren, B.M., Diczfalusy, E., 1981. Studies on ovarian and adrenal of fibroblast circadian gene expression varies widely among human individuals.
steroids at different phases of the menstrual cycle: II. A comparative assessment of PLoS Biol. 3 (10), e338.
the circadian variation in steroid and lutropin levels during the follicular, peri- Brown, S.A., Kunz, D., Dumas, A., Westermark, P.O., Vanselow, K., Tilmann-
ovulatory and luteal phases. Contraception 23 (4), 407–424. Wahnschaffe, A., Herzel, H., Kramer, A., 2008. Molecular insights into human
Ahima, R.S., Prabakaran, D., Mantzoros, C., Qu, D., Lowell, B., Maratos-Flier, E., daily behavior. Proc. Natl. Acad. Sci. U.S.A. 105 (5), 1602–1607.
Flier, J.S., 1996. Role of leptin in the neuroendocrine response to fasting. Nature Brown-Grant, K., Raisman, G., 1977. Abnormalities in reproductive function associated
382 (6588), 250–252. with the destruction of the suprachiasmatic nuclei in female rats. Proc. R. Soc.
Akhtar, R.A., Reddy, A.B., Maywood, E.S., Clayton, J.D., King, V.M., Smith, A.G., Lond. B Biol. Sci. 198 (1132), 279–296.
Gant, T.W., Hastings, M.H., Kyriacou, C.P., 2002. Circadian cycling of the mouse Buhr, E.D., Yoo, S.H., Takahashi, J.S., 2010. Temperature as a universal resetting cue
liver transcriptome, as revealed by cDNA microarray, is driven by the supra- for mammalian circadian oscillators. Science 330 (6002), 379–385.
chiasmatic nucleus. Curr. Biol. 12 (7), 540–550. Buijs, R.M., Hermes, M.H., Kalsbeek, A., 1998. The suprachiasmatic nucleus-
Albers, H.E., Yogev, L., Todd, R.B., Goldman, B.D., 1985. Adrenal corticoids in paraventricular nucleus interactions: a bridge to the neuroendocrine and auto-
hamsters: role in circadian timing. Am. J. Physiol. 248 (4 Pt 2), R434–R438. nomic nervous system. Prog. Brain Res. 119, 365–382.
Albrecht, U., 2012. Timing to perfection: the biology of central and peripheral circadian Buijs, R.M., Kalsbeek, A., 2001. Hypothalamic integration of central and peripheral
clocks. Neuron 74 (2), 246–260. clocks. Nat. Rev. Neurosci. 2 (7), 521–526.
Alstadhaug, K.B., Salvesen, R., Bekkelund, S.I., 2005. Seasonal variation in migraine. Buijs, R.M., van Eden, C.G., Goncharuk, V.D., Kalsbeek, A., 2003. The biological clock
Cephalalgia 25 (10), 811–816. tunes the organs of the body: timing by hormones and the autonomic nervous
Alvarez, J.D., Chen, D., Storer, E., Sehgal, A., 2003. Non-cyclic and developmental system. J. Endocrinol. 177 (1), 17–26.
stage-specific expression of circadian clock proteins during murine spermato- Buijs, R.M., Wortel, J., Van Heerikhuize, J.J., Feenstra, M.G., Ter Horst, G.J.,
genesis. Biol. Reprod. 69 (1), 81–91. Romijn, H.J., Kalsbeek, A., 1999. Anatomical and functional demonstration of
Alvarez, J.D., Hansen, A., Ord, T., Bebas, P., Chappell, P.E., Giebultowicz, J.M., a multisynaptic suprachiasmatic nucleus adrenal (cortex) pathway. Eur. J. Neu-
Williams, C., Moss, S., Sehgal, A., 2008. The circadian clock protein BMAL1 is rosci. 11 (5), 1535–1544.
necessary for fertility and proper testosterone production in mice. J. Biol. Rhythms Buijs, R.M., Wortel, J., Van Heerikhuize, J.J., Kalsbeek, A., 1997. Novel environment
23 (1), 26–36. induced inhibition of corticosterone secretion: physiological evidence for a supra-
Alvarez, J.D., Sehgal, A., 2005. The thymus is similar to the testis in its pattern of chiasmatic nucleus mediated neuronal hypothalamo-adrenal cortex pathway. Brain
circadian clock gene expression. J. Biol. Rhythms 20 (2), 111–121. Res. 758 (1–2), 229–236.
Amir, S., Lamont, E.W., Robinson, B., Stewart, J., 2004. A circadian rhythm in the Burgess, H.J., Revell, V.L., Molina, T.A., Eastman, C.I., 2010. Human phase response
expression of PERIOD2 protein reveals a novel SCN-controlled oscillator in the oval curves to three days of daily melatonin: 0.5 mg versus 3.0 mg. J. Clin. Endocrinol.
nucleus of the bed nucleus of the stria terminalis. J. Neurosci. 24 (4), 781–790. Metab. 95 (7), 3325–3331.
Antle, M.C., Foley, D.K., Foley, N.C., Silver, R., 2003. Gates and oscillators: a network Burke, T.M., Markwald, R.R., Chinoy, E.D., Snider, J.A., Bessman, S.C., Jung, C.M.,
model of the brain clock. J. Biol. Rhythms 18 (4), 339–350. Wright Jr., K.P., 2013. Combination of light and melatonin time cues for phase
Antle, M.C., Foley, N.C., Foley, D.K., Silver, R., 2007. Gates and oscillators II: zeit- advancing the human circadian clock. Sleep 36 (11), 1617–1624.
gebers and the network model of the brain clock. J. Biol. Rhythms 22 (1), 14–25. Butler, M.P., Rainbow, M.N., Rodriguez, E., Lyon, S.M., Silver, R., 2012. Twelve-hour
Antle, M.C., Silver, R., 2005. Orchestrating time: arrangements of the brain circadian days in the brain and behavior of split hamsters. Eur. J. Neurosci. 36 (4),
clock. Trends Neurosci. 28 (3), 145–151. 2556–2566.
Apter, D., Butzow, T.L., Laughlin, G.A., Yen, S.S., 1993. Gonadotropin-releasing Butler, M.P., Silver, R., 2011. Divergent photic thresholds in the non-image-forming
hormone pulse generator activity during pubertal transition in girls: pulsatile and visual system: entrainment, masking and pupillary light reflex. Proc. Biol. Sci. 278
diurnal patterns of circulating gonadotropins. J. Clin. Endocrinol. Metab. 76 (4), (1706), 745–750.
940–949. Cai, W., Rambaud, J., Teboul, M., Masse, I., Benoit, G., Gustafsson, J.A.,
Arendt, J., 2006. Melatonin and human rhythms. Chronobiol. Int. 23 (1–2), 21–37. Delaunay, F., Laudet, V., Pongratz, I., 2008. Expression levels of estrogen receptor
Armstrong, S.M., 1989. Melatonin and circadian control in mammals. Experientia 45 b are modulated by components of the molecular clock. Mol. Cell Biol. 28 (2),
(10), 932–938. 784–793.
Aschbacher, K., Rodriguez-Fernandez, M., van Wietmarschen, H., Tomiyama, A.J., Campbell, S.S., Murphy, P.J., 1998. Extraocular circadian phototransduction in
Jain, S., Epel, E., Doyle 3rd, F.J., van der Greef, J., 2014. The hypothalamic- humans. Science 279 (5349), 396–399.
pituitary-adrenal-leptin axis and metabolic health: a systems approach to resil- Carandente, F., Angeli, A., Candiani, G.B., Crosignani, P.G., Dammacco, F.,
ience, robustness and control. Interface Focus 4 (5), 20140020. De Cecco, L., Marrama, P., Massobrio, M., Martini, L., 1989. Rhythms in the
Asher, G., Sassone-Corsi, P., 2015. Time for food: the intimate interplay between ovulatory cycle. 2nd: LH, FSH, estradiol and progesterone. Chronobiologia 16 (4),
nutrition, metabolism, and the circadian clock. Cell 161 (1), 84–92. 353–363.
Balsalobre, A., Brown, S.A., Marcacci, L., Tronche, F., Kellendonk, C., Reichardt, H.M., Card, J.P., 2000. Pseudorabies virus and the functional architecture of the circadian
Schutz, G., Schibler, U., 2000. Resetting of circadian time in peripheral tissues by timing system. J. Biol. Rhythms 15 (6), 453–461.
glucocorticoid signaling. Science 289 (5488), 2344–2347. Cassone, V.M., Forsyth, A.M., Woodlee, G.L., 1990. Hypothalamic regulation of
Balsalobre, A., Damiola, F., Schibler, U., 1998. A serum shock induces circadian gene circadian noradrenergic input to the chick pineal gland. J. Comp. Physiol. A 167
expression in mammalian tissue culture cells. Cell 93 (6), 929–937. (2), 187–192.
Barinaga, M., 2002. Circadian clock. How the brain’s clock gets daily enlightenment. Chappell, P.E., White, R.S., Mellon, P.L., 2003. Circadian gene expression regulates
Science 295 (5557), 955–957. pulsatile gonadotropin-releasing hormone (GnRH) secretory patterns in the hypo-
Bartness, T.J., Song, C.K., Demas, G.E., 2001. SCN efferents to peripheral tissues: thalamic GnRH-secreting GT1-7 cell line. J. Neurosci. 23 (35), 11202–11213.
implications for biological rhythms. J. Biol. Rhythms 16 (3), 196–204. Chen, H., Zhao, L., Chu, G., Kito, G., Yamauchi, N., Shigeyoshi, Y., Hashimoto, S.,
Bechtel, W., 2016. Using computational models to discover and understand mecha- Hattori, M.A., 2013. FSH induces the development of circadian clockwork in rat
nisms. Stud. Hist. Philos. Sci. 56, 113–121. granulosa cells via a gap junction protein Cx43-dependent pathway. Am. J. Physiol.
Bellavia, S.L., Carpentieri, A.R., Vaque, A.M., Macchione, A.F., Vermouth, N.T., 2006. Endocrinol. Metab. 304 (6), E566–E575.
Pup circadian rhythm entrainment–effect of maternal ganglionectomy or pineal- Cho, K., 2001. Chronic ‘jet lag’ produces temporal lobe atrophy and spatial cognitive
ectomy. Physiol. Behav. 89 (3), 342–349. deficits. Nat. Neurosci. 4 (6), 567–568.
Bildt, C., Michelsen, H., 2002. Gender differences in the effects from working Cho, K., Ennaceur, A., Cole, J.C., Suh, C.K., 2000. Chronic jet lag produces cognitive
conditions on mental health: a 4-year follow-up. Int. Arch. Occup. Environ. Health deficits. J. Neurosci. 20 (6), RC66.
75 (4), 252–258. Christian, C.A., Mobley, J.L., Moenter, S.M., 2005. Diurnal and estradiol-dependent
Bittman, E.L., 2016. Timing in the testis. J. Biol. Rhythms 31 (1), 12–36. changes in gonadotropin-releasing hormone neuron firing activity. Proc. Natl.
Bittman, E.L., Doherty, L., Huang, L., Paroskie, A., 2003. Period gene expression in Acad. Sci. U.S.A. 102 (43), 15682–15687.
mouse endocrine tissues. Am. J. Physiol. 285 (3), R561–R569. Christian, C.A., Moenter, S.M., 2008. Vasoactive intestinal polypeptide can excite
Brown, S.A., 2016. Circadian metabolism: from mechanisms to metabolomics and gonadotropin-releasing hormone neurons in a manner dependent on estradiol and
medicine. Trends Endocrinol. Metab. 27 (6), 415–426. gated by time of day. Endocrinology 149 (6), 3130–3136.
Circadian Regulation of Endocrine Functions 363

Chu, A., Zhu, L., Blum, I.D., Mai, O., Leliavski, A., Fahrenkrug, J., Oster, H., Evans, J.A., Leise, T.L., Castanon-Cervantes, O., Davidson, A.J., 2013. Dynamic
Boehm, U., Storch, K.F., 2013. Global but not gonadotrope-specific disruption of interactions mediated by nonredundant signaling mechanisms couple circadian
Bmal1 abolishes the luteinizing hormone surge without affecting ovulation. Endo- clock neurons. Neuron 80 (4), 973–983.
crinology 154 (8), 2924–2935. Everett, J.W., Sawyer, C.H., 1950. A 24-hour periodicity in the “LH-release apparatus”
Colombo, J.A., Baldwin, D.M., Sawyer, C.H., 1974. Timing of the estrogen-induced of female rats, disclosed by barbiturate sedation. Endocrinology 47 (3), 198–218.
release of LH in ovariectomized rats under an altered lighting schedule. Proc. Fahrenkrug, J., Georg, B., Hannibal, J., Hindersson, P., Gras, S., 2006. Diurnal
Soc. Exp. Biol. Med. 145 (3), 1125–1127. rhythmicity of the clock genes Per1 and Per2 in the rat ovary. Endocrinology 147
Colwell, C.S., 2000. Rhythmic coupling among cells in the suprachiasmatic nucleus. (8), 3769–3776.
J. Neurobiol. 43 (4), 379–388. Fahrenkrug, J., Hannibal, J., Georg, B., 2008. Diurnal rhythmicity of the canonical
Conlon, M., Lightfoot, N., Kreiger, N., 2007. Rotating shift work and risk of prostate clock genes Per1, Per2 and Bmal1 in the rat adrenal gland is unaltered after
cancer. Epidemiology 18 (1), 182–183. hypophysectomy. J. Neuroendocrinol. 20 (3), 323–329.
Costa, G., 1996. The impact of shift and night work on health. Appl. Ergon. 27 (1), Fernandez, D.C., Chang, Y.T., Hattar, S., Chen, S.K., 2016. Architecture of retinal
9–16. projections to the central circadian pacemaker. Proc. Natl. Acad. Sci. U.S.A. 113
Cuninkova, L., Brown, S.A., 2008. Peripheral circadian oscillators: interesting mech- (21), 6047–6052.
anisms and powerful tools. Ann. N.Y. Acad. Sci. 1129. Fernandez-Guasti, A., Kruijver, F.P., Fodor, M., Swaab, D.F., 2000. Sex differences in
Czeisler, C.A., Klerman, E.B., 1999. Circadian and sleep-dependent regulation of the distribution of androgen receptors in the human hypothalamus. J. Comp.
hormone release in humans. Recent Prog. Horm. Res. 54, 97–130 discussion Neurol. 425 (3), 422–435.
130–132. Fitzgerald, K., Zucker, I., 1976. Circadian organization of the estrous cycle of the
Daan, S., Damassa, D., Pittendrigh, C.S., Smith, E.R., 1975. An effect of castration golden hamster. Proc. Natl. Acad. Sci. U.S.A. 73 (8), 2923–2927.
and testosterone replacement on a circadian pacemaker in mice (Mus musculus). Foradori, C.D., Coolen, L.M., Fitzgerald, M.E., Skinner, D.C., Goodman, R.L.,
Proc. Natl. Acad. Sci. U.S.A. 72 (9), 3744–3747. Lehman, M.N., 2002. Colocalization of progesterone receptors in parvicellular
Damiola, F., Le Minh, N., Preitner, N., Kornmann, B., Fleury-Olela, F., Schibler, U., dynorphin neurons of the ovine preoptic area and hypothalamus. Endocrinology
2000. Restricted feeding uncouples circadian oscillators in peripheral tissues from 143 (11), 4366–4374.
the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 14 (23), Funabashi, T., Shinohara, K., Mitsushima, D., Kimura, F., 2000. Gonadotropin-
2950–2961. releasing hormone exhibits circadian rhythm in phase with arginine-vasopressin
Davidson, A.J., Sellix, M.T., Daniel, J., Yamazaki, S., Menaker, M., Block, G.D., 2006. in co-cultures of the female rat preoptic area and suprachiasmatic nucleus.
Chronic jet-lag increases mortality in aged mice. Curr. Biol. 16 (21), R914–R916. J. Neuroendocrinol. 12 (6), 521–528.
De Koninck, J., 1991. Biological rhythms associated with sleep and psychological Gallagher, T.F., Yoshida, K., Roffwarg, H.D., Fukushima, D.K., Weitzman, E.D.,
adjustment. J. Psychiatry Neurosci. 16 (3), 115–122. Hellman, L., 1973. ACTH and cortisol secretory patterns in man. J. Clin. Endo-
de la Iglesia, H.O., Blaustein, J.D., Bittman, E.L., 1999. Oestrogen receptor-a- crinol. Metab. 36 (6), 1058–1068.
immunoreactive neurones project to the suprachiasmatic nucleus of the female Garcia-Segura, L.M., McCarthy, M.M., 2004. Minireview: role of glia in neuroendocrine
Syrian hamster. J. Neuroendocrinol. 11 (7), 481–490. function. Endocrinology 145 (3), 1082–1086.
de la Iglesia, H.O., Meyer, J., Carpino Jr., A., Schwartz, W.J., 2000. Antiphase Gerhold, L.M., Horvath, T.L., Freeman, M.E., 2001. Vasoactive intestinal peptide fibers
oscillation of the left and right suprachiasmatic nuclei. Science 290 (5492), innervate neuroendocrine dopaminergic neurons. Brain Res. 919 (1), 48–56.
799–801. Gerhold, L.M., Wise, P.M., 2006. Vasoactive intestinal polypeptide regulates dynamic
de la Iglesia, H.O., Meyer, J., Schwartz, W.J., 2003. Lateralization of circadian changes in astrocyte morphometry: impact on gonadotropin-releasing hormone
pacemaker output: activation of left- and right-sided luteinizing hormone-releasing neurons. Endocrinology 147 (5), 2197–2202.
hormone neurons involves a neural rather than a humoral pathway. J. Neurosci. 23 Gery, S., Virk, R.K., Chumakov, K., Yu, A., Koeffler, H.P., 2007. The clock gene Per2
(19), 7412–7414. links the circadian system to the estrogen receptor. Oncogene 26 (57),
de Roux, N., Genin, E., Carel, J.C., Matsuda, F., Chaussain, J.L., Milgrom, E., 2003. 7916–7920.
Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived Gibson, E., Humber, S.A., Jain, S., Williams, W.P.I., Zhao, S., Bentley, G.,
peptide receptor GPR54. Proc. Natl. Acad. Sci. U.S.A. 100 (19), 10972–10976. Tsutsui, K., Kriegsfeld, L., 2008. The preovulatory luteinizing hormone surge is
Dibner, C., Schibler, U., 2015. Circadian timing of metabolism in animal models and associated with deactivation of the gonadotropin-releasing hormone (GnIH)
humans. J. Intern. Med. 277 (5), 513–527. neuronal system. Endocrinology. http://dx.doi.org/10.1210/en.2008-0316.
Dickmeis, T., 2009. Glucocorticoids and the circadian clock. J. Endocrinol. 200 (1), Published online, 19 June.
3–22. Gillespie, J.M., Chan, B.P., Roy, D., Cai, F., Belsham, D.D., 2003. Expression of
Doi, M., Hirayama, J., Sassone-Corsi, P., 2006. Circadian regulator CLOCK is circadian rhythm genes in gonadotropin-releasing hormone-secreting GT1-7
a histone acetyltransferase. Cell 125 (3), 497–508. neurons. Endocrinology 144 (12), 5285–5292.
Drouyer, E., LeSauter, J., Hernandez, A.L., Silver, R., 2010. Specializations of gastrin- Gillette, M.U., Reppert, S.M., 1987. The hypothalamic suprachiasmatic nuclei:
releasing peptide cells of the mouse suprachiasmatic nucleus. J. Comp. Neurol. circadian patterns of vasopressin secretion and neuronal activity in vitro. Brain Res.
518 (8), 1249–1263. Bull. 19 (1), 135–139.
Dubey, A.K., Puri, C.P., Puri, V., Anand Kumar, T.C., 1983. Day and night levels of Golombek, D.A., Casiraghi, L.P., Agostino, P.V., Paladino, N., Duhart, J.M.,
hormones in male rhesus monkeys kept under controlled or constant environmental Plano, S.A., Chiesa, J.J., 2013. The times they’re a-changing: effects of circadian
light. Experientia 39 (2), 207–209. desynchronization on physiology and disease. J. Physiol. Paris 107 (4), 310–322.
Duffield, G.E., Best, J.D., Meurers, B.H., Bittner, A., Loros, J.J., Dunlap, J.C., 2002. Goodman, R.L., Lehman, M.N., Smith, J.T., Coolen, L.M., de Oliveira, C.V.,
Circadian programs of transcriptional activation, signaling, and protein turnover Jafarzadehshirazi, M.R., Pereira, A., Iqbal, J., Caraty, A., Ciofi, P., Clarke, I.J.,
revealed by microarray analysis of mammalian cells. Curr. Biol. 12 (7), 551–557. 2007. Kisspeptin neurons in the arcuate nucleus of the ewe express both
Dunkel, L., Alfthan, H., Stenman, U.H., Selstam, G., Rosberg, S., Albertsson- dynorphin A and neurokinin B. Endocrinology 148 (12), 5752–5760.
Wikland, K., 1992. Developmental changes in 24-hour profiles of luteinizing Gooley, J.J., Saper, C.B., 2007. Is food-directed behavior an appropriate measure of
hormone and follicle-stimulating hormone from prepuberty to midstages of puberty circadian entrainment to restricted daytime feeding? J. Biol. Rhythms 22 (6),
in boys. J. Clin. Endocrinol. Metab. 74 (4), 890–897. 479–483 discussion 484–477.
Dunn, J., Scheving, L., Millet, P., 1972. Circadian variation in stress-evoked increases Gottsch, M.L., Cunningham, M.J., Smith, J.T., Popa, S.M., Acohido, B.V.,
in plasma corticosterone. Am. J. Physiol. 223 (2), 402–406. Crowley, W.F., Seminara, S., Clifton, D.K., Steiner, R.A., 2004. A role for kiss-
Egli, M., Bertram, R., Sellix, M.T., Freeman, M.E., 2004. Rhythmic secretion of peptins in the regulation of gonadotropin secretion in the mouse. Endocrinology
prolactin in rats: action of oxytocin coordinated by vasoactive intestinal polypeptide 145 (9), 4073–4077.
of suprachiasmatic nucleus origin. Endocrinology 145 (7). Gray, G.D., Soderstein, P., Tallentire, D., Davidson, J.M., 1978. Effects of lesions in
Emens, J., Lewy, A., Kinzie, J.M., Arntz, D., Rough, J., 2009. Circadian misalignment various structures of the suprachiasmatic-preoptic region on LH regulation and
in major depressive disorder. Psychiatry Res. 168 (3), 259–261. sexual behavior in female rats. Neuroendocrinology 25 (3), 174–191.
Etchegaray, J.P., Machida, K.K., Noton, E., Constance, C.M., Dallmann, R., Di Green, D.J., Gillette, R., 1982. Circadian rhythm of firing rate recorded from single
Napoli, M.N., DeBruyne, J.P., Lambert, C.M., Yu, E.A., Reppert, S.M., cells in the rat suprachiasmatic brain slice. Brain Res. 245 (1), 198–200.
Weaver, D.R., 2009. Casein kinase 1 delta regulates the pace of the mammalian Groos, G., Hendriks, J., 1982. Circadian rhythms in electrical discharge of rat
circadian clock. Mol. Cell Biol. 29 (14), 3853–3866. suprachiasmatic neurones recorded in vitro. Neurosci. Lett. 34 (3), 283–288.
364 Circadian Regulation of Endocrine Functions

Grosbellet, E., Gourmelen, S., Pevet, P., Criscuolo, F., Challet, E., 2015. Leptin Iwahana, E., Karatsoreos, I., Shibata, S., Silver, R., 2008. Gonadectomy reveals sex
normalizes photic synchronization in male ob/ob mice, via indirect effects on the differences in circadian rhythms and suprachiasmatic nucleus androgen receptors
suprachiasmatic nucleus. Endocrinology 156 (3), 1080–1090. in mice. Horm. Behav. 53 (3), 422–430.
Grundschober, C., Delaunay, F., Puhlhofer, A., Triqueneaux, G., Laudet, V., Jiang, Z.G., Yang, Y.Q., Allen, C.N., 1997. Tracer and electrical coupling of rat
Bartfai, T., Nef, P., 2001. Circadian regulation of diverse gene products revealed suprachiasmatic nucleus neurons. Neuroscience 77 (4), 1059–1066.
by mRNA expression profiling of synchronized fibroblasts. J. Biol. Chem. 276 Jin, X., Shearman, L.P., Weaver, D.R., Zylka, M.J., de Vries, G.J., Reppert, S.M.,
(50), 46751–46758. 1999. A molecular mechanism regulating rhythmic output from the supra-
Guillaumond, F., Dardente, H., Giguere, V., Cermakian, N., 2005. Differential control of chiasmatic circadian clock. Cell 96 (1), 57–68.
Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors. J. Biol. Johnson, R.F., Moore, R.Y., Morin, L.P., 1988. Loss of entrainment and anatomical
Rhythms 20 (5), 391–403. plasticity after lesions of the hamster retinohypothalamic tract. Brain Res. 460 (2),
Guler, A.D., Ecker, J.L., Lall, G.S., Haq, S., Altimus, C.M., Liao, H.W., Barnard, A.R., 297–313.
Cahill, H., Badea, T.C., Zhao, H., Hankins, M.W., Berson, D.M., Lucas, R.J., Johnston, J.D., Skene, D.J., 2015. 60 YEARS OF NEUROENDOCRINOLOGY: regulation
Yau, K.W., Hattar, S., 2008. Melanopsin cells are the principal conduits for rod- of mammalian neuroendocrine physiology and rhythms by melatonin. J. Endocrinol.
cone input to non-image-forming vision. Nature 453 (7191), 102–105. 226 (2), T187–T198.
Gundlah, C., Kohama, S.G., Mirkes, S.J., Garyfallou, V.T., Urbanski, H.F., Bethea, C.L., Jolley, C.C., Ode, K.L., Ueda, H.R., 2012. A design principle for a posttranslational
2000. Distribution of estrogen receptor beta (ERb) mRNA in hypothalamus, biochemical oscillator. Cell Rep. 2 (4), 938–950.
midbrain and temporal lobe of spayed macaque: continued expression with Kalamatianos, T., Kallo, I., Goubillon, M.L., Coen, C.W., 2004. Cellular expression of
hormone replacement. Brain Res. Mol. Brain Res. 76 (2), 191–204. V1a vasopressin receptor mRNA in the female rat preoptic area: effects of oes-
Guo, H., Brewer, J.M., Champhekar, A., Harris, R.B., Bittman, E.L., 2005. Differential trogen. J. Neuroendocrinol. 16 (6), 525–533.
control of peripheral circadian rhythms by suprachiasmatic-dependent neural Kalsbeek, A., Buijs, R.M., 2002. Output pathways of the mammalian suprachiasmatic
signals. Proc. Natl. Acad. Sci. U.S.A. 102 (8), 3111–3116. nucleus: coding circadian time by transmitter selection and specific targeting. Cell
Guo, H., Brewer, J.M., Lehman, M.N., Bittman, E.L., 2006. Suprachiasmatic regulation Tissue Res. 309 (1), 109–118.
of circadian rhythms of gene expression in hamster peripheral organs: effects of Kalsbeek, A., Buijs, R.M., Engelmann, M., Wotjak, C.T., Landgraf, R., 1995. In vivo
transplanting the pacemaker. J. Neurosci. 26 (24), 6406–6412. measurement of a diurnal variation in vasopressin release in the rat supra-
Hakim, H., DeBernardo, A.P., Silver, R., 1991. Circadian locomotor rhythms, but not chiasmatic nucleus. Brain Res. 682 (1–2), 75–82.
photoperiodic responses, survive surgical isolation of the SCN in hamsters. J. Biol. Kalsbeek, A., Fliers, E., Franke, A.N., Wortel, J., Buijs, R.M., 2000. Functional
Rhythms 6 (2), 97–113. connections between the suprachiasmatic nucleus and the thyroid gland as
Hamada, T., LeSauter, J., Venuti, J.M., Silver, R., 2001. Expression of Period genes: revealed by lesioning and viral tracing techniques in the rat. Endocrinology 141
rhythmic and nonrhythmic compartments of the suprachiasmatic nucleus pace- (10), 3832–3841.
maker. J. Neurosci. 21 (19), 7742–7750. Kalsbeek, A., Ruiter, M., La Fleur, S.E., Van Heijningen, C., Buijs, R.M., 2003. The
Han, S.K., Gottsch, M.L., Lee, K.J., Popa, S.M., Smith, J.T., Jakawich, S.K., diurnal modulation of hormonal responses in the rat varies with different stimuli.
Clifton, D.K., Steiner, R.A., Herbison, A.E., 2005. Activation of gonadotropin- J. Neuroendocrinol. 15 (12), 1144–1155.
releasing hormone neurons by kisspeptin as a neuroendocrine switch for the Kalsbeek, A., van Heerikhuize, J.J., Wortel, J., Buijs, R.M., 1996. A diurnal rhythm of
onset of puberty. J. Neurosci. 25 (49), 11349–11356. stimulatory input to the hypothalamo-pituitary-adrenal system as revealed by timed
Hansen, J., 2006. Risk of breast cancer after night- and shift work: current evidence intrahypothalamic administration of the vasopressin V1 antagonist. J. Neurosci. 16
and ongoing studies in Denmark. Cancer Causes Control 17 (4), 531–537. (17), 5555–5565.
Harris, G.W., 1955. The function of the pituitary stalk. Bull. Johns Hopkins Hosp. 97 Karatsoreos, I.N., Bhagat, S., Bloss, E.B., Morrison, J.H., McEwen, B.S., 2011.
(5), 358–375. Disruption of circadian clocks has ramifications for metabolism, brain, and
Hattar, S., Lucas, R.J., Mrosovsky, N., Thompson, S., Douglas, R.H., Hankins, M.W., behavior. Proc. Natl. Acad. Sci. U.S.A. 108 (4), 1657–1662.
Lem, J., Biel, M., Hofmann, F., Foster, R.G., Yau, K.W., 2003. Melanopsin and Karatsoreos, I.N., Silver, R., 2007. Minireview: the neuroendocrinology of the supra-
rod-cone photoreceptive systems account for all major accessory visual functions chiasmatic nucleus as a conductor of body time in mammals. Endocrinology 148
in mice. Nature 424 (6944), 76–81. (12), 5640–5647.
He, P.J., Hirata, M., Yamauchi, N., Hashimoto, S., Hattori, M.A., 2007. The disruption Karatsoreos, I.N., Wang, A., Sasanian, J., Silver, R., 2007. A role for androgens in
of circadian clockwork in differentiating cells from rat reproductive tissues as regulating circadian behavior and the suprachiasmatic nucleus. Endocrinology 148
identified by in vitro real-time monitoring system. J. Endocrinol. 193 (3), 413–420. (11), 5487–5495.
Herzog, E.D., 2007. Neurons and networks in daily rhythms. Nat. Rev. Neurosci. 8 Karlsson, B., Alfredsson, L., Knutsson, A., Andersson, E., Toren, K., 2005. Total
(10), 790–802. mortality and cause-specific mortality of Swedish shift- and dayworkers in the pulp
Hileman, S.M., Handa, R.J., Jackson, G.L., 1999. Distribution of estrogen receptor- and paper industry in 1952-2001. Scand. J. Work Environ. Health 31 (1), 30–35.
b messenger ribonucleic acid in the male sheep hypothalamus. Biol. Reprod. 60 Kauffman, A.S., Clifton, D.K., Steiner, R.A., 2007. Emerging ideas about kisspeptin-
(6), 1279–1284. GPR54 signaling in the neuroendocrine regulation of reproduction. Trends Neu-
Hogenesch, J.B., Ueda, H.R., 2011. Understanding systems-level properties: timely rosci. 30 (10), 504–511.
stories from the study of clocks. Nat. Rev. Genet. 12 (6), 407–416. Kivimaki, M., Virtanen, M., Elovainio, M., Vaananen, A., Keltikangas-Jarvinen, L.,
Horvath, T.L., Cela, V., van der Beek, E.M., 1998. Gender-specific apposition between Vahtera, J., 2006. Prevalent cardiovascular disease, risk factors and selection out
vasoactive intestinal peptide-containing axons and gonadotrophin-releasing of shift work. Scand. J. Work Environ. Health 32 (3), 204–208.
hormone-producing neurons in the rat. Brain Res. 795 (1–2), 277–281. Klein, D.C., 1985. Photoneural regulation of the mammalian pineal gland. Ciba Found.
Hoyle, N.P., O’Neill, J.S., 2015. Oxidation-reduction cycles of peroxiredoxin proteins Symp. 117, 38–56.
and nontranscriptional aspects of timekeeping. Biochemistry 54 (2), 184–193. Klein, D.C., Moore, R.Y., 1979. Pineal N-acetyltransferase and hydroxyindole-O-
Hwang, S.Y., Lee, J.H., 2005. Comparison of cardiovascular risk profile clusters methyltransferase: control by the retinohypothalamic tract and the
among industrial workers. Taehan Kanho Hakhoe Chi 35 (8), 1500–1507. suprachiasmatic nucleus. Brain Res. 174 (2), 245–262.
Inouye, S.T., Kawamura, H., 1979. Persistence of circadian rhythmicity in Klein, D.C., Smoot, R., Weller, J.L., Higa, S., Markey, S.P., Creed, G.J.,
a mammalian hypothalamic “island” containing the suprachiasmatic nucleus. Proc. Jacobowitz, D.M., 1983. Lesions of the paraventricular nucleus area of the
Natl. Acad. Sci. U.S.A. 76 (11), 5962–5966. hypothalamus disrupt the suprachiasmatic leads to spinal cord circuit in the
Irwig, M.S., Fraley, G.S., Smith, J.T., Acohido, B.V., Popa, S.M., Cunningham, M.J., melatonin rhythm generating system. Brain Res. Bull. 10 (5), 647–652.
Gottsch, M.L., Clifton, D.K., Steiner, R.A., 2004. Kisspeptin activation of gonad- Koda, S., Yasuda, N., Sugihara, Y., Ohara, H., Udo, H., Otani, T., Hisashige, A.,
otropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat. Ogawa, T., Aoyama, H., 2000. Analyses of work-relatedness of health problems
Neuroendocrinology 80 (4), 264–272. among truck drivers by questionnaire survey. Sangyo Eiseigaku Zasshi 42 (1),
Isojima, Y., Nakajima, M., Ukai, H., Fujishima, H., Yamada, R.G., Masumoto, K.H., 6–16.
Kiuchi, R., Ishida, M., Ukai-Tadenuma, M., Minami, Y., Kito, R., Nakao, K., Koike, N., Yoo, S.H., Huang, H.C., Kumar, V., Lee, C., Kim, T.K., Takahashi, J.S.,
Kishimoto, W., Yoo, S.H., Shimomura, K., Takao, T., Takano, A., Kojima, T., 2012. Transcriptional architecture and chromatin landscape of the core circadian
Nagai, K., Sakaki, Y., Takahashi, J.S., Ueda, H.R., 2009. CKIepsilon/delta- clock in mammals. Science 338 (6105), 349–354.
dependent phosphorylation is a temperature-insensitive, period-determining Kolmodin-Hedman, B., Swensson, A., 1975. Problems related to shift work. A field
process in the mammalian circadian clock. Proc. Natl. Acad. Sci. U.S.A. 106 study of Swedish railroad workers with irregular work hours. Scand. J. Work
(37), 15744–15749. Environ. Health 1 (4), 254–262.
Circadian Regulation of Endocrine Functions 365

Kovanen, L., Saarikoski, S.T., Aromaa, A., Lonnqvist, J., Partonen, T., 2010. ARNTL Lewy, A., Bauer, V.K., Ahmed, S., Thomas, K.H., Cutler, N.L., Singer, C.M.,
(BMAL1) and NPAS2 gene variants contribute to fertility and seasonality. PLoS One Moffit, M.T., Sack, R.L., 1998. The human phase response curve (PRC) to
5 (4), e10007. melatonin is about 12 hours out of phase with the PRC to light. Chronobiol. Int. 15
Krajnak, K., Kashon, M.L., Rosewell, K.L., Wise, P.M., 1998. Sex differences in the (1), 71–83.
daily rhythm of vasoactive intestinal polypeptide but not arginine vasopressin Lewy, A., Sack, R., 1989. The dim light melatonin onset as a marker for circadian
messenger ribonucleic acid in the suprachiasmatic nuclei. Endocrinology 139 (10), phase position. Chronobiol. Int. 6 (1), 93–102.
4189–4196. Lewy, A.J., Lefler, B.J., Emens, J.S., Bauer, V.K., 2006. The circadian basis of winter
Krieger, D.T., Hauser, H., Krey, L.C., 1977. Suprachiasmatic nuclear lesions do not depression. Proc. Natl. Acad. Sci. U.S.A. 103 (19), 7414–7419.
abolish food-shifted circadian adrenal and temperature rhythmicity. Science 197 Lincoln, G.A., Messager, S., Andersson, H., Hazlerigg, D., 2002. Temporal expression
(4301), 398–399. of seven clock genes in the suprachiasmatic nucleus and the pars tuberalis of the
Krieger, D.T., Ossowski, R., Fogel, M., Allen, W., 1972. Lack of circadian periodicity of sheep: evidence for an internal coincidence timer. Proc. Natl. Acad. Sci. U.S.A. 99
human serum FSH and LH levels. J. Clin. Endocrinol. Metab. 35 (4), 619–623. (21), 13890–13895.
Kriegsfeld, L.J., 2006. Driving reproduction: RFamide peptides behind the wheel. Liston, C., Gan, W.B., 2011. Glucocorticoids are critical regulators of dendritic spine
Horm. Behav. 50 (5), 655–666. development and plasticity in vivo. Proc. Natl. Acad. Sci. U.S.A. 108 (38), 16074–
Kriegsfeld, L.J., 2013. Circadian regulation of kisspeptin in female reproductive 16079.
functioning. Adv. Exp. Med. Biol. 784, 385–410. Liu, A.C., Lewis, W.G., Kay, S.A., 2007. Mammalian circadian signaling networks and
Kriegsfeld, L.J., Leak, R.K., Yackulic, C.B., LeSauter, J., Silver, R., 2004. Organization therapeutic targets. Nat. Chem. Biol. 3 (10), 630–639.
of suprachiasmatic nucleus projections in Syrian hamsters (Mesocricetus auratus): Liu, R.Y., Unmehopa, U.A., Zhou, J.N., Swaab, D.F., 2006. Glucocorticoids suppress
an anterograde and retrograde analysis. J. Comp. Neurol. 468 (3), 361–379. vasopressin gene expression in human suprachiasmatic nucleus. J. Steroid Bio-
Kriegsfeld, L.J., LeSauter, J., Hamada, T., Pitts, S.M., Silver, S., 2002a. Circadian chem. Mol. Biol. 98 (4–5), 248–253.
rhythms in the endocrine system. In: Pfaff, D.W., Arnold, A.P., Etgen, A.M., Liu, X., Lee, K., Herbison, A.E., 2008. Kisspeptin excites gonadotropin-releasing
Fahrbach, S.E., Rubin, R.T. (Eds.), Hormones, Brain, and Behavior. Academic hormone neurons through a phospholipase C/calcium-dependent pathway regu-
Press, New York, pp. 33–92. lating multiple ion channels. Endocrinology 149 (9), 4605–4614.
Kriegsfeld, L.J., Mei, D.F., Bentley, G.E., Ubuka, T., Mason, A.O., Inoue, K., Ukena, K., Liu, Y., Johnson, B.P., Shen, A.L., Wallisser, J.A., Krentz, K.J., Moran, S.M.,
Tsutsui, K., Silver, R., 2006. Identification and characterization of a gonadotropin- Sullivan, R., Glover, E., Parlow, A.F., Drinkwater, N.R., Schuler, L.A.,
inhibitory system in the brains of mammals. Proc. Natl. Acad. Sci. U.S.A. 103 (7), Bradfield, C.A., 2014. Loss of BMAL1 in ovarian steroidogenic cells results in
2410–2415. implantation failure in female mice. Proc. Natl. Acad. Sci. U.S.A. 111 (39),
Kriegsfeld, L.J., Silver, R., 2006. The regulation of neuroendocrine function: timing is 14295–14300.
everything. Horm. Behav. 49 (5), 557–574. Lockley, S.W., Skene, D.J., Thapan, K., English, J., Ribeiro, D., Haimov, I.,
Kriegsfeld, L.J., Silver, R., Gore, A.C., Crews, D., 2002b. Vasoactive intestinal poly- Hampton, S., Middleton, B., von Schantz, M., Arendt, J., 1998. Extraocular light
peptide contacts on gonadotropin-releasing hormone neurones increase following exposure does not suppress plasma melatonin in humans. J. Clin. Endocrinol.
puberty in female rats. J. Neuroendocrinol. 14 (9), 685–690. Metab. 83 (9), 3369–3372.
Kubo, T., Ozasa, K., Mikami, K., Wakai, K., Fujino, Y., Watanabe, Y., Miki, T., Long, M.A., Jutras, M.J., Connors, B.W., Burwell, R.D., 2005. Electrical synapses
Nakao, M., Hayashi, K., Suzuki, K., Mori, M., Washio, M., Sakauchi, F., Ito, Y., coordinate activity in the suprachiasmatic nucleus. Nat. Neurosci. 8 (1), 61–66.
Yoshimura, T., Tamakoshi, A., 2006. Prospective cohort study of the risk of Loriaux, L., 2016. A Biographical History of Endocrinology. Wiley-Blackwell.
prostate cancer among rotating-shift workers: findings from the Japan collaborative Lowrey, P.L., Shimomura, K., Antoch, M.P., Yamazaki, S., Zemenides, P.D.,
cohort study. Am. J. Epidemiol. 164 (6), 549–555. Ralph, M.R., Menaker, M., Takahashi, J.S., 2000. Positional syntenic cloning and
Lamont, E.W., Robinson, B., Stewart, J., Amir, S., 2005. The central and basolateral functional characterization of the mammalian circadian mutation tau. Science 288
nuclei of the amygdala exhibit opposite diurnal rhythms of expression of the clock (5465), 483–492.
protein Period2. Proc. Natl. Acad. Sci. U.S.A. 102 (11), 4180–4184. Lucas, R.J., Hattar, S., Takao, M., Berson, D.M., Foster, R.G., Yau, K.W., 2003.
Landry, G.J., Mistlberger, R.E., 2007. Food entrainment: methodological issues. Diminished pupillary light reflex at high irradiances in melanopsin-knockout mice.
J. Biol. Rhythms 22 (6), 484–487. Science 299 (5604), 245–247.
Landry, G.J., Yamakawa, G.R., Webb, I.C., Mear, R.J., Mistlberger, R.E., 2007. The Maret, S., Dorsaz, S., Gurcel, L., Pradervand, S., Petit, B., Pfister, C.,
dorsomedial hypothalamic nucleus is not necessary for the expression of circadian Hagenbuchle, O., O’Hara, B.F., Franken, P., Tafti, M., 2007. Homer1a is a core
food-anticipatory activity in rats. J. Biol. Rhythms 22 (6), 467–478. brain molecular correlate of sleep loss. Proc. Natl. Acad. Sci. U.S.A. 104 (50),
Larsen, P.J., 1999. Tracing autonomic innervation of the rat pineal gland using viral 20090–20095.
transneuronal tracing. Microsc. Res. Tech. 46 (4–5), 296–304. Marpegan, L., Krall, T.J., Herzog, E.D., 2009. Vasoactive intestinal polypeptide entrains
Larsen, P.J., Enquist, L.W., Card, J.P., 1998. Characterization of the multisynaptic circadian rhythms in astrocytes. J. Biol. Rhythms 24 (2), 135–143.
neuronal control of the rat pineal gland using viral transneuronal tracing. Eur. J. Martino, T.A., Tata, N., Bjarnason, G.A., Straume, M., Sole, M.J., 2007. Diurnal
Neurosci. 10 (1), 128–145. protein expression in blood revealed by high throughput mass spectrometry pro-
Le Minh, N., Damiola, F., Tronche, F., Schutz, G., Schibler, U., 2001. Glucocorticoid teomics and implications for translational medicine and body time of day. Am. J.
hormones inhibit food-induced phase-shifting of peripheral circadian oscillators. Physiol. 293 (3), R1430–R1437.
EMBO J. 20 (24), 7128–7136. Masri, S., Orozco-Solis, R., Aguilar-Arnal, L., Cervantes, M., Sassone-Corsi, P., 2015.
Leak, R.K., Moore, R.Y., 2001. Topographic organization of suprachiasmatic nucleus Coupling circadian rhythms of metabolism and chromatin remodelling. Diabetes
projection neurons. J. Comp. Neurol. 433 (3), 312–334. Obes. Metab. 17 (Suppl. 1), 17–22.
Lee, H.S., Nelms, J.L., Nguyen, M., Silver, R., Lehman, M.N., 2003. The eye is Matsui, H., Takatsu, Y., Kumano, S., Matsumoto, H., Ohtaki, T., 2004. Peripheral
necessary for a circadian rhythm in the suprachiasmatic nucleus. Nat. Neurosci. 6 administration of metastin induces marked gonadotropin release and ovulation in
(2), 111–112. the rat. Biochem. Biophys. Res. Commun. 320 (2), 383–388.
Legan, S.J., Coon, G.A., Karsch, F.J., 1975. Role of estrogen as initiator of daily LH Maurel, D., Sage, D., Mekaouche, M., Bosler, O., 2000. Glucocorticoids up-regulate
surges in the ovariectomized rat. Endocrinology 96 (1), 50–56. the expression of glial fibrillary acidic protein in the rat suprachiasmatic nucleus.
Lehman, M.N., Coolen, L.M., Goodman, R.L., 2010. Minireview: kisspeptin/neurokinin Glia 29 (3), 212–221.
B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of Maywood, E.S., Chesham, J.E., O’Brien, J.A., Hastings, M.H., 2011. A diversity of
gonadotropin-releasing hormone secretion. Endocrinology 151 (8), 3479–3489. paracrine signals sustains molecular circadian cycling in suprachiasmatic nucleus
Lehman, M.N., Hileman, S.M., Goodman, R.L., 2013. Neuroanatomy of the kisspeptin circuits. Proc. Natl. Acad. Sci. U.S.A. 108 (34), 14306–14311.
signaling system in mammals: comparative and developmental aspects. Adv. Exp. McCarthy, J.J., Andrews, J.L., McDearmon, E.L., Campbell, K.S., Barber, B.K.,
Med. Biol. 784, 27–62. Miller, B.H., Walker, J.R., Hogenesch, J.B., Takahashi, J.S., Esser, K.A., 2007.
Lehman, M.N., Silver, R., Gladstone, W.R., Kahn, R.M., Gibson, M., Bittman, E.L., Identification of the circadian transcriptome in adult mouse skeletal muscle.
1987. Circadian rhythmicity restored by neural transplant. Immunocytochemical Physiol. Genomics 31 (1), 86–95.
characterization of the graft and its integration with the host brain. J. Neurosci. 7 Messager, S., Chatzidaki, E.E., Ma, D., Hendrick, A.G., Zahn, D., Dixon, J.,
(6), 1626–1638. Thresher, R.R., Malinge, I., Lomet, D., Carlton, M.B., Colledge, W.H., Caraty, A.,
Leonard, C., Fanning, N., Attwood, J., Buckley, M., 1998. The effect of fatigue, sleep Aparicio, S.A., 2005. Kisspeptin directly stimulates gonadotropin-releasing
deprivation and onerous working hours on the physical and mental wellbeing of hormone release via G protein-coupled receptor 54. Proc. Natl. Acad. Sci.
pre-registration house officers. Ir. J. Med. Sci. 167 (1), 22–25. U.S.A. 102 (5), 1761–1766.
366 Circadian Regulation of Endocrine Functions

Meyer-Bernstein, E.L., Jetton, A.E., Matsumoto, S.I., Markuns, J.F., Lehman, M.N., O’Leary, E.S., Schoenfeld, E.R., Stevens, R.G., Kabat, G.C., Henderson, K.,
Bittman, E.L., 1999. Effects of suprachiasmatic transplants on circadian rhythms of Grimson, R., Gammon, M.D., Leske, M.C., 2006. Shift work, light at night, and
neuroendocrine function in golden hamsters. Endocrinology 140 (1), 207–218. breast cancer on Long Island, New York. Am. J. Epidemiol. 164 (4), 358–366.
Miller, B.H., Olson, S.L., Levine, J.E., Turek, F.W., Horton, T.H., Takahashi, J.S., 2006. Oishi, K., Amagai, N., Shirai, H., Kadota, K., Ohkura, N., Ishida, N., 2005. Genome-
Vasopressin regulation of the proestrous luteinizing hormone surge in wild-type and wide expression analysis reveals 100 adrenal gland-dependent circadian genes
Clock mutant mice. Biol. Reprod. 75 (5), 778–784. in the mouse liver. DNA Res. 12 (3), 191–202.
Miller, B.H., Olson, S.L., Turek, F.W., Levine, J.E., Horton, T.H., Takahashi, J.S., 2004. Oishi, K., Miyazaki, K., Kadota, K., Kikuno, R., Nagase, T., Atsumi, G., Ohkura, N.,
Circadian clock mutation disrupts estrous cyclicity and maintenance of pregnancy. Azama, T., Mesaki, M., Yukimasa, S., Kobayashi, H., Iitaka, C., Umehara, T.,
Curr. Biol. 14 (15), 1367–1373. Horikoshi, M., Kudo, T., Shimizu, Y., Yano, M., Monden, M., Machida, K.,
Mirsky, H.P., Liu, A.C., Welsh, D.K., Kay, S.A., Doyle 3rd, F.J., 2009. A model of the Matsuda, J., Horie, S., Todo, T., Ishida, N., 2003. Genome-wide expression
cell-autonomous mammalian circadian clock. Proc. Natl. Acad. Sci. U.S.A. 106 analysis of mouse liver reveals CLOCK-regulated circadian output genes. J. Biol.
(27), 11107–11112. Chem. 278 (42), 41519–41527.
Mistlberger, R.E., 2006. Circadian rhythms: perturbing a food-entrained clock. Curr. Ono, D., Honma, K., Honma, S., 2015. Circadian and ultradian rhythms of clock gene
Biol. 16 (22), R968–R969. expression in the suprachiasmatic nucleus of freely moving mice. Sci. Rep. 5,
Miyamoto, Y., Sancar, A., 1999. Circadian regulation of cryptochrome genes in the 12310.
mouse. Brain Res. Mol. Brain Res. 71 (2), 238–243. Oster, H., Damerow, S., Hut, R.A., Eichele, G., 2006a. Transcriptional profiling in the
Model, Z., Butler, M.P., LeSauter, J., Silver, R., 2015. Suprachiasmatic nucleus as the adrenal gland reveals circadian regulation of hormone biosynthesis genes and
site of androgen action on circadian rhythms. Horm. Behav. 73, 1–7. nucleosome assembly genes. J. Biol. Rhythms 21 (5), 350–361.
Mong, J.A., Baker, F.C., Mahoney, M.M., Paul, K.N., Schwartz, M.D., Semba, K., Oster, H., Damerow, S., Kiessling, S., Jakubcakova, V., Abraham, D., Tian, J.,
Silver, R., 2011. Sleep, rhythms, and the endocrine brain: influence of sex and Hoffmann, M.W., Eichele, G., 2006b. The circadian rhythm of glucocorticoids is
gonadal hormones. J. Neurosci. 31 (45), 16107–16116. regulated by a gating mechanism residing in the adrenal cortical clock. Cell Metab.
Moore, R.Y., 1996. Entrainment pathways and the functional organization of the 4 (2), 163–173.
circadian system. Prog. Brain Res. 111, 103–119. Ottenweller, J.E., Tapp, W.N., Pitman, D.L., Natelson, B.H., 1987. Adrenal, thyroid,
Moore, R.Y., Eichler, V.B., 1972. Loss of a circadian adrenal corticosterone rhythm and testicular hormone rhythms in male golden hamsters on long and short days.
following suprachiasmatic lesions in the rat. Brain Res. 42 (1), 201–206. Am. J. Physiol. 253 (2 Pt 2), R321–R328.
Moore, R.Y., Klein, D.C., 1974. Visual pathways and the central neural control of Palm, I.F., Van Der Beek, E.M., Wiegant, V.M., Buijs, R.M., Kalsbeek, A., 1999.
a circadian rhythm in pineal serotonin N-acetyltransferase activity. Brain Res. 71 Vasopressin induces a luteinizing hormone surge in ovariectomized, estradiol-
(1), 17–33. treated rats with lesions of the suprachiasmatic nucleus. Neuroscience 93 (2),
Moore, R.Y., Lenn, N.J., 1972. A retinohypothalamic projection in the rat. J. Comp. 659–666.
Neurol. 146 (1), 1–14. Panda, S., Antoch, M.P., Miller, B.H., Su, A.I., Schook, A.B., Straume, M.,
Morikawa, Y., Nakagawa, H., Miura, K., Soyama, Y., Ishizaki, M., Kido, T., Naruse, Y., Schultz, P.G., Kay, S.A., Takahashi, J.S., Hogenesch, J.B., 2002a. Coordinated
Suwazono, Y., Nogawa, K., 2005. Shift work and the risk of diabetes mellitus transcription of key pathways in the mouse by the circadian clock. Cell 109 (3),
among Japanese male factory workers. Scand. J. Work Environ. Health 31 (3), 307–320.
179–183. Panda, S., Provencio, I., Tu, D.C., Pires, S.S., Rollag, M.D., Castrucci, A.M.,
Morin, L.P., 1980. Effect of ovarian hormones on synchrony of hamster circadian Pletcher, M.T., Sato, T.K., Wiltshire, T., Andahazy, M., Kay, S.A., Van Gelder, R.N.,
rhythms. Physiol. Behav. 24 (4), 741–749. Hogenesch, J.B., 2003. Melanopsin is required for non-image-forming photic
Morin, L.P., 2013. Neuroanatomy of the extended circadian rhythm system. Exp. responses in blind mice. Science 301 (5632), 525–527.
Neurol. 243, 4–20. Panda, S., Sato, T.K., Castrucci, A.M., Rollag, M.D., DeGrip, W.J., Hogenesch, J.B.,
Morin, L.P., Fitzgerald, K.M., Zucker, I., 1977. Estradiol shortens the period of hamster Provencio, I., Kay, S.A., 2002b. Melanopsin (Opn4) requirement for normal light-
circadian rhythms. Science 196 (4287), 305–307. induced circadian phase shifting. Science 298 (5601), 2213–2216.
Morris, C.J., Aeschbach, D., Scheer, F.A., 2012. Circadian system, sleep and Pando, M.P., Morse, D., Cermakian, N., Sassone-Corsi, P., 2002. Phenotypic rescue
endocrinology. Mol. Cell Endocrinol. 349 (1), 91–104. of a peripheral clock genetic defect via SCN hierarchical dominance. Cell 110 (1),
Morse, D., Cermakian, N., Brancorsini, S., Parvinen, M., Sassone-Corsi, P., 2003. No 107–117.
circadian rhythms in testis: period1 expression is Clock independent and devel- Partch, C.L., Green, C.B., Takahashi, J.S., 2014. Molecular architecture of the
opmentally regulated in the mouse. Mol. Endocrinol. 17 (1), 141–151. mammalian circadian clock. Trends Cell Biol. 24 (2), 90–99.
Munakata, M., Ichi, S., Nunokawa, T., Saito, Y., Ito, N., Fukudo, S., Yoshinaga, K., Patel, D., 2006. Shift work, light at night and risk of breast cancer. Occup. Med. (Lond)
2001. Influence of night shift work on psychologic state and cardiovascular and 56 (6), 433.
neuroendocrine responses in healthy nurses. Hypertens. Res. 24 (1), 25–31. Pauls, S., Foley, N.C., Foley, D.K., LeSauter, J., Hastings, M.H., Maywood, E.S.,
Nagoshi, E., Saini, C., Bauer, C., Laroche, T., Naef, F., Schibler, U., 2004. Circadian Silver, R., 2014. Differential contributions of intra-cellular and inter-cellular
gene expression in individual fibroblasts: cell-autonomous and self-sustained mechanisms to the spatial and temporal architecture of the suprachiasmatic
oscillators pass time to daughter cells. Cell 119 (5), 693–705. nucleus circadian circuitry in wild-type, cryptochrome-null and vasoactive intestinal
Nakajima, M., Imai, K., Ito, H., Nishiwaki, T., Murayama, Y., Iwasaki, H., Oyama, T., peptide receptor 2-null mutant mice. Eur. J. Neurosci. 40 (3), 2528–2540.
Kondo, T., 2005. Reconstitution of circadian oscillation of cyanobacterial KaiC Pauls, S.D., Honma, K., Honma, S., Silver, R., 2016. Deconstructing circadian
phosphorylation in vitro. Science 308 (5720), 414–415. rhythmicity with models and manipulations. Trends Neurosci. 39 (6), 405–419.
Nakamura, T.J., Sellix, M.T., Kudo, T., Nakao, N., Yoshimura, T., Ebihara, S., Perelis, M., Ramsey, K.M., Bass, J., 2015. The molecular clock as a metabolic
Colwell, C.S., Block, G.D., 2010. Influence of the estrous cycle on clock gene rheostat. Diabetes Obes. Metab. 17 (Suppl. 1), 99–105.
expression in reproductive tissues: effects of fluctuating ovarian steroid hormone Perreau-Lenz, S., Kalsbeek, A., Pevet, P., Buijs, R.M., 2004. Glutamatergic clock
levels. Steroids 75 (3), 203–212. output stimulates melatonin synthesis at night. Eur. J. Neurosci. 19 (2), 318–324.
Nakao, N., Yasuo, S., Nishimura, A., Yamamura, T., Watanabe, T., Anraku, T., Pevet, P., Challet, E., 2011. Melatonin: both master clock output and internal time-
Okano, T., Fukada, Y., Sharp, P.J., Ebihara, S., Yoshimura, T., 2007. Circadian giver in the circadian clocks network. J. Physiol. Paris 105 (4–6), 170–182.
clock gene regulation of steroidogenic acute regulatory protein gene expression in Piet, R., Fraissenon, A., Boehm, U., Herbison, A.E., 2015. Estrogen permits vaso-
preovulatory ovarian follicles. Endocrinology 148 (7), 3031–3038. pressin signaling in preoptic kisspeptin neurons in the female mouse. J. Neurosci.
Neill, J.D., 2005. Challis, J., de Kretser, D., Pfaff, D., et al. (Eds.). Knobil and Neill’s 35 (17), 6881–6892.
Physiology of Reproduction, vol. 2. Academic Press, Raven, New York, Pitts, S., Perone, E., Silver, R., 2003. Food-entrained circadian rhythms are sustained
pp. 487–540. in arrhythmic Clk/Clk mutant mice. Am. J. Physiol. Regul. Integr. Comp. Physiol.
Nelson, R.J., Zucker, I., 1981. Absence of extra-ocular photoreception in diurnal and 285 (1), R57–R67.
nocturnal rodents exposed to direct sunlight. Comp. Biochem. Physiol. A Physiol. Poole, C.J., Wright, A.D., Nattrass, M., 1992. Control of diabetes mellitus in shift
69 (1), 145–148. workers. Br. J. Ind. Med. 49 (7), 513–515.
Nishide, S.Y., Hashimoto, K., Nishio, T., Honma, K., Honma, S., 2014. Organ-specific Pouliot, W.A., Handa, R.J., Beck, S.G., 1996. Androgen modulates N-methyl-D-
development characterizes circadian clock gene Per2 expression in rats. Am. J. aspartate-mediated depolarization in CA1 hippocampal pyramidal cells. Synapse
Physiol. Regul. Integr. Comp. Physiol. 306 (1), R67–R74. 23 (1), 10–19.
Nunez, A.A., Stephan, F.K., 1977. The effects of hypothalamic knife cuts on drinking Price, D.A., Greenberg, M.J., 1977. Structure of a molluscan cardioexcitatory
rhythms and the estrus cycle of the rat. Behav. Biol. 20 (2), 224–234. neuropeptide. Science 197 (4304), 670–671.
Circadian Regulation of Endocrine Functions 367

Prosser, R.A., Edgar, D.M., Heller, H.C., Miller, J.D., 1994. A possible glial role in the Segall, L.A., Perrin, J.S., Walker, C.D., Stewart, J., Amir, S., 2006. Glucocorticoid
mammalian circadian clock. Brain Res. 643 (1–2), 296–301. rhythms control the rhythm of expression of the clock protein, Period2, in oval
Ralph, M.R., Foster, R.G., Davis, F.C., Menaker, M., 1990. Transplanted supra- nucleus of the bed nucleus of the stria terminalis and central nucleus of the
chiasmatic nucleus determines circadian period. Science 247 (4945), amygdala in rats. Neuroscience 140 (3), 753–757.
975–978. Segawa, K., Nakazawa, S., Tsukamoto, Y., Kurita, Y., Goto, H., Fukui, A., Takano, K.,
Ralph, M.R., Menaker, M., 1988. A mutation of the circadian system in golden 1987. Peptic ulcer is prevalent among shift workers. Dig. Dis. Sci. 32 (5),
hamsters. Science 241 (4870), 1225–1227. 449–453.
Ramanathan, C., Stowie, A., Smale, L., Nunez, A.A., 2010. Phase preference for the Sellix, M.T., 2015. Circadian clock function in the mammalian ovary. J. Biol. Rhythms
display of activity is associated with the phase of extra-suprachiasmatic nucleus 30 (1), 7–19.
oscillators within and between species. Neuroscience 170 (3), 758–772. Seminara, S.B., Messager, S., Chatzidaki, E.E., Thresher, R.R., Acierno Jr., J.S.,
Rash, J.E., Olson, C.O., Pouliot, W.A., Davidson, K.G., Yasumura, T., Furman, C.S., Shagoury, J.K., Bo-Abbas, Y., Kuohung, W., Schwinof, K.M., Hendrick, A.G.,
Royer, S., Kamasawa, N., Nagy, J.I., Dudek, F.E., 2007. Connexin36 vs. con- Zahn, D., Dixon, J., Kaiser, U.B., Slaugenhaupt, S.A., Gusella, J.F., O’Rahilly, S.,
nexin32, “miniature” neuronal gap junctions, and limited electrotonic coupling in Carlton, M.B., Crowley Jr., W.F., Aparicio, S.A., Colledge, W.H., 2003. The GPR54
rodent suprachiasmatic nucleus. Neuroscience 149 (2), 350–371. gene as a regulator of puberty. N. Engl. J. Med. 349 (17), 1614–1627.
Reddy, A.B., Maywood, E.S., Karp, N.A., King, V.M., Inoue, Y., Gonzalez, F.J., Shahab, M., Mastronardi, C., Seminara, S.B., Crowley, W.F., Ojeda, S.R., Plant, T.M.,
Lilley, K.S., Kyriacou, C.P., Hastings, M.H., 2007. Glucocorticoid signaling 2005. Increased hypothalamic GPR54 signaling: a potential mechanism for initi-
synchronizes the liver circadian transcriptome. Hepatology 45 (6), 1478–1488. ation of puberty in primates. Proc. Natl. Acad. Sci. U.S.A. 102 (6), 2129–2134.
Reiter, R.J., Tan, D.X., 2002. Role of CSF in the transport of melatonin. J. Pineal Res. Shibata, S., Oomura, Y., Kita, H., Hattori, K., 1982. Circadian rhythmic changes of
33 (1), 61. neuronal activity in the suprachiasmatic nucleus of the rat hypothalamic slice. Brain
Rey, G., Reddy, A.B., 2015. Interplay between cellular redox oscillations and circadian Res. 247 (1), 154–158.
clocks. Diabetes Obes. Metab. 17 (Suppl. 1), 55–64. Shinohara, K., Funabashi, T., Mitushima, D., Kimura, F., 2000. Effects of gap junction
Ripperger, J.A., Schibler, U., 2006. Rhythmic CLOCK-BMAL1 binding to multiple E-box blocker on vasopressin and vasoactive intestinal polypeptide rhythms in the rat
motifs drives circadian Dbp transcription and chromatin transitions. Nat. Genet. 38 suprachiasmatic nucleus in vitro. Neurosci. Res. 38 (1), 43–47.
(3), 369–374. Shinohara, K., Funabashi, T., Nakamura, T.J., Kimura, F., 2001. Effects of estrogen
Robertson, J.L., Clifton, D.K., de la Iglesia, H.O., Steiner, R.A., Kauffman, A.S., 2009. and progesterone on the expression of connexin-36 mRNA in the suprachiasmatic
Circadian regulation of Kiss1 neurons: implications for timing the preovulatory nucleus of female rats. Neurosci. Lett. 309 (1), 37–40.
gonadotropin-releasing hormone/luteinizing hormone surge. Endocrinology 150 (8), Shinohara, K., Tominaga, K., Isobe, Y., Inouye, S.T., 1993. Photic regulation of
3664–3671. peptides located in the ventrolateral subdivision of the suprachiasmatic nucleus of
Robinson, N., Yateman, N.A., Protopapa, L.E., Bush, L., 1990. Employment problems the rat: daily variations of vasoactive intestinal polypeptide, gastrin-releasing
and diabetes. Diabet. Med. 7 (1), 16–22. peptide, and neuropeptide Y. J. Neurosci. 13 (2), 793–800.
Roenneberg, T., Allebrandt, K.V., Merrow, M., Vetter, C., 2012. Social jetlag and Shughrue, P.J., Lane, M.V., Merchenthaler, I., 1997. Comparative distribution of
obesity. Curr. Biol. 22 (10), 939–943. estrogen receptor-a and -b mRNA in the rat central nervous system. J. Comp.
Rosenfeld, P., Van Eekelen, J.A., Levine, S., De Kloet, E.R., 1988. Ontogeny of the Neurol. 388 (4), 507–525.
type 2 glucocorticoid receptor in discrete rat brain regions: an immunocytochemical Silver, R., Lehman, M.N., Gibson, M., Gladstone, W.R., Bittman, E.L., 1990. Dispersed
study. Brain Res. 470 (1), 119–127. cell suspensions of fetal SCN restore circadian rhythmicity in SCN-lesioned adult
Rossmanith, W.G., Liu, C.H., Laughlin, G.A., Mortola, J.F., Suh, B.Y., Yen, S.S., hamsters. Brain Res. 525 (1), 45–58.
1990. Relative changes in LH pulsatility during the menstrual cycle: using data Silver, R., LeSauter, J., Tresco, P.A., Lehman, M.N., 1996. A diffusible coupling signal
from hypogonadal women as a reference point. Clin. Endocrinol. (Oxf.) 32 (5), from the transplanted suprachiasmatic nucleus controlling circadian locomotor
647–660. rhythms. Nature 382 (6594), 810–813.
Ruby, N.F., Brennan, T.J., Xie, X., Cao, V., Franken, P., Heller, H.C., O’Hara, B.F., Silver, R., Sookhoo, A.I., LeSauter, J., Stevens, P., Jansen, H.T., Lehman, M.N., 1999.
2002. Role of melanopsin in circadian responses to light. Science 298 (5601), Multiple regulatory elements result in regional specificity in circadian rhythms of
2211–2213. neuropeptide expression in mouse SCN. Neuroreport 10 (15), 3165–3174.
Russell, G.M., Kalafatakis, K., Lightman, S.L., 2015. The importance of biological Skinner, D.C., Caraty, A., 2002. Measurement and possible function of GnRH in
oscillators for hypothalamic-pituitary-adrenal activity and tissue glucocorticoid cerebrospinal fluid in ewes. Reprod. Suppl. 59, 25–39.
response: coordinating stress and neurobehavioural adaptation. J. Neuroendocrinol. Skinner, D.C., Malpaux, B., 1999. High melatonin concentrations in third ventricular
27 (6), 378–388. cerebrospinal fluid are not due to Galen vein blood recirculating through the choroid
Russo, K.A., La, J.L., Stephens, S.B., Poling, M.C., Padgaonkar, N.A., Jennings, K.J., plexus. Endocrinology 140 (10), 4399–4405.
Piekarski, D.J., Kauffman, A.S., Kriegsfeld, L.J., 2015. Circadian control of the Skipper Jr., J.K., Jung, F.D., Coffey, L.C., 1990. Nurses and shiftwork: effects on
female reproductive Axis through gated responsiveness of the RFRP-3 system to physical health and mental depression. J. Adv. Nurs. 15 (7), 835–842.
VIP signaling. Endocrinology 156 (7), 2608–2618. Slominski, R.M., Reiter, R.J., Schlabritz-Loutsevitch, N., Ostrom, R.S., Slominski, A.T.,
Sack, R.L., Brandes, R.W., Kendall, A.R., Lewy, A.J., 2000. Entrainment of free- 2012. Melatonin membrane receptors in peripheral tissues: distribution and
running circadian rhythms by melatonin in blind people. N. Engl. J. Med. 343 functions. Mol. Cell Endocrinol. 351 (2), 152–166.
(15), 1070–1077. Smith, J.T., 2008. Kisspeptin signalling in the brain: steroid regulation in the rodent
Sanborn, C., Currie, A.C., Bailey, C.C., 1982. Shift work: how to adjust patterns of and ewe. Brain Res. Rev. 57 (2), 288–298.
diabetes care. Occup. Health Nurs. 30 (12), 25–28. Smith, J.T., Clarke, I.J., 2010. Gonadotropin inhibitory hormone function in mammals.
Sarvari, M., Hrabovszky, E., Kallo, I., Galamb, O., Solymosi, N., Liko, I., Molnar, B., Trends Endocrinol. Metab. 21 (4), 255–260.
Tihanyi, K., Szombathelyi, Z., Liposits, Z., 2010. Gene expression profiling iden- Smith, M.J., Jiennes, L., Wise, P.M., 2000. Localization of the VIP2 receptor protein
tifies key estradiol targets in the frontal cortex of the rat. Endocrinology 151 (3), on GnRH neurons in the female rat. Endocrinology 141 (11), 4317–4320.
1161–1176. Spies, H.G., Mahoney, C.J., Norman, R.L., Clifton, D.K., Resko, J.A., 1974. Evidence
Schirman-Hildesheim, T.D., Ben-Aroya, N., Koch, Y., 2006. Daily GnRH and GnRH- for a diurnal rhythm in ovarian steroid secretion in the rhesus monkey. J. Clin.
receptor mRNA expression in the ovariectomized and intact rat. Mol. Cell Endocrinol. Metab. 39 (2), 347–351.
Endocrinol. 252 (1–2), 120–125. Spiga, F., Liu, Y., Aguilera, G., Lightman, S.L., 2011. Temporal effect of adrenocor-
Schulz, P., Walker, J.P., Peyrin, L., Soulier, V., Curtin, F., Steimer, T., 1996. Lower sex ticotrophic hormone on adrenal glucocorticoid steroidogenesis: involvement of the
hormones in men during anticipatory stress. Neuroreport 7 (18), 3101–3104. transducer of regulated cyclic AMP-response element-binding protein activity.
Schwartz, W.J., Coleman, R.J., Reppert, S.M., 1983. A daily vasopressin rhythm in rat J. Neuroendocrinol. 23 (2), 136–142.
cerebrospinal fluid. Brain Res. 263 (1), 105–112. Spiga, F., Walker, J.J., Terry, J.R., Lightman, S.L., 2014. HPA axis-rhythms. Compr.
Schwartz, W.J., Reppert, S.M., 1985. Neural regulation of the circadian vasopressin Physiol. 4 (3), 1273–1298.
rhythm in cerebrospinal fluid: a pre-eminent role for the suprachiasmatic nuclei. Spratt, D.I., O’Dea, L.S., Schoenfeld, D., Butler, J., Rao, P.N., Crowley Jr., W.F., 1988.
J. Neurosci. 5 (10), 2771–2778. Neuroendocrine-gonadal axis in men: frequent sampling of LH, FSH, and testos-
Scott, C.J., Jansen, H.T., Kao, C.C., Kuehl, D.E., Jackson, G.L., 1995. Disruption of terone. Am. J. Physiol. 254 (5 Pt 1), E658–E666.
reproductive rhythms and patterns of melatonin and prolactin secretion following Stephan, F.K., Zucker, I., 1972. Rat drinking rhythms: central visual pathways and
bilateral lesions of the suprachiasmatic nuclei in the ewe. J. Neuroendocrinol. 7 (6), endocrine factors mediating responsiveness to environmental illumination. Physiol.
429–443. Behav. 8 (2), 315–326.
368 Circadian Regulation of Endocrine Functions

Stieg, M.R., Sievers, C., Farr, O., Stalla, G.K., Mantzoros, C.S., 2015. Leptin: Van Reeth, O., Sturis, J., Byrne, M.M., Blackman, J.D., L’Hermite-Baleriaux, M.,
a hormone linking activation of neuroendocrine axes with neuropathology. Psy- Leproult, R., Oliner, C., Refetoff, S., Turek, F.W., Van Cauter, E., 1994. Nocturnal
choneuroendocrinology 51, 47–57. exercise phase delays circadian rhythms of melatonin and thyrotropin secretion in
Stokkan, K.A., Yamazaki, S., Tei, H., Sakaki, Y., Menaker, M., 2001. Entrainment of normal men. Am. J. Physiol. 266 (6 Pt 1), E964–E974.
the circadian clock in the liver by feeding. Science 291 (5503), 490–493. Veldhuis, J.D., Rogol, A.D., Evans, W.S., Iranmanesh, A., Lizarralde, G.,
Storch, K.F., Lipan, O., Leykin, I., Viswanathan, N., Davis, F.C., Wong, W.H., Johnson, M.L., 1986. Spectrum of the pulsatile characteristics of LH release in
Weitz, C.J., 2002. Extensive and divergent circadian gene expression in liver and normal men. J. Androl. 7 (2), 83–92.
heart. Nature 417 (6884), 78–83. Venuta, M., Barzaghi, L., Cavalieri, C., Gamberoni, T., Guaraldi, G.P., 1999. Effects of
Suzuki, T., Sato, F., Kondo, J., Liu, Y., Kusumi, T., Fujimoto, K., Kato, Y., Sato, T., shift work on the quality of sleep and psychological health based on a sample of
Kijima, H., 2008. Period is involved in the proliferation of human pancreatic MIA- professional nurses. G. Ital. Med. Lav. Ergon. 21 (3), 221–225.
PaCa2 cancer cells by TNF-a. Biomed. Res. 29 (2), 99–103. Vida, B., Hrabovszky, E., Kalamatianos, T., Coen, C.W., Liposits, Z., Kallo, I., 2008.
Swann, J.M., Turek, F.W., 1985. Multiple circadian oscillators regulate the timing of Oestrogen receptor alpha and beta immunoreactive cells in the suprachiasmatic
behavioral and endocrine rhythms in female golden hamsters. Science 228 (4701), nucleus of mice: distribution, sex differences and regulation by gonadal hormones.
898–900. J. Neuroendocrinol. 20 (11), 1270–1277.
Takahashi, J.S., 2015. Molecular components of the circadian clock in mammals. Vollmers, C., Gill, S., DiTacchio, L., Pulivarthy, S.R., Le, H.D., Panda, S., 2009. Time of
Diabetes Obes. Metab. 17 (Suppl. 1), 6–11. feeding and the intrinsic circadian clock drive rhythms in hepatic gene expression.
Takahashi, Y., Okamura, H., Yanaihara, N., Hamada, S., Fujita, S., Ibata, Y., 1989. Proc. Natl. Acad. Sci. U.S.A. 106 (50), 21453–21458.
Vasoactive intestinal peptide immunoreactive neurons in the rat suprachiasmatic Vrang, N., Larsen, P.J., Mikkelsen, J.D., 1995. Direct projection from the supra-
nucleus demonstrate diurnal variation. Brain Res. 497 (2), 374–377. chiasmatic nucleus to hypophysiotrophic corticotropin-releasing factor immunore-
Tavakoli-Nezhad, M., Schwartz, W.J., 2005. c-Fos expression in the brains of active cells in the paraventricular nucleus of the hypothalamus demonstrated by
behaviorally “split” hamsters in constant light: calling attention to a dorsolateral means of Phaseolus vulgaris-leucoagglutinin tract tracing. Brain Res. 684 (1),
region of the suprachiasmatic nucleus and the medial division of the lateral 61–69.
habenula. J. Biol. Rhythms 20 (5), 419–429. Walker, J.J., Spiga, F., Waite, E., Zhao, Z., Kershaw, Y., Terry, J.R., Lightman, S.L.,
Teclemariam-Mesbah, R., Ter Horst, G.J., Postema, F., Wortel, J., Buijs, R.M., 1999. 2012. The origin of glucocorticoid hormone oscillations. PLoS Biol. 10 (6),
Anatomical demonstration of the suprachiasmatic nucleus-pineal pathway. e1001341.
J. Comp. Neurol. 406 (2), 171–182. Walker, J.J., Terry, J.R., Lightman, S.L., 2010. Origin of ultradian pulsatility in the
Tessonneaud, A., Locatelli, A., Caldani, M., Viguier-Martinez, M.C., 1995. Bilateral hypothalamic-pituitary-adrenal axis. Proc. Biol. Sci. 277 (1688), 1627–1633.
lesions of the suprachiasmatic nuclei alter the nocturnal melatonin secretion in Weaver, D.R., 1998. The suprachiasmatic nucleus: a 25-year retrospective. J. Biol.
sheep. J. Neuroendocrinol. 7 (2), 145–152. Rhythms 13 (2), 100–112.
Tonsfeldt, K.J., Goodall, C.P., Latham, K.L., Chappell, P.E., 2011. Oestrogen induces Weick, R.F., Stobie, K.M., 1995. Role of VIP in the regulation of LH secretion in the
rhythmic expression of the Kisspeptin-1 receptor GPR54 in hypothalamic female rat. Neurosci. Biobehav. Rev. 19 (2), 251–259.
gonadotrophin-releasing hormone-secreting GT1-7 cells. J. Neuroendocrinol. 23 Weitzman, E.D., Fukushima, D., Nogeire, C., Roffwarg, H., Gallagher, T.F., Hellman, L.,
(9), 823–830. 1971. Twenty-four hour pattern of the episodic secretion of cortisol in normal
Tricoire, H., Moller, M., Chemineau, P., Malpaux, B., 2003. Origin of cerebrospinal subjects. J. Clin. Endocrinol. Metab. 33 (1), 14–22.
fluid melatonin and possible function in the integration of photoperiod. Reprod. Welsh, D.K., Logothetis, D.E., Meister, M., Reppert, S.M., 1995. Individual neurons
Suppl. 61, 311–321. dissociated from rat suprachiasmatic nucleus express independently phased
Tsutsui, K., Saigoh, E., Ukena, K., Teranishi, H., Fujisawa, Y., Kikuchi, M., Ishii, S., circadian firing rhythms. Neuron 14 (4), 697–706.
Sharp, P.J., 2000. A novel avian hypothalamic peptide inhibiting gonadotropin Welsh, D.K., Yoo, S.H., Liu, A.C., Takahashi, J.S., Kay, S.A., 2004. Bioluminescence
release. Biochem. Biophys. Res. Commun. 275 (2), 661–667. imaging of individual fibroblasts reveals persistent, independently phased circadian
Tsutsui, K., Ubuka, T., Son, Y.L., Bentley, G.E., Kriegsfeld, L.J., 2015. Contribution of rhythms of clock gene expression. Curr. Biol. 14 (24), 2289–2295.
GnIH research to the progress of reproductive neuroendocrinology. Front. Endo- Williams 3rd, W.P., Jarjisian, S.G., Mikkelsen, J.D., Kriegsfeld, L.J., 2011. Circadian
crinol. (Lausanne) 6, 179. control of kisspeptin and a gated GnRH response mediate the preovulatory lutei-
Ueda, H.R., Chen, W., Adachi, A., Wakamatsu, H., Hayashi, S., Takasugi, T., nizing hormone surge. Endocrinology 152 (2), 595–606.
Nagano, M., Nakahama, K., Suzuki, Y., Sugano, S., Iino, M., Shigeyoshi, Y., Williams 3rd, W.P., Kriegsfeld, L.J., 2012. Circadian control of neuroendocrine circuits
Hashimoto, S., 2002. A transcription factor response element for gene expression regulating female reproductive function. Front. Endocrinol. (Lausanne) 3, 60.
during circadian night. Nature 418 (6897), 534–539. Wintermantel, T.M., Campbell, R.E., Porteous, R., Bock, D., Grone, H.J., Todman, M.G.,
Ueda, H.R., Chen, W., Minami, Y., Honma, S., Honma, K., Iino, M., Hashimoto, S., Korach, K.S., Greiner, E., Perez, C.A., Schutz, G., Herbison, A.E., 2006. Definition of
2004. Molecular-timetable methods for detection of body time and rhythm estrogen receptor pathway critical for estrogen positive feedback to gonadotropin-
disorders from single-time-point genome-wide expression profiles. Proc. Natl. releasing hormone neurons and fertility. Neuron 52 (2), 271–280.
Acad. Sci. U.S.A. 101 (31), 11227–11232. Wittmann, M., Dinich, J., Merrow, M., Roenneberg, T., 2006. Social jetlag:
Ungar, F., Halberg, F., 1962. Circadian rhythm in the in vitro response of mouse misalignment of biological and social time. Chronobiol. Int. 23 (1–2), 497–509.
adrenal to adrenocorticotropic hormone. Science 137, 1058–1060. Wolk, R., Gami, A.S., Garcia-Touchard, A., Somers, V.K., 2005. Sleep and cardio-
Van Cauter, E., Refetoff, S., 1985. Multifactorial control of the 24-hour secretory vascular disease. Curr. Probl. Cardiol. 30 (12), 625–662.
profiles of pituitary hormones. J. Endocrinol. Invest 8 (4), 381–391. Wong, C.C., Dohler, K.D., Geerlings, H., von zur Muhlen, A., 1983. Influence of age,
van der Beek, E.M., Horvath, T.L., Wiegant, V.M., Van den Hurk, R., Buijs, R.M., strain and season on circadian periodicity of pituitary, gonadal and adrenal
1997. Evidence for a direct neuronal pathway from the suprachiasmatic nucleus hormones in the serum of male laboratory rats. Horm. Res. 17 (4), 202–215.
to the gonadotropin-releasing hormone system: combined tracing and light and Wright Jr., K.P., Czeisler, C.A., 2002. Absence of circadian phase resetting in
electron microscopic immunocytochemical studies. J. Comp. Neurol. 384 (4), response to bright light behind the knees. Science 297 (5581), 571.
569–579. Yagita, K., Tamanini, F., van Der Horst, G.T., Okamura, H., 2001. Molecular mech-
van der Beek, E.M., Swarts, H.J., Wiegant, V.M., 1999. Central administration of anisms of the biological clock in cultured fibroblasts. Science 292 (5515),
antiserum to vasoactive intestinal peptide delays and reduces luteinizing hormone 278–281.
and prolactin surges in ovariectomized, estrogen-treated rats. Neuroendocrinology Yamamoto, T., Nakahata, Y., Soma, H., Akashi, M., Mamine, T., Takumi, T., 2004.
69 (4), 227–237. Transcriptional oscillation of canonical clock genes in mouse peripheral tissues.
van der Beek, E.M., Wiegant, V.M., van der Donk, H.A., van den Hurk, R., BMC Mol. Biol. 5, 18.
Buijs, R.M., 1993. Lesions of the suprachiasmatic nucleus indicate the presence Yamazaki, S., Numano, R., Abe, M., Hida, A., Takahashi, R., Ueda, M., Block, G.D.,
of a direct vasoactive intestinal polypeptide-containing projection to Sakaki, Y., Menaker, M., Tei, H., 2000. Resetting central and peripheral circadian
gonadotrophin-releasing hormone neurons in the female rat. J. Neuroendocrinol. oscillators in transgenic rats. Science 288 (5466), 682–685.
5 (2), 137–144. Yamazaki, S., Takahashi, J.S., 2005. Real-time luminescence reporting of circadian
van der Horst, G.T., Muijtjens, M., Kobayashi, K., Takano, R., Kanno, S., Takao, M., de gene expression in mammals. Methods Enzymol. 393, 288–301.
Wit, J., Verkerk, A., Eker, A.P., van Leenen, D., Buijs, R., Bootsma, D., Yan, L., Karatsoreos, I., Lesauter, J., Welsh, D.K., Kay, S., Foley, D., Silver, R., 2007.
Hoeijmakers, J.H., Yasui, A., 1999. Mammalian Cry1 and Cry2 are essential for Exploring spatiotemporal organization of SCN circuits. Cold Spring Harb. Symp.
maintenance of circadian rhythms. Nature 398 (6728), 627–630. Quant. Biol. 72, 527–541.
Circadian Regulation of Endocrine Functions 369

Yoo, S.H., Yamazaki, S., Lowrey, P.L., Shimomura, K., Ko, C.H., Buhr, E.D., Zhao, S., Kriegsfeld, L.J., 2009. Daily changes in GT1-7 cell sensitivity to GnRH
Siepka, S.M., Hong, H.K., Oh, W.J., Yoo, O.J., Menaker, M., Takahashi, J.S., secretagogues that trigger ovulation. Neuroendocrinology 89 (4), 448–457.
2004. PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals Zylka, M.J., Shearman, L.P., Weaver, D.R., Reppert, S.M., 1998. Three period
persistent circadian oscillations in mouse peripheral tissues. Proc. Natl. Acad. Sci. homologs in mammals: differential light responses in the suprachiasmatic
U.S.A. 101 (15), 5339–5346. circadian clock and oscillating transcripts outside of brain. Neuron 20 (6),
Zhang, E.E., Kay, S.A., 2010. Clocks not winding down: unravelling circadian 1103–1110.
networks. Nat. Rev. Mol. Cell Biol. 11 (11), 764–776.
Zhang, R., Lahens, N.F., Ballance, H.I., Hughes, M.E., Hogenesch, J.B., 2014.
A circadian gene expression atlas in mammals: implications for biology and
medicine. Proc. Natl. Acad. Sci. U.S.A. 111 (45), 16219–16224.
This page intentionally left blank
1.13 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates
Tyler J Stevenson, Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
Brian J Prendergast, University of Chicago, Chicago, IL, USA
Randy J Nelson, The Ohio State University Wexner Medical Center, Columbus, OH, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by B.J. Prendergast, R.J. Nelson, I. Zucker, volume 1, pp. 507–540, Ó 2009, Elsevier Inc.

1.13.1 Introduction 371


1.13.2 Evolution of Seasonal Rhythms 372
1.13.3 Formal Classifications of Seasonal Rhythms 373
1.13.3.1 Type I Rhythms 373
1.13.3.1.1 Induction and Termination of Photorefractoriness 373
1.13.3.1.2 Critical Day Lengths 374
1.13.3.1.3 Puberty 375
1.13.3.1.4 Sexual Behavior and Physiology 375
1.13.3.2 Type II Rhythms 376
1.13.3.2.1 Entrainment of Type II Rhythms 377
1.13.3.2.2 Circannual Modulation of Circadian Organization 377
1.13.4 Endocrine Transduction of Photoperiod Signals 377
1.13.4.1 Duration versus Phase of the Melatonin Signal 377
1.13.4.2 Decoding Melatonin Signals 378
1.13.5 Neural Bases for Photoperiodism 378
1.13.5.1 Pathways for Melatonin Secretion and Hypothalamic-Binding Sites 378
1.13.5.2 Localized Thyroid Hormone Synthesis Induces Morphological Changes in the CNS 380
1.13.5.3 Photoperiod and Hormone Regulation of GnRH, Kisspeptin, and RFRP-3 380
1.13.6 Maternal–Fetal Communication of Day Length 381
1.13.7 Seasonal Rhythms in Primates 382
1.13.7.1 Nonhuman Primates 382
1.13.7.2 Humans 383
1.13.7.3 Evolution and Human Seasonality 383
1.13.7.4 Human Seasonal Reproductive Rhythms 383
1.13.7.5 Melatonin and Human Reproduction 384
1.13.7.6 What Can Animal Studies Tell Us about Human Seasonality? 384
1.13.8 Seasonal Rhythms in Nonreproductive Traits 384
1.13.8.1 Immune Function 384
1.13.8.2 Energetics and Immune Function 385
1.13.8.3 Social Organization: Affiliation and Aggression 387
1.13.8.4 Brain Development 388
References 391

Glossary
Circannual rhythm An endogenous self-sustained Photoperiodism The ability of an organism to measure
oscillation with a period of approximately 12 months. environmental day length.
Melatonin An indoleamine hormone secreted by the
pineal gland.

1.13.1 Introduction time in the same months on successive years. Seasonality can
be inferred or distinguished from mere synchrony when more
Seasonal phenotypes in behavior and physiology are pervasive; than a single cycle has been tracked. Strong seasonality generally
many reflect adaptations to matching environmental periodic- implies interanimal synchrony within a population, but the
ities and presumably have their highest fitness in the seasons converse is not necessarily true (Di Bitetti and Janson, 2000).
in which they occur (Brakefield, 1996). Seasonality is appropri- Pronounced seasonal variations in day length, temperature,
ately conferred upon traits that recur during a relatively limited and humidity derive from the yearly orbit of Earth about the

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00013-4 371


372 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

Sun. The amount and intensity of solar radiation varies with reproductive, immunological, and select behavioral and cogni-
latitude; the greater the distance from the equator, the more tive rhythms, largely in rodent models, reflects our own
pronounced interseasonal differences in ambient temperature research backgrounds and is not intended to diminish the
and solar radiation. Plant growth, which is also affected by importance of nonreproductive rhythms in the overall
other abiotic factors such as seasonal winds and patterns of economy of mammals, nor the significance of other mamma-
rain and snowfall, determines food availability and reproduc- lian and nonmammalian orders.
tion in herbivores and, consequently, in carnivores too.
Climatological shaping of animal structure and function is
a neglected aspect of mammalian neuroendocrinology research; 1.13.2 Evolution of Seasonal Rhythms
this is a major limitation, considering that the annual variation
in mean temperature in North America exceeds the glacial– Successful reproduction is profoundly influenced by food avail-
interglacial changes in mean annual temperature during the ability. Foraging effort, ambient temperatures, presence of
Pleistocene (Potts, 1998). Nonclimatological factors also predators, and quality and quantity of food located during
contribute to the evolution of seasonality, not least in humans, foraging determine whether or not reproduction will succeed
where social customs, religious practices, and legal regulations (Bronson, 1989). Two different modes of responding to food
account for some seasonal rhythms (Farrell and Pease, 1994; abundance have been identified. At the time of mating, food
Brewis et al., 1996; Dickert-Conlin and Chandra, 1999). This and energy reserves may be relatively low for income breeders,
chapter updates the article published in the second edition of and natural selection favors individual females that produce
this series (Prendergast et al., 2009). Our aim is to review recent offspring coincident with an abundance of high-quality food
seasonality research in the context of established frameworks. during lactation and at the time of weaning (income breeders;
The selective approach taken in this chapter does not Figure 1(a)). The latter stage of lactation is an extreme energetic
provide an in-depth treatment of formal models of photoperi- bottleneck for small mammals (Bronson, 1985). An alternative
odism (Goldman, 2001), nor does it address literatures on mating strategy is capital breeding; mating is triggered by an
nonmammalian vertebrates or invertebrates, each of which abundance of food and/or prior energy storage; lactation and
informs analyses of mammalian seasonality (see Nelson weaning of young occur during declines in food availability
et al., 2010). The extensive literature on endocrine changes (Figure 1(a)). Animals resident in highly seasonal environ-
unrelated to behavior is not considered. The emphasis on ments with short growing seasons are more likely to evolve

Figure 1 (a) Two distinct modes of responding to seasonality in the abundance of preferred foods. In A1, females are selected to give birth before
the annual food peak; they have evolved responsiveness to environmental cues that lead to conception one gestation period before this optimal
timing (income breeding). In A2, females respond directly to increases in food abundance as cues for conception, leading to births one gestation
period after that event, often coinciding with the decline in food availability (capital breeding). Line arrows indicate timing of conceptions; outlined
arrows indicate timing of births. (b) Timing of births among Soay sheep on the island of St Kilda in relation to availability and digestibility of food.
Births are restricted to a period early enough for the ewe and lambs to capitalize on abundant food of the spring pasture. Data from Lincoln, G.A.,
Short, R.V., 1980. Seasonal breeding: nature’s contraceptive. Recent Prog. Horm. Res. 36, 1–52.
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 373

seasonal phenotypes than conspecifics whose niches provide and boreal mammals, this has been argued to reflect an ances-
more evenly distributed food resources throughout the year. tral form of mammalian seasonality from which type II
Food generalists, such as baboons, monitor and exploit many rhythms evolved (Farner, 1985). Type II rhythms are endoge-
plant and fruit species, switching to less preferred items in nous but persist for two or more cycles even when day length,
times of scarcity (Hemingway and Bynum, 2005) and are less temperature, humidity, and food availability are held constant
subject to seasonal pressures than specialists reliant on a few throughout the year (Figure 2(b); Pengelley and Asmundson,
seasonally variable foods (e.g., a local population of vervets 1974; Gwinner, 1986). Also referred to as ‘circannual’ rhythms,
went extinct when their main food source, fever trees, died type II rhythms are characteristic of longer-lived mammals
off (Alberts et al., 2005). from several orders (primates, bats, carnivores, ungulates,
Small mammals have higher metabolic rates, higher costs rodents); their period is usually <12 months, generated by bio-
of thermoregulation, reduced fat stores with which to bridge logical oscillators that have eluded localization (Zucker, 2001),
intervals of food scarcity, and shorter life spans than do larger although recent molecular evidence points to coordinated
animals. They tend to breed more opportunistically than large seasonal activation of molecular events in the pars tuberalis
species, with greater year-to-year and locale-to-locale vari- (PT) that may constitute output signals from the circannual
ability. Although seasonal timing of food availability is oscillator(s) (Sáenz de Miera et al., 2014). Seasonal changes
usually generally constant from year to year, animals that in day length entrain both type I and II rhythms to a period
rigidly commit to reproduction at a specified calendar date of 12 months. Syrian (Mesocricetus auratus) and Siberian (Pho-
in disregard of local temperature variations, time of the first dopus sungorus) hamsters and golden-mantled ground squirrels
frost, or snow melt are disadvantaged (Lee and Gorman, (Spermophilus lateralis) are the most thoroughly studied species
2000). Day length, by far the most accurate predictor of phase with type I and II rhythms, respectively.
within the geophysical cycle, is sufficient to synchronize most Type III rhythms, which have been neglected by chronobio-
seasonal rhythms, and in many environments it is an accurate logists, are based on a strict stimulus-response system. Typi-
predictor of food availability (Figure 1(b)), but mammalian cally, environmental signals present at a particular time of
reproduction in the wild is typically affected by multiple year control effector systems with little or no modulation
cues (Bronson, 1989). The neglect of interactions among the from time-keeping mechanisms (Figure 2(c)).
several proximate determinants of seasonal rhythms is Below we provide an overview of type I and II rhythms, with
a feature of all but a handful of laboratory investigations. an emphasis on hormone–behavior interactions and recent
Also typically missing is the distinction between ‘strategic’ work in the field. The version of this chapter in the first edition
(photoperiod-dependent) versus ‘tactical’ (supplemental) of this series addressed these issues in greater detail.
cues in seasonality (Follett, 2015) and differential neural
circuits that may be involved in these two approaches
1.13.3.1 Type I Rhythms
(Migaud et al., 2015). Seasonal breeding may be abandoned
or relaxed in animals approaching the end of their lives; their Many North Temperate Zone female rodents restrict reproduc-
residual reproductive value is reduced (Horton and Yellon, tion to the months of April–August, during which they
2001), and presumably the cost–benefit ratio of reproduction produce one or more litters. The seasonal rhythm for such
versus survival favors out-of-season reproduction. long-day breeders can be replicated by exposing animals to
The above views of seasonality emphasize post-hoc adap- natural photocycles, even if all other factors are held constant.
tive scenarios of considerable surface plausibility. This adaptive For sexually mature individuals, the decreasing day lengths of
hypothesis approach has been criticized because it stops short mid- to late summer trigger the onset of gonadal involution,
of direct field validation (Huey and Berrigan, 1996), and eventually eliminating spermatogenesis, ovulation, and
assumes that physiological adjustments to the immediate envi- mating behavior. Reproductive quiescence endures for
ronment must increase fitness. A given seasonal phenotype can 5 months, yielding in mid-winter to gonadal regrowth
reflect random drift, factors other than natural selection, and (recrudescence); approximately 6 weeks later animals mate
adaptation to past rather than present environmental condi- (Butler et al., 2007a,b).
tions (Huey and Berrigan, 1996).
The mediation of mammalian seasonal breeding by day 1.13.3.1.1 Induction and Termination of Photorefractoriness
length and photoperiodic changes in melatonin production is Late-winter initiation of gonadal recrudescence reflects devel-
well established. Goldman et al. (2003) noted that the similar opment of refractoriness to short day lengths and is indepen-
actions of pineal melatonin in various mammals support dent from the small increases in day length after the winter
a single ancestral origin for photoperiodism early in the history solstice (Gorman and Zucker, 1995). Mid-winter gonadal reju-
of the mammalian lineage. venation is termed ‘spontaneous recrudescence’ because it
occurs in the absence of long day lengths; it defines the onset
of the refractory state in which reproduction is disinhibited
1.13.3 Formal Classifications of Seasonal Rhythms and no longer depends on the presence of long days
(Figure 2(a)), and it is triggered by an interval timer set during
Two principal forms of seasonal rhythms have been distin- the first few weeks of short days (Prendergast et al., 2000a; Park
guished (Zucker et al., 1991). Type I rhythms are based on an et al., 2006). The interval timer exhibits considerable inter-
endogenous interval timer and do not persist for more than sexual and interindividual variability; males typically initiate
a single cycle in the absence of environmental resetting recrudescence 4–6 weeks in advance of females (Prendergast
(Figure 2(a)). Common among many short-lived temperate et al., 2004b; Butler et al., 2007a,b). Refractoriness to short
374 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

(a) Type I rhythm


Mixed: Driven by endogenous and exogenous components
(e.g., Syrian hamster reproduction)

Reproductively Reproductively
active active

e al
ce o n ad
Te
sti

nc
cu

rud us g
es
Ce us c

lar
tro
ss

es
rec neo
Termination of

reg
ati yclic
photorefractoriness

ta
on

res

on
of y

sio

Sp
it

n
Reproductive
quiescence
Photorefractory Photosensitive

M A M J J A S O N D J F M A M J J A S O N

(b) Type II rhythm


Endogenous: Product of circannual clock(s)
(e.g., ground squirrel body weight)

Events analogous to type I rhythms,


but no phase transition is dependent
on photoperiod.

M A M J J A S O N D J F M A M J J A S O N

(c) Type III rhythm


Exogenous: Environmentally triggered
(e.g., hay fever)

Events analogous to type I & type II rhythms,


but all phase transitions are dependent
on stimuli in the environment.

M A M J J A S O N D J F M A M J J A S O N

Figure 2 Schematic representation of seasonal rhythms. (a) Type I (mixed) rhythms involve exogenous (photoperiod) and endogenous (interval
timer) components. Decreasing day lengths (1) initiate gonadal regression in the late summer and (2) trigger an interval timer that eventually renders
animals refractory to short days 20 weeks later leading to gonadal recrudescence. Exposure to long day lengths in the spring and summer ‘breaks’
refractoriness and resensitizes the neuroendocrine axis to short days. (b) Type II (circannual) rhythms are entirely endogenous and typically have
a period less than 12 months. Seasonal changes in day length synchronize type II rhythms to a period of exactly 1 year. (c) Type III rhythms are
entirely exogenous and driven by environmental stimuli.

days persists until animals are exposed for several weeks to the entrained by natural photoperiods but not evident under static
relatively longer days of spring and summer (Kauffman et al., exposure to short days (Butler et al., 2010). The foregoing
2003); this allows overwintered animals to undergo a second description applies to several reproductive and nonreproduc-
gonadal regression when day lengths again decrease in late tive type I photoperiodic traits.
summer. Although sufficient in this regard, recent work indi-
cated that melatonin is not mandatory for the ‘breaking’ of 1.13.3.1.2 Critical Day Lengths
refractoriness. Under a simulated natural photoperiod, The critical day length was initially proposed as a value above
hamsters that were pinealectomized at the spring equinox and below which long-day breeders exhibit the summer and
(prior to the appearance of categorically long photoperiods) winter phenotypes, respectively (Elliott, 1976). Critical day
nevertheless exhibited gonadal regression in response to length values were originally established with a protocol that
long-duration melatonin signals delivered weeks later, suggest- transferred animals between invariant long or short photope-
ing an endogenous component to the seasonal cycle that is riods (e.g., 16–10 L) in a single day, an event that bears no
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 375

resemblance to the gradual changes in day length experienced steroid-independent mechanisms may also participate in the
in nature. The critical day length concept generally refers to termination of the breeding season.
a fixed value at which a trait reverts between seasonal pheno-
types under conditions of thermoneutrality, access to unlim- 1.13.3.1.4.1 Males
ited free food, inescapable exposure to light, absence of Mating behavior declines in Syrian hamsters transferred from
conspecifics, and other conditions that likewise seldom prevail long to short days (Morin and Zucker, 1978; Campbell et al.,
in nature. Several lines of evidence have compelled 1978). Male hamsters undergo gonadal regression after 6–
modification of the strict critical day length framework, 10 weeks of exposure to short photoperiods and circulating
including photoperiod history effects (Duncan et al., 1985; testosterone (T) declines over this interval (Reiter, 1980). Defi-
Darrow and Goldman, 1986), graded effects of photoperiods cits in male sexual behavior are apparent after several weeks of
below the critical day length (Powers et al., 1997), and robust exposure to short days (Powers et al., 1989). Castrated
effects of simulated natural photoperiods (Gorman and hamsters in short days copulated less than their long-day coun-
Zucker, 1995, 1998). The pattern of change in day length codes terparts during replacement therapy with testosterone (Morin
important information, and in some instances changes in day and Zucker, 1978). In agreement with other studies (Powers
length are more salient than the absolute length of the day et al., 1989), short days apparently reduce responsiveness to
(Nicholls et al., 1988; Gorman and Zucker, 1995). Although hormones in neural substrates that activate copulatory
useful as an analytic tool, its value in the exegesis of seasonality behavior and thus may elevate thresholds for hormonal activa-
in photoperiodic rodents has likely been overemphasized. tion of copulation. However, conclusions derived from
hormone replacement studies can be questioned on grounds
that hormone implants fail to replicate the episodic pattern
1.13.3.1.3 Puberty
of testicular androgen secretion. For example, the presence of
Rates of somatic and reproductive development differ mark-
T in the circulation during the circadian interpulse interval is
edly in beginning cohorts versus end-of-season cohorts; rapid
not required to maintain male hamster sexual behavior (Park
somatic growth and puberty occur in early-born offspring of
et al., 2007).
several species (Worth et al., 1973), whereas those born several
In Syrian hamsters, there is little indication that a steroid-
weeks after the solstice forego reproduction in the season of
independent decrease in motivation to mate contributes to
birth, overwinter, and first attain sexual competence and adult
the seasonal suppression of reproductive behavior. In Siberian
body mass at 6 months of age or greater. Variations in day
hamsters, however, copulatory behavior is sustained for
length directly control whether puberty occurs in the season
months following castration but can be inhibited during this
of birth. Hamsters housed in simulated natural photoperiods
interval by exposure to short days, implicating gonadal
and born 8 or more weeks after the summer solstice fails to
hormone-independent effects of photoperiod in this species
undergo sexual maturation in the year of their birth; rather,
(Park et al., 2004).
they initiate synchronous puberty in the following spring, irre-
Part of the decline in mating among short-day male
spective of birth date (Butler et al., 2007a,b). The interval timer
hamsters may be caused by altered responses to vaginal chemo-
underlying vernal spontaneous development is set by the pre-
attractants, although the data are inconsistent across species
vailing photoperiod shortly after weaning (3–9 weeks of
(Miernicki et al., 1990). In some reports, chemosensory stimu-
age) and is dependent on melatonin signals generated during
lation of luteinizing hormone (LH) by heterosexual conspe-
this interval (Gorman, 2003). The subset of individuals
cifics was categorically abolished by exposure to short days
born 8 weeks after the summer solstice likely forms the
(Anand et al., 2002); in others, investigation of estrous females
main breeding nucleus the following spring (Butler et al.,
was greater in short-day males than in long-day males
2007a,b). Depending on species, individuals born before the
(Honrado et al., 1991).
summer solstice may not survive for more than a single
Social stimuli interact with seasonal changes in day length
breeding season (Sadleir, 1969).
to influence gonadal status. In short-day housed deer mice
(Peromyscus maniculatus), growth of the testes and seminal
1.13.3.1.4 Sexual Behavior and Physiology vesicles, indicators of increased androgen secretion, was stim-
Changes in photoperiod are potent regulators of male and ulated by the presence of a long-day female in each male’s
female sex behaviors in the laboratory. In principle, only one cage (Whitsett and Lawton, 1982). Recovery of sexual
sex needs to terminate reproductive activity to prevent out-of- behavior in short days was also accelerated in photorefractory
season breeding. In Syrian hamsters, the inhibitory effect of male Syrian hamsters provided with opportunities to interact
short days on mating behavior occurs several weeks earlier in with long-day females (Honrado and Fleming, 1996). Cohab-
females relative to males; in both sexes, a seasonal decline in itation with heterosexual conspecifics inhibited gonadal size
fertility is evident prior to a measurable decrease in sex in Siberian hamsters housed in intermediate-duration day
behavior (Beery et al., 2007). Because copulation in most lengths (Paul et al., 2009). Testicular regression was blocked
mammals is strictly tied to gonadal hormones, photoperiodic in Siberian hamsters transferred to short days in tandem
changes in sex behavior could be achieved by a reduction in with a gonad-intact summer-phenotype female (Hegstrom
hormone secretion several weeks before conditions become and Breedlove, 1999b) but was unaffected by cohabitation
unfavorable for offspring survival. Alternatively, substrates with an ovariectomized female (Weil et al., 2007). In this
that mediate mating activity could become unresponsive species, maintenance of LH production in males requires
to steroid hormones several weeks before the decline of pheromones contained in the female feces (Anand et al.,
gonadal steroidogenesis eliminates copulatory behavior. Lastly, 2004). Females do not elicit LH surges in males adapted to
376 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

short days, indicating that photoperiod gates LH responsive- is reduced but not eliminated in short day lengths and, as in
ness to pheromones (Anand et al., 2002). The extent to which males, may reflect an elevation of the threshold for activation
interactions with females prevent males in the field from of specific behavioral components (cf. Morin and Zucker,
undergoing gonadal involution as the end of the breeding 1978).
season approaches is unknown. There is disagreement about whether photoperiodic inhibi-
An intact pineal gland is essential for inhibition of sexual tion of female sex behavior in Syrian hamsters is mediated by
behavior in short day lengths (Miernicki et al., 1990). Pinealec- the pineal gland. Karp and Powers (1993) implicated the pineal
tomized male–female pairs of Syrian hamsters, unlike intact gland, whereas Badura and Nunez (1989) did not. Differences
controls, produced offspring during the winter when housed between the two studies in the interval between pinealectomy
in a natural photocycle (Reiter, 1974), again emphasizing the and behavioral assessments may explain the discrepant claims,
essential role of the pineal gland in transducing effects of short as residual effects of pineal hormones can persist for as long as
day lengths on the reproductive axis. a month (Ruby et al., 1989), perhaps accounting for the dimin-
Activation of male sex behavior in Syrian hamsters involves ished responsiveness to steroid hormones in recently pinealec-
conversion of T to estradiol via aromatization in neural tissue tomized females. Pinealectomy accelerated resumption of
(Wood, 1996). Aromatase activity in the preoptic area(POA) estrous cycles in anestrous Siberian hamsters previously housed
was not affected by day length or castration but was decreased in short days (Schlatt et al., 1993). The pineal gland mediates
in the anterior hypothalamus of short-day males and may effects of day length on intrasexual aggressive behavior of
contribute to behavioral deficits (Hutchison et al., 1991). female hamsters (Fleming et al., 1989).
Callard et al. (1986) documented decreased aromatase activity Reduced behavioral responsiveness to estradiol in short-day
in whole hypothalamus of both intact and castrated males female hamsters is accompanied by reduced estrogen receptor
housed in short days; androstenedione and T replacement in immunoreactivity (ER-ir) in the medial POA and by increases
castrated hamsters lowered aromatase activity and induced in ER-ir positive cells in the amygdala. Induction of
fewer estradiol receptors in short than long days. The sugges- progesterone receptors by estradiol is significantly reduced in
tion that aromatase activity is an important rate limiting step the hypothalamic ventromedial nucleus (Mangels et al.,
that determines the number of estrogen receptors in dience- 1998), a target tissue for hormonal induction of lordosis
phalic tissues is a plausible mechanism by which day length (Pleim et al., 1990).
may control androgen-dependent processes. In some rodents, day length may affect female fecundity
Low ambient temperature does not by itself induce gonadal indirectly, via changes in male physiology and behavior. Wean-
regression in some long-day breeding rodents (e.g., Desjardins ling female Peromyscus caged together with adult males in short
and Lopez, 1980), but it is effective in Turkish hamsters (Hall days sustained greater uterine weights than social isolates
et al., 1982). Maintenance at 5  C in conjunction with short (Garcia and Whitsett, 1983). Pheromones or the behavior of
day lengths markedly accelerated gonadal regression in several one sex may override inhibitory effects of short days on the
rodent species (Steinlechner et al., 1991; Ruf et al., 1997; Larkin other sex (Walkden-Brown et al., 1999).
et al., 2002). Conversely, higher ambient temperatures delayed Almost all female meadow voles are pregnant during the
appearance of short-day gonadal responses (Larkin et al., summer but fewer than 50% are pregnant during the winter
2002). The day length at which testicular regression occurs in (Meek and Lee, 1993a). Complex behavioral changes
Siberian hamsters is 2 h longer at 5 than 22  C (15 vs contribute to the decreased incidence of winter pregnancy.
13 L; Steinlechner et al., 1991). At 50 N latitude, well within Virgin female voles maintained in short days for 8 weeks all
the home range of this species, this would accelerate gonadal mated when paired with long-day males, but fewer short-day
regression by 5 weeks (Steinlechner et al., 1991). We are females produced litters (Meek and Lee, 1993a). Female
unaware of studies of mating behavior of photoperiodic responsiveness to male pheromones and male responsiveness
rodents under conditions of elevated or reduced ambient to the female interact to affect winter pregnancy rates in this
temperature. species. Additional complexity is indicated by the finding that
prior sexual experience inhibited reproductive responsiveness
1.13.3.1.4.2 Females to short days (Meek and Lee, 1993b).
Syrian hamsters are rendered anovulatory within 6 weeks of
transfer from long to short days (Seegal and Goldman,
1.13.3.2 Type II Rhythms
1975). They cease to display lordosis following transfer to short
days (Badura and Nunez, 1989). In Siberian hamsters, estrous Endogenous annual rhythms have been documented in a wide
cyclicity is eliminated for 20 weeks beginning 7 weeks into variety of animals (Paul et al., 2008). In the golden-mantled
short-day exposure (Schlatt et al., 1993). ground squirrel, these rhythms persist for many cycles irrespec-
Short day lengths reduced behavioral responsiveness to tive of environmental lighting conditions (Zucker, 2001); in
estradiol and progesterone in female Syrian hamsters (Karp others, only certain photoperiodic or temperature conditions
and Powers, 1993; Bittman et al., 1990; Honrado et al., 1991; are compatible with rhythm expression (Gwinner, 1986), and
Badura and Nunez, 1989; Mangels et al., 1998). This effect in still others the rhythm fades after several years.
was only observed at relatively low doses of estradiol (Badura The annual sequence of gonadal growth, a restricted
et al., 1987; Karp and Powers, 1993). In one instance, sexual breeding season, and gonadal involution are superficially
receptivity was attenuated in response to treatment with estra- similar in type I and II cycles. Closer examination reveals signif-
diol but not to estradiol followed by progesterone (Badura icant differences: reproduction in type II species is not main-
et al., 1987). Behavioral responsiveness to steroid hormones tained indefinitely during exposure to any sequence of
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 377

constant or variable day lengths; either gonadal recrudescence cycle and with high-amplitude seasonal changes in body
is not accompanied by refractoriness or, if refractoriness occurs, temperature, circannual rhythms in circadian organization
it terminates spontaneously (Paul et al., 2008). This feature persist independent of reproductive (Lee and Zucker, 1995)
permits a self-sustaining annual rhythm. Recent high- and thermoregulatory rhythms (Freeman and Zucker, 2000).
throughput screening in the 13-lined ground squirrel identified
scores of plasma metabolites that change significantly over the
hibernation cycle, indicating widespread changes in metabo- 1.13.4 Endocrine Transduction of Photoperiod
lism that accompany the profound heterothermy exhibited Signals
by hibernating rodents (Epperson et al., 2011).
An alternative view is that type II rhythms emerge from Initial clues that the pineal gland played a role in seasonality
sequential refractoriness to short and long days, respectively. came out of the work of Czyba et al. (1964): excision of the
Ferrets and sheep develop refractoriness to both long and short pineal gland reduced the rate and magnitude of seasonal
day lengths (Herbert and Klinowska, 1978; Almeida and gonadal regression in male Syrian hamsters. This outcome
Lincoln, 1984; Robinson et al., 1985). This feature may be char- anticipated results obtained under more controlled laboratory
acteristic of many type II rhythms but is not universally present; conditions by Hoffman and Reiter (1965) who reported that
ground squirrels make repeated, spontaneous transitions from gonadal regression in response to blinding or exposure to short
winter to summer phenotypes even when housed continuously photoperiods was absent in pinealectomized hamsters. Mela-
in an unvarying day length (Zucker, 2001). tonin is secreted by the pineal gland in a circadian pattern,
with peak gland and blood concentrations occurring during
1.13.3.2.1 Entrainment of Type II Rhythms subjective night. The light-entrainable circadian clock in the
The period of Type II rhythms deviates from 12 months in the suprachiasmatic nuclei (SCN) drives the melatonin secretory
absence of entraining signals; endogenous periods of body rhythm; separate circadian oscillators, entrained to lights-off
mass, hibernation and reproductive rhythms of squirrels are and lights-on, regulate the onset and offset of nocturnal
10.5 months, necessitating a phase delay of 1.5 months hormone secretion, respectively (Illnerova, 1991). Over the
each year to synchronize with the annual geophysical cycle. annual cycle, the duration of nocturnal melatonin varies
Simulated annual variations in day length effect this correction inversely with day length (Darrow and Goldman, 1986).
(Lee and Zucker, 1991). Pinealectomy eliminates melatonin
from the circulation and obliterates most type I rhythms, which
1.13.4.1 Duration versus Phase of the Melatonin Signal
invariably revert to the summer or long-day phenotype (Reiter,
1980). Type II rhythms, in contrast, persist after pinealectomy The circadian phase during which melatonin is elevated and
(Zucker, 1985; Woodfill et al., 1991), but the ability to entrain the duration of nocturnal elevation are two features which
to a simulated natural photoperiod is lost (Hiebert et al., could account for its gonadotropic effects. Syrian hamsters
2000). Infusions of melatonin that mimic durations of housed in 14 L exhibited gonadal regression when injected
nocturnal melatonin during the interval from the summer with melatonin during late afternoon (shortly before the onset
solstice to the autumnal equinox entrain the LH rhythm of of darkness) but not when treated earlier in the light phase
pinealectomized ewes to a period of 12 months (Woodfill (Reiter et al., 1974; Tamarkin et al., 1976b). This initially sug-
et al., 1994). In ground squirrels, melatonin treatments initi- gested a circadian rhythm in reproductive responsiveness to
ated in late summer produced durable phase shifts of body melatonin (Tamarkin et al., 1977), but subsequent experi-
mass and reproductive rhythms; identical treatments in the ments indicated that, for regulation of seasonal traits, the dura-
spring were ineffective (Zucker, 2001). Circannual oscillators tion of the nocturnal melatonin peak is paramount (Carter and
in ground squirrels and sheep evidently respond differently Goldman, 1983; Bittman and Karsch, 1984). Timed melatonin
to melatonin at discrete phases of the annual cycle. infusions induced gonadal regression in pinealectomized
hamsters irrespective of the stage of the circadian cycle of its
1.13.3.2.2 Circannual Modulation of Circadian Organization administration; only the duration of the signal was relevant
Home range size and the amount of wheel-running activity to its antigonadotropic effects (Goldman et al., 1984; Gorman,
vary seasonally in several rodents. In type I species (e.g., voles, 2003; but see Gunduz and Stetson (2001) for a contrary view
hamsters), these transitions in activity are driven by seasonal that emphasizes coincidence of melatonin secretion with
changes in day length, temperature, and food availability; in a circadian rhythm of sensitivity to the hormone). Melatonin
type II species, however, seasonal variations in locomotor infusions 8 h per day suppressed serum gonadotropin and
activity are evident in the absence of environmental change. prolactin (PRL) concentrations and provoked gonadal regres-
The period of the squirrel free-running circadian locomotor sion, whereas infusions 6 h per day stimulated the reproduc-
rhythm is less than 24 h during the subjective summer and tive axis. As few as 4 weeks of daily long-duration melatonin
greater than 24 h during the subjective winter (Zucker et al., signals induced gonadal regression in adult Siberian hamsters,
1983); locomotor activity begins earlier and ends later in the and 6 weeks of such treatments sufficed in Syrian hamsters
day during subjective spring and summer than in autumn (Maywood and Hastings, 1995; Prendergast et al., 2000a).
and winter (Lee et al., 1990; Lee and Zucker, 1995). The interval From this and other work, the ‘duration hypothesis’ has
between successive recurrences of summer or winter patterns of emerged. The apparent circadian rhythm in responsiveness to
activity is 10.5 months and is driven by an endogenous cir- melatonin injections in pineal-intact hamsters reflected the
cannual mechanism. Although increases in the duration and summation of exogenous and endogenous melatonin to result
distribution of activity are correlated with the reproductive in a longer daily signal. A version of the circadian sensitivity
378 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

hypothesis was resurrected to account for the observation that ventricle (3rdV) and a series of discrete, interconnected nuclei
Syrian hamsters exhibited gonadal regression when provided (Paul et al., 2008; Wood and Loudon, 2014; see Figure 3 for
with two 2.5-h melatonin infusions per day separated by a 5- current working model). The primary hypothalamic nuclei
h melatonin-free interval (Pitrosky et al., 1995); however, this include the arcuate nucleus (Arc), anteroventricular periventicu-
interpretation has been challenged (Hastings, 1996). The lar nucleus (AvPv), dorsomedial nucleus (DMH), and the POA.
viability of the duration hypothesis has been established in These core structures are critical for the generation and mainte-
several rodent and ungulate species and at present best nance of the constellation of genomic, molecular, and neuroen-
accounts for the way in which melatonin mediates photoperi- docrine changes that govern the photoperiodic regulation of
odism (Arendt, 1995). seasonal rhythms (Weems et al., 2015). As described in the prior
section, the pineal hormone melatonin is a key internal signal
that encodes day length; thus the duration of melatonin secre-
1.13.4.2 Decoding Melatonin Signals
tion provides the physiological mechanism for photoperiodic
Over the course of a year, the duration of nightly melatonin time measurement. Seasonal changes in melatonin binding
secretion neither varies markedly nor remains static from within core hypothalamic structures are essential to trigger
night-to-night; rather, melatonin signals either gradually genomic and morphological plasticity that underlies the photo-
increase or decrease as days get shorter and longer, respectively. period information essential for generation (type I) and/or
Just as photoperiod information is contained in the direction entrainment (types I and II) of seasonal rhythms. The role of
of change in day length, independently of absolute day length melatonin on seasonal rhythms has been most extensively
(Gorman and Zucker, 1995; Butler et al., 2007a,b), the direc- investigated using reproductive endpoints, and this section
tion of change in melatonin secretion conveys seasonal infor- will focus primarily on this literature.
mation to the neuroendocrine system. Melatonin infusions
gradually decreasing in duration from 10 to 7.5 h per night eli-
cited gonadal growth in Siberian hamsters, whereas those grad-
1.13.5.1 Pathways for Melatonin Secretion
ually increasing from 5 to 7.5 h per night induced regression
and Hypothalamic-Binding Sites
(Gorman and Zucker, 1997). Seasonal changes in day length impinge on the neuroendocrine
Delivery of ‘unnatural’ melatonin signals has unveiled substrates via a retinal–hypothalamic–pineal pathway (Morin
formal properties of the melatonin signal-processing system. and Allen, 2006). Light entrains circadian oscillators and alters
Short-duration melatonin signals that occur every other night gene expression in the SCN, which in turn initiate and termi-
fail to induce gonadal growth; likewise, long-duration mela- nate nightly melatonin synthesis (Illnerova et al., 1984; Elliott
tonin infusions at 48 h intervals do not trigger regression and Tamarkin, 1994). Melatonin is secreted at night, in propor-
(Elliott et al., 1989; Prendergast and Hugenberger, 1999). tion to the duration of the scotophase. Consistent with the
Neuroendocrine responsiveness to melatonin is frequency- essential role of melatonin in mammalian photoperiodism,
tuned: only signals recurring at frequencies approximating lesions along the retina–SCN–pineal pathway disrupt mela-
the organism’s endogenous frequency (in most cases 24 h) tonin secretion and reproductive responses to day length
elicit duration-appropriate responses. When challenged with (Rusak and Morin, 1976; Pickard and Turek, 1983; Bittman
a series of melatonin signals that vary in duration by several and Lehman, 1987).
hours from night-to-night, hamster reproductive responses sug- Two classes of melatonin receptors have been identified in
gested that a signal-averaging algorithm is used to abstract the mammalian CNS: membrane-bound, G protein-coupled
seasonal information (Piatkowska et al., 2003). high-affinity receptors (MT1 and MT2; Reppert et al., 1994)
Just as prior photoperiod history modifies the critical day and isoforms of the RZR/ROR family of orphan nuclear recep-
length for reproductive stimulation and inhibition, photope- tors, which bind melatonin with nanomolar affinity (Becker-
riod history likewise determines whether melatonin signals of Andre et al., 1994). There is no evidence that nuclear receptors
intermediate duration are regarded as long- or short-day cues. participate in control of seasonal changes by melatonin
For example, 7-h melatonin infusions elicited gonadal growth (Hazlerigg et al., 1996); the consensus is that these actions
in hamsters previously housed in short days but caused are mediated by MT1 receptors (Weaver et al., 1996; Prender-
gonadal regression in hamsters from long days (Prendergast gast, 2010). Although inbred laboratory mice are not reproduc-
et al., 2000a). Photoperiodic mammals encode a representation tively photoperiodic in the laboratory, Yasuo et al. (2009)
of recent prior day lengths which may provide a continuously reported that such mice exhibit molecular hypothalamic
changing context for the interpretation of intermediate day responses to melatonin treatment but failed to do so following
lengths. In nature, the encoding and eventual decay of such targeted deletion of the MT1 melatonin receptor. 125I mela-
photoperiodic history information may ensure that hamsters’ tonin binds with high affinity in the SCN of several rodents
responses to ambiguous photoperiod cues are influenced (Weaver et al., 1989). In Siberian hamsters, the thalamic para-
only by relatively recent day lengths. ventricular nucleus (PVN) and reunions nuclei (NRe) also
exhibit high-density melatonin binding (Duncan et al.,
1989). Among photoperiodic mammals, there is considerable
1.13.5 Neural Bases for Photoperiodism interspecific variability in the neural locations of high-affinity
melatonin-binding sites (Bittman et al., 1994; Bittman and
The neuroendocrine control of photoperiodism involves Weaver, 1990), ranging from diffuse (Peromyscus species: Kor-
an evolutionarily conserved cluster of hypothalamic brain ytko et al., 1995; Heideman et al., 1999) to nonexistent (ferrets:
regions that includes the PT, ependymal layer along the third Weaver and Reppert, 1990).
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 379

(a) (b)
Long day Short day

S
Caudal
lH
GnRH

ME
Tanycyte
Light
DIO2/3


Rostral PT TSHβ
NSt Thyrotropes

T3 MEL
PVN PT
RN ME
DM

SCN
↓ Gonadotropes

LH/FSH

Figure 3 Light-dependent regulation of seasonal rhythms. The nocturnal secretion of the pineal gland hormone melatonin (MEL) codes night length
in all known vertebrate species. Annual changes in day length lead to short (long days in summer) or long (short days in winter) durations in MEL
secretion. MEL acts both centrally and peripheral by binding to MEL receptors. (a) MEL receptor is widely distributed in the brain; this schematic
provides the general distribution using a rodent brain. (b) The pars tuberalis (PT) and median eminence (ME) in the ventral hypothalamus are the key
brain regions for the seasonal regulation of physiology and behavior in many species. Long day lengths (>12 h; left side) stimulate thyrotropes in
the PT to produce thyrotropin-stimulating hormone subunit-b (TSHb). Increased TSHb alters the ratio of thyroid hormone enzymes (deiodinase type
II and III, DIO2, and DIO3) leading to the increased local synthesis in triiodothyronine (T3). T3 then permits the release of gonadotropin-releasing
hormone1 (GnRH) which in turn stimulates gonadotropes to release luteinizing hormone (LH) and follicle-stimulating hormone (FSH). LH and FSH
induce gametogenesis and steroidogenesis facilitating reproductive physiology and behavior. In short days (right side), increased MEL duration
inhibits TSHb expression, reversing the ratio of DIO2/3, reducing local T3 concentrations and inducing gonadal involution. AvPv, anteroventral peri-
ventricular nucleus; DM, dorsomedial nucleus; lH, lateral habenula; NSt, nucleus of the stria terminalus; PVN, paraventricular nucleus; RN, reunions
nucleus; S, subiculum; SCN, suprachiasmatic nucleus. Adapted from Stevenson, T.J., Visser, M.E., Arnold, W., Parrett, P., Biello, S., Dawson, A.,
Denlinger, D.L., Dominoni, D., Ebling, F.J., Elton, S., Evans, N., Ferguson, H.M., Foster, R.G., Hau, M., Haydon, D.T., Hazlerigg, D.G., Heideman, P.,
Hopcraft, J.G., Jonsson, N.N., Kronfeld-Schor, N., Kumar, V., Lincoln, G.A., MacLeod, R., Martin, S.A., Martinez-Bakker, M., Nelson, R.J., Reed, T.,
Robinson, J.E., Rock, D., Schwartz, W.J., Steffan-Dewenter, I., Tauber, E., Thackeray, S.J., Umstatter, C., Yoshimura, T., Helm, B., 2015. Disrupted
seasonal biology impacts health, food security and ecosystems. Proc. R. Soc. B 282, 1–10.

The PT is the predominant neuroendocrine substrate (Lewis et al., 2002). Lesions of the SCN eliminated the antigo-
located within the core evolutionarily conserved hypothalamic nadal effects of long-duration melatonin signals in Siberian
nuclei implicated in seasonal rhythms that binds melatonin in (Bartness et al., 1991) but were without effect in Syrian
mammals. MT1 receptors are highly expressed in the PT and the hamsters (Bittman et al., 1979, 1989). DMH and VMH lesions
absence of melatonin binding facilitates the secretion of in Syrian hamsters (Maywood and Hastings, 1995; Bae et al.,
thyrotropin-stimulating hormone subunit-b (TSHb) from PT 1999; Lewis et al., 2002) and SCN lesions in Siberian hamsters
thyrotropes (reviewed below). Increased TSHb synthesis in (Bartness et al., 1991) spared the lactotropic (PRL) response to
long-day conditions triggers a cascade of events that include short-day melatonin signals. Melatonin signals are transduced
thyroid hormone-mediated neuroendocrine plasticity that into the hypothalamo–pituitary axis via multiple pathways
permits gonadotropin secretion from the pituitary gland that may be trait specific (Lincoln, 1990; Bartness et al.,
(Wood and Loudon, 2014). 1991; Maywood and Hastings, 1995). In hypothalamic–pitui-
Lesions of hypothalamic substrates enabled the necessity of tary disconnected sheep, melatonin implants in the PT-
melatonin receptors in anatomically localized regions to be inhibited PRL secretion in a manner that mimicked the cycle
examined. PT lesions are very difficult due to the small and of responsiveness and refractoriness to short days, even as
elongated structure of the tissue; however, lesions of several gonadal responses to melatonin were absent (Lincoln and
other hypothalamic nuclei involved in seasonal rhythmicity Clarke, 1997). In common with sheep, the hamster PT contains
(e.g., DMH, VMH) reveal these regions are critical for the main- substrates sufficient to decode melatonin signals that control
tenance of annual reproductive cycles. In male Syrian hamsters, PRL and gonadotropin secretion.
ablation of melatonin targets in the DMH eliminated the There is interspecific variability and intraspecific redun-
gonadal response to long-duration melatonin infusions dancy in the neural substrates that decode the nightly duration
(Maywood and Hastings, 1995; Maywood et al., 1996); in of melatonin signals. In pinealectomized sheep, Siberian
female hamsters, similar lesions centered on the junction of hamsters, and white-footed mice, melatonin implants or infu-
the DMH and the ventromedial nuclei (VMH) abolished sions in the anterior hypothalamus, SCN, MBH, NRe or PVT
the suppressive effect of short days on estrous cycles are sufficient to engage seasonal reproductive responses
380 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

(Glass and Lynch, 1982; Badura and Goldman, 1992; Lincoln provokes gonadal growth in photoinhibited hamsters and
and Maeda, 1992). In pinealectomized Siberian hamsters, blocks short day-induced gonadal regression (Freeman et al.,
melatonin implants localized to the SCN, NRe, or PVT were 2007; Barrett et al., 2007). In addition to a T3-dependent facil-
each sufficient to induce gonadal regression. The eventual itation of gonadotropin release from the pituitary, T3 has also
loss of gonadal responsiveness to melatonin in any one of these been implicated in the direct regulation of neuropeptides
nuclei was indicative of tissue-specific development of located in many hypothalamic nuclei (i.e., Arc, DMH, AvPv;
refractoriness and did not preclude responsiveness to Henson et al., 2013).
melatonin by other tissues (Freeman and Zucker, 2001). The eventual induction of photorefractoriness and loss of
Refractoriness appears to occur independently at several mela- responsiveness to short days may also depend on hypotha-
tonin target tissues. Other melatonin-mediated effects of lamic regulation of thyroid hormone signaling. Expression of
photoperiod in hamsters occur in a more target-specific thyroxine binding proteins, along with hypothalamic T4
manner: elimination of melatonin signaling in the NRe, but uptake, was each lower in photorefractory relative to responsive
not the SCN or PVT interfered with long-day stimulation of hamsters (Prendergast et al., 2002a). DIO2 expression
the testes, whereas obscuring melatonin signals to the SCN remained low after the development of refractoriness, but
and NRe but not the PVT disrupted acquisition of photoperi- DIO3 returned to long day-like levels (Watanabe et al.,
odic histories (Teubner and Freeman, 2007). 2007). Thus in recent years, it has become apparent that T3
can affect reproductive physiology in seasonal breeders via
morphological changes in tanycytes and via the expression of
1.13.5.2 Localized Thyroid Hormone Synthesis Induces
key neuropeptides involved in the neuroendocrine control of
Morphological Changes in the CNS
reproduction.
During the 1990s, thyroid hormones were clearly demon-
strated to play an essential role for mammalian photoperi-
1.13.5.3 Photoperiod and Hormone Regulation of GnRH,
odism (Parkinson and Follett, 1994, 1995; Moenter et al.,
Kisspeptin, and RFRP-3
1991). Given the consistent observation of thyroid-
dependent regulation of seasonal reproductive rhythms across As described in Section 1.13.4, a series of long-duration mela-
vertebrates, thyroid hormone signaling may reflect an evolu- tonin signals suppresses anterior pituitary gonadotropin secre-
tionarily conserved system (Paul et al., 2008). Photoperiod tion (Bartness et al., 1993; Goldman and Nelson, 1993).
itself has little effect on circulating T3 or T4 concentrations in Melatonin may act in the hypothalamus to control GnRH
Siberian hamsters (O’Jile and Bartness, 1992; Prendergast secretion, the pituitary to influence follicle-stimulating
et al., 2002a), although short days inhibited secretion of these hormone (FSH) and LH release, the gonads to affect respon-
hormones in Syrian hamsters (Vriend, 1983). Hypothyroidism siveness to gonadotropins, or at a combination of these sites.
does not abolish gonadal regression (Vriend and Wasserman, In hamsters, negative-feedback sensitivity of LH to testosterone
1986; Prendergast et al., 2002a) in short-day exposed hamsters. is enhanced by exposure to short days (Tamarkin et al., 1976a;
The recent identification of enzymes involved in thyroid Ellis and Turek, 1980) and attenuated by the development of
hormone catabolism has led to a significant advancement in photorefractoriness (Ellis et al., 1979; Matt and Stetson,
our understanding of the molecular mechanisms that govern 1979; Prendergast et al., 2006); the neural sites and mecha-
the internal representation of seasonal time information. Two nisms by which these effects occur are not known. There is
key enzymes called deiodinase type II and III (DIO2 and no consistent evidence that day length or melatonin alters
DIO3, respectively) are expressed in cells along the ependymal direct pituitary responsiveness to GnRH independent of steroid
layer in the third ventricle (Wood and Loudon, 2014). DIO2 feedback effects (e.g., Martin et al., 1977; Jetton et al., 1994). In
functions to convert the prohormone thyroxine (T4) into the ruminants a significant modulatory role for photoperiod
bioactive triiodothyronine (T3). Conversely, DIO3 reduces signals in this regard has been documented (Xu et al., 1992).
the bioactive forms of thyroid hormone by converting T4 into In some studies of Syrian and Siberian hamsters, the
reverse T3, and T3 into diiodothyronine (T2). Adaptation to number of hypothalamic GnRH-immunoreactive (GnRH-ir)
short days or treatment with melatonin yields decreases in neurons was increased in short day lengths (Ronchi et al.,
the expression of DIO2 in Syrian hamsters (Watanabe et al., 1992; Bernard et al., 1999). This effect was not found in
2004, 2007), and a single afternoon injection of melatonin other studies (Urbanski et al., 1991; Yellon, 1994; Bittman
inhibits DIO2 production (Yasuo et al., 2007). In contrast, et al., 1996). Interpretation of these data is further
DIO3 increases under short photoperiod (Watanabe et al., complicated by the fact that decreased GnRH-ir may reflect
2007; Barrett et al., 2007), an effect that is paralleled by less peptide production or more peptide release. It is not
decreases in methylation of the promoter region of the gene known whether melatonin affects GnRH neurons directly or
that encodes DIO3 (Stevenson and Prendergast, 2013). indirectly, although in some species there is overlap between
Changes in T3 concentrations induce tanycyte end-feet along brain nuclei that exhibit high-density melatonin binding and
the ependymal layer to retract (Yamamura et al., 2004); these those that contain GnRH-ir neurons (Glass, 1986; Korytko
morphological changes facilitate exposure of gonadotropin- et al., 1995).
releasing hormone (GnRH) neurosecretory terminals to the In contrast to GnRH, two recently discovered neuropep-
basal membrane of the median eminence. This response likely tides: kisspeptin (KiSS) and RFamide-related peptide-3
facilitates the release of gonadotropins from the pituitary gland (RFRP3, also referred to as gonadotropin-inhibitory hormone;
and, ultimately, drives photoperiodic changes in reproductive GnIH) exhibit marked seasonal variation in hypothalamic
physiology. In support of this conjecture, exogenous T3 expression (Weems et al., 2015). KiSS, is expressed in two
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 381

hypothalamic nuclei (Arc, AvPv), and stimulates GnRH secre- Some of these genes change in a manner that cannot be dis-
tion via GnRH neurons expressing the GPR54 receptor counted as masking effects of steroid feedback or entrainment
(Dungan et al., 2006). Adaptation to short days in Syrian (Ross et al., 2005; Watanabe et al., 2007), leaving viable the
hamsters inhibited kisspeptin-ir and mRNA expression in the hypothesis that their protein products may constitute critical
Arc (Revel et al., 2006). This effect was not a consequence of intermediates between melatonin-mediated day length infor-
changes in circulating testosterone but was abolished by pine- mation and the HPG axis or are output signals of an endoge-
alectomy, thereby implicating photoperiod-driven changes in nous circannual interval timer.
melatonin secretion, as the mediator of seasonal kisspeptin Cyclic expression of ‘clock genes’ in the central daily pace-
rhythms (Revel et al., 2006). In ovariectomized ewes, seasonal maker, the SCN and extra-SCN tissues are a key component
anestrus is accompanied by lower ARC kisspeptin mRNA of an autoregulatory feedback loop that confers circadian peri-
expression (Smith et al., 2007). By contrast, kisspeptin-ir was odicity on these cells (Takahashi, 2004). Because several of
increased in the ARC and decreased in the anteroventral peri- these genes are phase-locked to dusk and dawn, their expres-
ventricular nucleus following adaptation to short days in sion provides a neural representation of ambient photoperiod,
male and female Siberian hamsters (Greives et al., 2007; Mason which may complement the endocrine representation of day
et al., 2007). Exogenous kisspeptin is sufficient to stimulate length contained in the nightly melatonin signal (Lincoln,
gonadal hormone secretion in both hamster species (Revel 2002; Hastings and Herzog, 2004). Expression of at least
et al., 2006; Greives et al., 2007). Reports that kisspeptin also some clock genes rises and falls in response to melatonin
responds to changes in energy balance (Castellano et al., (Morgan et al., 1998; Messager et al., 1999, 2000; Johnston
2005) suggest it may transduce masking effects of food avail- et al., 2003, 2005). Melatonin-proficient Per2 mutant mice
ability on seasonal reproductive rhythms. KiSS neurons in the exhibited photoperiod-driven changes in Tshb, Dio2, and
Arc and AvPv do not appear to coexpress with melatonin recep- Dio3 gene expression, suggesting that Per2 is not mandatory
tors (Clarke and Caraty, 2013) suggesting that melatonin does for initial molecular responses to photoperiod change (Ikegami
not directly regulate seasonal variation in KiSS expression. In et al., 2013). The role of clock gene expression in the induction
Siberian hamsters, daily peripheral injections of T3 in short of photorefractoriness has also been investigated. Clock gene
day significantly increased the number of cells expressing expression in the short-day refractory hamster brain (Johnston
KiSS-ir in the AvPv, and a reduction was observed in the Arc et al., 2003) and the long-day refractory sheep PT (Lincoln
(Henson et al., 2013). These findings indicate a potential et al., 2005) continued to track dusk and dawn, and the
dual role for T3: in addition to the regulating tanycyte onset/offset of a melatonin signal to which the tissues were
morphology (and thus GnRH neurosecretory terminal no longer responsive.
exposure), the local synthesis of T3 in the hypothalamus Lesion and histological analyses over annual time scales
regulates KiSS neuronal function leading to enhanced GnRH suggest that the SCN and circadian clock genes are not neces-
activity. sary for the generation and maintenance of some seasonal
RFRP3 was originally identified in birds and shown in vivo rhythms. Annual oscillations in body mass persist in many
and in vitro to inhibit gonadotropin release in a dose- golden-mantled ground squirrels subjected to SCN lesions
dependent manner (reviewed in Bentley et al., 2006). RFRP3 (Dark et al., 1985). Moreover, Arctic reindeer exhibit robust
is localized to the DMH and directly interacts with GnRH to seasonal variation in melatonin duration that does not appear
regulate GnRH release. RFRP3 exhibits robust seasonal varia- to be entrained by circadian clock genes (Lu et al., 2010). These
tion in expression with greater mRNA and protein expression findings indicate the integrity of the SCN as well as cellular
in long-day conditions (Revel et al., 2008; Mason et al., oscillations in circadian clock genes are not critical components
2010; Ubuka, 2012). Daily melatonin injections are sufficient of the proximate mechanisms for seasonal rhythms in some
to inhibit RFRP3 expression and pinealectomized hamsters species. This raises the tantalizing conjecture for endogenous
maintain high DMH content after transfer to short-day photo- circannual clock timers for the expression of seasonal rhythms.
periods (Revel et al., 2008). Moreover, T3 has also been shown Indeed, annual patterns in the coastal dinoflagellate, Alexan-
to stimulate RFRP3 expression, as 2 weeks of afternoon injec- drium tamarense, suggest that even unicellular organisms
tions to short day-regressed hamsters stimulated more immu- contain endogenous circannual molecular machinery for the
noreactive RFRP3 (Henson et al., 2013). These data indicate timing of seasonal rhythms (Andersen and Keafer, 1987).
that photoperiodic regulation of RFRP3 plasticity may lie With the advent of ‘omics’ technologies, the ability to sequence
downstream of melatonin-driven changes in PT function (via entire genomes, delineate cellular and tissue transcriptomes,
T3 mechanisms) as well as the potential for direct effects on and examine the biological effectors contained within proteo-
expression. mic analyses offers opportunities to generate meaningful new
This report has primarily focused on the role of melatonin, insights into the neural mechanisms that govern seasonal
thyroid hormones, and neuropeptides (i.e., GnRH, KiSS, and rhythms.
RFRP3) for seasonal rhythms in reproductive physiology. It is
important to note that several other modulatory systems are
involved, and these have been covered in detail elsewhere. In 1.13.6 Maternal–Fetal Communication of Day
brief, transcription of several hypothalamic genes is affected Length
by photoperiod and the induction of photorefractoriness
(GPR50, VGF, histamine H3 receptor, CRBP1 and CRABP2, An earlier version of this chapter reviewed the literature
RXR/RAR nuclear receptors; Barrett et al., 2005; Ross et al., documenting the ability of rodent dams to communicate to
2005; Watanabe et al., 2004, 2007; Bechtold et al., 2012). their fetuses’ photoperiods prevailing during gestation. In
382 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

a laboratory setting, prevailing day lengths around the time of Squirrel monkeys housed indoors for 3 years in a 14-L photo-
weaning arrest or accelerate somatic and reproductive growth period sustained marked annual rhythms; full-term pregnan-
in several photoperiodic rodents (Johnston and Zucker, 1980; cies usually terminated in the months of August to October.
Horton, 1984; Stetson et al., 1989). When weanlings encounter Striking annual body mass rhythms and variations in testicular
intermediate day lengths (12–14 h light per day) at weaning size also persisted under constant conditions (Clewe, 1969).
(intended to simulate early spring or late summer), reproduc- These rhythms suggest a type II circannual organization.
tive development is markedly affected by day length during The rhesus macaque (Macaca mulatta) is perhaps the best
gestation. Comparison of pre- and postnatal day lengths exemplar of seasonality among Old World monkeys. At lati-
provides pups with directional information regarding changes tudes between 24 and 31 N, approximately 80% of rhesus
in photoperiod duration, and thereby either promotes or births occur between March and May and none between
retards growth and development. November and March (reviewed in Lindburg, 1987). Clear
The view that photoperiodic rodents are obliged to compare seasonal rhythms in plasma testosterone secretion persisted
prenatal with postnatal photoperiods in order to accurately in males housed in a 14-L photoperiod for up to 3 years (Plant
trigger or delay puberty has been modified by recent experi- et al., 1974; Michael and Bonsall, 1977). Individually housed
ments on Siberian hamsters maintained during gestation and males maintained in a 13-h day length for 14 months gener-
postnatally on simulated natural photoperiods. These incre- ated clear rhythms in serum concentrations of PRL, LH, and
mentally changing photoperiods exert a strong organizing testosterone but not FSH (Beck and Wuttke, 1979). Marked cir-
effect on seasonal rhythms by providing hamsters with a richer cannual variations in spermatogenesis, testicular volume, spon-
source of environmental timing cues than are available in taneous and provoked ejaculations, and sperm output, with
simple static day lengths. Butler et al. (2007b) found no maximum values in the autumn and winter months that
evidence that prenatal photoperiod history influenced post- encompass the mating season in the natural habitat, were
natal gonadal development of female hamsters in the presence documented in males (Wickings and Nieschlag, 1980). Female
of simulated natural photoperiods, and concluded that the first rhesus monkeys sustain seasonal anovulatory rhythms when
animals to delay puberty may do so predominantly on the housed in a fixed 13-h day length (Riesen et al., 1971).
basis of postnatal decreases in day length, whereas in later Changes in day length are sufficient to entrain annual
cohorts, a comparison of postnatal day length to gestational rhythms of rhesus monkeys. Males exposed to alternating
day length may contribute to arrested development. 16-week cycles of 16 and 8 L manifested reproductive rhythms
with a period of 31  1.3 weeks, without evidence of 52-week
periodicity. Day length variation is also posited as the proxi-
1.13.7 Seasonal Rhythms in Primates mate synchronizer of rhythms of testosterone, dihydrotestos-
terone, frequencies of mounts, ejaculations, number of
1.13.7.1 Nonhuman Primates
female partners, courtship displays, and aggression in Japanese
Discrete birth seasons have been conclusively established or are macaques (Macaca fuscata; Rostal et al., 1986).
probable for some two dozen species of nonhuman primates Ovulation was restricted to the months of August to March
(Lindburg, 1987). Other primates, though they have birth in a group of rhesus females housed outdoors in Atlanta, GA,
peaks, produce some young at all times of year. The volume USA; estradiol failed to induce gonadotropin surges during
edited by Brockman and van Schaik (2005) provides a compre- the summer months at a time when endogenous concentra-
hensive overview of primate seasonality. The editors note that tions of gonadotropins and ovarian steroids are low (Riesen
90% of extant primates live in tropical forests, with limited et al., 1971). Estradiol lowered basal LH concentrations to
seasonal variation in ambient temperature and day length a greater extent in ovariectomized females treated during the
but often striking seasonal variations in rainfall and sunshine summer anovulatory season than during the fall-winter season
(van Schaik and Brockman, 2005). Most primates have diverse during which ovulation ordinarily occurs. As in hamsters and
diets composed of over 50 species of plants and generally vary sheep, negative feedback sensitivity of gonadotropin secretion
food types ingested over the course of the year (Hemingway to steroid hormones increases during the nonbreeding season
and Bynum, 2005). Dietary diversity, reliance on specific and steroid-independent seasonal regulation of gonadotropin
food types, and readiness with which animals switch between secretion is documented for untreated ovariectomized females
food items may contribute to the evolution of seasonal who generate LH pulses with a lower frequency in summer
rhythms in reproduction (e.g., Bercovitch and Harding, 1993; than fall (Wilson et al., 1987).
Jablonski et al., 2000). Dietary diversity may be a necessary Behavioral responsiveness to estradiol varies seasonally in
but not sufficient condition for nonseasonal reproduction rhesus macaques. Ovariectomized females treated with estra-
(Bercovitch and Harding, 1993; van Schaik et al., 1993). diol engaged in heterosexual behavior during the breeding
Birth seasonality is lowest for foliovores and more season but not during the nonbreeding season (Pope et al.,
pronounced among omnivores or frugivore-insectivores of 1987). The duration of female proximity to males was influ-
equivalent body mass (Janson and Verdolin, 2005). There is enced by season but not by estradiol and the percentage of
little effect of latitude on birth seasonality between 0 and time spent with nonkin was increased by estradiol, regardless
15 . Among new world primates, squirrel monkeys (Saimiri of time-of-year. Females were not attracted to males during
sciureus) are highly seasonal, with births restricted to a 3- to the nonbreeding season, nor were males interested in females
5-month interval each year. In a captive colony exposed to during this time of year, despite the induction of high out-of
natural day length variations in California, 87% of births season testosterone secretion. Males as well as females may
were recorded between May and September (Kaplan, 1977). be refractory to behavioral actions of steroids during the
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 383

nonbreeding season, as previously established for Syrian boreal regions (Foley, 1993). Modern humans, dating from
hamsters (see Section 1.13.3). 40 000 to 50 000 years ago, colonized the entire Earth not by
There is little evidence for seasonality of reproduction habitat-specific adaptations but by relying on increased cogni-
among the apes (Lindburg, 1987; Watts, 1998). Savanna tive and behavioral capacity. The use of fire, improved weapons
baboons at the equator in Kenya experience two rainy seasons for hunting, language for communication, and deployment of
with two intervening dry seasons each year but display no animal skins for warmth, all contributed to this development
significant birth seasonality; nor is the number of births per (Foley, 1993; Potts, 1998) which presumably involved selec-
year significantly correlated with rainfall (Bercovitch and tion for expanded, more complex neural machinery.
Harding, 1993). No single pattern of seasonality is discernable The past 18 000 years have seen some of the largest climatic
among the many nonhuman primates. In Nepal, ovulatory changes in the Earth’s history (Webb et al., 1993) with large
cycles and estrous behavior were restricted to the period from temperature changes in mid- to high latitudes accompanied
July to October, during which most conceptions occurred by significant alterations in atmospheric circulation and mois-
(Ziegler et al., 2000). In contrast, Struhsaker (1997) found no ture balance. The heightened seasonality of northern hemi-
evidence of a restricted birth season in five monkey species in sphere solar radiation from 12 000 to 6000 years ago
a tropical rain forest in Kimbale. In some species, conception increased seasonality of northern hemisphere climates,
is associated with the presence of abundant food, and at the bringing warmer summers and cooler winters (Kutzback and
time of parturition both food availability and quality are at Webb, 1993). The development of agriculture some
their worst (Ziegler et al., 2000; capital breeders). In other 12 000 years ago and domestication of animals led to the even-
species the opposite relation obtains (Struhsaker, 1997). tual worldwide settlement of agrarian societies 8000–
3000 years before the present. During this time the climate of
the Earth was highly variable relative to preceding and succeed-
1.13.7.2 Humans
ing millennia (Sandweiss et al., 1999). Potts (1998) has
Given humans’ ability to buffer themselves from the environ- proposed that human traits were shaped by a succession of
ment, the number and diversity of human traits that vary habitat changes over the course of the late Pliocene and early
seasonally are impressive. Depression, stroke, suicide, sudden Pleistocene epochs, with strong selection for genes that permit
infant death, cardiac arrest, infections, homicides, hip fractures adaptation to multiple habitats (discussion in Alberts et al.,
among the newborn, multiple sclerosis, intracerebral hemor- 2005).
rhage, and autism, as well as nonpathological functions such
as breast feeding, hormone secretion, food consumption, and
1.13.7.4 Human Seasonal Reproductive Rhythms
energy expenditure are a subset of human traits that vary
seasonally (Stevenson et al., 2015). The underlying mecha- Environmental factors contribute to seasonal variations in
nisms are essentially unknown. The following discussion human births (Roenneberg and Aschoff, 1990a,b; Becker,
focuses on historical and evolutionary trends that contribute 1991; Bronson, 2004). The amplitude of these rhythms pales
to the blunting of human seasonality. in comparison to those of most other mammals; peak to trough
variations usually are in the range of 10–20%. Reproduction in
many populations occurs throughout the year and typically is
1.13.7.3 Evolution and Human Seasonality
not markedly seasonal, unless driven by food shortages or reli-
Early hominids originated in East Africa, with current evidence gious and cultural customs. With the advent of ‘big-data,’ the
localized to the Rift Valley (Maslin et al., 2014). The develop- assessment of seasonal rhythms in human reproduction can
ment of humanlike attributes coincided with a decline in easily be examined with impressive statistical power. Recent
global temperatures during the middle Pliocene and the emer- work assessing historical and modern month-of-birth estab-
gence of mosaic habitats far more seasonal than previously lishes a significant reduction in seasonal amplitudes from
thought (Foley, 1993). The ability to contend with drought pre-Baby Boom (1931–45) through Baby Boom Era
and associated nutritional stress is considered a key feature in (1946–64) to Modern Era (1965–2008) (Martinez-Bakker
hominid evolution (Foley, 1993; Jablonski, 2005; Maslin et al., 2013). A strong latitudinal gradient in seasonal human
et al., 2014). The savanna occupied by our early ancestors is birth was observed: countries located at higher latitudes
marked by striking seasonal variation in plant and animal (35–65 ) were found to exhibit peak births in spring–
resources that have experienced marked changes in climate summer months, while lower latitude countries (0–35 )
over the past 4 million years (Maslin et al., 2014). For extended have peaks in the fall–winter periods (Martinez-Bakker et al.,
periods, each year hunting and scavenging were likely insuffi- 2013). Modern assessments of large survey data sets of approx-
cient to provide adequate sustenance, in part because deteriora- imately 450 000 individuals indicated marked seasonality of
tion of grazing and browsing matter during drought renders the birth on weight, age at menarche, and height (Day et al.,
meat of game animals marginally nutritious (Stewart, 1994). 2015). For example, individuals born in summer experienced
Hominids from the late Pleistocene may have relied heavily later pubertal development. Given such effects of birth month
on fish when other foods were scarce or of low quality (Stewart, on measures such as reproduction and immunity (Stevenson
1994). They may also have built up fat reserves to ameliorate et al., 2015), there likely exists a fetal programming effect
seasonal food shortages (Speth, 1987). that varies by season. One consistently reported finding is the
Thermoregulatory stress from extreme winter conditions is seasonal variation in human semen parameters, such as
thought to have limited expansion of African hominids from motility and morphology (Proctor et al., 2004; Levitas et al.,
tropical and warmer mid-latitudes to more temperate and 2013). Sperm motility is significantly increased seasonally,
384 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

with greater ‘normal’ morphology during fall and winter 1.13.7.6 What Can Animal Studies Tell Us about Human
periods compared to samples analyzed in spring and summer Seasonality?
(Levitas et al., 2013; Proctor et al., 2004). However, in vitro
Indeed humans exhibit seasonal rhythms over multiple levels
fertilization–embryo transfer procedures do not have seasonal
of analyses, how these rhythms are generated and maintained
outcomes; indicating that in the ovary, fertilization and
at a mechanistic level remains poorly defined. Animal research
implantation processes may not be affected by seasonal
provides a conceptual framework for the exegesis and manipu-
patterns in sperm (Revelli et al., 2005).
lation of human seasonal rhythms. Lessons learned from
Identifying the factors (i.e., environmental/internal) that
research on rodents and monkeys raise the possibility that
mediate effects of birth season has been challenging. Evidence
hormone treatments will be effective in modifying behavior
suggests that shortages in metabolic fuels suppress reproduc-
at one but not another stage of the annual cycle. The demon-
tion in female mammals, with puberty as the most sensitive
stration that drugs and hormones phase shift annual rhythms
phase of the life cycle to such resources (Bronson, 1989; Bron-
at some but not other stages of the annual cycle can open the
son and Heideman, 1994). Shortages in energy impact the
way for rational drug treatments for human rhythm dysfunc-
GnRH pulse generator, which stops functioning when energy
tion. Over the past few years it has become clear that disrup-
supplies are restricted or expenditures are very great, as in
tions to daily cycles, via jet-lag or shift work, has a significant
anorectic women or athletes (Bronson, 1995, 2000). Studies
and negative impact on many traits, at the genetic, physiolog-
of subsistence foragers, agriculturists and pastoralists suggest
ical, behavioral, and cognitive levels. One particularly good
this mechanism impacts human reproduction (Bronson,
example is the impact of dim light at night on immunity
1995, 2000; Bentley, 1999). Among the Ache people, contem-
(Scheer et al., 2009; Fonken et al., 2010). Animal models
porary hunter-gatherers in eastern Paraguay, the low food
have been extremely valuable for directing research efforts to
period is associated with lowest conception rates (Hill and
understand the effects of daily disruptions on humans.
Hurtado, 1996). In a far ranging review, Bronson (2004)
However, we have a rudimental understanding of the impact
contends there is no evidence that photoperiod plays an adap-
of seasonal disruption, such as climate change, on wild and
tive role in modern humans, a view shared by Ellison et al.
laboratory animal species (Stevenson et al., 2015). There are
(2005). The evidence supporting the hypothesis that human
simply no data available in humans. Rectifying this lack of
reproduction is sensitive to seasonal variation in day length is
knowledge is particularly important given the hypotheses of
inconsistent (but see Roenneberg, 2004). The suggestion that
climate change driven evolution of hominidae (Trauth et al.,
individual humans are to different degrees seasonally photores-
2005). Advances in ‘omics’ research and the availability of
ponsive (Bronson, 2004) has parallels in an extensive rodent
‘big-data’ resources will undoubtedly yield significant insights
literature.
into the effects of disruption in daily and seasonal rhythms.
Limits to the usefulness of animal models (Zucker, 1988) are
1.13.7.5 Melatonin and Human Reproduction apparent, however, when we consider that the many social, reli-
gious and cultural customs that influence human seasonality
Humans, in common with all other mammals, secrete mela-
have no parallels in the animal kingdom.
tonin nearly exclusively at night and exposure to light at night
interrupts melatonin secretion (Wehr, 1997). In urban environ-
ments with artificial lighting, people differ in the extent to
which duration of nocturnal melatonin secretion corresponds 1.13.8 Seasonal Rhythms in Nonreproductive Traits
to seasonal changes in day length (Wehr, 1997; Vondrasova
et al., 1997). Vondrasova and colleagues suggest that during Several nonreproductive traits that fluctuate seasonally are
the middle ages, when people worked the fields from dawn mediated by reproductive steroid hormones, whereas others
to dusk, summer versus winter melatonin durations may are modulated by melatonin or other seasonal events.
have differed by as many as 4 h at 50 N. If so, then uncoupling Many seasonal changes conserve energy during the fall and
of melatonin secretion from gonadotropin regulatory mecha- winter. Some species accomplish this goal by migrating to
nisms must have been in place for many centuries in Homo better conditions; others cope with winter by undergoing
sapiens. Otherwise seasonal rhythms in human reproduction torpor or hibernation. Individuals that brave winter use
would be far more pronounced in preindustrial societies than changing photoperiod signals and decreases in steroid
indicated by the historical record (Lummaa et al., 1998). hormone secretion to inhibit food intake, increase or
Furthermore, molecular evidence indicates that the disconnect decrease body mass, increase pelage density (Park et al.,
of pituitary responsiveness from melatonin signaling preceded 2007), or transition from nocturnal to diurnal locomotor
the hominidae lineage. Female baboon pituitary cells cultured activity to conserve energy. These topics were reviewed exten-
with melatonin did not impact long-day, FSH or TSH expres- sively in the previous version of this chapter (Prendergast
sion nor release (Ibanez-Costa et al., 2015). Melatonin only et al., 2002b) and are not addressed here. We focus on
stimulated increased growth hormone and PRL expression in immune function, social interactions, and brain size as key
a dose-dependent manner. These data indicate that mela- nonreproductive adaptations.
tonin-driven seasonal physiology in primate physiology is
localized to the pars distalis and regulates GH and PRL, but
1.13.8.1 Immune Function
not components of steroidogenesis or gametogenesis. There is
presently no convincing evidence for a functional relation of Reduced food availability, in common with many other ener-
human reproduction and melatonin (Reiter, 1998). getic challenges, is a potent stressor. The number and types of
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 385

stressors vary seasonally (Breuner et al., 1999) and are corre- In response to a simulated bacterial infection, Siberian
lated with seasonal changes in immune function (Nelson, hamsters maintained in long day lengths that mimic summer
2004). The role of temporal fluctuations in response to stress conditions have significantly higher levels of interleukin
has been well reviewed elsewhere (e.g., Chrousos, 1998; (IL)-6 in circulation and produce higher fevers compared to
Dallman et al., 1993). animals maintained in short days that mimic winter condi-
Seasonal cycles of illness and death, as well as birth, are tions (Bilbo et al., 2002a, Figure 4(a)). Both IL-6 production
common among many mammals (e.g., Bolinger et al., 1996; and febrile responses are mediated in part by macrophages
Bradley et al., 1980; Lochmiller et al., 1994). Mammals may (Medzhitov, 2007). In addition, long-day, more than short-
have evolved mechanisms to combat seasonal stress-induced day, hamsters increase macrophage-mediated phagocytosis
reductions in immune function (Nelson and Drazen, 1999; activity and increase oxidative burst activity (Yellon et al.,
Yellon et al., 1999; Sinclair and Lochmiller, 2000; Nelson, 1999). These photoperiod differences may be mediated by
2004). It is plausible that fitness is increased in individuals activation of Toll-like receptors, which during infection stim-
that bolster immune function in advance of seasonally recur- ulate macrophages to produce the proinflammatory cytokine,
ring immunosuppression. tumor necrosis factor a (TNFa) (Aderem and Ulevitch, 2000)
Numerous studies have now documented marked changes among other acute-phase cytokines. Elicited peritoneal
in immune function in response to photoperiod change in macrophages cultured from long-day hamsters produced
the laboratory. Here we review broad concepts related to the significantly more TNFa after a challenge with a bacterial
adaptive significance of seasonal immunomodulation and component known to stimulate specific Toll-like receptors,
highlight classic studies along with select, recent developments. lipopolysaccharide (LPS) compared to macrophages cultured
The previous version of this chapter reviewed this material from short-day hamsters (Prendergast et al., 2003; Navara
extensively, as have several recent review papers (Prendergast et al., 2007, Figure 4(a)). However, TLR2 and TLR4 expres-
et al., 2010; Stevenson and Prendergast, 2015; Weil et al., sion was similar between photoperiods and unrelated to
2015). macrophage responsiveness (Navara et al., 2007); other
Consistent with laboratory analyses, many field studies downstream intracellular mechanisms may be responsible
reveal enhanced immune function and decreased disease for photoperiod-evoked variation in macrophage responsive-
prevalence during winter as compared to summer. Several ness. Photoperiodic changes in immune function in, e.g.,
reports, however, document the opposite pattern of results Siberian hamsters occur in parallel with photoperiodic
(Nelson and Demas, 1996; Nelson et al., 1995, 1998); i.e., changes in reproductive physiology. Several lines of evidence
immune function is compromised during the short days of suggest that the suite of changes evident in the hamster
winter. Additional environmental factors, not usually manip- immune system during adaptation to short days is both
ulated in laboratory studies, may account for the discrepancy. gonadal hormone (i.e., sex steroid)-dependent and gonadal
For example, winter-associated stressors such as restricted hormone-independent, to varying degrees, according to the
food availability and low ambient temperatures counteract trait (reviewed in Stevenson and Prendergast, 2015).
short-day enhancement of immune function in the laboratory Gonadal hormone-independent effects of photoperiod on
(reviewed in Demas and Nelson, 1996). Thus, immune func- immune function in hamsters may be mediated by changes
tion may be enhanced and compromised, respectively, during in thyroid hormone availability to immune tissues. Exoge-
mild and severe winters. Long-term field studies are required nous T3 enhanced several aspects of hamster immune func-
to test this hypothesis (see, e.g., Sinclair and Lochmiller, tion, and photoperiod-driven changes in deiodinase
2000). Although the effects of melatonin on immunity are expression in immune cells may mediate these effects
well-established (see Nelson, 2004 for review), an under- in vivo (Stevenson et al., 2014). However, other hormones,
standing of how the ecological context affects immune func- including glucocorticoids (Bilbo et al., 2002a), PRL (Weil
tion is necessary to appreciate that these actions of et al., 2015), and leptin (Carlton and Demas, 2014) appear
melatonin may be adaptive, rather than merely physiological to mediate other aspects of photoperiod-induced changes
oddities. in immunity in photoperiodic rodents.
The cascade of cellular events during the acute phase
immune response and inflammation, and the elevation of
1.13.8.2 Energetics and Immune Function
body temperature in response to cytokine activation, presum-
ably requires substantial energy, although precise quantifica- Maintaining optimal immune function is energetically expen-
tion is lacking (Henken and Brandsma, 1982; Maier et al., sive; the cascades of dividing immune cells, the onset and
1994). Cytokine activation elevates body temperature, and maintenance of inflammation and fever, and the production
the energy requirements of inflammation and acute phase of humoral immune factors require substantial energy
immune responses may increase metabolic rate by more than (Demas et al., 1997; Spurlock, 1997). Mounting an immune
10% for each degree of increase in body temperature (reviewed response likely requires resources that could otherwise be
in Grimble, 1994). In house mice (Mus musculus) injection of allocated to other functions (Sheldon and Verhulst, 1996;
keyhole limpet hemocyanin, a specific antigen that evokes an Demas, 2004). Thus, it is reasonable to consider immune
antibody response without inducing fever or symptoms of function from the perspective of energetic trade-offs (Martin
illness resulted in increased oxygen consumption and meta- et al., 2008). Individuals may ‘optimize’ immune
bolic heat production. Animals with energy deficits that impair function and allocate energy resources to immune function,
the ability to sustain immune function display elevated risk of other maintenance functions, or reproduction (Demas,
infection and mortality. 2004; Martin et al., 2008). Seasonal changes in immune
386 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

(a)

20 80
10 *
70

TNFα (pg ml–1)


16 * 60

IL-1β (pg ml–1)

IL-6 (pg ml–1)


*
50
12
1 40
8 30
20
4
10
0.1 0 0
LD SD LD SD LD SD

(b)
0–24 h 24–48 h 48–72 h 0–24 h 24–48 h 48–72 h

0 0

Decrease in food intake (%)


Decrease in milk intake (%)

* *
−20
*
−20
*
−40 −40

−60 −60

−80 −80

Figure 4 Exposure to short photoperiods for 10–12 weeks attenuates lipopolysaccharide (LPS)-induced cytokine production of Siberian hamsters
(top panel; TNFa production from peritoneal macrophages; interleukin (IL)-1b and IL-6 production from lymphocytes), and LPS-induced sickness
behaviors (bottom panel). Milk and food intake are commonly used dependent measures for sickness symptoms. Sweetened-condensed milk is
highly palatable and reflects both a hedonic and an energetic measure. The impact of LPS on milk intake is attenuated, indicating that these sickness
symptoms (anorexia and/or anhedonia) are resolved faster in short days. Data from Bilbo, S.D., Drazen, D.L., Quan, N., He, L., Nelson, R.J., 2002b.
Short day lengths attenuate the symptoms of infection in Siberian hamsters. Proc. R. Soc. B 269, 447–454.

function are likely driven by seasonal changes in energy (Beutler and Cerami, 1988; Waage et al., 1989; Kelley and
requirements and availability (Nelson and Demas, 1996). Dantzer, 1990; Kwiatkowski et al., 1990). Consequently,
According to this hypothesis, in an environment where energy a balanced release of cytokines must be achieved for maximal
needs and demands fluctuate, individuals maintain the high- effectiveness in protecting the host from infection.
est level of immune function that is energetically possible Day length and ambient temperature interact to affect
(without evoking autoimmune diseases) subject to immune function. In deer mice maintained at 20  C, serum
constraints of other survival needs. A complete understanding immunoglobulin G (IgG) concentrations were elevated in
of seasonal cycles in human immune function requires short-day mice as compared to long-day mice (Demas and
consideration of how energetic and other stressors impinge Nelson, 1996). At 8  C, long-day deer mice had reduced IgG
on immunocompetence. Animal models illustrate these concentrations and short-day animals had IgG concentrations
relations. comparable to those of long-day mice at 20  C; i.e., IgG
Among domesticated mammals, reduced food intake is concentrations were higher in short than long days, and low
associated with compromised immune function (reviewed in temperatures significantly reduced IgG concentrations. Short
Spurlock, 1997). For example, proinflammatory cytokines, day lengths and low temperatures thus appear to produce
particularly IL-1, IL-6, and TNFa, interrupt anabolic processes opposite effects on IgG concentrations, in essence restoring
and alter nutrient uptake and utilization (Grimble, 1994; values to those of animals in long days, which may help
Johnson, 1997). These cytokines also cause the release of animals cope with seasonal stress of winter and ultimately
glucocorticoids, catecholamines, and prostaglandins, which increase reproductive fitness (Demas and Nelson, 1996).
in turn modulate metabolism and immune function. Although 2-deoxy-D-glucose (2-DG) induces glucoprivation and is
proinflammatory cytokines are critical in the responses to a metabolic stressor that increases serum corticosterone
infection, an excess of cytokines can damage tissue and concentrations in male rats (Weidenfeld et al., 1984) and
compromise survival. Excess or inappropriate secretion of cyto- inhibits murine splenic T lymphocyte proliferation in a dose-
kines has been linked to morbidity in malaria, sepsis, menin- dependent fashion in laboratory mice (Miller et al., 1994). In
gitis, inflammatory bowel disease, and rheumatoid arthritis long days, corticosterone concentrations were elevated in
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 387

mice injected with 2-DG versus saline; an effect that was absent changes in photoperiod may be antagonized by such artificial
in short days (Demas et al., 1997a), again underscoring impor- stimuli (see Weil et al., 2015 for discussion).
tance of understanding interactions between stressors and
photoperiod in the effect on immune responses. 2-DG was
1.13.8.3 Social Organization: Affiliation and Aggression
associated with reduced mitogen-stimulated splenocyte prolif-
eration in long-day mice, but short-day mice, regardless of Many rodent species shift from highly territorial asocial
experimental treatment, exhibited enhanced immune function behavior during the breeding season to a social, highly interac-
in response to 2-DG treatment (Demas et al., 1997a). 2-DG tive existence during winter. These species typically undergo
treatment of long-day hamsters decreased antibody response reproductive quiescence at the end of the breeding season in
compared with control animals but did not affect antibody response to short days; the resulting decrease in androgen secre-
responses in short-day hamsters (Zysling and Demas, 2007). tion may be necessary for and permissive of the seasonal shift
These data suggest that short day lengths buffer animals in sociality, although in wood rats (Neotoma fuscipes) nonste-
against metabolic stress. Reduced corticosterone concentra- roidal mechanisms mediate the seasonal transition in social
tions in animals maintained on short days or treated with behavior (Caldwell et al., 1984). During the breeding season,
melatonin likely reflect improved metabolic function (Nelson rodents control resources that promote their own survival
and Demas, 1996; Sinclair and Lochmiller, 2000). Accordingly, and that of their offspring and often aggressively exclude non-
enhancements in several aspects of immune function in short kin from access to resources. During the winter, however, this
days are likely one component of the winter adaptation medi- strategy is abandoned in favor of group living which conserves
ated by melatonin. The expectation is that natural perturba- energy and enhances survival in the face of low temperatures
tions of energy associated with pregnancy, lactation, torpor, and reduced food availability. Many species of rodents
and hibernation will affect immune function in predictable conserve energy during the winter by forming aggregations of
directions (Lochmiller and Deerenberg, 2000; Nelson et al., huddling animals (West and Dublin, 1984). In these aggrega-
2002). tions, different sexes and even different species commingle
Exposure to pathogens or to inflammatory stimuli such as (Madison et al., 1984).
LPS activates the immune system and initiates physiological Even in the absence of huddling behavior, animals may
and behavioral responses collectively termed the acute phase tolerate one another better in close quarters during the winter
response. These so-called sickness responses, including fever, than during the breeding season. For example, male meadow
may be constrained by energy availability; if so, then cytokine voles are highly territorial in the spring and summer and
production, fever, and anorexia should be attenuated in occupy open meadows whereas red-backed voles (Clethriono-
infected Siberian hamsters housed under short days (simulated mys gapperii) breed in forest habitats. During the winter
winter). Indeed, short days attenuated the response to LPS by months, meadow voles move into the spruce forest habitats
diminishing the duration of fever and anorexia (Bilbo et al., occupied by the red-backed voles, presumably to take advan-
2002b). Remarkably, decreased ingestion of dietary iron, tage of the protective cover provided by trees. In some cases
a nutrient vital to bacterial replication, was reduced in short- they share nests with other rodent species (Madison et al.,
day hamsters. These changes in sickness responses corre- 1984). Individual meadow voles trapped during the winter
sponded with decreasing production of IL-6 and IL-1b (Bilbo tested in paired encounters in a small neutral arena exhibit
et al., 2002b; Figure 4(a)). Taken together, short days attenu- less interspecific aggression than voles trapped in summer
ated the symptoms of infection, presumably to optimize energy (Turner et al., 1975). The winter reduction in aggressiveness
expenditure and facilitate survival (Figure 4(b)). Similar results permits energy-saving habitat sharing. As the animals enter
were attained with melatonin treatment (Bilbo and Nelson, breeding condition in the spring, they reestablish mutually
2002), suggesting a therapeutic use of melatonin or its receptor exclusive territories.
agonists. Some males do not undergo reproductive regression and
Although much of the research on photoperiodic effects on maintain testicular function and produce sperm and androgens
immune responses has been conducted in adult animals, early- during simulated winter conditions (Nelson, 1987; Prendergast
life photoperiods are also important sources of seasonal infor- et al., 2001). The advantages of continuous breeding evidently
mation that establish a developmental trajectory and have incur substantial hidden costs, because only a minority of each
profound consequences for the immune system. Early-life population adopts this strategy. Reproductively competent
photoperiods appear to program adult immune responses, as males, because of unusual aggressiveness during the winter,
Siberian hamsters enhance immune responses as adults if are unable to participate in communal huddling and thus
they were housed in short days both pre- and postnatally may incur greater energetic costs in overwintering. High behav-
(simulating early winter births) relative to hamsters that were ioral and energetic costs associated with maintenance of the
in long days during any developmental period (Weil et al., reproductive system in winter may explain why nonregressive
2006). Perinatal and adult photoperiods interact to determine types do not normally predominate in temperate or boreal
the adult immunological phenotype in developing animals zone populations of rodents (Nelson, 1987; Nelson et al.,
(Prendergast et al., 2004a,b). 1989). This contention is supported by a field study of winter
Lastly, recent evidence is emerging indicating that exposure nesting behavior of prairie voles (McShea, 1990). Most voles
to dim light at night, a phenomenon that is becoming more in the population studied were reproductively inactive during
prevalent in modern times, can impair multiple aspects of the winter and formed groups of huddling individuals. Two
immune function (Bedrosian et al., 2011). The aforementioned males, however, remained in breeding condition and were
enhancements in select aspects of immunity conferred by never observed to huddle with other animals. In pair-wise tests
388 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

of aggression, these two males were much more aggressive than coordinates a ‘seasonal switch’ from gonadal to adrenal regula-
reproductively quiescent individuals. In another study, repro- tion of aggression by direct action on the adrenal glands.
ductive status also influenced odor preferences of meadow In common with hamsters, short day lengths increase
voles maintained in simulated winter day lengths (Gorman aggression in beach mice (Peromyscus polionotus) (Trainor
et al., 1993). Males that retained reproductive capability in et al., 2007). This effect is apparently mediated by estrogens.
winter day lengths preferred the odors of females that also Remarkably, estrogens have the opposite effects depending
failed to inhibit reproduction during short days. This prefer- on day length (Figure 6). The ERa agonist PPT
ence may facilitate the sporadic occurrences of winter breeding (propylpyrazole-triol) and the ERb agonist DPN (diarylpropio-
frequently reported for this species (reviewed in Nelson, 1987). nitrile) increased aggression in ‘short-day’ P. polionotus and
The decrease in circulating androgens associated with cessa- decreased aggression in ‘long-day’ mice (Trainor et al., 2007).
tion of reproduction may mediate the increase in male affilia- These results indicate that photoperiod controls processes
tive interactions during winter; males that maintain ‘summer’ that occur after estrogens bind to their appropriate receptors.
reproductive status may thereby also maintain ‘summer’ Steroid hormones, including estrogens, can affect physiological
patterns of aggression. This hypothesis is supported by the and behavioral processes via slow genomic (hours to days) or
observation that castration results in decreased aggressiveness. rapid nongenomic (seconds to minutes) pathways (Vasudevan
Although the relation between castration and reduced male and Pfaff, 2007). Estradiol injections of beach mice evoked
aggression is well established, anecdotal observations of male rapid (<15 min) increases in aggression in short-day, but not
prairie voles suggest that low circulating testosterone concen- long-day mice (Trainor et al., 2007). These data suggest that
trations are not sufficient to reduce aggressive behavior. Castra- estradiol increases aggression via nongenomic actions in
tion did not reduce the frequency of male aggression in tests short-day, but not long day mice. Estrogen-dependent expres-
that involved resident–intruder, grouped aggression, or aggres- sion of genes with estrogen response elements in their
sion against a lactating female (Demas et al., 1999). Similarly, promoters was decreased in the bed nucleus of the stria termi-
postpubertal castration of male dusky-footed wood rats also nalis (BNST) of short-day mice compared with that of long-day
did not eliminate the marked vernal increase in aggression in mice. Environmental factors may regulate the effects of steroid
laboratory encounters with castrated or gonadally intact oppo- hormones on aggression in P. polionotus by determining the
nents (Caldwell et al., 1984). Aggressive behavior may be inde- molecular pathways activated by steroid receptors (Nelson
pendent of gonadal steroid hormones in some adult male and Trainor, 2007).
rodents; further studies will be needed to establish the gener- Male song sparrows (Melospiza melodia) display estrogen-
ality of the notion that aggression is maintained by testicular dependent territorial aggression throughout virtually the entire
androgens. year (Wingfield and Soma, 2002). Although territorial aggres-
Individuals of many, if not most, species reduce aggression sion in response to a territorial intrusion appears similar
outside of the breeding season. For some species, however, throughout the year, the neuroendocrine responses after terri-
aggression must be maintained throughout the year indepen- torial aggression vary seasonally. After a territorial intrusion
dent of breeding season. In such cases, aggression would likely during the breeding season, territorial residents continue
be modulated by mechanisms unrelated to reproduction. In patrolling their territories and display spontaneous song for
hamsters, short days increase male resident–intruder aggression hours, whereas during the nonbreeding season, resident birds
(P. sungorus, Demas et al., 2004; M. auratus, Garrett and stop behaving aggressively within minutes (Wingfield, 1994).
Campbell, 1980). This effect is paradoxical because increased Thus, breeding territorial aggression is persistent, whereas
aggression occurs when testosterone concentrations are often nonbreeding territorial aggression is transient. It appears that
undetectable. Despite the lack of plasma androgens, estrogens estrogen varies in function by season as in Peromyscus described
may still be important (see below). Adrenalectomy prevents above. To test this hypothesis, captive song sparrows in either
increased aggression in short-day Siberian hamsters (Demas breeding or nonbreeding conditions were provided with wax
et al., 2004). moth larvae dosed with either estradiol or the vehicle. Birds
Adrenal steroid hormones appear to be involved in the provided with estradiol-laced larvae increased aggressive
short-day aggressive phenotype in several species of rodents behaviors within 20 min of treatment indicating that estrogens
and birds (Soma et al., 2015). Both male and female Siberian increase aggression during the nonbreeding season via a rapid
hamsters housed in short day lengths undergo gonadal regres- nongenomic mechanism of action (Heimovics et al., 2015).
sion and reductions in circulating gonadal steroids, but Birds in breeding condition did not show this rapid onset of
substantial increases in aggression, compared with animals in aggression in response to estradiol. In song sparrows and other
long days (Jasnow et al., 2000; Scotti et al., 2007). Replacement small animals, transient expression of aggressive behavior
of gonadal steroid hormones such as 17b-estradiol in short-day during the nonbreeding season is presumably highly adaptive
females (to mimic long-day concentrations) does not reduce as it minimizes energy expenditure and maximizes the amount
aggression to summer levels (Scotti et al., 2007). Thus, another of time available for foraging (Heimovics et al., 2015).
neuroendocrine mediator was sought. Recently, it was reported
that melatonin bolsters DHEA secretion from adrenal glands in
1.13.8.4 Brain Development
culture (Rendon et al., 2015; Figure 5). Furthermore, long-day
hamsters provided with short-day melatonin doses displayed Seasonal changes in brain weight have been documented in
increased aggression and elevated DHEA concentrations rodents and shrews (e.g., Clethrionomys glareolus, Clethrionomys
(Rendon et al., 2015). These results suggest that DHEA is rutilus, Microtus oeconomus, Microtus gregalis, Sorex araneus, Sorex
a key peripheral regulator of aggression and that melatonin minutus; Bielak and Pucek, 1960, 1965; Yaskin, 1984). Brain
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 389

(a) 30 (b) 2.0

25 ∗ ∗

Serum DHEA (ng ml–1)


1.5
20

No. of attacks
15 1.0

10
0.5
5

0 0
Control Melatonin Control Melatonin

(c) 1.0 Control


Long days
Melatonin-induced in vitro DHEA

b Short days
0.8 b

0.6
(ng ml–1)

d
0.4

a a
0.2
c

0
Adrenal Ovarian

Figure 5 Melatonin-induced changes in aggression and DHEA. In vivo-induced (a) number of attacks, (b) serum DHEA concentrations in melatonin
and control-treated female Siberian hamsters. (c) In vitro DHEA response to melatonin or control treatment. Data from Rendon, N.M., Rudolph, L.M.,
Sengelaub, D.R., Demas, G.E., 2015. The agonistic adrenal: melatonin elicits female aggression via regulation of adrenal androgens. Proc. R. Soc. B
282, 20152080.

weights tend to be higher in summer than winter (Yaskin,


1984). Seasonal variations in brain weight may decrease energy
Saline Estradiol expenditure (Jacobs, 1996). Although the brain constitutes
20
only 2–3% of the total body mass in rodents, it consumes
*
Offensive attacks (freq)

a somewhat higher percentage of total energy expenditure


(Mink et al., 1981). It has been suggested that minor reductions
in brain mass could result in substantial energetic savings
(Jacobs, 1996). A significant part of the seasonal change in
10
brain weight may be attributable to differences in water
content; however, the neocortex and the basal portion of the
brains (i.e., the corpus striatum) of rodents and shrews show
seasonal cytoarchitectural changes. The relative weight of the
forebrain declines during the winter, the relative weight of
0
Long day Short day the hippocampus increases from winter to summer, and the
relative weight of the olfactory bulbs, myelencephalon, and
Figure 6 Photoperiod alters nongenomic actions of estradiol on cerebellum increases during the winter (Yaskin, 1984). A sex
aggressive behavior. Beach mice (Peromyscus polionotus) were housed difference in brain weight is observed among bank voles
in long or short photoperiods for 8 weeks. One week prior to behavioral (C. glareolus) only during the winter months when male brains
testing, mice were injected daily with the aromatase inhibitor fadrozole,
are heavier than those of females. The absolute and relative
mice were then injected with saline or cyclodextrin-conjugated estradiol
weight of the hippocampus is significantly higher in males
(cE2), a water-soluble, rapid-acting estrogen, and tested in resident–
intruder tests 15 min later. Data from Trainor, B.C., Lin, S., Finy, M.S., than in females throughout the year, but the difference is
Rowland, M.R., Nelson, R.J., 2007. Photoperiod reverses the effects of most pronounced during the winter (Yaskin, 1984). Meadow
estrogens on male aggression via genomic and nongenomic pathways. voles also show seasonal changes in brain weight that are
Proc. Natl. Acad. Sci. U.S.A. 104, 9840–9845. Copyright (2007) controlled by photoperiod (Dark et al., 1987); males kept
National Academy of Sciences, U.S.A. under short-day conditions have lighter brains than long-day
390 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

animals. Meadow voles born into long day lengths have greater the olfactory bulb (Walton et al., 2012); a potential functional
brain growth (i.e., more cells) than cohorts born in short days consequence of this difference was demonstrated in photoperi-
(Dark et al., 1990). Furthermore, long days enhance myelina- odic changes in habituation to odor cues. Effects of photope-
tion of both midbrain and hindbrain regions of developing riod on adult mammalian neurogenesis have been reported
male meadow voles (Spears et al., 1990). in other species as well (e.g., sheep) and have been extensively
Photoperiodic effects on cell division in the dentate gyrus reviewed by Migaud et al. (2015).
and subependymal zone of adult mammals were noted by Sex differences in hippocampal size were evident in male
Huang et al. (1998). Long-term exposure to short days in Syrian polygamous meadow voles (Microtus pennsylvanicus), but not
hamsters doubled the number of new neurons produced in in monogamous pine voles (Microtus pinetorum; Jacobs et al.,
these brain regions, as well as in the hypothalamus and cingu- 1990). Similar sex differences, but not seasonal changes, were
late/retrosplenial cortex. There were no appreciable photoperi- also reported for eastern gray squirrels (Sciurus carolinensis;
odic differences in the brain volume of either the granule cell Lavenex et al., 2000). In both cases, it was suggested that the
layer of the hippocampus or the dentate gyrus. And in white- larger hippocampal complex in males was related to their
footed mice, exposure to short days increased progenitor cell increased reliance on spatial memory in foraging and mainte-
survival in the posterior plexiform and granulae cell layers of nance of territories (Jacobs, 1996). These results suggest that

Figure 7 Photoperiod alters spatial learning and hippocampal activity. (a) Male white-footed mice (Peromyscus leucopus) were housed in long or
short photoperiods for 13 weeks and trained on a Morris water maze. Latency to reach the hidden platform over successive blocks of training. (b)
Impairment of long-term potentiation of fEPSPs in the Schaffer collateral–CA1 pathway of white-footed mouse hippocampal slices after 10 weeks of
exposure to short days. Upper left and middle: representative traces of fEPSPs (1) pretetanus and (2) 50 min posttetanus. Calibration bar: 0.2 mV;
10 ms. Upper right: location of stimulating (black arrow) and recording (white arrow) electrodes. Lower panel: comparison of LTP between long-day
(open box) and short-day (black box) mice. Tetanus stimulation was delivered at the time indicated by two arrows. Data from Pyter, L.M., Reader,
B.F., Nelson, R.J., 2005. Short photoperiods impair spatial learning and alter hippocampal dendritic morphology in adult male white-footed mice
(Peromyscus leucopus). J. Neurosci. 25, 4521–4526; Walton, J.C., Chen, Z., Weil, Z.M., Pyter, L.M., Travers, J.B., Nelson, R.J., 2011. Photoperiod-
mediated impairment of long-term potention and learning and memory in male white-footed mice. Neuroscience 175, 127–132.
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 391

developmental effects of steroids might mediate changes in Anand, S., Turek, F.W., Horton, T.H., 2004. Chemosensory stimulation of luteinizing
hippocampal size. There was, however, no overall sex differ- hormone secretion in male Siberian hamsters (Phodopus sungorus). Biol. Reprod.
70, 1033–1040.
ence in hippocampal volume or dentate gyrus width in wild-
Andersen, D.M., Keafer, B.A., 1987. An endogenous annual clock in the toxic marine
captured male and female meadow voles (Galea et al., 1999). dinoflagellate Gonyaulax tamarensis. Nature 325, 616–617.
A sex difference in hippocampal volume emerged if only the Arendt, J., 1995. Melatonin and the Mammalian Pineal Gland. Chapman and Hall,
males with relatively high circulating testosterone concentra- London.
tions (top 50th percentile) were compared to females with rela- Badura, L.L., Goldman, B.D., 1992. Central sites mediating reproductive responses to
melatonin in juvenile male Siberian hamsters. Brain Res. 598, 98–106.
tively low circulating estradiol concentrations (bottom 50th Badura, L.L., Nunez, A.A., 1989. Photoperiodic modulation of sexual and aggressive
percentile; Galea et al., 1999). behavior in female golden hamsters (Mesocricetus auratus): role of the pineal
Despite the evidence for seasonal changes in brain weight in gland. Horm. Behav. 23, 27–42.
rodents, there has been relatively little research investigating Badura, L.L., Yant, W.R., Nunez, A.A., 1987. Photoperiodic modulation of steroid-
induced lordosis in golden hamsters. Physiol. Behav. 40, 551–554.
seasonal changes in learning among mammalian species (but
Bae, H.H., Mangels, R.A., Cho, B.S., Dark, J., Yellon, S.M., Zucker, I., 1999.
see Jacobs, 1996). Voles trapped in winter make more errors Ventromedial hypothalamic mediation of photoperiodic gonadal responses in male
and require longer to learn mazes than do summer-captured Syrian hamsters. J. Biol. Rhythms 14, 391–401.
voles (Jacobs, 1996). Barrett, P., Ross, A.W., Balik, A., Littlewood, P.A., Mercer, J.G., Moar, K.M.,
Similarly, short-day white-footed mice (Peromyscus leucopus) Sallmen, T., Kaslin, J., Panula, P., Schuhler, S., Ebling, F.J., Ubeaud, C.,
Morgan, P.J., 2005. Photoperiodic regulation of histamine H3 receptor and VGF
require longer to learn the Morris spatial water maze (Pyter messenger ribonucleic acid in the arcuate nucleus of the Siberian hamster.
et al., 2005; Figure 7(a)). Short days also decreased brain Endocrinology 146, 1930–1939.
mass and hippocampal volume compared to mice housed in Barrett, P., Ebling, F.J., Schuhler, S., Wilson, D., Ross, A.W., Warner, A.,
long days (Pyter et al., 2005). Despite impairments in spatial Jethwa, P., Boelen, A., Visser, T.J., Ozanne, D.M., Archer, Z.A., Mercer, J.G.,
Morgan, P.J., 2007. Hypothalamic thyroid hormone catabolism acts as a gate-
learning and memory, there were no photoperiodic effects on
keeper for the seasonal control of body weight and reproduction. Endocrinology
sensory discrimination or other types of memory. Short days 148, 3608–3617.
decreased apical (stratum lacunosum-moleculare) CA1 spine Bartness, T.J., Goldman, B.D., Bittman, E.L., 1991. SCN lesions block responses to
density, as well as increased basilar (stratum oriens) CA3 spine systemic melatonin infusions in Siberian hamsters. Am. J. Physiol. 260,
density (Pyter et al., 2005). Short days also decreased LTP in R102–R112.
Bartness, T.J., Powers, J.B., Hastings, M.H., Bittman, E.L., Goldman, B.D., 1993. The
the Schaffer collateral–CA1 pathway and impaired spatial timed infusion paradigm for melatonin delivery: what has it taught us about the
learning and memory in the Barnes maze (Walton et al., melatonin signal, its reception, and the photoperiodic control of seasonal
2011; Figure 7(b)). responses? J. Pineal Res. 15, 161–190.
The motoneurons of the spinal nucleus of the bulbocaver- Bechtold, D.A., Sidibe, A., Saer, B.R.C., Li, J., Hand, L.E., Ivanova, E.A., Darras, V.M.,
Dam, J., Jockers, R., Luckman, S.M., Loudon, A.S.I., 2012. A role for the
nosus and their target muscles, the bulbocavernosus and
melatonin-related receptor GPR50 in leptin signaling, adaptive thermogenesis and
levator ani are sexually dimorphic and control penile erection torpor. Curr. Biol. 22, 70–77.
in rodents (Breedlove, 1992). Short days reduce the size and Beck, W., Wuttke, W., 1979. Annual rhythms of luteinizing hormone, follicle-
neuronal numbers in these regions in white-footed mice and stimulating hormone, prolactin and testosterone in the serum of male rhesus
Siberian hamsters (Forger and Breedlove, 1987; Hegstrom monkeys. J. Endocrinol. 82, 131–139.
Becker, S., 1991. Seasonal patterns of births and conception throughout the world.
and Breedlove, 1999a). These neuronal changes may help Adv. Exp. Med. Biol. 286, 59–72.
conserve energy during winter. Becker-Andre, M., Wiesenberg, I., Schaeren-Wiemers, N., Andre, E., Missbach, M.,
The interaction among sex and seasonal differences is Saurat, J.H., Carlberg, C., 1994. Pineal gland hormone melatonin binds and
complex but may be important in typical development. For activates an orphan of the nuclear receptor superfamily. J. Biol. Chem. 269,
28531–28534.
example, androgenic induction of sexual dimorphism in
Bedrosian, T.A., Fonken, L.K., Walton, J.C., Nelson, R.J., 2011. Chronic exposure to
meadow voles proceeds under long, but not short, days (Kelly, dim light at night suppresses immune responses in Siberian hamsters. Biol. Lett. 7,
1993). The slight, but significant, increases in neurodevelop- 468–471.
mental disorders associated with autumn conceptions may Beery, A.K., Trumbull, J.J., Tsao, J.M., Costantini, R.M., Zucker, I., 2007. Sex
involve subtle interactions among sex steroid hormones, differences in the onset of seasonal reproductive quiescence in hamsters. Proc. R.
Soc. B 274, 281–286.
photoperiod, and brain development in humans (Liederman Bentley, G.E., Kriegsfeld, L.J., Osugi, T., Ukena, K., O’Brien, S., Perfito, N., Moore, I.T.,
and Flannery, 1994). Tsutsui, K., Wingfield, J.C., 2006. Interactions of gonadotropin-releasing hormone
(GnRH) and gonadotropin-inhibitory hormone (GnIH) in birds and mammals. J. Exp.
Zool. 305A, 807–814.
Bentley, G.R., 1999. Aping our ancestors: comparative aspects of reproductive
References ecology. Evol. Anthropol. 7, 175–185.
Bercovitch, F.B., Harding, R.S., 1993. Annual birth patterns of savanna baboons
Aderem, A., Ulevitch, R.J., 2000. Toll-like receptors in the induction of the innate (Papio cynocephalus anubis) over a ten-year period at Gilgil, Kenya. Folia Primatol.
immune response. Nature 406, 782–787. (Basel) 61, 115–122.
Alberts, S.C., Hollister-Smith, J.A., Mututua, R.S., Sayialel, S.N., Muruthi, P.M., Bernard, D.J., Abuav-Nussbaum, R., Horton, T.H., Turek, F.W., 1999. Photoperiodic
Wurutere, M., Altmann, J., 2005. Seasonality and long-term change in a savanna effects on gonadotropin-releasing hormone (GnRH) content and the GnRH-
environment. In: Brockman, D.K., van Schaik, C.P. (Eds.), Seasonality in Primates: immunoreactive neuronal system of male Siberian hamsters. Biol. Reprod. 60,
Studies of Living and Extinct Human and Non-human Primates. Cambridge 272–276.
University Press, Cambridge, pp. 157–195. Beutler, B., Cerami, A., 1988. Cachectin, cachexia, and shock. Annu. Rev. Med. 39,
Almeida, O.F., Lincoln, G.A., 1984. Reproductive photorefractoriness in rams and 75–83.
accompanying changes in the patterns of prolactin and melatonin secretion. Biol. Bielak, T., Pucek, Z., 1960. Seasonal changes in the brain weight of the
Reprod. 30, 143–158. common shrew (Sorez araneus araneur Linnaeus, 1758). Acta Theriol. 3,
Anand, S., Losee-Olson, S., Turek, F.W., Horton, T.H., 2002. Differential regulation of 297–300.
luteinizing hormone and follicle-stimulating hormone in male Siberian hamsters by Bilbo, S.D., Nelson, R.J., 2002. Melatonin regulates energy balance and attenuates
exposure to females and photoperiod. Endocrinology 143, 2178–2188. fever in Siberian hamsters. Endocrinology 143, 2527–2533.
392 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

Bilbo, S.D., Dhabhar, F.S., Viswanathan, K., Saul, A., Yellon, S.M., Nelson, R.J., Caldwell, G.S., Glickman, S.E., Smith, E.R., 1984. Seasonal aggression independent
2002a. Short day lengths augment stress-induced leukocyte trafficking and stress- of seasonal testosterone in wood rats. Proc. Natl. Acad. Sci. U.S.A. 81,
induced enhancement of skin immune function. Proc. Natl. Acad. Sci. U.S.A. 99, 5255–5257.
4067–4072. Callard, G.V., Mak, P., Solomon, D.J., 1986. Effects of short days on aromatization and
Bilbo, S.D., Drazen, D.L., Quan, N., He, L., Nelson, R.J., 2002b. Short day lengths attenuate accumulation of nuclear estrogen receptors in the hamster brain. Biol. Reprod. 35,
the symptoms of infection in Siberian hamsters. Proc. R. Soc. B 269, 447–454. 282–291.
Bittman, E.L., Karsch, F.J., 1984. Nightly duration of pineal melatonin secretion Campbell, C.S., Finkelstein, J.S., Turek, F.W., 1978. The interaction of photoperiod
determines the reproductive response to inhibitory day length in the ewe. Biol. and testosterone on the development of copulatory behavior in castrated male
Reprod. 30, 585–593. hamsters. Physiol. Behav. 21, 409–415.
Bittman, E.L., Lehman, M.N., 1987. Paraventricular neurons control hamster photo- Carlton, E.D., Demas, G.E., 2014. Leptin mediates seasonal variation in some but not
periodism by a predominantly uncrossed descending pathway. Brain Res. Bull. 19, all symptoms of sickness in Siberian hamsters. Horm. Behav. 66, 802–811.
687–694. Carter, D.S., Goldman, B.D., 1983. Antigonadal effects of timed melatonin infusion in
Bittman, E.L., Weaver, D.R., 1990. The distribution of melatonin binding sites in pinealectomized male Djungarian hamsters (Phodopus sungorus sungorus):
neuroendocrine tissues of the ewe. Biol. Reprod. 43, 986–993. duration is the critical parameter. Endocrinology 113, 1261–1267.
Bittman, E.L., Goldman, B.D., Zucker, I., 1979. Testicular responses to melatonin are Castellano, J.M., Navarro, V.M., Fernandez-Fernandez, R., Nogueiras, R., Tovar, S.,
altered by lesions of the suprachiasmatic nuclei in golden hamsters. Biol. Reprod. Roa, J., Vazquez, M.J., Vigo, E., Casanueva, F.F., Aguilar, E., Pinilla, L.,
21, 647–656. Dieguez, C., Tena-Sempere, M., 2005. Changes in hypothalamic KiSS-1 system
Bittman, E.L., Crandell, R.G., Lehman, M.N., 1989. Influences of the paraventricular and restoration of pubertal activation of the reproductive axis by kisspeptin in
and suprachiasmatic nuclei and olfactory bulbs on melatonin responses in the undernutrition. Endocrinology 146, 3917–3925.
golden hamster. Biol. Reprod. 40, 118–126. Chrousos, G.P., 1998. Ultradian, circadian, and stress-related hypothalamic-pituitary-
Bittman, E.L., Hegarty, C.M., Layden, M.Q., Jonassen, J.A., 1990. Influences of adrenal axis activity–a dynamic digital-to-analog modulation. Endocrinology 139,
photoperiod on sexual behaviour, neuroendocrine steroid receptors and adenohy- 437–440.
pophysial hormone secretion and gene expression in female golden hamsters. Clarke, I.J., Caraty, A., 2013. Kisspeptin and seasonality of reproduction. Adv. Exp.
J. Mol. Endocrinol. 5, 15–25. Med. Biol. 784, 411–430.
Bittman, E.L., Thomas, E.M., Zucker, I., 1994. Melatonin binding sites in sciurid and Clewe, T.H., 1969. Observations on reproduction of squirrel monkeys in captivity.
hystricomorph rodents: studies on ground squirrels and guinea pigs. Brain Res. J. Reprod. Fertil. 151–156.
648, 73–79. Czyba, J.C., Girod, C., Durland, N., 1964. On epiphyso-hypophysial antagonism and
Bittman, E.L., Jetton, A.E., Villalba, C., Devries, G.J., 1996. Effects of photoperiod and seasonal spermatogenesis variations in the golden hamster. Comptes Rendus des
androgen on pituitary function and neuropeptide staining in Siberian hamsters. Am. Seances de la Soc. de Biol. de ses Fil. 158, 742–745.
J. Physiol. 271, R64–R72. Dallman, M.F., Strack, A.M., Akana, S.F., Bradbury, M.J., Hanson, E.S., Scribner, K.A.,
Bolinger, M., Olson, S.L., Delagrange, P., Turek, F.W., 1996. Melatonin agonist Smith, M., 1993. Feast and famine: critical role of glucocorticoids with insulin in
attenuates a stress response and permits growth hormone release in male golden daily energy flow. Front. Neuroendocrinol. 14, 303–347.
hamsters. In: Fifth Meeting of the Society for Research on Biological Rhythms, Dark, J., Pickard, G.E., Zucker, I., 1985. Persistence of circannual rhythms in ground
Amelia Island FL. squirrels with lesions of the suprachiasmatic nuclei. Brain Res. 332, 201–207.
Bradley, A.J., McDonald, I.R., Lee, A.K., 1980. Stress and mortality in a small Dark, J., Whaling, C.S., Zucker, I., 1987. Androgens exert opposite effects on body
marsupial (Antechinus stuartii Macleay). General Comp. Endocrinol. 40, 188–200. mass of heavy and light meadow voles. Horm. Behav. 21, 471–477.
Brakefield, P.M., 1996. Seasonal polyphenism in butterflies and natural selection. Dark, J., Spears, N., Whaling, C.S., Wade, G.N., Meyer, J.S., Zucker, I., 1990. Long day
Trends Ecol. Evol. 11, 275–277. lengths promote brain growth in meadow voles. Dev. Brain Res. 53, 264–269.
Breedlove, S.M., 1992. Sexual dimorphism in the vertebrate nervous system. Darrow, J.M., Goldman, B.D., 1986. Circadian regulation of pineal melatonin and
J. Neurosci. 12, 4133–4142. reproduction in the Djungarian hamster. J. Biol. Rhythms 1, 39–54.
Breuner, C.W., Wingfield, J.C., Romero, L.M., 1999. Diel rhythms of basal and stress- Day, F.R., Forouhi, N.G., Ong, K.K., Perry, J.R.B., 2015. Season of birth is associated
induced corticosterone in a wild, seasonal vertebrate, Gambel’s white-crowned with birth weight, pubertal timing, adult body size and educational attainment: a UK
sparrow. J. Exp. Zool. 284, 334–342. Biobank study. Heliyon 1, e00031.
Brewis, A., Laycock, J., Huntsman, J., 1996. Birth non-seasonality on the Pacific Demas, G.E., 2004. The energetics of immunity: a neuroendocrine link between energy
equator. Curr. Anthropol. 37, 842–851. balance and immune function. Horm. Behav. 45, 173–180.
Brockman, D.K., van Schaik, C.P., 2005. Seasonality and reproductive function. In: Demas, G.E., Nelson, R.J., 1996. The effects of photoperiod and temperature on
Brockman, D.K., van Schaik, C.P. (Eds.), Seasonality in Primates: Studies of Living immune function of adult male deer mice (Peromyscus maniculatus). J. Biol.
and Extinct Human and Non-human Primates. Cambridge University Press, Cam- Rhythms 11, 94–102.
bridge, UK, pp. 269–305. Demas, G.E., Chefer, V., Talan, M.C., Nelson, R.J., 1997a. Metabolic costs of an
Bronson, F.H., 1985. Mammalian reproduction, an ecological perspective. Biol. antigen-stimulated immune response in adult and aged C57BL/6J mice. Am. J.
Reprod. 32, 1–26. Physiol. 273, R1631–R1637.
Bronson, F.H., 1989. Mammalian Reproductive Biology. University of Chicago Press, Demas, G.E., DeVries, A.C., Nelson, R.J., 1997b. Effects of photoperiod and 2-deoxy-
Chicago. D-glucose-induced metabolic stress on immune function in female deer mice. Am.
Bronson, F.H., 1995. Seasonal variation in human reproduction: environmental factors. J. Physiol. 272, R1762–R1767.
Q. Rev. Biol. 70, 141–164. Demas, G.E., Moffatt, C.A., Drazen, D.L., Nelson, R.J., 1999. Castration does not
Bronson, F.H., 2000. Puberty and energy reserves: a walk on the wild side. In: inhibit aggressive behavior in adult male prairie voles (Microtus ochrogaster).
Wallen, K., Schneider, J.E. (Eds.), Reproduction in Context. MIT Press, Cambridge, Physiol. Behav. 66, 59–62.
pp. 15–33. Demas, G.E., Polacek, K.M., Durazzo, A., Jasnow, A.M., 2004. Adrenal hormones
Bronson, F.H., 2004. Are humans seasonally photoperiodic? J. Biol. Rhythms 19, mediate melatonin-induced increases in aggression in male Siberian hamsters
180–192. (Phodopus sungorus). Horm. Behav. 46, 582–591.
Bronson, F.H., Heideman, P.D., 1994. Seasonal regulation of reproduction in Desjardins, C., Lopez, M.J., 1980. Sensory and nonsensory modulation of testis
mammals. In: Knobil, E., Neill, J.D. (Eds.), Physiology of Reproduction. Raven function. In: Steinberger, A., Steinberger, E. (Eds.), Testicular Development,
Press, New York, pp. 541–583. Structure and Function. Raven Press, New York, pp. 381–388.
Butler, M.P., Trumbull, J.J., Turner, K.W., Zucker, I., 2007a. Timing of puberty and Di Bitetti, M.S., Janson, C.H., 2000. When will the stork arrive? Patterns of birth
synchronization of seasonal rhythms by simulated natural photoperiods in female seasonality in neotropical primates. Am. J. Primatol. 50, 109–130.
Siberian hamsters. Am. J. Physiol. 293, R413–R420. Dickert-Conlin, S., Chandra, A., 1999. Taxes and the timing of births. J. Political Econ.
Butler, M.P., Turner, K.W., Park, J.H., Butler, J.P., Trumbull, J.J., Dunn, S.P., 107, 161–177.
Villa, P., Zucker, I., 2007b. Simulated natural day lengths synchronize seasonal Duncan, M.J., Goldman, B.D., DiPinto, M.N., Stetson, M.H., 1985. Testicular function
rhythms of asynchronously born male Siberian hamsters. Am. J. Physiol. 293, and pelage color have different critical daylengths in the Djungarian hamster,
R402–R412. Phodopus sungorus sungorus. Endocrinology 116, 424–430.
Butler, M.P., Turner, K.W., Park, J.H., Schoomer, E.E., Zucker, I., Gorman, M.R., Duncan, M.J., Takahashi, J.S., Dubocovich, M.L., 1989. Characteristics and autora-
2010. Seasonal regulation of reproduction: altered role of melatonin under natu- diographic localization of 2-[125I]iodomelatonin binding sites in Djungarian hamster
ralistic conditions in hamsters. Proc. R. Soc. B 277, 2867–2874. brain. Endocrinology 125, 1011–1018.
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 393

Dungan, H.M., Clifton, D.K., Steiner, R.A., 2006. Minireview: kisspeptin neurons as Goldman, B.D., Gwinner, E., Karsch, F.J., Saunders, D., Zucker, I., Ball, G.F., 2003.
central processors in the regulation of gonadotropin-releasing hormone secretion. Circannual rhythms and photoperiodism. In: Dunlap, J., Loros, J., DeCoursey, P.
Endocrinology 147, 1154–1158. (Eds.), Chronobiology: Biological Timekeeping. Sinauer Associates, Sunderland,
Elliott, J.A., 1976. Circadian rhythms and photoperiodic time measurement in MA, pp. 107–142.
mammals. Fed. Proc. 35, 2339–2346. Gorman, M.R., 2003. Melatonin implants disrupt developmental synchrony regulated
Elliott, J.A., Tamarkin, L., 1994. Complex circadian regulation of pineal melatonin and by flexible interval timers. J. Neuroendocrinol. 15, 1084–1094.
wheel-running in Syrian hamsters. J. Comp. Physiol. A 174, 469–484. Gorman, M.R., Zucker, I., 1995. Seasonal adaptations of Siberian hamsters. II. Pattern
Elliott, J.A., Bartness, T.J., Goldman, B.D., 1989. Effect of melatonin infusion duration of change in day length controls annual testicular and body weight rhythms. Biol.
and frequency on gonad, lipid, and body mass in pinealectomized male Siberian Reprod. 53, 116–125.
hamsters. J. Biol. Rhythms 4, 439–455. Gorman, M.R., Zucker, I., 1997. Environmental induction of photononresponsiveness in
Ellis, G.B., Turek, F.W., 1979. Time course of the photoperiod-induced change in the Siberian hamster (Phodopus sungorus). Am. J. Physiol. 272, R887–R895.
sensitivity of the hypothalamic-pituitary axis to testosterone feedback in castrated Gorman, M.R., Zucker, I., 1998. Mammalian seasonal rhythms: new perspectives
male hamsters. Endocrinology 104, 625–630. gained from the use of simulated natural photoperiods. In: Touitou, Y. (Ed.),
Ellis, G.B., Turek, F.W., 1980. Photoperiod-induced change in responsiveness Biological Clocks: Mechanisms and Applications. Elsevier, Amsterdam,
of the hypothalamic-pituitary axis to exogenous 5-alpha-dihydrotestosterone pp. 195–204.
and 17ß-estradiol in castrated male hamsters. Neuroendocrinology 31, Gorman, M.R., Ferkin, M.H., Nelson, R.J., Zucker, I., 1993. Reproductive status
205–209. influences odor preferences of the meadow vole, Microtus pennsylvanicus, in
Ellis, G.B., Losee, S.H., Turek, F.W., 1979. Prolonged exposure of castrated male winter day lengths. Can. J. Zool. 71, 1748–1754.
hamsters to a nonstimulatory photoperiod: spontaneous change in sensitivity of the Greives, T.J., Mason, A.O., Scotti, M.A., Levine, J., Ketterson, E.D., Kriegsfeld, L.J.,
hypothalamic-pituitary axis to testosterone feedback. Endocrinology 104, 631–635. Demas, G.E., 2007. Environmental control of kisspeptin: implications for seasonal
Ellison, P.T., Valeggia, C.R., Sherry, D.S., 2005. Human birth seasonality. In: reproduction. Endocrinology 148, 1158–1166.
Brockman, D.K., van Schaik, C.P. (Eds.), Seasonality in Primates: Studies of Living Grimble, R.F., 1994. Malnutrition and the immune response. 2. Impact of nutrients on
and Extinct Human and Non-human Primates. Cambridge University Press, cytokine biology in infection. Trans. R. Soc. Trop. Med. Hyg. 88, 615–619.
Cambridge, UK, pp. 379–399. Gunduz, B., Stetson, M.H., 2001. A test of the coincidence and duration models of
Epperson, L.E., Karimpour-Fard, A., Hunter, L.E., Martin, S.L., 2011. Metabolic cycles melatonin action in Siberian hamsters: the effects of 1-hr melatonin infusions on
in a circannual hibernator. Physiol. Genomics 43, 799–807. testicular development in intact and pinealectomized prepubertal Phodopus sun-
Farner, D.S., 1985. Annual rhythms. Annu. Rev. Physiol. 47, 65–82. gorus. J. Pineal Res. 30, 97–107.
Farrell, G., Pease, K., 1994. Crime seasonality. Br. J. Criminol. 34, 487–498. Gwinner, E., 1986. Circannual Rhythms. Springer-Velag, Berlin.
Fleming, A.S., Cheung, U., Myhal, N., Kessler, Z., 1989. Effects of maternal hormones Hall, V.D., Bartke, A., Goldman, B.D., 1982. Role of the testis in regulating the duration
on ‘timidity’ and attraction to pup-related odors in female rats. Physiol. Behav. 46, of hibernation in the Turkish hamster, Mesocricetus brandti. Biol. Reprod. 27,
449–453. 802–810.
Foley, R.A., 1993. The influence of seasonality on hominid evolution. In: Ulijaszek, S.J., Hastings, M., 1996. Melatonin and seasonality: filling the gap. J. Neuroendocrinol. 8,
Strickland, S.S. (Eds.), Seasonality and Human Ecology. Cambridge University 482–483.
Press, Cambridge, pp. 17–37. Hastings, M.H., Herzog, E.D., 2004. Clock genes, oscillators, and cellular networks in
Follett, B.K., 2015. “Seasonal changes in the neuroendocrine system”: some reflec- the suprachiasmatic nuclei. J. Biol. Rhythms 19, 400–413.
tions. Front. Neuroendocrinol. 37, 3–12. Hazlerigg, D.G., Barrett, P., Hastings, M.H., Morgan, P.J., 1996. Are nuclear receptors
Fonken, L.K., Workman, J.L., Walton, J.C., Weil, Z.M., Morris, J.S., Haim, A., involved in pituitary responsiveness to melatonin? Mol. Cell. Endocrinol. 123,
Nelson, R.J., 2010. Light at night increases body mass by shifting the time of food 53–59.
intake. Proc. Natl. Acad. Sci. U.S.A. 107, 18664–18669. Hegstrom, C.D., Breedlove, S.M., 1999a. Seasonal plasticity of neuromuscular junc-
Forger, N.G., Breedlove, S.M., 1987. Seasonal variation in mammalian striated muscle tions in adult male Siberian hamsters (Phodopus sungorus). Brain Res. 819,
mass and motoneuron morphology. J. Neurobiol. 18, 155–165. 83–88.
Freeman, D.A., Zucker, I., 2000. Temperature-independence of circannual variations Hegstrom, C.D., Breedlove, S.M., 1999b. Social cues attenuate photoresponsiveness
in circadian rhythms of golden-mantled ground squirrels. J. Biol. Rhythms 15, of the male reproductive system in Siberian hamsters (Phodopus sungorus).
336–343. J. Biol. Rhythms 14, 54–61.
Freeman, D.A., Zucker, I., 2001. Refractoriness to melatonin occurs independently at Heideman, P.D., Kane, S.L., Goodnight, A.L., 1999. Differences in hypothalamic 2-
multiple brain sites in Siberian hamsters. Proc. Natl. Acad. Sci. U.S.A. 98, [125I]iodomelatonin binding in photoresponsive and non-photoresponsive white-
6447–6452. footed mice, Peromyscus leucopus. Brain Res. 840, 56–64.
Freeman, D.A., Teubner, B.J., Smith, C.D., Prendergast, B.J., 2007. Exogenous Heimovics, S.A., Ferris, L.K., Somac, K.K., 2015. Non-invasive administration of
T3 mimics long day lengths in Siberian hamsters. Am. J. Physiol. 292, 17b-estradiol rapidly increases aggressive behavior in non-breeding, but not
R2368–R2372. breeding, male song sparrows. Horm. Behav. 69, 31–38.
Galea, L.A.M., Perrot-Sinal, T.S., Kavaliers, M., Ossenkopp, K.P., 1999. Relations of Hemingway, C.A., Bynum, N., 2005. The influence of seasonality on primate diet and
hippocampal volume and dentate gyrus width to gonadal hormone levels in male ranging. In: Brockman, D.K., van Schaik, C.P. (Eds.), Seasonality in Primates:
and female meadow voles. Brain Res. 821, 383–391. Studies of Living and Extinct Human and Non-human Primates. Cambridge
Garcia, I.M., Whitsett, J.M., 1983. Influence of photoperiod and social environment on University Press, Cambridge, UK, pp. 57–104.
sexual maturation in female deer mice (Peromyscus maniculatus bairdii). J. Comp. Henken, A.M., Brandsma, H.A., 1982. The effect of environmental temperature on
Psychol. 97, 127–134. immune response and metabolism of the young chicken. 2. Effect of the immune
Garrett, J.W., Campbell, C.S., 1980. Changes in social behavior of the male golden response to sheep red blood cells on energy metabolism. Poult. Sci. 61,
hamster accompanying photoperiodic changes in reproduction. Horm. Behav. 14, 1667–1677.
303–318. Henson, J.R., Carter, S.N., Freeman, D.A., 2013. Exogenous T3 elicits long day-like
Glass, J.D., 1986. Short photoperiod-induced gonadal regression: effects on the alterations in testis size and the RFamides Kisspeptin and gonadotropin inhibi-
gonadotropin-releasing hormone (GnRH) neuronal system of the white-footed tory hormone in short-day Siberian hamsters. J. Biol. Rhythms 28, 193–200.
mouse, Peromyscus leucopus. Biol. Reprod. 35, 733–743. Herbert, J., Klinowska, M., 1978. Day length and the annual reproductive cycle in the
Glass, J.D., Lynch, G.R., 1982. Evidence for a brain site of melatonin action in the ferret (Mustela furo): the role of the pineal body. In: Environmental Endocrinology.
white-footed mouse, Peromyscus leucopus. Neuroendocrinology 34, 1–6. Springer-Verlag, Berlin, pp. 87–93.
Goldman, B.D., 2001. Mammalian photoperiodic system: formal properties and Hiebert, S.M., Thomas, E.M., Lee, T.M., Pelz, K.M., Yellon, S.M., Zucker, I., 2000.
neuroendocrine mechanisms of photoperiodic time measurement. J. Biol. Rhythms Photic entrainment of circannual rhythms in golden-mantled ground squirrels: role
16, 283–301. of the pineal gland. J. Biol. Rhythms 15, 126–134.
Goldman, B.D., Nelson, R.J., 1993. Melatonin and seasonality in mammals. In: Hill, K., Hurtado, A.M., 1996. Ache Life History. Aldine/De Gruyter, New York.
Yu, H.S., Reiter, R.J. (Eds.), Melatonin: Biosynthesis, Physiological Effects, and Hoffman, R.A., Reiter, R.J., 1965. Pineal gland: influence on gonads of male hamsters.
Clinical Applications. CRC Press, Boca Raton, Florida, pp. 225–252. Science 148, 1609–1611.
Goldman, B.D., Darrow, J.M., Yogev, L., 1984. Effects of timed melatonin infusions on Honrado, G.I., Fleming, A.S., 1996. Chemical and behavioral stimulation from females
reproductive development in the Djungarian hamster (Phodopus sungorus). accelerates recrudescence in male Syrian hamsters exposed to short days. J. Biol.
Endocrinology 114, 2074–2083. Rhythms 11, 103–112.
394 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

Honrado, G.I., Bird, M., Fleming, A.S., 1991. The effects of short day exposure on Kelly, K.K., 1993. Androgenic induction of brain sexual dimorphism depends on
seasonal and circadian reproductive rhythms in male golden hamsters. Physiol. photoperiod in meadow voles. Physiol. Behav. 53, 245–249.
Behav. 49, 277–287. Korytko, A.I., Marcelino, J., Blank, J.L., 1995. Differential testicular responses to short
Horton, T.H., 1984. Growth and reproductive development of male Microtus montanus daylength in deer mice are reflected by regional and morphological differences in
is affected by the prenatal photoperiod. Biol. Reprod. 31, 499–504. the GnRH neuronal system. Brain Res. 685, 135–142.
Horton, T.H., Yellow, S.M., 2001. Aging, reproduction and the melatonin rhythm in the Kutzback, J.E., Webb, T., 1993. Conceptual basis for understanding late quaternary
Siberian hamster. J. Biol. Rhythms 16, 243–253. climates. In: Wright Jr., H.E. (Ed.), Global Climates since the Last Glacial Maximum.
Huang, L.Y., DeVries, G.J., Bittman, E.L., 1998. Photoperiod regulates neuronal Univesity of Minnesota Press, Minneapolis, pp. 5–11.
bromodeoxyuridine labeling in the brain of a seasonally breeding mammal. Kwiatkowski, D., Hill, A.V., Sambou, I., Twumasi, P., Castracane, J., Manogue, K.R.,
J. Neurobiol. 36, 410–420. Cerami, A., Brewster, D.R., Greenwood, B.M., 1990. TNF concentration in fatal
Huey, R.B., Berrigan, D., 1996. Testing evolutionary hypotheses of acclimation. In: cerebral, non-fatal cerebral, and uncomplicated Plasmodium falciparum malaria.
Society for Experimental Biology Seminar Series No. 59: Animals and Temperature: Lancet 336, 1201–1204.
Phenotypic and Evolutionary Adaptation. Cambridge University Press, New York, Larkin, J.E., Jones, J., Zucker, I., 2002. Temperature dependence of gonadal
pp. 205–237. regression in Syrian hamsters exposed to short day lengths. Am. J. Physiol. Regul.
Hutchison, R.E., Hutchison, J.B., Steimer, T., Steel, E., Powers, J.B., Walker, A.P., Integr. Comp. Physiol. 282, R744–R752.
Herbert, J., Hastings, M.H., 1991. Brain aromatization of testosterone in the male Lavenex, P., Steele, M.A., Jacobs, L.F., 2000. Sex differences, but no seasonal
Syrian hamster: effects of androgen and photoperiod. Neuroendocrinology 53, variations in the hippocampus of food-caching squirrels: a stereological study.
194–203. J. Comp. Neurol. 425, 152–166.
Ibanez-Costa, A., Cordoba-Chacon, J., Gahete, M.D., Kineman, R.D., Castano, J.P., Lee, T.M., Gorman, M.R., 2000. In: Wallen, K., Schneider, J.S. (Eds.), Reproduction
Lugue, R.M., 2015. Melatonin regulates somatotrope and lactotrope function in Context. Massachusetts Institute of Technology Press, Cambridge, MA,
through common and distinct signaling pathways in cultured primary pituitary cells pp. 191–218, 1999.
from female primates. Endocrinology 156, 1100–1110. Lee, T.M., Zucker, I., 1991. Suprachiasmatic nucleus and photic entrainment of cir-
Ikegami, K., Iigo, M., Yoshimura, T., 2013. Circadian clock gene Per2 is not necessary cannual rhythms in ground squirrels. J. Biol. Rhythms 6, 315–330.
for the photoperiodic response in mice. PLoS One 8, e58482. Lee, T.M., Zucker, I., 1995. Seasonal variations in circadian rhythms persist in
Illnerova, H., 1991. The suprachiasmatic nucleus and rhythmic pineal melatonin gonadectomized golden-mantled ground squirrels. J. Biol. Rhythms 10, 188–195.
production. In: Klein, D.C., Moore, R.Y., Reppert, S.M. (Eds.), Suprachiasmatic Lee, T.M., Holmes, W.G., Zucker, I., 1990. Temperature dependence of circadian
Nucleus: The Mind’s Clock. Oxford University Press, New York. rhythms in golden-mantled ground squirrels. J. Biol. Rhythms 5, 25–34.
Illnerova, H., Hoffmann, K., Vanecek, J., 1984. Adjustment of pineal melatonin Levitas, E., Lunenfeld, E., Weisz, N., Friger, M., Har-Vardi, I., 2013. Seasonal variation
and N-acetyltransferase rhythms to change from long to short photoperiod in of human sperm cells among 6455 semen samples: a plausible explanation of
the Djungarian hamster Phodopus sungorus. Neuroendocrinology 38, a seasonal birth pattern. Am. J. Obstetrics Gynecol. 208, 406.e1–6.
226–231. Lewis, D., Freeman, D.A., Dark, J., Wynne-Edwards, K.E., Zucker, I., 2002. Photo-
Jablonski, N.G., 2005. Primate diversity and environmental seasonality in historical periodic control of oestrous cycles in Syrian hamsters: mediation by the mediobasal
perspective. In: Brockman, D.K., van Schaik, C.P. (Eds.), Seasonality in Primates: hypothalamus. J. Neuroendocrinol. 14, 294–299.
Studies of Living and Extinct Human and Non-human Primates. Cambridge Liederman, J., Flannery, K.A., 1994. Fall conception increases the risk of neuro-
University Press, Cambridge, UK, pp. 465–486. developmental disorder in offspring. J. Clin. Exp. Neuropsychol. 16, 754–768.
Jablonski, N.G., Whitfort, M.J., Roberts-Smith, N., Qinqi, X., 2000. The influence of life Lincoln, G.A., 1990. Correlation with changes in horns and pelage, but not repro-
history and diet on the distribution of catarrhine primates during the Pleistocene in duction, of seasonal cycles in the secretion of prolactin in rams of wild, feral and
eastern Asia. J. Hum. Evol. 39, 131–157. domesticated breeds of sheep. J. Reprod. Fertil. 90, 285–296.
Jacobs, L.F., 1996. The economy of winter: phenotypic plasticity in behavior and brain Lincoln, G.A., 2002. Neuroendocrine regulation of seasonal gonadotrophin and
structure. Biol. Bull. 191, 92–100. prolactin rhythms: lessons from the Soay ram model. Reprod. Suppl. 59,
Jacobs, L.F., Gaulin, S.J., Sherry, D.F., Hoffman, G.E., 1990. Evolution of spatial 131–147.
cognition: sex-specific patterns of spatial behavior predict hippocampal size. Proc. Lincoln, G.A., Clarke, I.J., 1997. Refractoriness to a static melatonin signal develops in
Natl. Acad. Sci. U.S.A. 87, 6349–6352. the pituitary gland for the control of prolactin secretion in the ram. Biol. Reprod. 57,
Janson, C., Verdolin, J., 2005. Seasonality of primate births in relation to climate. In: 460–467.
Brockman, D.K., van Schaik, C.P. (Eds.), Seasonality in Primates: Studies of Living Lincoln, G.A., Maeda, K., 1992. Effects of placing micro-implants of melatonin in the
and Extinct Human and Non-human Primates. Cambridge University Press, Cam- mediobasal hypothalamus and preoptic area on the secretion of prolactin and beta-
bridge, UK, pp. 307–350. endorphin in rams. J. Endocrinol. 134, 437–448.
Jasnow, A.M., Huhman, K.L., Bartness, T.J., Demas, G.E., 2000. Short-day Lincoln, G.A., Short, R.V., 1980. Seasonal breeding: nature’s contraceptive. Recent
increases in aggression are inversely related to circulating testosterone Prog. Horm. Res. 36, 1–52.
concentrations in male Siberian hamsters (Phodopus sungorus). Horm. Behav. Lincoln, G.A., Johnston, J.D., Andersson, H., Wagner, G., Hazlerigg, D.G., 2005.
38, 102–110. Photorefractoriness in mammals: dissociating a seasonal timer from the circadian-
Jetton, A.E., Turek, F.W., Schwartz, N.B., 1994. Effects of melatonin and time of day based photoperiod response. Endocrinology 146, 3782–3790.
on in vitro pituitary gonadotropin basal secretion and GnRH responsiveness in the Lindburg, D.G., 1987. Seasonality of reproduction in primates. In: Mitchell, G.,
male golden hamster. Neuroendocrinology 60, 527–534. Erwin, J. (Eds.), Comparative Primate Biology, Vol. 2, Part B, Behavior, Cognition
Johnson, R.W., 1997. Inhibition of growth by pro-inflammatory cytokines: an integrated and Motivation. A.R. Liss, New York.
view. J. Anim. Sci. 75, 1244–1255. Lochmiller, R.L., Vestey, M.R., McMurry, S.T., 1994. Temporal variation in humoral
Johnston, J.D., Cagampang, F.R., Stirland, J.A., Carr, A.J., White, M.R., Davis, J.R., and cell-mediated immune response in a Sigmodon hispidus population. Ecology
Loudon, A.S., 2003. Evidence for an endogenous per1- and ICER-independent 75, 236–245.
seasonal timer in the hamster pituitary gland. FASEB J. 17, 810–815. Lochmiller, R.L., Deerenberg, C., 2000. Trade-offs in evolutionary immunology: just
Johnston, J.D., Ebling, F.J., Hazlerigg, D.G., 2005. Photoperiod regulates multiple what is the cost of immunity? Oikos 88, 87–98.
gene expression in the suprachiasmatic nuclei and pars tuberalis of the Siberian Lu, W., Meng, Q.J., Tyler, N.J., Stokkan, K.A., Loudon, A.S., 2010. A circadian clock
hamster (Phodopus sungorus). Eur. J. Neurosci. 21, 2967–2974. is not required in an arctic mammal. Curr. Biol. 20, 533–537.
Johnston, P.G., Zucker, I., 1980. Photoperiodic regulation of reproductive development Lummaa, V., Lemmetyinen, R., Haukioja, E., Pikkola, M., 1998. Seasonality of births in
in white-footed mice (Peromyscus leucopus). Biol. Reprod. 22, 983–989. Homo sapiens in pre-industrial Finland: maximisation of offspring survivorship?
Kaplan, J.N., 1977. Breeding and rearing squirrel monkeys (Saimiri sciureus) in J. Evol. Biol. 11, 147–157.
captivity. Lab. Anim. Sci. 27, 557–567. Madison, D.M., FitzGerald, R.W., McShea, W.J., 1984. Dynamics of social nesting in
Karp, J.D., Powers, J.B., 1993. Photoperiodic and pineal influences on estrogen- overwintering meadow voles (Microtus pennsylvanicus): possible consequences for
stimulated behaviors in female Syrian hamsters. Physiol. Behav. 54, 19–28. population cycling. Behav. Ecol. Sociobiol. 15, 9–17.
Kauffman, A.S., Freeman, D.A., Zucker, I., 2003. Termination of neuroendocrine Maier, S.F., Watkins, L.R., Fleshner, M., 1994. Psychoneuroimmunology: the interface
refractoriness to melatonin in Siberian hamsters (Phodopus sungorus). between behavior, brain, and immunity. Am. Psychol. 49, 1004–1017.
J. Neuroendocrinol. 15, 191–196. Mangels, R.A., Powers, J.B., Blaustein, J.D., 1998. Effect of photoperiod on neural
Kelley, K.W., Dantzer, R., 1990. Neuroendocrine-immune interactions. Adv. Veterinary estrogen and progestin receptor immunoreactivity in female Syrian hamsters. Brain
Sci. Comp. Med. 35, 283–305. Res. 796, 63–74.
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 395

Martin, J.E., Engel, J.N., Klein, D.C., 1977. Inhibition of the in vitro pituitary response Nelson, R.J., 1987. Photoperiod-nonresponsive morphs: a possible variable in
to luteinizing hormone-releasing hormone by melatonin, serotonin, and microtine population-density fluctuations. Am. Nat. 130, 350–369.
5-methoxytryptamine. Endocrinology 100, 675–680. Nelson, R.J., 2004. Seasonal immune function and sickness responses. Trends
Martin, L.B., Weil, Z.M., Nelson, R.J., 2008. Seasonal changes in vertebrate immune Immunol. 25, 187–192.
activity: mediation by physiological trade-offs. Phil. Trans. R. Soc. Lond. B 363, Nelson, R.J., Demas, G.E., 1996. Seasonal changes in immune function. Q. Rev. Biol.
321–339. 71, 511–548.
Martinez-Bakker, M., Bakker, K.M., King, A.A., Rohani, P., 2013. Human birth sea- Nelson, R.J., Drazen, D.L., 1999. Melatonin mediates seasonal adjustments in immune
sonality: latitudinal gradient and interplay with childhood disease dynamics. Proc. function. Reprod. Nutr. Dev. 39, 383–398.
R. Soc. B 281, 20132438. Nelson, R.J., Trainor, B.C., 2007. Neural mechanisms of aggression. Nat. Rev.
Maslin, M.A., Brierley, C.M., Milner, A., Shultz, S., Trauth, M.H., Wilson, K.E., 2014. Neurosci. 8, 536–546.
East African climate pulses and early human evolution. Q. Sci. Rev. 101, 1–17. Nelson, R.J., Frank, D., Smale, L., Willoughby, S.B., 1989. Photoperiod and
Mason, A.O., Duffy, S., Zhao, S., Ubuka, T., Bentley, G.E., Tsutsui, K., Silver, R., temperature affect reproductive and nonreproductive functions in male prairie voles
Kriegsfeld, L.J., 2010. Photoperiod and reproductive condition are associated with (Microtus ochrogaster). Biol. Reprod. 40, 481–485.
changes in RFamide-related peptide (RFRP) expression in Syrian hamsters. J. Biol. Nelson, R.J., Demas, G.E., Klein, S.L., Kriegsfeld, L.J., 1995. The influence of season,
Rhythms 25, 176–185. photoperiod, and pineal melatonin on immune function. J. Pineal Res. 19,
Mason, A.O., Greives, T.J., Scotti, M.A., Levine, J., Frommeyer, S., Ketterson, E.D., 149–165.
Demas, G.E., Kriegsfeld, L.J., 2007. Suppression of kisspeptin expression and Nelson, R.J., Demas, G.E., Klein, S.L., 1998. Photoperiodic mediation of seasonal
gonadotropic axis sensitivity following exposure to inhibitory day lengths in female breeding and immune function in rodents: a multi-factorial approach. Am. Zool. 38,
Siberian hamsters. Horm. Behav. 52, 492–498. 226–237.
Matt, K.S., Stetson, M.H., 1979. Hypothalamic-pituitary-gonadal interactions during Nelson, R.J., Demas, G.E., Klein, S.L., Kriegsfeld, L.J., 2002. Seasonal Cycles in
spontaneous testicular recrudescence in golden hamsters. Biol. Reprod. 20, Immune Function and Disease Processes. Cambridge University Press,
739–746. New York.
Maywood, E.S., Hastings, M.H., 1995. Lesions of the iodomelatonin-binding sites of Nelson, R.J., Denlinger, D.L., Somers, D.E., 2010. Photoperiodism: Biological
the mediobasal hypothalamus spare the lactotropic, but block the gonadotropic Calendar. Oxford University Press, New York.
response of male Syrian hamsters to short photoperiod and to melatonin. Endo- Nicholls, T.J., Goldsmith, A.R., Dawson, A., 1988. Photorefractoriness in birds and
crinology 136, 144–153. comparison with mammals. Physiol. Rev. 68, 133–176.
Maywood, E.S., Bittman, E.L., Hastings, M.H., 1996. Lesions of the melatonin- and O’Jile, J.R., Bartness, T.J., 1992. Effects of thyroxine on the photoperiodic control of
androgen-responsive tissue of the dorsomedial nucleus of the hypothalamus block energy balance and reproductive status in Siberian hamsters. Physiol. Behav. 52,
the gonadal responses of male Syrian hamsters to programmed infusions of 267–270.
melatonin. Biol. Reprod. 54, 470–477. Park, J.H., Kauffman, A.S., Paul, M.J., Butler, M.P., Beery, A.K., Costantini, R.M.,
McShea, W.J., 1990. Social tolerance and proximate mechanisms of dispersal among Zucker, I., 2006. Interval timer control of puberty in photoinhibited Siberian
winter groups of meadow voles, Microtus pennsylvanicus. Anim. Behav. 39, hamsters. J. Biol. Rhythms 21, 373–383.
346–351. Park, J.H., Paul, M.J., Butler, M.P., Villa, P., Burke, M., Kim, D.P., Routman, D.M.,
Medzhitov, R., 2007. Recognition of microorganisms and activation of the immune Schoomer, E.E., Zucker, I., 2007. Short duration testosterone infusions maintain
response. Nature 449, 819–826. male sex behavior in Syrian hamsters. Horm. Behav. 52, 169–176.
Meek, L.R., Lee, T.M., 1993a. Female meadow voles have a preferred mating pattern Park, J.H., Takasu, N., Alvarez, M.I., Clark, K., Aimaq, R., Zucker, I., 2004. Long-term
predicted by photoperiod, which influences fertility. Physiol. Behav. 54, persistence of male copulatory behavior in castrated and photo-inhibited Siberian
1201–1210. hamsters. Horm. Behav. 45, 214–221.
Meek, L.R., Lee, T.M., 1993b. Prediction of fertility by mating latency and photoperiod Parkinson, T.J., Follett, B.K., 1994. Effect of thyroidectomy upon seasonality in rams.
in nulliparous and primiparous meadow voles (Microtus pennsylvanicus). J. Reprod. Fertil. 101, 51–58.
J. Reprod. Fertil. 97, 353–357. Parkinson, T.J., Follett, B.K., 1995. Thyroidectomy abolishes seasonal testicular cycles
Messager, S., Hazlerigg, D.G., Mercer, J.G., Morgan, P.J., 2000. Photoperiod differen- of Soay rams. Proc. R. Soc. B 259, 1–6.
tially regulates the expression of Per1 and ICER in the pars tuberalis and the Paul, M.J., Zucker, I., Schwartz, W.J., 2008. Tracking the seasons: the internal
suprachiasmatic nucleus of the Siberian hamster. Eur. J. Neurosci. 12, 2865–2870. calendars of vertebrates. Phil. Trans. R. Soc. Lond. B 363, 341–361.
Messager, S., Ross, A.W., Barrett, P., Morgan, P.J., 1999. Decoding photoperiodic Paul, M.J., Galang, J., Schwartz, W.J., Prendergast, B.J., 2009. Intermediate-duration
time through Per1 and ICER gene amplitude. Proc. Natl. Acad. Sci. U.S.A. 96, day lengths unmask reproductive responses to nonphotic environmental cues. Am.
9938–9943. J. Physiol. Regul. Integr. Comp. Physiol. 296, 1613–1619.
Michael, R.P., Bonsall, R.W., 1977. A 3-year study of an annual rhythm in plasma Pengelley, E.T., Asmundson, S.J., 1974. Circannual rhythmicity in hibernating
androgen levels in male rhesus monkeys (Macaca mulatta) in a constant laboratory mammals. In: Pengelley, E.T. (Ed.), Circannual Clocks. Academic Press, New York,
environment. J. Reprod. Fertil. 49, 129–131. pp. 95–160.
Miernicki, M., Karp, J.D., Powers, J.B., 1990. Pinealectomy prevents short photo- Piatkowska, J.M., Prendergast, B.J., Gorman, M.R., 2003. Temporal integration of
period inhibition of male hamster sexual behavior. Physiol. Behav. 47, 293–299. melatonin infusion duration: signal averaging versus frequency dependence.
Migaud, M., Butrille, L., Batailler, M., 2015. Seasonal regulation of structural plasticity J. Pineal Res. 35, 91–97.
and neurogenesis in the adult mammalian brain: focus on the sheep hypothalamus. Pickard, G.E., Turek, F.W., 1983. The hypothalamic paraventricular nucleus mediates
Front. Neuroendocrinol. 37, 146–157. the photoperiodic control of reproduction but not the effects of light on the
Miller, E.S., Klinger, J.C., Akin, C., Koebel, D.A., Sonnenfeld, G., 1994. Inhibition of circadian rhythm of activity. Neurosci. Lett. 43, 67–72.
murine splenic T lymphocyte proliferation by 2-deoxy-D-glucose-induced metabolic Pitrosky, B., Kirsch, R., Vivien-Roels, B., Georg-Bentz, I., Canguilhem, B., Pevet, P.,
stress. J. Neuroimmunol. 52, 165–173. 1995. The photoperiodic response in Syrian hamster depends upon a melatonin-
Mink, J.W., Blumenschine, R.J., Adams, D.B., 1981. Ratio of central nervous system driven circadian rhythm of sensitivity to melatonin. J. Neuroendocrinol. 7,
to body metabolism in vertebrates: its constancy and functional basis. Am. J. 889–895.
Physiol. 241, R203–R212. Plant, T.M., Zumpe, D., Sauls, M., Michael, R.P., 1974. An annual rhythm in the
Moenter, S.M., Woodfill, C.J., Karsch, F.J., 1991. Role of the thyroid gland in seasonal plasma testosterone of adult male rhesus monkeys maintained in the laboratory.
reproduction: thyroidectomy blocks seasonal suppression of reproductive neuro- J. Endocrinol. 62, 403–404.
endocrine activity in ewes. Endocrinology 128, 1337–1344. Pleim, E.T., Lisciotto, C.A., DeBold, J.F., 1990. Facilitation of sexual receptivity in
Morgan, P.J., Ross, A.W., Graham, E.S., Adam, C., Messager, S., Barrett, P., 1998. hamsters by simultaneous progesterone implants into the VMH and ventral
Per1 is an early response gene under photoperiodic regulation in the ovine pars mesencephalon. Horm. Behav. 24, 139–151.
tuberalis. J. Neuroendocrinol. 10, 319–323. Pope, N.S., Wilson, M.E., Gordon, T.P., 1987. The effect of season on the induction of
Morin, L.P., Zucker, I., 1978. Photoperiodic regulation of copulatory behaviour in the sexual behavior by estradiol in female rhesus monkeys. Biol. Reprod. 36, 1047–1054.
male hamster. J. Endocrinol. 77, 249–258. Potts, R., 1998. Environmental hypotheses of hominin evolution. Yearb. Phys.
Morin, L.P., Allen, C.N., 2006. The circadian visual system. Brain Res. Rev. 51, 1–60. Anthropol. 41, 93–136.
Navara, K.J., Trainor, B.C., Nelson, R.J., 2007. Photoperiod alters macrophage Powers, J.B., Steel, E.A., Hutchison, J.B., Hastings, M.H., Herbert, J., Walker, A.P.,
responsiveness, but not expression of Toll-like receptors in Siberian hamsters. 1989. Photoperiodic influences on sexual behavior in male Syrian hamsters.
Comp. Biochem. Physiol. A Mol. Integr. Physiol. 148, 354–359. J. Biol. Rhythms 4, 61–78.
396 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

Powers, J.B., Jetton, A.E., Mangels, R.A., Bittman, E.L., 1997. Effects of photoperiod Roenneberg, T., 2004. The decline in human seasonality. J. Biol. Rhythms 19,
duration and melatonin signal characteristics on the reproductive system of male 193–195.
Syrian hamsters. J. Neuroendocrinol. 9, 451–466. Roenneberg, T., Aschoff, J., 1990a. Annual rhythm of human reproduction: I. Biology,
Prendergast, B.J., Hugenberger, J.L., 1999. Frequency coding of melatonin signals sociology, or both? J. Biol. Rhythms 5, 195–216.
sufficient to induce testicular growth in photoregressed Siberian hamsters. Roenneberg, T., Aschoff, 1990b. Annual rhythm of human reproduction: II. Environ-
J. Neuroendocrinol. 11, 237–241. mental correlations. J. Biol. Rhythms 5, 217–239.
Prendergast, B.J., Bilbo, S.D., Dhabhar, F.S., Nelson, R.J., 2004a. Effects of photo- Ronchi, E., Krey, L.C., Pfaff, D.W., 1992. Steady state analysis of hypothalamic GnRH
period history on immune responses to intermediate day lengths in Siberian mRNA levels in male Syrian hamsters: influences of photoperiod and androgen.
hamsters (Phodopus sungorus). J. Neuroimmunol. 149, 31–39. Neuroendocrinology 55, 146–155.
Prendergast, B.J., Renstrom, R.A., Nelson, R.J., 2004b. Genetic analyses of Ross, A.W., Bell, L.M., Littlewood, P.A., Mercer, J.G., Barrett, P., Morgan, P.J., 2005.
a seasonal interval timer. J. Biol. Rhythms 19, 298–311. Temporal changes in gene expression in the arcuate nucleus precede seasonal
Prendergast, B.J., Flynn, A.K., Zucker, I., 2000a. Triggering of neuroendocrine responses in adiposity and reproduction. Endocrinology 146, 1940–1947.
refractoriness to short-day patterns of melatonin in Siberian hamsters. Rostal, D.C., Glick, B.B., Eaton, G.G., Resko, J.A., 1986. Seasonality of adult male
J. Neuroendocrinol. 12, 303–310. Japanese macaques (Macaca fuscata): androgens and behavior in a confined troop.
Prendergast, B.J., Gorman, M.R., Zucker, I., 2000b. Establishment and persistence of Horm. Behav. 20, 452–462.
photoperiodic memory in hamsters. Proc. Natl. Acad. Sci. U.S.A. 97, 5586–5591. Ruby, N.F., Ibuka, N., Barnes, B.M., Zucker, I., 1989. Suprachiasmatic nuclei influence
Prendergast, B.J., Hotchkiss, A.K., Bilbo, S.D., Kinsey, S.G., Nelson, R.J., 2003. torpor and circadian temperature rhythms in hamsters. Am. J. Physiol. 257,
Photoperiodic adjustments in immune function protect Siberian hamsters from R210–R215.
lethal endotoxemia. J. Biol. Rhythms 18, 51–62. Ruf, T., Korytko, A.I., Stieglitz, A., Lavenburg, K.R., Blank, J.L., 1997. Phenotypic
Prendergast, B.J., Hotchkiss, A.K., Wen, J., Horton, T.H., Nelson, R.J., 2006. Refractoriness variation in seasonal adjustments of testis size, body weight, and food intake in
to short day lengths augments tonic and gonadotrophin-releasing hormone-stimulated deer mice: role of pineal function and ambient temperature. J. Comp. Physiol. B
lutenising hormone secretion. J. Neuroendocrinol. 18, 339–348. 167, 185–192.
Prendergast, B.J., Kriegsfeld, L.J., Nelson, R.J., 2001. Photoperiodic polyphenisms in Rusak, B., Morin, L.P., 1976. Testicular responses to photoperiod are blocked by lesions
rodents: neuroendocrine mechanisms, costs and functions. Q. Rev. Biol. 76, of the suprachiasmatic nuclei in golden hamsters. Biol. Reprod. 15, 366–374.
293–325. Sadleir, R.M.F.S., 1969. The Ecology of Reproduction in Wild and Domestic Mammals.
Prendergast, B.J., Mosinger Jr., B., Kolattukudy, P.E., Nelson, R.J., 2002a. Hypo- Methuen, London.
thalamic gene expression in reproductively photoresponsive and photorefractory Saenz de Miera, C., Monecke, S., Bartzen-Sprauer, J., Laran-Chich, M.P., Pevet, P.,
Siberian hamsters. Proc. Natl. Acad. Sci. U.S.A. 99, 16291–16296. Hazlerigg, D.G., Simonneaux, V., 2014. A circannual clock drives expression of
Prendergast, B.J., Nelson, R.J., Zucker, I., 2002b. Mammalian seasonal rhythms: gene central for seasonal reproduction. Curr. Biol. 24, 1500–1506.
behavior and neuroendocrine substrates. In: Pfaff, D.W., et al. (Eds.), Hormones, Sandweiss, D.H., Maasch, K.A., Anderson, D.G., 1999. Climate and culture – transitions
Brain, and Behavior. Academic Press, San Diego, pp. 93–156. in the mid-Holocene. Science 283, 499–500.
Prendergast, B.J., Nelson, R.J., Zucker, I., 2009. Mammalian seasonal rhythms: Scheer, F.A.J.L., Hilton, M.F., Mantzoros, C.S., Shea, S.A., 2009. Adverse metabolic
behavior and neuroendocrine substrates. In: Pfaff, D.W., Arnold, A.P., Etgen, A.M., and cardiovascular consequences of circadian misalignment. Proc. Natl. Acad. Sci.
Fahrbach, S.E., Rubin, R.T. (Eds.), Hormones, Brain and Behavior, second ed. U.S.A. 106, 4453–4458.
Academic Press, San Diego, pp. 507–540. Schlatt, S., Niklowitz, P., Hoffmann, K., Nieschlag, E., 1993. Influence of short photo-
Prendergast, B.J., 2010. MT1 melatonin receptors mediate somatic, behavioral and periods on reproductive organs and estrous cycles of normal and pinealectomized
reproductive neuroendocrine responses to photoperiod and melatonin in Siberian female Djungarian hamsters, Phodopus sungorus. Biol. Reprod. 49, 243–250.
hamsters (Phodopus sungorus). Endocrinology 151, 714–721. Scotti, M.A.L., Place, N.J., Demas, G.E., 2007. Short-day increases in aggression are
Proctor, J.G., Blackhurst, D.W., Boone, W.R., 2004. Does seasonality alter intrauterine independent of circulating gonadal steroids in female Siberian hamsters (Phodopus
insemination outcomes: a 5 year study. J. Assisted Reprod. Genet. 21, 263–270. sungorus). Horm. Behav. 52, 183–190.
Pucek, Z., 1965. Water contents and seasonal changes of the brain weight in shrews. Seegal, R.F., Goldman, B.D., 1975. Effects of photoperiod on cyclicity and serum
Acta Theriol. 10, 353–367. gonadotropins in the Syrian hamster. Biol. Reprod. 12, 223–231.
Pyter, L.M., Reader, B.F., Nelson, R.J., 2005. Short photoperiods impair spatial Sheldon, B.C., Verhulst, S., 1996. Ecological immunology: costly parasite defences
learning and alter hippocampal dendritic morphology in adult male white-footed and trade-offs in evolutionary ecology. Trends Ecol. Evol. 11, 317–321.
mice (Peromyscus leucopus). J. Neurosci. 25, 4521–4526. Sinclair, J.A., Lochmiller, R.L., 2000. The winter immunoenhancement hypothesis:
Reiter, R.J., 1974. Influence of pinealectomy on the breeding capability of hamsters associations among immunity, density, and survival in prairie vole (Microtus
maintained under natural photoperiodic and temperature conditions. Neuroendo- ochrogaster) populations. Can. J. Zool. 78, 254–264.
crinology 13, 366–370. Smith, J.T., Clay, C.M., Caraty, A., Clarke, I.J., 2007. KiSS-1 messenger ribonucleic
Reiter, R.J., 1980. Photoperiod: its importance as an impeller of pineal and seasonal acid expression in the hypothalamus of the ewe is regulated by sex steroids and
reproductive rhythms. Int. J. Biometeorol. 24, 57–63. season. Endocrinology 148, 1150–1157.
Reiter, R.J., 1998. Melatonin and human reproduction. Annu. Med. 30, 103–108. Soma, K.K., Rendon, N.M., Boonstra, R., Albers, H.E., Demas, G.E., 2015. DHEA
Reiter, R.J., Vaughn, M.K., Blask, D.E., Johnson, L.Y., 1974. Melatonin: its inhibition of effects on brain and behavior: insights from comparative studies of aggression.
pineal antigonadotrophic activity in male hamsters. Science 185, 1169–1171. J. Steroid Biochem. Mol. Biol. 145, 261–272.
Rendon, N.M., Rudolph, L.M., Sengelaub, D.R., Demas, G.E., 2015. The agonistic Spears, N., Meyer, J.S., Whaling, C.S., Wade, G.N., Zucker, I., Dark, J., 1990. Long
adrenal: melatonin elicits female aggression via regulation of adrenal androgens. day lengths enhance myelination of midbrain and hindbrain regions of developing
Proc. R. Soc. B 282, 20152080. meadow voles. Dev. Brain Res. 55, 103–108.
Reppert, S.M., Weaver, D.R., Ebisawa, T., 1994. Cloning and characterization of Speth, J.D., 1987. Early hominid subsistence strategies in seasonal habitats.
a mammalian melatonin receptor that mediates reproductive and circadian J. Archaeol. Sci. 14, 13–30.
responses. Neuron 13, 1177–1185. Spurlock, M.E., 1997. Regulation of metabolism and growth during immune challenge:
Revel, F.G., Saboureau, M., Masson-Pevet, M., Pevet, P., Mikkelsen, J.D., an overview of cytokine function. J. Animal Sci. 75, 1773–1783.
Simonneaux, V., 2006. Kisspeptin mediates the photoperiodic control of repro- Steinlechner, S., Stieglitz, A., Ruf, T., Heldmaier, G., Reiter, R.J., 1991. Integration of
duction in hamsters. Curr. Biol. 16, 1730–1735. environmental signals by the pineal gland and its significance for seasonality in
Revel, F.G., Saboureau, M., Pevet, P., Simmoneaux, V., Mikkelsen, J.D., 2008. small mammals. In: Fraschini, F., Reiter, R.J. (Eds.), Role of Melatonin and Pineal
RFamide-related peptide gene is a melatonin driven photoperiodic gene. Endo- Peptides in Neuroimmunoregulation. Plenum Press, New York, pp. 159–163.
crinology 149, 902–912. Stetson, M.H., Ray, S.L., Creyaufmiller, N., Horton, T.H., 1989. Maternal transfer of
Revelli, A., La Sala, G.B., Gernnarelli, G., Scatigna, L., Racca, C., Massobrio, M., photoperiodic information in Siberian hamsters. II. The nature of the maternal
2005. Seasonality and human in vitro fertilization outcome. Gynecol. Endocrinol. signal, time of signal transfer, and the effect of the maternal signal on peripubertal
21, 12–17. reproductive development in the absence of photoperiodic inputs. Biol. Reprod. 40,
Riesen, J.W., Meyer, R.K., Wolf, R.C., 1971. The effect of season on occurrence of 458–465.
ovulation in the rhesus monkey. Biol. Reprod. 5, 111–114. Stevenson, T.J., Prendergast, B.J., 2013. Reversible DNA methylation regulates seasonal
Robinson, J.E., Wayne, N.L., Karsch, F.J., 1985. Refractoriness to inhibitory day photoperiodic time measurement. Proc. Natl. Acad. Sci. 110, 16651–16656.
lengths initiates the breeding season of the Suffolk ewe. Biol. Reprod. 32, Stevenson, T.J., Prendergast, B.J., 2015. Photoperiodic time measurement and
1024–1030. seasonal immunological plasticity. Front. Neuroendocrinol. 37, 76–88.
Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates 397

Stevenson, T.J., Onishi, K.G., Bradley, S.P., Prendergast, B.J., 2014. Cell-autonomous Watanabe, T., Yamamura, T., Watanabe, M., Yasuo, S., Nakao, N., Dawson, A.,
iodothyronine deiodinase expression mediates seasonal plasticity in immune Ebihara, S., Yoshimura, T., 2007. Hypothalamic expression of thyroid hormone-
function. Brain Behav. Immun. 36, 61–70. activating and -inactivating enzyme genes in relation to photorefractoriness in
Stevenson, T.J., Visser, M.E., Arnold, W., Parrett, P., Biello, S., Dawson, A., birds and mammals. Am. J. Physiol. 292, R568–R572.
Denlinger, D.L., Dominoni, D., Ebling, F.J., Elton, S., Evans, N., Ferguson, H.M., Watanabe, M., Yasuo, S., Watanabe, T., Yamamura, T., Nakao, N., Ebihara, S.,
Foster, R.G., Hau, M., Haydon, D.T., Hazlerigg, D.G., Heideman, P., Yoshimura, T., 2004. Photoperiodic regulation of type 2 deiodinase gene in
Hopcraft, J.G., Jonsson, N.N., Kronfeld-Schor, N., Kumar, V., Lincoln, G.A., Djungarian hamster: possible homologies between avian and mammalian photo-
MacLeod, R., Martin, S.A., Martinez-Bakker, M., Nelson, R.J., Reed, T., periodic regulation of reproduction. Endocrinology 145, 1546–1549.
Robinson, J.E., Rock, D., Schwartz, W.J., Steffan-Dewenter, I., Tauber, E., Watts, D.P., 1998. Seasonality in the ecology and life histories of mountain gorillas
Thackeray, S.J., Umstatter, C., Yoshimura, T., Helm, B., 2015. Disrupted (Gorilla gorilla beringei). Int. J. Primatol. 19, 929–948.
seasonal biology impacts health, food security and ecosystems. Proc. R. Soc. B Weaver, D.R., Reppert, S.M., 1990. Melatonin receptors are present in the ferret pars
282, 1–10. tuberalis and pars distalis, but not in brain. Endocrinology 127, 2607–2609.
Stewart, K.M., 1994. Early hominid utilisation of fish resources and implications for Weaver, D.R., Liu, C., Reppert, S.M., 1996. Nature’s knockout: the Mel1b receptor is
seasonality and behaviour. J. Hum. Evol. 27, 229–245. not necessary for reproductive and circadian responses to melatonin in Siberian
Struhsaker, T.T., 1997. Ecology of an African Rain Forest: Logging in Kibale and the hamsters. Mol. Endocrinol. 10, 1478–1487.
Conflict between Conservation and Exploitation. University Press of Florida, Weaver, D.R., Rivkees, S.A., Reppert, S.M., 1989. Localization and characterization of
Gainesville. melatonin receptors in rodent brain by in vitro autoradiography. J. Neurosci. 9,
van Schaik, C.P., Terborgh, J.W., Wright, S.J., 1993. The phenology of tropical 2581–2590.
forests – adaptive significance and consequences for primary consumers. Annu. Weems, P.W., Goodman, R.L., Lehmann, M.N., 2015. Neural mechanisms controlling
Rev. Ecol. Syst. 24, 353–377. seasonal reproduction: principles derived from the sheep model and its comparison
van Schaik, C.P., Brockman, D.K., 2005. Seasonality in primate ecology, reproduction, with hamsters. Front. Neuroendocrinol. 37, 43–51.
and life history: an overview. In: Brockman, D.K., van Schaik, C.P. (Eds.), Sea- Webb, T., 1993. Vegetation, lake levels, and climate in eastern North America for the
sonality in Primates: Studies of Living and Extinct Human and Non-human past 18,000 years. In: Wright, H.E., Kutzbach, J.E., Webb, T., Ruddiman, W.F.,
Primates. Cambridge University Press, Cambridge, UK, pp. 3–20. Street-Perrott, F.A., Bartlein, P.J. (Eds.), Global Climates since the Last Glacial
Takahashi, J.S., 2004. Finding new clock components: past and future. J. Biol. Maximum. University of Minnesota Press, Minneapolis, USA, pp. 415–467.
Rhythms 19, 339–347. Wehr, T.A., 1997. Melatonin and seasonal rhythms. J. Biol. Rhythms 12, 518–527.
Tamarkin, L., Hutchison, J.S., Goldman, B.D., 1976a. Regulation of serum gonado- Weidenfeld, J., Siegel, R.A., Corcos, A.P., Heled, V., Conforti, N., Chowers, I., 1984.
tropins by photoperiod and testicular hormone in the Syrian hamster. Endocrinology ACTH and corticosterone secretion following 2-deoxyglucose administration in
99, 1528–1533. intact and in hypothalamic deafferentated male rats. Brain Res. 305, 109–113.
Tamarkin, L., Westrom, W.K., Hamill, A.I., Goldman, B.D., 1976b. Effect of melatonin Weil, Z.M., Pyter, L.M., Martin, L.B., Nelson, R.J., 2006. Perinatal photoperiod
on the reproductive systems of male and female Syrian hamsters: a diurnal rhythm organizes adult immune responses in Siberian hamsters (Phodopus sungorus).
in sensitivity to melatonin. Endocrinology 99, 1534–1541. Am. J. Physiol. 290, R1714–R1719.
Tamarkin, L., Lefebvre, N.G., Hollister, C.W., Goldman, B.D., 1977. Effect of melatonin Weil, Z.M., Workman, J.L., Nelson, R.J., 2007. Housing condition alters immunological
administered during the night on reproductive function in the Syrian hamster. and reproductive responses to day length in Siberian hamsters (Phodopus sun-
Endocrinology 101, 631–634. gorus). Horm. Behav. 52, 261–266.
Teubner, B.J., Freeman, D.A., 2007. Different neural melatonin-target tissues are Weil, Z.M., Borniger, J.C., Cisse, Y.M., Abi Salloum, B.A., Nelson, R.J., 2015.
critical for encoding and retrieving day length information in Siberian hamsters. Neuroendocrine control of photoperiodic changes in immune function. Front.
J. Neuroendocrinol. 19, 102–108. Neuroendocrinol. 37, 108–118.
Trainor, B.C., Lin, S., Finy, M.S., Rowland, M.R., Nelson, R.J., 2007. Photoperiod West, S.D., Dublin, H.T., 1984. Behavioral strategies of small mammals under winter
reverses the effects of estrogens on male aggression via genomic and nongenomic conditions solitary or social. Carnegie Mus. Nat. Hist. Special Publ. 10, 293–300.
pathways. Proc. Natl. Acad. Sci. U.S.A. 104, 9840–9845. Whitsett, J.M., Lawton, A.D., 1982. Social stimulation of reproductive development in
Trauth, M.H., Maslin, M.A., Deino, A., Strecker, M.R., 2005. Late Cenozoic moisture male deer mice housed on a short-day photoperiod. J. Comp. Physiol. Psychol. 96,
history of East Africa. Science 309, 2051–2053. 416–422.
Turner, B.N., Perrin, M.R., Iverson, S.L., 1975. Winter coexistence of voles in spruce Wickings, E.J., Nieschlag, E., 1980. Seasonality in endocrine and exocrine testicular
forest: relevance of seasonal changes in aggression. Can. J. Zool. 53, function of the adult rhesus monkey (Macaca mulatta) maintained in a controlled
1004–1011. laboratory environment. Int. J. Androl. 3, 87–104.
Ubuka, T., Inoue, K., Fukuda, Y., Mizuno, T., Ukena, K., Kriegsfeld, L.J., Tsutsui, K., Wilson, M.E., Pope, N.S., Gordon, T.P., 1987. Seasonal modulation of luteinizing-
2012. Identification, expression and physiological functions of Siberian hamster hormone secretion in female rhesus monkeys. Biol. Reprod. 36, 975–984.
gonadotropin-inhibitory hormone. Endocrinology 153, 373–385. Wingfield, J.C., 1994. Regulation of territorial behavior in the sedentary song sparrow,
Urbanski, H.F., Doan, A., Pierce, M., 1991. Immunocytochemical investigation of Melospiza melodia morphna. Horm. Behav. 28, 1–15.
luteinizing hormone-releasing hormone neurons in Syrian hamsters maintained Wingfield, J.C., Soma, K.K., 2002. Spring and autumn territoriality in song sparrows:
under long or short days. Biol. Reprod. 44, 687–692. same behavior, different mechanisms? Integr. Comp. Biol. 42, 11–20.
Vasudevan, N., Pfaff, D.W., 2007. Membrane-initiated actions of estrogens in Wood, R.I., 1996. Estradiol, but not dihydrotestosterone, in the medial amygdala
neuroendocrinology: emerging principles. Endocr. Rev. 28, 1–19. facilitates male hamster sex behavior. Physiol. Behav. 59, 833–841.
Vondrasova, D., Hajek, I., Illnerova, H., 1997. Exposure to long summer days affects Wood, S., Loudon, A., 2014. Clocks for all seasons: unwinding the roles and
the human melatonin and cortisol rhythms. Brain Res. 759, 166–170. mechanisms of circadian and interval timers in the hypothalamus and pituitary.
Vriend, J., 1983. Evidence for pineal gland modulation of the neuroendocrine-thyroid J. Endocrinol. 222, R39–R59.
axis. Neuroendocrinology 36, 68–78. Woodfill, C.J., Robinson, J.E., Malpaux, B., Karsch, F.J., 1991. Synchronization of the
Vriend, J., Wasserman, R.A., 1986. Effects of afternoon injections of melatonin in circannual reproductive rhythm of the ewe by discrete photoperiodic signals. Biol.
hypothyroid male Syrian hamsters. Neuroendocrinology 42, 498–503. Reprod. 45, 110–121.
Waage, A., Brandtzaeg, P., Halstensen, A., Kierulf, P., Espevik, T., 1989. The complex Woodfill, C.J., Wayne, N.L., Moenter, S.M., Karsch, F.J., 1994. Photoperiodic
pattern of cytokines in serum from patients with meningococcal septic shock. synchronization of a circannual reproductive rhythm in sheep: identification of
Association between interleukin 6, interleukin 1, and fatal outcome. J. Exp. Med. season-specific time cues. Biol. Reprod. 50, 965–976.
169, 333–338. Worth, R.W., Charlton, H.M., Mackinnon, P.C., 1973. Field and laboratory studies on
Walkden-Brown, S.W., Martin, G.B., Restall, B.J., 1999. Role of male-female inter- the control of luteinizing hormone secretion and gonadal activity in the vole,
action in regulating reproduction in sheep and goats. J. Reprod. Fertil. Suppl. 154, Microtus agrestis. J. Reprod. Fertil. 19, 89–99.
243–257. Xu, Z.Z., McDonald, M.F., McCutcheon, S.N., Blair, H.T., 1992. Effects of season and
Walton, J.C., Chen, Z., Weil, Z.M., Pyter, L.M., Travers, J.B., Nelson, R.J., 2011. testosterone treatment on gonadotrophin secretion and pituitary responsiveness to
Photoperiod-mediated impairment of long-term potention and learning and memory gonadotrophin-releasing hormone in castrated Romney and Poll Dorset rams.
in male white-footed mice. Neuroscience 175, 127–132. J. Reprod. Fertil. 95, 183–190.
Walton, J.C., Pyter, L.M., Weil, Z.M., Nelson, R.J., 2012. Photoperiod mediated Yamamura, T., Hirunagi, K., Ebihara, S., Yoshimura, T., 2004. Seasonal morphological
changes in olfactory bulb neurogenesis and olfactory behavior in male white-footed changes in the neuro-glial interaction between gonadotropin-releasing hormone nerve
mice. PLoS One 7, e42743. terminals and glial endfeet in Japanese quail. Endocrinology 145, 4264–4267.
398 Mammalian Seasonal Rhythms: Behavior and Neuroendocrine Substrates

Yaskin, V.A., 1984. Seasonal changes in brain morphology in small mammals. Car- Zucker, I., 1985. Pineal gland influences period of circannual rhythms of ground
negie Mus. Nat. Hist. Special Publ. 10, 183–192. squirrels. Am. J. Physiol. 249, R111–R115.
Yasuo, S., Yoshimura, T., Ebihara, S., Korf, H.W., 2007. Temporal dynamics of type 2 Zucker, I., 1988. Seasonal affective disorders: animal models non fingo. J. Biol.
deiodinase expression after melatonin injections in Syrian hamsters. Endocrinology Rhythms 3, 209–223.
148, 4385–4392. Zucker, I., 2001. Circannual rhythms: mammals. In: Takahashi, J., Turek, F.W.,
Yasuo, S., Yoshimura, T., Ebihara, S., Korf, H.W., 2009. Melatonin transmits photope- Moore, R.Y. (Eds.), Handbook of Behavioral Neurobiology, Circadian Clocks, vol. 12.
riodic signals through the MT2 melatonin receptor. J. Neurosci. 29, 2885–2889. Kluwer Academic/Plenum, New York, pp. 509–528.
Yellon, S.M., 1994. Effects of photoperiod on reproduction and the gonadotropin- Zucker, I., Boshes, M., Dark, J., 1983. Suprachiasmatic nuclei influence cir-
releasing hormone-immunoreactive neuron system in the postpubertal male cannual and circadian rhythms of ground squirrels. Am. J. Physiol. 244,
Djungarian hamster. Biol. Reprod. 50, 368–372. R472–R480.
Yellon, S.M., Fagoaga, O.R., Nehlsen-Cannarella, S.L., 1999. Influence of photoperiod Zucker, I., Lee, T.M., Dark, J., 1991. The suprachiasmatic nucleus and annual rhythms
on immune cell functions in the male Siberian hamster. Am. J. Physiol. 276, of mammals. In: Klein, D.C., Moore, R.Y., Reppert, S.M. (Eds.), The Suprachiasmatic
R97–R102. Nucleus: The Mind’s Clock. Oxford University Press, New York, pp. 246–259.
Ziegler, T., Hodges, K., Winkler, P., Heistermann, M., 2000. Hormonal correlates of Zysling, D.A., Demas, G.E., 2007. Metabolic stress suppresses humoral immune
reproductive seasonality in wild female Hanuman langurs (Presbytis entellus). Am. function in long-day, but not short-day, Siberian hamsters (Phodopus sungorus).
J. Primatol. 51, 119–134. J. Comp. Physiol. B 177, 339–347.
1.14 Sex Steroids, Learning and Memory
SE Nielsen and AY Herrera, University of Southern California, Los Angeles, CA, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by G. Dohanich, D. Korol, T. Shors, volume 1, pp. 539–578, Ó 2009, Elsevier Inc.

1.14.1 Gonadal Steroids, Learning and Memory 400


1.14.1.1 History 400
1.14.1.2 Brief Introduction into Ovarian Hormones and the Hypothalamic–Pituitary–Gonadal Axis 400
1.14.1.3 Ovarian Hormone Cycling 401
1.14.1.3.1 Rodents and the Estrous Cycle 401
1.14.1.3.2 Humans and the Menstrual Cycle 401
1.14.1.3.3 Birth Control and the HPG Axis 401
1.14.1.3.4 Summary 402
1.14.1.4 Ovarian Steroids and Working Memory 402
1.14.1.4.1 Ovarian Hormones and Working Memory Paradigms in Animals 402
1.14.1.4.2 Ovarian Hormones and Working Memory in Humans 403
1.14.1.4.3 Ovarian Hormones and Working Memory in Postmenopausal Women 403
1.14.1.4 Summary 404
1.14.1.5 Ovarian Steroids and Reference Memory in Animals 404
1.14.1.5.1 Water and Radial Arm Mazes 404
1.14.1.6 Ovarian Steroids and Spatial Learning in Humans 406
1.14.1.6.1 Influence of Ovarian Steroids on Spatial Learning in Women 406
1.14.1.6.2 Sex Differences in Spatial Learning in Humans 406
1.14.1.7 Ovarian Steroids and Associated Learning in Animals 406
1.14.1.7.1 Avoidance Learning and Ovarian Steroids 406
1.14.1.7.2 Stress and Conditioning Paradigms 407
1.14.1.8 Summary 407
1.14.2 Mechanisms of Steroid Action 407
1.14.2.1 Cellular Mechanisms of Steroid Action 407
1.14.2.2 Effects of Ovarian Steroids on Electrical Properties 407
1.14.2.3 A Model of Estrogen Action 408
1.14.2.3.1 Estrogen Receptor Subtypes 408
1.14.2.3.2 Estrogen as a Neuroprotective Steroid 409
1.14.2.4 A Model of Progesterone Action 409
1.14.2.4.1 Progesterone and the Stress Response 409
1.14.2.4.2 Progesterone Action and Cognition 410
1.14.3 Interaction between Steroid Hormones, Learning and Memory 410
1.14.3.1 Overview 410
1.14.3.2 Glucocorticoids, Learning and Memory 410
1.14.3.2.1 Factors Influencing the Relationship between Stress and Learning 410
1.14.3.2.2 Impairing Effects of Stress on Learning and Memory in Animals 410
1.14.3.2.3 Impairing Effects of Stress on Learning and Memory in Humans 411
1.14.3.2.4 Sex Steroid Hormones and Stress Response Interactions 411
1.14.3.3 Summary 412
1.14.3.4 Steroid Hormone Interactions in Learning and Memory of Arousing Information 412
1.14.3.4.1 Ovarian Steroids, Stress and Emotional Memory in Animals 412
1.14.3.4.2 Ovarian Steroids and Emotional Memory in Humans 412
1.14.3.4.3 Sex Differences in Emotional Memory Paradigms 413
1.14.3.4.4 Shifts in Emotional Memory across the Menstrual Cycle 413
1.14.3.4.5 Hormonal Contraceptive Use and Emotional Memory 415
1.14.4 Ovarian Steroids and Clinical Implications for Human Neuropathologies 415
1.14.4.1 Overview 415
1.14.4.2 Gonadal Steroids and AD 415
1.14.4.3 Gonadal Steroids and Disorders of Emotional Memory 416
References 416

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00014-6 399


400 Sex Steroids, Learning and Memory

Glossary
Alzheimer’s disease The most common form of dementia Hippocampus Located in the medial temporal lobe of the
that leads to issues with memory, cognition, and behavior. brain and plays an important role in consolidation of
The disease worsens over time, and although the greatest information from short-term to long-term memory.
risk factor is increasing age, up to 5% of people with the Hormonal contraception A form of birth control
disease have early onset Alzheimer’s. composed of synthetic steroid hormones that acts on the
Amygdala Almond-shaped structure located in the medial endocrine system.
temporal lobe of the brain and has been shown to be critical Reference memory Knowledge for aspects of a task,
for processing emotions, fear responses, and pleasure. typically spatial, that remain constant between trials.
Emotional memory Storage and recall of events and details Working memory Short-term memory that allows for
that are coupled with physiological responses present when keeping information ‘online’ and readily available for
the event occurred. access.

1.14.1 Gonadal Steroids, Learning and Memory To better understand how ovarian hormones might affect
the onset and/or treatment of neurological disorders, many
1.14.1.1 History
learning and memory researchers have focused their work on
Evidence collected over the past 25 years has shown there are how these hormones might alter the underlying neurobiology
profound sex differences in learning and memory. Research of different memory processes. The multiple memory systems
with both animals and humans indicates that these sex approach (Sherry and Schacter, 1987) accounts for differential
differences are mediated in large part by ovarian steroid effects of ovarian hormones on certain forms of memory,
hormones, namely estradiol and progesterone. Work demon- including working memory, reference memory, and even
strating these effects of sex steroids on learning and memory emotional memory. As we explore here, ovarian hormones
was largely prompted by reports showing that estradiol sometimes have impairing or enhancing effects on learning
promoted neurogenesis in the dentate gyrus of the hippo- and memory, or even no effect at all, depending on the type
campus and subventricular zone (Fowler et al., 2008). In of memory being tested.
these early studies, the highest number of new cells in the In this chapter, we will explore the physiological mecha-
dentate gyrus of female rats was identified during proestrus, nisms underlying steroid hormone production and how steroid
when there is a significant surge in estradiol levels (Tanapat hormones influence different types of learning and memory in
et al., 1999). Moreover, ovariectomy decreases neurogenesis both animals and humans. We will also provide information
while estradiol replacement restores it (Tanapat et al., about how hormone manipulations, such as hormonal contra-
1999). Similarly, when female meadow voles receive acute ception in humans, can alter learning and memory functions in
treatment with estradiol, they also show a transient increase adults. Finally, we will address the implications of steroid
in the number of new neurons in the dentate gyrus hormone effects on learning and memory in the clinical setting.
(Ormerod and Galea, 2001), suggesting that estradiol may
facilitate neurogenesis. These early studies of hormones
1.14.1.2 Brief Introduction into Ovarian Hormones and the
and neurogenesis promoted future research on the relation-
Hypothalamic–Pituitary–Gonadal Axis
ship between ovarian hormones and learning and memory
processes in both animal and human models. Additionally, The hypothalamic–pituitary–gonadal (HPG) axis is primarily
the early ovarian hormone work encouraged the exploration responsible for regulating reproductive activity and the release
of sex differences in the underlying neurobiology of learning of ovarian hormones in animals and humans (Couse et al.,
and memory, particularly with regard to brain structures such 2003; Meethal and Atwood, 2005). In this way, the HPG axis
as the amygdala and hippocampus (for review, see Andreano also plays a key role in promoting healthy brain function, as
and Cahill, 2009). ovarian hormones exert neuroprotective effects and facilitate
Research has also focused on how ovarian hormones may neurogenesis, neuronal differentiation and survival, and cogni-
play a role in the onset of different neurological disorders, tive function (Blair et al., 2015).
such as Alzheimer’s disease (AD), a common type of dementia The HPG axis is responsible for orchestrating the release
in older adults (Paganini-Hill and Henderson, 1994) and of both centrally and peripherally produced ovarian
disorders of emotional memory, such as anxiety, depression, hormones. Centrally produced regulatory hormones include
and posttraumatic stress disorder (PTSD). For example, early gonadotropin-releasing hormone (GnRH) from the hypothal-
studies in the 1990s provided evidence that hormone replace- amus and gonadotropins from the pituitary, specifically lutei-
ment therapy (HT) in older women may offset the risk of devel- nizing hormone (LH) and follicle-stimulating hormone
oping AD (Paganini-Hill and Henderson, 1994). However, (FSH) (Meethal and Atwood, 2005). Recently, researchers iden-
follow-up studies have had mixed findings, indicating that tified a central regulator of GnRH; kisspeptin was identified as
more work is still needed for researchers to truly elucidate the a neuromodulator that modulates GnRH release and subse-
relationship between ovarian hormones and neurological quently, controls activity of the gonadotropins and the HPG
disorders. axis (Skorupskaite et al., 2014). The other gonadotropins,
Sex Steroids, Learning and Memory 401

LH and FSH, are instrumental in modulating behavioral and the luteal phase. These phases can be further subdivided into
neuronal changes (Meethal and Atwood, 2005). early follicular, late follicular and midluteal based on notable
Receptors for estradiol, progesterone, and testosterone are changes in the hormonal milieu.
widely distributed throughout the brain and are highly concen- The onset of menstruation marks the first day of the cycle
trated in regions outside of the hypothalamus and pituitary, and this period lasts 4–5 days on average. The menstrual cycle
such as the amygdala and cerebral cortex (Meethal and begins with the follicular phase, which lasts from the first day
Atwood, 2005). Additionally, estradiol is locally generated of menstruation until ovulation (days 1–13) (Reilly, 2000).
within the hippocampus (Mukai et al., 2006), and levels of Throughout menstruation, estradiol, progesterone, and LH
estradiol in this region can fluctuate based on circulating levels circulate at relatively low levels while FSH levels are slightly
(Barker and Galea, 2009). Since estradiol is generated in the elevated relative to baseline in order to stimulate the develop-
hippocampus, it not only has an effect on the negative- ment of follicles. After menstruation, the generated follicles
feedback loop regulating HPG axis activity, but estradiol, as begin to secrete estradiol, gradually increasing the levels of
well as progesterone and testosterone, can also have effects this hormone during the early follicular phase (days 1–6).
on different neuronal pathways (McEwen, 2002). In addition Simultaneously, one follicle develops into the dominant
to ovarian hormones, there are peripherally produced protein follicle destined for ovulation; this follicle typically has
complexes that are responsible for regulating the activity of a high concentration of FSH receptors.
the HPG axis. For example, activins and inhibins are dimeric In the latter half of the follicular phase (days 7–13),
paracrine protein complexes with opposing physiological increasing levels of estradiol facilitate a gradual decrease of
effects on FSH secretion; activins are produced in all tissues, FSH levels as estradiol inhibits FSH production. Simulta-
whereas inhibins are generated in the gonads (Meethal and neously, the dominant follicle outcompetes other follicles for
Atwood, 2005). Activins are critical for stimulating FSH secre- the remaining FSH, thereby maturing and secreting increasing
tion, activating the HPG axis, and facilitating neuronal plas- amounts of estradiol. Between days 10 and 13, there is
ticity, whereas inhibins are responsible for inhibiting FSH a preovulatory surge of estradiol. Concurrently, the pituitary
secretion and inactivating and modulating the effects of acti- initiates positive feedback, resulting in a preovulatory surge
vins. A third protein complex, follistatin, also works with of FSH and LH and subsequently, ovulation. Ovulation usually
inhibin to regulate the activin activity. Together, these protein occurs on day 14 of the menstrual cycle, and at this point, the
complexes help regulate the onset and offset of HPG axis mature dominant follicle forces out the oocyte into the perito-
activity (Meethal and Atwood, 2005) in coordination with neal cavity. During ovulation, LH, FSH, and estradiol decrease
circulating levels of the ovarian hormones. to levels slightly above those observed during menstruation,
and FSH and LH circulate at low levels for the remainder of
the menstrual cycle.
1.14.1.3 Ovarian Hormone Cycling
The second half of the menstrual cycle is known as the luteal
Given that this chapter will focus on the influences of ovarian phase (days 15–28), which is initiated by the LH surge and is
hormones on learning and memory in rodents and humans, characterized by the luteinization of follicle elements that
only the ovarian hormone cycling in these species will be dis- were not ovulated. Different cells and tissues, namely the gran-
cussed. The following section addresses ovarian hormone ulosa cells, theca cells, and connective tissue, are transformed
cycling in females of both the rodent and human species. into the corpus luteum, which is responsible for secreting
progesterone and estradiol. Progesterone and estradiol secre-
1.14.1.3.1 Rodents and the Estrous Cycle tion by the corpus luteum typically peaks between days 22
In rodents, there are four phases in the estrous cycle, and the and 24 of the menstrual cycle. Notably, progesterone levels
average cycle length is 4–5 days. In metestrus (day 1), proges- in the menstrual cycle reach their highest peak during this mid-
terone and estradiol circulate at relatively low levels. The luteal phase. Estradiol levels, while elevated, are lower than the
following day (day 2), diestrus ensues, and early on in this levels observed during the preovulatory surge. At day 24 of the
postovulation phase, progesterone increases sharply and then cycle, the corpus luteum starts to evolve into the corpus albi-
drops quickly by the end of day (Butcher et al., 1974). At cans and no longer secretes estradiol and progesterone. The
1200 h on day 3, proestrus is initiated by a significant surge decrease in estradiol and progesterone levels facilitates an
in estradiol levels. This marked increase in estradiol causes increase in FSH levels and subsequently, the initiation of the
a rapid peak of LH and FSH between 1600 and 1800 h as next menstrual cycle.
well as an increased progesterone secretion. Finally, in estrus,
ovulation occurs and all hormones return to baseline, save 1.14.1.3.3 Birth Control and the HPG Axis
for a temporary peak in estradiol in the evening of estrus on The menstrual cycle can be manipulated and regulated by
day 4 (Butcher et al., 1974). synthetic analogs of estradiol and progesterone. To better
understand how hormone manipulations such as hormonal
1.14.1.3.2 Humans and the Menstrual Cycle contraception can alter learning and memory processes in
The average menstrual cycle in women lasts 28 days, though it women, it is important to know how hormonal contraception
can vary between 23 and 38 days depending on the individual. affects HPG axis activity. These synthetic hormone formula-
Most women have monthly menstrual cycling starting at tions have become a widely used and well-known form of
menarche around the age of 11–13 years old until the onset reversible contraception for women. Since its introduction in
of menopause (Reilly, 2000). Usually, the menstrual cycle 1960, hormonal contraception has undergone many changes
consists of three phases: the follicular phase, ovulation, and to its synthetic hormone elements and dosage as well as the
402 Sex Steroids, Learning and Memory

method of administration. In spite of these alterations, 1.14.1.4 Ovarian Steroids and Working Memory
however, the original utility of the drugs has been preserved.
Working memory is considered a critical cognitive function
The most common method of hormonal contraception is
(Janowsky et al., 2000) and is classified as a form of short-
known as a combination formulation; here, ethinyl estradiol
term memory in which information is stored for the period
and a synthetic progestin work synergistically to prevent ovula-
of time that it remains useful and relevant. Retention intervals
tion by inhibiting different components of the HPG axis (Lobo
for working memory experiments vary from seconds to hours,
and Stanczyk, 1994). Although interference at either the hypo-
depending on the parameters of the task as defined by the
thalamus or the pituitary is thought to be sufficient to prevent
experimenter; working memory load can be further intensified
ovulation (Lobo and Stanczyk, 1994; Rapkin et al., 2006),
by increasing the quantity or complexity of information. This
combined contraceptive formulations target both regions to
section will cover how steroid hormones modulate working
ensure maximum effectiveness. It is well established that
memory in rodents and humans.
ethinyl estradiol/progestin contraceptives utilize negative feed-
back to the hypothalamus to inhibit the midcycle GnRH surge
(Lobo and Stanczyk, 1994). In addition, these synthetic 1.14.1.4.1 Ovarian Hormones and Working Memory Paradigms
hormones suppress the release of LH and FSH from the pitui- in Animals
tary to inhibit the gonadal production of endogenous estrogen 1.14.1.4.1.1 Land Mazes in Animals
and progesterone. Thus, hormonal contraception significantly In animals, estradiol has been shown to facilitate learning and
suppresses endogenous sex hormone levels in women, and memory processes. For example, if ovariectomized (OVX)
this hormone suppression has been shown to be involved in mammals were treated with estradiol, they made fewer working
different forms of learning and memory processes in women memory errors during a radial arm maze than OVX-females not
(Warren et al., 2014). treated with estradiol (Daniel et al., 1997; Bimonte and
Denenberg, 1999; Fader et al., 1999). Likewise, females
1.14.1.3.4 Summary receiving only a sham surgery exhibited a greater number of
The HPG axis is responsible for the production of ovarian and consecutive correct choices on a radial arm maze, compared
testicular hormones. Additionally, these steroids are locally with OVX-alone females (Wilson et al., 1999).
produced in brain regions that are critical for learning and 17b-estradiol also modulates working memory perfor-
memory, such as the hippocampus. Circulating levels of mance on radial arm mazes, even when maintained at low
ovarian hormone levels fluctuate throughout ovarian monthly levels. For example, subcutaneous implantation of continuous
cycling in female rodents and humans, and importantly, these release 17b-estradiol capsules decreased errors during the
levels can be further modified in humans by the use of acquisition of the radial arm maze in OVX-females (Daniel
hormonal contraceptives. In induced ovulators, that do not et al., 1997; Luine et al., 1998). Please refer to Figure 1.
have an estrous cycle (e.g., rabbits, musk shrews), ovulation This improvement in working memory performance was
is induced by mating-related sensory stimulation (Bakker and also observed in a food-reinforced T-maze; OVX-females
Baum, 2000). systemically treated with estradiol benzoate over days or

(a) 6.6 (b) 7.4

7.2
6.4
Correct choices in first 8 visits

7.0
Correct choices in first 8 visits

6.8 Estradiol
6.2 *
6.6

6.4
6.0
6.2

6.0
5.8
5.8 Cholesterol

5.6 5.6
1 2 3 4 5 6
E CH
4-day blocks

Figure 1 Effect of estradiol administered prior to and during training on a radial arm maze. Ovariectomized rats were implanted with 5-mm silastic
capsules containing 25% estradiol (E, n ¼ 14) or 100% cholesterol (CH, n ¼ 16) for 30 days prior to and continuing throughout 24 days of maze
training. (a) Mean number of correct arm choices in first eight visits (SEM) averaged over 24 days of training (*p < 0.01; ANOVA). (b) Mean
number of correct arm choices in the first eight visits (SEM) presented as 4-day blocks over 24 days of training. Reproduced from Daniel, J.M.,
Fader, A.J., Spencer, A.L., Dohanich, G.P., 1997. Estrogen enhances performance of female rats during acquisition of a radial arm maze. Horm.
Behav. 32, 215–225, with permission from Elsevier.
Sex Steroids, Learning and Memory 403

weeks improved performance on a nonmatching-to-position to those of an older woman postmenopause (Grigorova et al.,
maze (Fader et al., 1998), and those treated with 2006), and working memory performance also decreased
17b-estradiol improved on a matching-to-position task significantly as estradiol decreased.
(Gibbs et al., 1998). Other studies explored the effect of endogenous hormone
levels on working memory in young women. In a working
1.14.1.4.1.2 Water Mazes in Animals memory task with visual stimuli (i.e., faces), women performed
Benefits of estradiol also have been observed in other tests of better on the task during high-estradiol (late follicular and mid-
working memory, such as water mazes. In a delayed match- luteal) compared to the low-estradiol (early follicular) phases
to-place task, ovariectomized animals treated with estradiol (Vranic and Hromatko, 2008). However, this study was unable
benzoate displayed better retention for the location of a hidden to determine a direct effect of estradiol since the high-estradiol
platform compared to OVX-alone females; this performance group was comprised of two cycle phases with different
enhancement was stable across 10-, 30-, and 100-min delay hormone profiles. In particular, the high-estradiol group con-
intervals (Sandstrom and Williams, 2001). Likewise, sisted of the late follicular phase, where progesterone levels
estradiol-treated animals also required less time to reach are still very low but estradiol peaks to initiate ovulation, and
learning criteria of a delayed-match-to-position task than the midluteal phase, where estradiol is moderately high but
OVX-alone animals (Gibbs, 1999). progesterone is also reaching its peak levels. A more recent
study in naturally cycling women focused explicitly on estra-
1.14.1.4.1.3 Object Recognition and Placement Tasks in Animals diol; when salivary estradiol levels were elevated, women
The same pattern of estrogens improving working memory has committed fewer working memory errors on a spatial working
also been observed in an object recognition task; here, animals memory task compared to when their estradiol levels were low
were exposed to a total of four novel objects. They were first (Hampson and Morley, 2013). Additionally, estradiol nega-
allowed to explore a pair of two novel objects, and after tively correlated with a number of working memory errors.
some delay they were exposed to another two objects (one Although most studies have focused on the relationship
object from pair #1 and the third novel object); after a second between estradiol and working memory, some studies have
delay period the animals were exposed to two additional explored how progesterone may affect working memory perfor-
objects (one being the second object from pair #1 and the mance in women. These studies indicate that progesterone may
fourth novel object). Correct recognition was scored when have a negative effect on working memory performance in
the animal spent less time exploring the familiar object women (Man et al., 1999; Schmitt et al., 2005) given that
compared with time spent exploring the novel object. Ovariec- performance on working memory and short-term memory
tomy followed by 21 days of estradiol treatment enhanced tasks were worse in the midluteal relative to the follicular
retention for the first-tested object after a 3-h delay and of phase. However, effects of progesterone on working memory
the second-tested object after a 6-h delay, compared with in women remain relatively understudied and unclear at this
nonestradiol-treated mice (Vaucher et al., 2002). These data point.
suggest that in female animals, estradiol enhances working In summary, progesterone and estradiol both appear to
memory performance on a variety of different paradigms. modulate working memory performance in young women;
however, they may have opposing effects. A number of studies
1.14.1.4.2 Ovarian Hormones and Working Memory in Humans support a positive influence of estradiol on working memory,
1.14.1.4.2.1 Young Adults but more work is needed to determine the true role of proges-
Research on working memory in humans largely supports the terone. As research continues in this domain, researchers
findings from the animal literature. Several studies have found should focus on how hormone levels and activity in different
a positive relationship between estradiol and working memory brain regions relate to different kinds of working memory in
in young, healthy women. One study tested how performance women.
on a verbal span task varied over the course of the menstrual
cycle. Naturally cycling women were tested four times 1.14.1.4.3 Ovarian Hormones and Working Memory
throughout the menstrual cycle, and researchers found that in Postmenopausal Women
performance was enhanced at days 7 and 14 (high-estradiol 1.14.1.4.3.1 Endogenous Hormones
periods in a regular cycle) (Rosenberg and Park, 2002). Another Few studies have examined the cognitive effects of naturally
study indicated that women tested at menstruation (low estra- declining estradiol levels during menopause in the absence of
diol period) made significantly more working memory errors hormone replacement. One study showed that postmeno-
on the test of spatial working memory than age- and pausal women performed significantly worse than pre- and
education-matched women tested at other phases of the cycle perimenopausal women on delayed verbal memory tasks;
(Hampson and Moffat, 2004). These studies suggest that estra- additionally, these postmenopausal women performed signifi-
diol has a positive effect on working memory processes; cantly worse than perimenopausal women on phonemic verbal
however, each of these studies relied on self-report and failed fluency tasks (Weber et al., 2013).
to measure hormone levels in the women (Rosenberg and Another study showed no differences between women
Park, 2002; Hampson and Moffat, 2004). within 5 years of menopause and beyond 5 years of meno-
Later studies focused on assessing the correlational relation- pause on measures of sustained attention, category generation,
ship between hormone levels and working memory perfor- or episodic memory. However, higher estradiol levels in
mance and women. For example, when women were treated women within 5 years of menopause were associated with
with a GnRH analog for 4 weeks, estradiol levels were reduced better performance in measures of mental flexibility and
404 Sex Steroids, Learning and Memory

planning (i.e., executive function) compared with the steady Maki, 2013). In line with the critical window hypothesis,
low-estradiol levels in the women beyond 5 years of meno- Brinton (2005) proposed the healthy cell bias of estradiol
pause (Elsabagh et al., 2007; refer to Figure 2), an effect main- action as a mechanism for why such disparities exist in the
tained after controlling for age and IQ of women within and research canon (Brinton, 2005). Work in the Brinton lab sup-
beyond 5 years of menopause. Endogenous levels of estradiol ported the healthy cell bias theory, showing that introduction
in postmenopausal women not taking hormone supplements of estradiol after 5 days of beta-amyloid exposure exacerbated
also were positively correlated with performance on the beta-amyloid-induced damage, but estradiol pretreatment
Stroop task, another measure of executive function (Wolf and before beta-amyloid exposure protected the cells from damage
Kirschbaum, 2002). and death (Chen et al., 2006).

1.14.1.4.3.2 Exogenous Hormones and the Women’s Health


1.14.1.4 Summary
Initiative Studies
Postmenopausal women exhibit improved working memory Estradiol treatment after menopause appears to facilitate cogni-
performance with exogenous estradiol treatment. For example, tion, although certain limitations exist. The primary of these
when women received 2 weeks of transdermal estradiol treat- limitations appears to be the timing of treatment initiation.
ment, those experiencing higher endogenous estradiol levels When considering this point, treatment should be initiated as
in response to the dosage performed better on the delayed close to the final menses as possible and the patient should
recall portion of a verbal memory test than women experi- be in good health prior to beginning any treatment regimen.
encing lower dose-induced endogenous estradiol levels (Wolf
et al., 1999). Additionally, women randomly assigned to trans-
1.14.1.5 Ovarian Steroids and Reference Memory in Animals
dermal hormone replacement experienced improved perfor-
mance on the Stroop task, spatial working memory (Krug Reference memory refers to knowledge for aspects of a task that
et al., 2006; Baker et al., 2012), digit ordering (Krug et al., remain constant between trials. Rodent researchers often test
2006), and delayed recall (Baker et al., 2012). Similarly, reference memory in paradigms similar to those used to test
women already using hormone replacement also made fewer working memory (i.e., radial arm maze and Morris water
errors on nonspatial working memory (sequential memory of maze), but reference memory is distinguishable by its memory
an unfamiliar story, digit ordering, and digit span backward) consolidation processes. It is difficult to map reference memory
and spatial working memory tasks (Duff and Hampson, processes onto humans; however, it appears to resemble
2000), performed better on measures of verbal fluency and knowledge that is consistent across episodes rather than
working memory (Miller et al., 2002), and displayed better specific to one event (Nadel and Hardt, 2011). Thus, the
performance on a nonverbal memory task (Smith et al., following section will focus on how ovarian hormones modu-
2001) compared with women not taking any hormone late reference memory specifically in rodents.
supplements.
However, others report no effect (Maki et al., 2001; Smith 1.14.1.5.1 Water and Radial Arm Mazes
and Zubieta, 2001; Yaffe et al., 2006; Pefanco et al., 2007) or Reference memory is typically assessed using either water or
negative effects of estradiol replacement (Mulnard et al., radial arm maze. For the radial arm maze, studies will utilize
2000; Rapp et al., 2003; Shumaker et al., 2003; Espeland eight arms, four of which are baited on all trials while the other
et al., 2004; Resnick et al., 2006). The most widely publicized four arms are never baited (Hodges, 1996). When rodents enter
negative findings come from the Women’s Health Initiative a maze arm that is never baited, this is considered a reference
(WHI) studies. At the time of inception, the Women’s Health memory error (RME). In the case of the water maze, rodents
Initiative Memory Study (WHIMS) and Women’s Health Initia- learn that the submerged platform which represents an escape
tive Study of Cognitive Aging (WHISCA) were the largest is always in the same location (Hodges, 1996); here, an RME
randomized placebo-controlled trials testing claims that occurs when a rodent does not acquire the escape response.
hormone replacement after menopause maintained cognitive Studies have found that ovarian hormones modulate refer-
function otherwise observed to decline after menopause. ence memory differently at acquisition, consolidation, and
However, the WHIMS and WHISCA studies found that conju- retrieval. In the case of acquisition, high levels of estradiol
gated equine estrogens, a form of synthetic estrogen used in appear to be impairing. Studies have shown that elevated levels
some HTs, negatively affected performance on various cogni- of estradiol impair acquisition of the escape response in the
tive measures, including the California Verbal Learning Test Morris water maze (Galea et al., 1994, 1995, 2000; Frye,
(test of verbal retention and recall; Resnick et al., 2006) and 1995; Galea et al., 1995; Warren and Juraska, 1997). Proges-
the Modified Mini Mental State Exam (a measure of global terone also seems to have an impairing effect on acquisition
cognitive function; Rapp et al., 2003; Espeland et al., 2004), as the combination of estrogen and progesterone replacement
as well as increased risk of developing dementia (Shumaker in OVX-females impaired acquisition of the place and cued
et al., 2003). version of the Morris water maze (Chesler and Juraska, 2000).
In order to reconcile these reports of estradiol-induced Although gonadal hormones appear to have detrimental
cognitive impairment and basic science reports of estradiol- effects on acquisition in reference memory, there are mixed
related protection of the central nervous system (Pike, 1999; findings in how they affect consolidation and retrieval.
Brinton et al., 2000; Hosoda et al., 2001; Chen et al., 2006), For example, researchers found that posttraining injections
some have proposed a critical window for estradiol treatment of estrogen enhanced the consolidation and retention of
initiation (for review, see Resnick and Henderson, 2002; the Morris water maze (Packard and Teather, 1997a,b).
Sex Steroids, Learning and Memory 405

Figure 2 Executive function (scores are mean  SEM) in early and late postmenopausal stages. (a) In the IDED task: stages completed; extra-
dimensional shift (EDS) errors; trials to criterion; errors to criterion. (b) In the stockings of Cambridge planning task: number of moves; subse-
quent thinking time (ms). ***p < 0.001, **p < 0.01, *p < 0.05 significant difference between postmenopausal stages. Reproduced from
Elsabagh, S., Hartley, D.E., File, S.E., 2007. Cognitive function in late versus early postmenopause stage. Maturitas 56, 84–93, with permission
from Elsevier.
406 Sex Steroids, Learning and Memory

Other studies have found the complete opposite with both best (Hampson and Kimura, 1988), particularly when
endogenous and exogenous sex steroid hormones. For compared to midluteal women (Phillips and Silverman, 1997).
example, intact rats exhibited worse performance on a spatial
reference memory version of the Morris water maze during 1.14.1.6.2 Sex Differences in Spatial Learning in Humans
proestrus (high hormone) compared to estrus (low hormone) The impairing effect of estradiol on spatial learning in humans
(Warren and Juraska, 1997) or simply when levels of ovarian may account for the prevalent sex differences in this type of
hormones were high (Markham et al., 2002). Other work using learning. Early on, studies used two-dimensional maps to
exogenous estradiol treatment found that estradiol-treated assess spatial learning differences between men and women.
females made significantly more RMEs on a radial arm maze One study used a two-dimensional map of a fictional town
compared to the vehicle-treated animals (Galea et al., 2001). and found that males required fewer trials and made fewer
Thus, although there is some evidence to support enhancing errors when learning a new route compared to females;
effects of ovarian hormones on reference memory perfor- researchers also noted that men retained more information
mance, most of the findings indicate that estradiol, and likely about Euclidean geometric properties of the map, while
progesterone, has impairing effects on acquisition and retrieval females recalled more landmarks (Galea and Kimura, 1993).
of reference memory. Other work supported these findings by showing a significant
Biological sex can also be a mediating factor on reference male advantage for spatial route learning through an unfa-
memory performance. Across a number of studies, male miliar environment (Moffat et al., 1998). In this study, males
rodents are far superior to females in acquiring the reference learned a novel route through a simulated maze more quickly
memory component of the water-escape radial arm maze and accurately than females (Moffat et al., 1998).
(Gresack and Frick, 2003). Males tended to make fewer initial Subsequent studies expanded upon this work by using
RMEs and repeated RMEs compared to females (Williams virtual mazes. Some studies used a virtual water maze to assess
et al., 1990; Williams and Meck, 1991; LaBuda et al., 2002). sex differences in spatial learning. In these tests, men navigate
This sex difference in reference memory performance is likely to a hidden platform better than females (Astur et al., 1998,
linked to elevated levels of estradiol and progesterone in female 2004), and when availability of landmarks and room geometry
rodents compared with males. were manipulated, men were able to use either landmarks or
room geometry to find the platform, but women were impaired
when landmarks were removed (Sandstrom et al., 1998). Other
1.14.1.6 Ovarian Steroids and Spatial Learning in Humans
work supported this notion that women rely much more
Spatial learning and memory pertains to the ability to encode heavily on landmarks to navigate through a maze compared
and subsequently recall the spatial layout of a particular envi- to men (Andersen et al., 2012). Additionally, men tend to
ronment (Postma et al., 1999). As with other memory systems, explore more space initially in the virtual water maze compared
the hippocampus is a critical brain region for the encoding and to females, whereas females spend a greater amount of time
subsequent storage of relevant spatial information, evidenced fixating on one location (Mueller et al., 2008).
by the structure’s role in the construction of cognitive maps These data suggest that men utilize multiple strategies to
of space (O’keefe and Nadel, 1978). Given that this region is navigate new spaces and acquire knowledge about their space
modulated by ovarian hormones, the following section will more quickly than women. Sex differences in spatial learning
review the influence of ovarian hormones on the processing may be related to differences in estradiol, and possibly proges-
of spatial information and sex differences in spatial learning. terone and testosterone, levels at the time of learning. Given
that women perform better on spatial tasks when their ovarian
1.14.1.6.1 Influence of Ovarian Steroids on Spatial Learning in hormone levels are lower, it may be the case that low levels of
Women circulating estradiol and progesterone in males facilitate the
Although some studies have found no relation between encoding and memory of spatial information.
menstrual cycle phase in women and performance on spatial
tasks (Gordon et al., 1986; Ho et al., 1986; McBurney et al.,
1.14.1.7 Ovarian Steroids and Associated Learning
1997), some studies have found a relationship between cycle
in Animals
phase and spatial abilities that may be estradiol-mediated
(Silverman and Phillips, 1993). For example, on a spatial test The following section will address ovarian hormone effects on
battery (Rod and Frame, Space Relations, Hidden Figures), avoidance learning and how these hormones interact with
Hampson (1990a) found that women in the menstrual phase stress to alter learning processes in animals. Specifically, we
of the cycle (low estradiol and progesterone) performed better will focus on classical conditioning paradigms.
than women in the midluteal phase (higher estradiol and
progesterone). A within-subjects study using this paradigm 1.14.1.7.1 Avoidance Learning and Ovarian Steroids
found similar effects of phase in that women performed best There are two types of tasks that assess avoidance learning. In
on the spatial tasks when they were in their menstrual phase active avoidance tasks, rodents learn over multiple trials to
compared to the preovulatory phase (high estradiol and low avoid a mild presignaled shock by actively moving into a safe
progesterone) (Hampson, 1990b). Interestingly, women on location within the testing space. In a test of two-way active
oral contraceptives did not differ in their performance on the avoidance, intact female rats in proestrus showed facilitation
tasks from menstrual women. Subsequent studies with of avoidance acquisition compared to OVX-females (Sfikakis
different spatial tests found similar effects of estrogen; et al., 1978); compared to OVX-alone females, estradiol
menstrual women with lower levels of estradiol performed benzoate-treated OVX-females showed the same facilitation,
Sex Steroids, Learning and Memory 407

particularly after long-term, chronic estradiol-benzoate example, exposure to stress facilitated the acquisition of
treatment (Singh et al., 1994). Interestingly, however, when a conditioned response in males, but impaired acquisition in
OVX-females received acute estradiol benzoate treatment, females (Wood and Shors, 1998). Females’ performance under
they showed impaired acquisition of two-way avoidance stress was further affected by ovariectomy. When sham-OVX-
behavior compared to untreated OVX-females (Diaz-Veliz females were exposed to stress, they exhibited a significant
et al., 1989). These data suggest that long-term exposure to impairment in their conditioning relative to unstressed control
estrogens facilitates active avoidance learning, but acute animals; this impairment was not observed in stressed OVX
exposure to the hormone may have an impairing effect or no animals (Wood and Shors, 1998). Thus, these data suggest
effect on acquisition at all (Farr et al., 1995). that estrogen mediates the stress-induced impairment observed
Estrogen is not the only ovarian hormone that modulates in female acquisition.
active avoidance learning. Progesterone may also play a critical
role in this type of learning. For example, Farr et al. (1995)
1.14.1.8 Summary
showed that mice in estrus, with elevated levels of proges-
terone, needed more trials to reach acquisition and retention Ovarian hormones exert a robust influence on cognitive
criteria on a T-maze avoidance task compared to diestrous processes in female mammals and may account for sex differ-
mice, when progesterone increases sharply and then drops ences observed in spatial and associative learning although
quickly; similar impairing effects of exogenous progesterone the extent and direction of such influence appears to depend
were observed in OVX-female mice (Farr et al., 1995). on the duration of hormone exposure and the cognitive para-
Other studies of ovarian hormone effects on avoidance digm used.
learning focus on passive avoidance, where rodents learn to
avoid a mild shock in a particular location in one training trial.
Research indicates that elevated levels of ovarian hormones, 1.14.2 Mechanisms of Steroid Action
particularly estrogen, may have an impairing effect on passive
1.14.2.1 Cellular Mechanisms of Steroid Action
avoidance learning. For example, in intact female rats, passive
avoidance was inhibited during proestrus, when there is a surge Neuronal plasticity and synaptic remodeling appear to be inte-
in estradiol levels; this phenomenon was also observed in gral to learning and memory processes (Garcia-Segura et al.,
OVX-females treated with 17b-estradiol and/or progesterone 1994), and recent theories of memory formation are often
(Rivas-Arancibia and Vazquez-Pereyra, 1994; Mora et al., based on the concept of synaptic plasticity, particularly in the
1996; Foster et al., 2003). hippocampus. Ovarian hormones play a key role in modu-
lating synaptic plasticity in the hippocampus, particularly the
1.14.1.7.2 Stress and Conditioning Paradigms density and number of synaptic inputs to the hippocampal
In the absence of stress, ovarian hormones can alter perfor- pyramidal cells (Woolley, 1998). The following section will
mance on a hippocampal-dependent learning task. For review the effects of ovarian hormones on cellular mechanisms
example, when OVX-females are trained on an eye-blink underlying learning and memory processes, specifically electri-
conditioning task, acute exposure can enhance performance cal properties within the hippocampus and estrogen and
(Leuner et al., 2004). Furthermore, results indicated that progesterone effects at the receptor level.
removal of estrogen does not necessarily prevent female
rodents from acquiring trace memory or extinguishing
1.14.2.2 Effects of Ovarian Steroids on Electrical Properties
a learned association, rather the exposure to highly elevated
levels of estradiol actually enhance trace conditioning, and Given that ovarian hormones modulate synaptic plasticity in
subsequently, memory performance (Leuner et al., 2004; the hippocampus (e.g., Woolley, 1998), a subset of research
Jasnow et al., 2006). has focused on how estrogens alter the morphology of neurons.
Ovarian hormones also interact with stress to further modu- For example, research indicates that estradiol-induced changes
late performance on conditioning tasks. In the case of fear in the density of dendritic spines occur without changes in
conditioning, estradiol benzoate-treated mice froze signifi- overall dendritic length or the number of dendritic branches
cantly more to a conditioned tone 24 h after training compared (Woolley and McEwen, 1994), suggesting that the hormone-
to vehicle-treated mice (Morgan and Pfaff, 2001). Other work induced alterations in spine density are due to changes in the
showed that when intact female rats were trained and tested number of spines rather than an expansion and shrinkage of
in proestrus, when estradiol levels are surging, they acquired the dendritic tree (Woolley and McEwen, 1994). Studies with
the classically conditioned eye-blink response faster than fetal primary hippocampal neurons exposed to conjugated-
females in other stages of the estrous cycle (Shors et al., equine estrogens (Brinton et al., 2000) or estradiol (Chen
1998), and the most prominent sex differences in the acquisi- et al., 2006) showed similar effects of estrogens on neurites;
tion of the eye-blink conditioning response were observed here, studies found that both forms of estrogen increased the
when female rats commenced training in proestrus (Wood number and length of neurites, as well as secondary branching
and Shors, 1998; Shors, 2001; Hodes and Shors, 2005; and branch length when compared to estrogen-free cultures.
Bangasser and Shors, 2007). Figure 3 illustrates this point. Other studies tested how exogenous estradiol in OVX-female
Thus, estrogen appears to facilitate both fear and conditioning rats modified dendritic spines. Gould et al. (1990) found that
response learning. 6 days after female rats were ovariectomized, there was
Exposure to external stressors also changes how condi- a significant decrease in the density of dendritic spines on
tioned responses are acquired in male and female rats. For lateral dendritic branches on CA1 pyramidal cells; however,
408 Sex Steroids, Learning and Memory

Figure 3 Effects of stress of trace eyeblink conditioning before, during, and after puberty. For (a)–(c), the bars represent the mean (SEM)
percentage of conditioned responses (CRs) across 300 trails of training. For (d)–(f), the bars represent the mean (SEM) percentage of CRs on
conditioned stimulus (CS)-alone trails. (a) Prior to puberty, there were no differences in conditioning between males and females and no effect of
stress on trace conditioning as measured 24 h after exposure to an acute stressor of brief, periodic tailshocks. (b) During puberty, there were no sex
differences in trace conditioning, although exposure to the stressful event enhanced levels of conditioning in males and females. (c) In adulthood,
females emitted more CRs than did males and exposure to the stressful event enhanced conditioned responding in males and impaired responding in
females. (d) Prior to puberty, there are no sex differences or effects of stress on the responses to the CS on CS-alone trials. (e) During puberty,
stress enhanced these responses regardless of sex. (f) In adulthood, females emitted more CRs than did males but in response to stressor emitted
fewer. In contrast, males emitted more CRs on CS-alone trials if they had been exposed to the acute stressor. Reproduced from Hodes, G.E., Shors,
T.J., 2005. Distinctive effects on learning during puberty. Horm. Behav. 48, 163–171, with permission from Elsevier.

estradiol administration prevented the OVX-induced decrease enhanced LTP. A more recent study assessed whether estradiol
in spine density (Gould et al., 1990). A subsequent study affected LTP differently in male and female rats (Vierk et al.,
found that estradiol administration actually reversed the 2014). In females, synaptic loss emerged after inhibiting estra-
OVX-induced reduction in spine density (Woolley and diol synthesis in hippocampal neurons. This synaptic loss was
McEwen, 1993). Thus, estradiol seems to enhance spine attributed to impaired LTP. However, this effect was not
density and branching in neurons within the hippocampus, observed in males. Thus, it is likely that estradiol synthesis in
a critical region for learning and memory processing. the hippocampus occurs exclusively in females, and this local
Given that hippocampal dendrite spine density (Gould estradiol synthesis assists in the maintenance of LTP and
et al., 1990; Woolley et al., 1990; Woolley and McEwen, synapses; therefore, inhibition of estradiol synthesis results in
1993) and synapse density (Woolley and McEwen, 1992) are impaired LTP and eventually synapse loss in females only.
modulated by estradiol levels, as a function of NMDA receptor
activation (Woolley and McEwen, 1994), estradiol should be
1.14.2.3 A Model of Estrogen Action
able to modulate the ability of the hippocampus to exhibit
long-term potentiation (LTP). LTP of synaptic transmission in 1.14.2.3.1 Estrogen Receptor Subtypes
this region is considered the primary candidate for major Estrogens are a class of steroid hormones comprised of estrone,
synaptic changes underlying learning and memory processes estriol, and estradiol; each estrogen binds to a series of
(Malenka and Nicoll, 1999), and estradiol decreases the receptors including ERɑ, ERb, and G-protein-coupled ER 1
threshold for LTP induction (Cordoba Montoya and Carrer, (GPER1) (Almey et al., 2015). ERɑ and ERb are both found
1997). For example, studies showed that electrophysiological in the cell membrane, nuclei, and cytoplasm. These receptors
sensitivity of CA1 pyramidal cells is modified by exogenous are expressed in different tissues and organs throughout the
estradiol treatment (Montoya and Carrer, 1997) and changes body, including the uterus, breast tissue, testicles, prostate,
in ovarian levels across the estrous cycle (Warren et al., the cardiovascular system (Rollerova and Urbancikova,
1995); in both cases, elevated levels of estradiol are related to 2000), and importantly for learning and memory, the brain.
Sex Steroids, Learning and Memory 409

ERɑ and ERb have been identified in brain regions critical for performance on the task; a pattern not observed in women
learning and memory, such as the hippocampus, prefrontal who had never used hormone treatment (Berent-Spillson
cortex, and amygdala (Almey et al., 2015). et al., 2010). Women using combination hormone treatment
GPER1 was discovered more recently and was first identified also exhibited higher cholinergic activity in the hippocampus
in breast tissue (Carmeci et al., 1997). This G-protein-coupled and posterior cingulate at rest, compared with never-users
receptor is found at the endoplasmic reticulum in neurons and (Smith et al., 2011), which may contribute to the positive asso-
plasma membrane in cells of the hippocampus and ciation between hormone use and cognition, as the neurotrans-
hypothalamus (Almey et al., 2015); binding at GPER1 is mitter is intimately involved in attention, learning, and
thought to modulate effects of estrogens at ERɑ and ERb memory (for reviews, see Klinkenberg et al., 2011; Micheau
(Revankar et al., 2005; Sakamoto et al., 2007) and have and Marighetto, 2011).
direct effects on neuronal transmission in the hippocampus Research also indicates that estradiol can act as a neuropro-
and hypothalamus (Funkoshi et al., 2006; Prossnitz et al., tective steroid in males. For example, intracerebroventricular
2008; Waters et al., 2015). injection of estradiol 10 min prior to quinolinic acid injection
protected the CA1 region of the hippocampus in males (Kuroki
1.14.2.3.2 Estrogen as a Neuroprotective Steroid et al., 2001). Male mice pretreated with estradiol were also pro-
Of the class of estrogens, much of the work on estrogen action tected from stereotypical motor deficits associated with methyl-
in learning and memory has focused on estradiol since, in addi- mercury exposure and exhibited reduced lipid peroxidation in
tion to promoting neurogenesis and maintaining neural integ- the cerebellum (Malagutti et al., 2009); these effects were
rity, estradiol is also neuroprotective. For example, in vitro thought to be the result of estradiol’s protection of the antiox-
studies found that when hippocampal fetal neurons were idant glutathione (Malagutti et al., 2009).
pretreated with either estradiol (Chen et al., 2006) or The aforementioned findings indicate that estradiol protects
conjugated-equine estrogens (Brinton et al., 2000), neurode- the brain via regulation of excitatory ion influx, antioxidant
generation caused by beta-amyloid exposure was blocked, the systems, and maintaining proper energy metabolism despite
degree of apoptosis decreased, and beta-amyloid-induced facing trauma. On a molecular level, estradiol utilizes subme-
decrease in ATP levels was attenuated. In vivo studies also report chanisms to modulate these systemic neuroprotective effects,
patterns of neuroprotection by estradiol in hippocampus and including upregulation of extracellular signal-regulated kinase
entorhinal cortex, as well as neurogenesis in prefrontal cortex. (ERK) phosphorylation in the face of quinolinic acid exposure
For example, when the entorhinal cortex and the hippocampus (Kuroki et al., 2001), reduction of oxidative stress and mainte-
were pretreated with estradiol in OVX-alone females, the nance of respiratory capacity of mitochondria faced with
marked dose-dependent neuron loss in response to kainic various mitochondrial-specific toxins (Yao et al., 2011), and
acid was blocked (Hoffman et al., 2003). Estradiol may also blockade of glutamate-induced and hydrogen peroxide-
protect the brain from age-related declines in females. In induced cell loss by increasing nitric oxide synthesis (Mannella
young-adult female rhesus monkeys, ovariectomy followed et al., 2009; Andozia et al., 2010).
by cyclic estradiol replacement increased dendrite spine
density in layer III of dorsolateral prefrontal cortex; estradiol
1.14.2.4 A Model of Progesterone Action
also restored density in aged female monkeys, compared
with female rhesus monkeys not treated with estradiol (Hao Progesterone is synthesized by different regions, including the
et al., 2007). adrenal cortex, the ovaries, and Leydig’s cells, and binds to
Neuroimaging studies in women support findings of progesterone receptors (Barros et al., 2015) throughout the
estrogenic neuroprotection (Smith and Zubieta, 2001). For uterus, ovaries, brain, and bones (Graham and Clarke,
example, in a structural magnetic resonance imaging (MRI) 1997). Progesterone can have both excitatory and inhibitory
study, postmenopausal women using unopposed estradiol effects (Mahesh et al., 1996) and is considered a neurosteroid
for over 20 years experienced less ventricular enlargement with protective effects on the central and peripheral nervous
and white matter loss than women who did not use estradiol system (Barros et al., 2015). Research indicates that release
(Ha et al., 2007), possibly resulting from estrogenic protection of progesterone is related to a myriad of biological functions,
of oligodendrocytes and subsequent promotion of myelination including reproduction and stress; the sex steroid hormone
(Gerstner et al., 2007). is also important for modulating cognition (McEwen and
Functional magnetic resonance imaging (fMRI) studies Alves, 1999).
have also reported greater activity in brain regions associated
with the task of interest. In one study, women who started 1.14.2.4.1 Progesterone and the Stress Response
a combination estradiol/progestin treatment during perimeno- Work with animals and humans has shown that progesterone
pause performed better on a verbal recognition task; perfor- secreted by the ovaries and adrenals may relate to stress-
mance on the task was subsequently associated with greater related cortisol increases. The effect of stress on adrenal output
activation in the hippocampus compared with women not in animals and men has shown that the adrenal glands secrete
using the hormone treatment (Maki et al., 2011). When not only cortisol, but also progesterone, in response to stress
women started combination hormone treatment within 2 years (Fajer et al., 1971; Brown et al., 1976; Deis et al., 1989; Breier
of menopause and continued uninterrupted for 10 years, they and Buchanan, 1992; Cooper et al., 1995; Elman and Breier,
exhibited greater frontal cortex and hippocampal activation 1997; Duncan et al., 1998; Romeo et al., 2004, 2006) with
during a visual working memory task performed during an limited work examining the effect in women (Childs et al.,
fMRI scan. This increased activity positively correlated with 2010; Gaffey and Wirth, 2014; Herrera et al., 2016).
410 Sex Steroids, Learning and Memory

In addition to stress-induced adrenal release of proges- peripheral details of a scene under stress (Nielsen et al.,
terone, baseline levels of the hormone have been associated 2014). Thus, although progesterone can have different effects
with greater salivary cortisol response to stress. For instance, on cognition, research does clearly indicate that progesterone
the high-progesterone luteal phase of menstrual cycle is asso- is responsible for modulating cognitive processes related to
ciated with greater cortisol responses to social stress, information processing and memory.
compared with the response observed during the low-
progesterone follicular phase (Kirschbaum et al., 1999).
More recent studies show that hormonal contraceptives also 1.14.3 Interaction between Steroid Hormones,
attenuate the salivary cortisol response to physical stress Learning and Memory
compared to naturally cycling women (Nielsen et al.,
1.14.3.1 Overview
2013b) and more specifically, luteal women (Nielsen et al.,
2014), suggesting the inhibition of ovarian hormone release Steroid hormones have been shown to exert robust effects on
exerted by birth control pills contributes to the reduced cognition. Depending on the hormone under investigation,
cortisol response to stress. This reduction with ovarian inhibi- the course of exposure, and the cognitive domain of interest,
tion is likely a result of reduced ovarian progesterone, as these steroid hormones can influence cognitive processes in
women with induced hypogonadism (via administration of a negative or positive fashion. The following sections cover
the GnRH agonist Lupron) exhibited amplified cortisol the effects of glucocorticoids and ovarian hormones on various
responses to exercise stress when also administered proges- cognitive domains.
terone but not estradiol (Roca et al., 2003).
In spite of these compelling data, the interpretation that
1.14.3.2 Glucocorticoids, Learning and Memory
progesterone amplifies the cortisol response to stress fails to
acknowledge that the adrenal glands also secrete progesterone. Glucocorticoids, a class of stress hormone, are necessary for
Importantly, the purported influence of progesterone fluctua- proper neuronal and cognitive functioning. Much focus is
tions during the menstrual cycle on cortisol response to stress placed on the detrimental effects of stress on cognition since
may be masking whether and how adrenal progesterone may long-term or extreme exposure to glucocorticoids or stress
be responding to stress. If women also experience adrenal release can impair learning and memory. However, stress can also
of progesterone in response to stress, then this may contribute to have enhancing effects on learning and memory as well. The
the pattern of greater bioavailability of cortisol in response to following section will address impairing and enhancing effects
stress during high-progesterone phases of the menstrual cycle of glucocorticoids and stress on learning and memory in
(Kirschbaum et al., 1999; Kudielka and Kirschbaum, 2005). animals and humans.
There are multiple purposes stress-induced progesterone
release may serve, such as recruitment of the progesterone 1.14.3.2.1 Factors Influencing the Relationship between
metabolite allopregnanolone in the shutting down the HPA Stress and Learning
axis (Patchev et al., 1994), reducing feelings of anxiety Different factors mediate these potential effects of stress. For
(Dennerstein et al., 1985), and/or exerting a slight sedative example, the magnitude of the stress applied is extremely
effect (Freeman et al., 1992, 1993; Söderpalm et al., 2004). important in determining how stress affects memory. The rela-
Additionally, social closeness has been shown to raise proges- tionship typically follows an inverted-U-shaped curve, with too
terone levels (Brown et al., 2009). This effect may prove bene- little or too much being related to poor performance. However,
ficial during times of stress, as feelings of social support a moderate amount of stress tends to be related to peak perfor-
increase well-being in the face of stress (Cohen and Wills, mance on a variety of tasks, including shock avoidance, spatial
1985), and might promote individuals to seek out beneficial learning, and working memory (Yerkes and Dodson, 1908;
social support systems (Wirth, 2011). Mateo, 2008; Salehi et al., 2010). It is important to note
that there is no defined ‘optimal’ level of stress; this can
1.14.2.4.2 Progesterone Action and Cognition shift depending on the difficulty of the task (Dodson, 1915;
In relation to cognition, recent studies have focused on the rela- Anderson, 1994; Shors, 2004; Salehi et al., 2010).
tionship between progesterone levels and cognitive domains
such as information processing and memory. In studies exam- 1.14.3.2.2 Impairing Effects of Stress on Learning
ining the relationship between progesterone and the processing and Memory in Animals
of emotional or biologically salient stimuli, higher proges- There are numerous reports of glucocorticoid- or stress-
terone levels during the luteal phase are negatively correlated induced cognitive impairment related to chronic or excessive
with recognition of emotional facial expressions (Derntl stress (for review, see Holmes and Wellman, 2009). Some
et al., 2008), while oral progesterone administration reduced studies have used exogenous administration of corticosterone
recognition of faces and brain activation associated with to test the effects of stress on working memory and reference
successful performance of the task (Van Wingen et al., 2007). memory. For example, 8 weeks of daily corticosterone injec-
Other studies found that progesterone levels during the tions resulted in male rodents committing significantly
menstrual cycle are associated with enhanced memory for more working memory errors during a Y-maze task
emotional items (Ertman et al., 2011; Felmingham et al., compared to control animals (Coburn-Litvak et al., 2003).
2012a) with heightened memory for peripheral details of Similarly, 9 weeks of corticosterone treatment induced more
a scene rather than its general theme in a nonstressful context working memory errors during a radial-arm-maze task
(Nielsen et al., 2013a), and with enhanced memory for (Arbel et al., 1994), and 3 weeks of daily corticosterone
Sex Steroids, Learning and Memory 411

injections impaired male rodents’ reference memory on the recognition task (Oei et al., 2006), and the N-back task
Barnes maze (Coburn-Litvak et al., 2003). (Schoofs et al., 2008). Similarly, when tested during the
To test the effects of stress on learning in animals, social stressor, men and women made fewer correct responses
researchers also use psychological and physical stressors. For during the compound remote associates test and took
instance, male rats exposed to a combination of predator and significantly longer to complete anagrams (Alexander et al.,
social stress exhibited significant working memory impair- 2007). The TSST was also related to impaired performance
ments on a radial arm water maze (Park et al., 2001). Compa- on a go/no-go task in men (Scholz et al., 2009). These
rably, males exposed to an unfamiliar environment during impairments are likely related to stressor-induced elevated
a testing delay period for 21 days made more working memory cortisol levels, as men also showed impairments on other
errors on a radial arm maze compared with animals housed in tasks (i.e., a mental arithmetic task) when they exhibited the
their home cages during the delay period (Diamond et al., highest cortisol responses during the task or after giving
1996). Males exposed to the physical and psychological stress a surprise speech (Al’Absi et al., 2002).
of restraint also displayed working memory impairments on Other acute laboratory stressors, such as the cold pressor
a Y-maze (Kleen et al., 2006) and a delayed alternation task task (CPT), are more physical in nature. The CPT reliably
in a T-maze (Lee and Goto, 2015) compared with rats not induces a stress response by requiring participants to hold
exposed to restraint stress. one of their hands in ice water for an extended period of
Exposure to brief periods of stress, rather than long-term time (Bullinger et al., 1984; Edelson and Robertson, 1986;
exposure, can also cause cognitive impairments in animals. Lighthall et al., 2009, 2011; Mather et al., 2010). Like the
For example, while 4 weeks of unpredictable stress resulted in TSST, stress associated with the CPT modulates learning and
impaired acquisition, working, and reference memory and memory. For example, following exposure to the CPT, men
impaired reversal learning in male rodents, shorter regimens had impaired performance on working memory measures
of 6 days and 3 days of unpredictable stress also negatively such as the operation span and digit span backward tasks
impacted reference memory and reversal learning or just refer- (Schoofs et al., 2009); both men and women exhibited
ence memory, respectively (Cerqueira et al., 2007). Even impaired performance on the Sternberg item recognition task
shorter exposure can lead to decrements in performance. For (Duncko et al., 2009).
example, 15 min of tail pinch stress increased error rates during Although stress effects on learning and memory are quite
a delayed win-shift task in a radial arm maze, an effect blocked consistent in the laboratory, the ecological validity of labora-
in animals given a glucocorticoid antagonist (Butts et al., tory studies is sometimes difficult to determine. However,
2011). Such short-term exposure also applies to exogenous research indicates that stressors encountered outside of the
application, such that corticosterone 30 min prior to testing laboratory can interfere with cognition in both men and
impaired performance on a delayed alternation task in T- women. For example, a meta-analysis revealed that interme-
maze in male rodents (Roozendaal, 2004). Thus, even short- diate intensity exercise led to declines in performance on
term and acute exposures to stress can negatively impact a variety of working memory tasks (McMorris et al., 2011).
learning and memory in animals. In other instances, attention-shifting was impaired in
students preparing for a major academic exam; those
1.14.3.2.3 Impairing Effects of Stress on Learning and students’ scores of chronic stress on the Perceived Stress Scale
Memory in Humans predicted their performance on the attention-shift task
Although much of the animal literature focuses on how chronic (Liston et al., 2009). Decision making on the game of dice
glucocorticoid or stress exposure affects cognitive processes, task was also impaired in participants experiencing anticipa-
research with humans focuses on how acute stress modulates tory stress after being told they would need to give a surprise
cognitive processes. Nevertheless, the impairing effects of stress speech (Starcke et al., 2008). Additionally, another cohort
observed in animals are also seen in humans using acute phar- with higher perceived chronic stress showed poorer working
macological, psychological, and physical stress manipulations. memory than those rating lower perceived chronic stress
For example, men treated with acute intravenous hydrocorti- (Öhman et al., 2007). Even within a population of patients
sone showed greater working memory errors during a visuospa- suffering from unipolar major depression, morning cortisol
tial sketchpad task, as well as higher error rates in a paired levels were negatively correlated with performance on the
associate task (Young et al., 1999). Comparably, chronic oral Wisconsin Card Sorting Task, a measure of executive function
hydrocortisone administration in men caused similar impair- (Egeland et al., 2005).
ments on a Sternberg item recognition task (Lupien et al.,
1999), suggesting that elevated cortisol levels impaired 1.14.3.2.4 Sex Steroid Hormones and Stress Response
working memory processes in men. Interactions
Acute psychological stressors, such as the Trier Social Stress Thus far, we have addressed how stress alone affects learning;
Test (TSST), can also have impairing effects on learning. When however, there is also research showing how stress interacts
men completed the TSST, which reliably elevates participants’ with ovarian hormones to modulate learning and memory
cortisol levels by requiring them to give a speech and perform processes in animals and humans. Although comparisons
mental arithmetic in front of an audience (Elzinga and Roelofs, have been made between the animal and human literature
2005; Oei et al., 2006; Schoofs et al., 2008; Wolf et al., 2009), thus far, the following discussion will be limited to studies in
they displayed increased reaction times and other impairments humans. This limitation is because the physiological response
on tests of working memory given 10–45 min later, such as the to stress differs between female rodents and women. For
reading span task (Luethi et al., 2009), the Sternberg item example, female rodents exhibit larger glucocorticoid responses
412 Sex Steroids, Learning and Memory

to stressors than males (Handa et al., 1994; Suzuki et al., 2001; 1.14.3.3 Summary
McCormick et al., 2002; Seale et al., 2004a,b), likely due to
Pharmacological, physical, psychological, and nonlaboratory
excitatory actions of estradiol on the female stress response
stressors can interfere with normal, or optimal, cognitive func-
system (Burgess and Handa, 1992; Suzuki et al., 2001; Weiser
tion in animal models and humans. In particular, the evidence
and Handa, 2009) and inhibitory actions of testosterone on
suggests that chronic or excessive glucocorticoid levels impair
the male stress response system (Bingaman et al., 1994; Suzuki
cognition in tasks requiring executive resources, such as
et al., 2001).
working memory. The following section will address how
In humans, however, women display smaller glucocorticoid
ovarian steroids interact with stress to modulate memory for
responses than men (Kirschbaum et al., 1992, 1999; Kudielka
arousing material and also how these hormones generally
et al., 1998, 2004; Dixon et al., 2004; Kudielka and Kirsch-
affect emotional memory.
baum, 2005). The contradicting glucocorticoid response
profiles of rodents and humans limit the translational general-
ization of stress response profiles and suggest the way in which 1.14.3.4 Steroid Hormone Interactions in Learning and
the two systems may differ across species. Thus, the way Memory of Arousing Information
ovarian hormones influence the stress response and cognition,
and implications for peri- and postmenopausal women, will be 1.14.3.4.1 Ovarian Steroids, Stress and Emotional Memory
limited to studies with humans. in Animals
Rodent studies have explored how ovarian hormones modu-
1.14.3.2.4.1 Estradiol and Stress Response Interactions late emotional memory using a series of acutely arousing tasks.
in Humans For example, when cycling female rats were stressed prior to
In humans, there is evidence of estrogenic blunting of the training, eye-blink conditioning was impaired; the impairment
hypothalamic–pituitary–adrenal (HPA) axis activation, as was prevented if females were ovariectomized or treated with
women display dampened HPA responses to stressors. Natu- the estrogen antagonist, tamoxifen (Wood and Shors, 1998).
rally cycling young-adult women show lower adrenocortico- Similarly, Gupta et al. (2001) found that in a fear conditioning
tropic hormone (ACTH) (Kudielka et al., 1998; Kirschbaum paradigm, ovariectomized female rodents significantly
et al., 1999; Faraday et al., 2005) and biologically active free increased their freezing behavior, an enhanced response similar
cortisol (Davis and Emory, 1995; Kudielka et al., 1998, 2004; to males (Gupta et al., 2001). Please refer to Figure 4 for graph-
Kirschbaum et al., 1999) responses compared with young- ical representation of this behavior.
adult men. Likewise, women tested during the high-estradiol Another study examined differences between male and
phase of the menstrual cycle showed reduced stress-related acti- female rats in the effects of restraint stress on spatial recogni-
vation of different brain regions compared to men, including tion memory, and results showed that 1 h of restraint impaired
the anterior cingulate gyrus, orbitofrontal cortex, medial and spatial memory in males, but had the opposite effect in female
ventromedial prefrontal cortex, amygdala, and hippocampus animals (Conrad et al., 2004). A second experiment revealed
(Goldstein et al., 2010), with similar activational differences that acute stress facilitated spatial memory in females regardless
in women tested during the low-estradiol versus higher- of the estrous cycle phase. These aforementioned studies indi-
estradiol phases of the menstrual cycle (Goldstein et al., cate that sex differences in emotional memory may be
2005). Furthermore, women using combined formulations of estrogen-mediated.
hormonal contraception show blunted cortisol responses Other work has shown that ovarian hormone effects on
compared with naturally cycling women, possibly as a result memory for arousing material may depend on the phase of
of the ethinyl estradiol component (Kirschbaum et al., 1999). the estrous cycle. For example, one study assessed effects of
Evidence in young-adult men also suggests that estradiol the estrous cycle on memory for a spatial water maze task
dampens sympathetic responses to a stressor, with just 1 day when the water temperature was varied (Rubinow et al.,
of estradiol treatment resulting in blunted systolic blood pres- 2004). Results indicated that ovarian hormones enhanced or
sure, pulse rate, epinephrine, and norepinephrine responses to impaired performance on the task depending on the water
a mental stressor (Del Rio et al., 1994). temperature, and plasma corticosterone levels were higher in
Similar dampening of the stress response has been proestrous, when there is surging estradiol levels, rats
observed in postmenopausal women on estradiol treatment. compared to estrous rats. Shors et al. (2000) found similar
Transdermal or oral estradiol interventions spanning 1 day results in that the impairing effect of stress on delay and trace
(Del Rio et al., 1998), 3 weeks (Ceresini et al., 2000), 1 month conditioning was more pronounced in female rats in proestrus.
(Puder et al., 2001), 6 weeks (Lindheim et al., 1992), and Other work also supports these findings (Shansky et al., 2006;
8 weeks (Komesaroff et al., 1999) reduced the epinephrine Dalla et al., 2009), which strongly implicate ovarian hormones
(Del Rio et al., 1998; Ceresini et al., 2000), norepinephrine in the mediation of stress effects on emotional memory in
(Ceresini et al., 2000), diastolic blood pressure (Lindheim animals.
et al., 1992; Komesaroff et al., 1999; Ceresini et al., 2000),
systolic blood pressure (Lindheim et al., 1992; Komesaroff 1.14.3.4.2 Ovarian Steroids and Emotional Memory in Humans
et al., 1999), ACTH and cortisol (Puder et al., 2001) responses Emotionally arousing stimuli tend to be better remembered than
to various stressors, such as mental stressors (Lindheim et al., neutral stimuli (McGaugh, 2000). Studies have shown that
1992; Del Rio et al., 1998; Ceresini et al., 2000), the CPT although men and women exhibit emotional memory enhance-
(Lindheim et al., 1992), and an endotoxin challenge (Puder ments, their memory patterns can differ. In a number of studies,
et al., 2001). sex has been shown to modulate the encoding, consolidation,
Sex Steroids, Learning and Memory 413

have shown that women recall more emotional information


than men (Davis, 1999) and that women outperform men
on emotional memory assessments (Fujita et al., 1991; Bloise
and Johnson, 2007). These behavioral studies indicate that
women recall emotionally arousing material better than men;
however, these studies do not provide evidence for how sex
modulates the underlying neurobiology of emotional memory.
Additional neuroimaging studies have explored sex effects
on brain regions implicated in the processing of emotional
memory, namely the amygdala (McGaugh, 2000). Several
studies have reported a sex-related hemispheric lateralization
of amygdala activity in relation to long-term emotional
memory (Andreano and Cahill, 2009). Cahill et al. (2001)
found that activity of the right amygdala in men viewing
emotional stimuli was significantly correlated with long-term
recall of the stimuli; however, there was no relationship with
the left amygdala. However, activity in the amygdala of the
left hemisphere of the brain while viewing emotional stimuli
was significantly related to long-term recall in women; this rela-
tionship was not observed between activity in the right hemi-
sphere amygdala and memory. This relationship was
replicated in different paradigms investigating sex influences
on emotional memory (Canli et al., 2002; Cahill et al., 2004;
Mackiewicz et al., 2006). A subsequent study demonstrated
that a sex-related hemispheric lateralization of amygdala func-
tion exists even when men and women are resting (Kilpatrick
et al., 2006); these results have been strongly confirmed (Savic
and Lindström, 2008).
Several studies expanded upon this sex-related hemisphere
laterality of amygdala function to examine sex influences on
emotional information processing. There is evidence for
a hemispheric specialization in the processing of global (gist)
versus local (detail) information from an event; the right hemi-
Figure 4 (a) Mean (SEM) percentage of freezing on the conditioning sphere is associated with the processing of gist, whereas the left
day in female rats that received sham surgery (filled), female rats that hemisphere is associated with detail processing (Fink et al.,
were ovariectomized (open), and male rats (hatched). The values were 1996, 1999). One study integrated these hemispheric laterality
normalized to preshock freezing and averages across the three 1-min findings to test the hypothesis that a b-adrenergic blocker
postshock periods. (b) Mean (SEM) percentage of freezing collapsed should impair the amygdala’s modulatory effects on memory.
across the 8-min of the context extinction test over the four consecutive
That is, the drug would impair memory for gist of an emotion-
days of testing for groups described in (a). Reproduced from Gupta,
ally arousing story in men (right amygdala/hemisphere bias)
R.R., Sen, S., Diepenhorst, L.L., Rudick, C.N., Mare, S., 2001. Estrogen
modulates sexually dimorphic contextual fear conditioning and hippo- but memory for details of the same story in women (left amyg-
campal long-term potentiation (LTP) in rats. Brain Res. 888, 356–365, dala/hemisphere bias) (Cahill and van Stegeren, 2003). Results
with permission from Elsevier. from the study supported the hypothesis – emotional arousal
appears to enhance memory for gist in men via activation of
the right amygdala/hemisphere and memory for detail in
and/or retrieval of emotional material (Andreano and Cahill, women via activation of the left amygdala/hemisphere. This
2009). The following section will address sex differences in point is illustrated in Figure 5.
emotional memory, shifts in emotional memory across the
menstrual cycle, and how emotional memory is influenced by 1.14.3.4.4 Shifts in Emotional Memory across the Menstrual
hormonal contraceptive use. Cycle
Sex differences in emotional memory appear to be driven by
1.14.3.4.3 Sex Differences in Emotional Memory Paradigms sex steroid hormone influences on emotional memory
The study of sex differences in memory has received increasing processes, particularly the ovarian hormones estradiol and
interest over the last 20 years. Researchers identified a female progesterone (e.g., Andreano et al., 2008). In women, memory
advantage in autobiographical memory; women are more accu- for emotional material can shift with changes in sex hormone
rate in their recall (Pohl et al., 2005; Bloise and Johnson, 2007) levels, particularly across the menstrual cycle. Some studies of
and provide more details (Seidlitz and Diener, 1998). The emotional memory shifts across the menstrual cycle have inves-
female advantage in autobiographical memory may be linked tigated interactions between sex and stress hormones
to a female advantage in recall of emotional experiences (Andreano et al., 2008). For example, when female participants
(Andreano and Cahill, 2009). For example, some studies read a mildly arousing story and were then administered
414 Sex Steroids, Learning and Memory

Figure 5 Multiple-choice test scores do each experimental group and story phase. (a) Values for questions defined as pertaining to central informa-
tion. (b) Values for questions defined as pertaining to peripheral detail. Values represent means percent correct (SEM) on the multiple-choice test
in each experimental group. P1, P2, and P3 indicate story phases 1, 2, and 3, respectively. Emotional story elements were introduced in P2.
*p < 0.01 placebo compared with corresponding P2 propranolol group. Reproduced from Cahill, L., van Stegeren, A., 2003. Sex-related impairment
of memory for emotional events with b-adrenergic blockade. Neurobiol. Learn. Mem. 79, 81–88, with permission from Elsevier.

a physical stressor (cold pressor stress), stress-induced cortisol examined whether naturally cycling women in hormonally
increases positively correlated with story recall for women in distinct phases of the menstrual cycle differentially recalled
the midluteal phase of the menstrual cycle (Andreano et al., emotional images 1 week after encoding. Enhanced recall of
2008). In the late follicular phase, cortisol responses tended emotional images was only observed in women in the luteal
to negatively correlate with story recall, and women in the early (high hormone) phase of the menstrual cycle; furthermore,
follicular phase exhibited no relationship between cortisol and regression analyses revealed a positive correlation between
memory performance. A more recent study showed similar memory and progesterone levels at encoding (Ertman et al.,
effect of cortisol on memory in women with high and low 2011). Nielsen et al. (2013a) found similar results using an
progesterone; high-progesterone women displayed greater emotional story paradigm. Women in the luteal phase recalled
recall of negative images under stress compared to low- significantly more details, but not gist, from the negatively
progesterone women under stress (Felmingham et al., valenced emotional story 1 week after encoding, whereas
2012b). Other research with arousing and nonarousing words women in the follicular phase did not exhibit enhanced reten-
found that when women were administered a posttraining cold tion for gist or detail in the emotional story.
pressor stress after learning these words, those in the follicular Although these behavioral studies provide evidence for
phase had enhanced long-term memory for arousing words shifts in emotional memory across the menstrual cycle, they
relative to nonarousing words (Zoladz et al., 2015). The find- do not speak to the underlying neurobiological mechanisms.
ings from these studies indicate that the relationship between However, over the past two decades, human neuroimaging
cortisol and memory changes depending upon levels of circu- studies have revealed potential mechanisms modulating sex
lating sex hormones in naturally cycling women. hormone influences on emotional memory. For example,
Other studies have focused on how the menstrual cycle and Goldstein et al. (2005) demonstrated that in women viewing
sex hormones modulate emotional memory in the absence of negative, emotionally arousing material, activity in several
a posttraining stressor. For example, Ertman et al. (2011) limbic, frontal, and hypothalamic regions was related to
Sex Steroids, Learning and Memory 415

menstrual cycle phase; women in the late follicular phase stressors using long-term memory paradigms. A recent study
showed significantly decreased responses in these areas as showed that the use of hormonal contraception altered
compared to those in the early follicular phase. In another memory for information from an emotional story (Nielsen
study, researchers administered synthetic progesterone to et al., 2011). Hormonal contraceptive users exhibited enhanced
young women and found that high levels of synthetic proges- memory for gist, but not details of an emotional story, whereas
terone increased amygdala responses to emotional images naturally cycling women exhibited enhanced emotional
relative to neutral images (Van Wingen et al., 2008). These memory for details, but not gist (Nielsen et al., 2011).
findings were further supported by a study where women in
the midluteal phase exhibited an enhanced response to
emotional images in the hippocampus and amygdala as 1.14.4 Ovarian Steroids and Clinical Implications
compared to women in the early follicular phase (Andreano for Human Neuropathologies
and Cahill, 2010). These findings suggest that sex hormones,
1.14.4.1 Overview
particularly progesterone, modulate the responsiveness of
a key node in the brain’s emotional memory circuitry – the The topics discussed throughout this chapter demonstrate
amygdala. that ovarian steroids have a profound effect on learning and
memory processes in both animal and human models. In
1.14.3.4.5 Hormonal Contraceptive Use and Emotional addition, a subset of research has identified interactions
Memory between ovarian and stress hormones to modulate these
There are limited studies on how hormonal contraception cognitive processes. The final section of the chapter will
influences learning and memory of emotional stimuli. Some discuss implications of ovarian hormones in the clinical
of these studies have focused on how stress and hormonal setting, particularly with respect to AD and disorders of
contraceptives interact to modulate emotional memory emotional memory.
processes. For example, Nielsen et al. (2013b) found that post-
training glucocorticoid release enhanced memory for positive
1.14.4.2 Gonadal Steroids and AD
images in hormonal contraceptive users, but not in naturally
cycling women. In a follow-up study, posttraining stress Women are at greater risk to develop AD compared to men due
enhanced memory for gist and detail from a negative to the substantial loss of estrogens and progesterone at meno-
emotional story in luteal women, but did not affect retention pause (for review, see Pike et al., 2009). If the decrease of
for the emotional story in hormonal contraceptive users ovarian hormones at menopause contributes to women’s
(Nielsen et al., 2014). These studies demonstrate that stress increased risk of AD, then it should follow that treatment
and hormonal contraception interact to modulate emotional with HT would be effective at preventing or treating the disease.
memory in ways not observed in naturally cycling women. However, results to date are inconclusive. Some studies have
Other studies have utilized pharmacological manipulations found that women who used HT had a reducing risk of AD
to explore the relationship between hormonal contraceptive compared to nonusers (Henderson and Buckwalter, 1994;
use, stress, and memory. Kuhlmann and Wolf (2005) tested Paganini-Hill and Henderson, 1994, 1996; Tang et al., 1996;
the influence of cortisol on verbal retrieval in oral contraceptive Kawas et al., 1997; Carlson et al., 2001). These findings support
users and nonusers; results showed that exogenous cortisol estrogen as a protective agent against the development of AD in
administration impaired verbal retrieval in naturally cycling women postmenopause.
women, but not in those using oral contraception (Kuhlmann However, other studies have found that estrogen supple-
and Wolf, 2005). More recent studies with exogenous cortisol mentation can negatively affect the development of AD. For
administration found that cortisol enhanced fear learning in example, the AD Cooperative Study, a randomized, double-
oral contraceptive users compared to men and naturally cycling blind, placebo-controlled, clinical trial, found that older
women (Merz et al., 2012). women with preexisting mild to moderate AD had a faster
Other studies have used classical conditioning paradigms to progression to AD when they were on estradiol supplementa-
explore learning and memory of arousing stimuli in hormonal tion (Mulnard et al., 2000). Other studies found that HT use
contraceptive users. Several studies using classical conditioning was not associated with a reduced risk of AD or that HT use
found that hormonal contraceptive use was associated with did not yield any significant benefits on cognition (Wang
facilitated acquisition of classical conditioning responses et al., 2000; Almeida et al., 2006).
(Beck et al., 2008; Holloway et al., 2011), and this facilitated Although studies have found opposing effects of HT on AD
learning persisted even under stress conditions (Merz et al., development in postmenopausal women, there is a growing
2013a). Imaging work with a classical fear conditioning para- consensus that estrogen-based HT can positively affect AD
digm also revealed that in oral contraceptive users and men, development depending on the time of initiation and length
but not in women in the luteal phase, there was a positive of time that women are on HT relative to the onset of meno-
correlation between basal cortisol levels and differential activa- pause. If women initiate HT in the perimenopausal period,
tion in the amygdala (Merz et al., 2013b). Together, these research indicates that HT is more likely to reduce the risk of
studies suggest that fear learning, even under conditions of AD and improve age-related deficits in cognition (for review,
stress, is significantly altered in women using hormonal Pike et al., 2009). This effect may be due to the healthy cell
contraception. bias (Brinton, 2005) discussed above in section Exogenous
Finally, other work has explored emotional memory in Hormones, Working Memory, and the Women’s Health
hormonal contraceptive users in the absence of external Initiative Studies.
416 Sex Steroids, Learning and Memory

1.14.4.3 Gonadal Steroids and Disorders of Emotional Although there are mixed findings with ovarian hormones
Memory and disorders of emotional memory in women, all studies indi-
cate that these steroids affect depression and anxiety in some
Over the course of their lifetime, women suffer from disorders
manner.
of emotional memory at a significantly higher rate than men
(Risbrough and Stein, 2006; Toufexis et al., 2006). For
example, compared to men, women are more likely to suffer
References
from depressive disorders such as dysthymia, depression,
atypical depression, and seasonal depression (Risbrough Al’Absi, M., Hugdahl, K., Lovallo, W.R., 2002. Adrenocortical stress responses and
and Stein, 2006; Toufexis et al., 2006). Not only are women altered working memory performance. Psychophysiology 39 (1), 95–99.
more likely to suffer from depressive disorders, but their Alexander, J.K., Hillier, A., Smith, R.M., Tivarus, M.E., Beversdorf, D.Q., 2007.
depressive symptoms can last longer, be more severe, tend Beta-adrenergic modulation of cognitive flexibility during stress. J. Cogn. Neurosci.
19 (3), 468–478.
to recur (Nolen-Hoeksema, 1987).
Almeida, O.P., Lautenschlager, N.T., Vasikaran, S., Leedman, P., Gelavis, A.,
Research has also shown that women significantly Flicker, L., 2006. A 20-week randomized controlled trial of estradiol replacement
outnumber men in anxiety disorders. For example, women therapy for women aged 70 years and older: effect on mood, cognition and quality
are twice as likely as men to suffer from generalized anxiety of life. Neurobiol. Aging 27 (1), 141–149.
disorder, PTSD, and panic disorder. Additionally, they are 1.5 Almey, A., Milner, T.A., Brake, W.G., 2015. Estrogen receptors in the central nervous
system and their implication for dopamine-dependent cognition in females. Horm.
times as likely to develop obsessive compulsive disorder and Behav. 74, 125–138.
social anxiety disorder (Pigott, 2003). Andersen, N.E., Dahmani, L., Konishi, K., Bohbot, V.D., 2012. Eye tracking, strategies,
The increased prevalence of anxiety and depressive disorders and sex differences in virtual navigation. Neurobiol. Learn. Mem. 97 (1), 81–89.
in women is likely related to ovarian hormone levels. Some Anderson, K.J., 1994. Impulsivity, caffeine, and task difficulty: a within-subjects test of
the Yerkes-Dodson law. Pers. Individ. Differ. 16 (6), 813–829.
research indicates that lower levels of ovarian steroids facilitate
Andozia, M.B., Vieira, C.S., Franceschini, S.A., Tolloi, M.R.T., de Sá, M.F.S.,
the onset of these disorders. For instance, in humans, reduced Ferriani, R.A., 2010. Ethinylestradiol and estradiol have different effects on
estrogen levels have been associated with the onset of depres- oxidative stress and nitric oxide synthesis in human endothelial cell cultures. Fertil.
sion, sleep disturbance, irritability, anxiety, panic disorders, Steril. 94 (5), 1578–1582.
and cognitive dysfunction (Campbell and Whitehead, 1977; Andreano, J.M., Cahill, L., 2009. Sex influences on the neurobiology of learning and
memory. Learn. Mem. 16 (4), 248–266.
Arpels, 1996; Sherwin, 1998). Additionally, estrogen supple- Andreano, J.M., Cahill, L., 2010. Menstrual cycle modulation of medial temporal
mentation can improve symptoms of these emotional memory activity evoked by negative emotion. Neuroimage 53 (4), 1286–1293.
disorders. Research found that in postmenopausal women, Andreano, J.M., Arjomandi, H., Cahill, L., 2008. Menstrual cycle modulation of the
estrogen replacement therapy was associated with improved relationship between cortisol and long-term memory. Psychoneuroendocrinology
33 (6), 874–882.
mood, increased energy levels, feelings of general well-being,
Arbel, I., Kadar, T., Silberman, M., Levy, A., 1994. The effects of long-term corti-
and improved learning (Halbreich, 1997; Sherwin, 1998). costerone administration on hippocampal morphology and cognitive performance of
Other work suggests that increased levels of gonadal middle-aged rats. Brain Res. 657, 227–235.
hormones actually perpetuate disorders of emotional memory. Arpels, J.C., 1996. The female brain hypoestrogenic continuum from the premenstrual
Some studies have linked levels of estrogens and progestins to syndrome to menopause. A hypothesis and review of supporting data. J. Reprod.
Med. 41 (9), 633–639.
increased vulnerability to anxiety disorders in women (Pigott, Astur, R.S., Ortiz, M.L., Sutherland, R.J., 1998. A characterization of performance by
2003), and other work indicates that hypo- and hyperestrogen- men and women in a virtual Morris water task: a large and reliable sex difference.
ism and the pre-ovulatory estrogen surge are linked to anxiety Behav. Brain Res. 93 (1), 185–190.
and increased nervous energy in women (Arpels, 1996). Astur, R.S., Tropp, J., Sava, S., Constable, R.T., Markus, E.J., 2004. Sex differences
and correlations in a virtual Morris water task, a virtual radial arm maze, and mental
A subset of research has focused on how major hormonal
rotation. Behav. Brain Res. 151 (1), 103–115.
changes throughout a woman’s lifetime affect the onset of Baker, L.D., Asthana, S., Cholerton, B.A., Wilkinson, C.W., Plymate, S.R., Green, P.S.,
emotional memory disorders. Sex differences in anxiety tend Merriam, G.R., Fishel, M.A., Watson, G.S., Cherrier, M.M., Kletke, M.L.,
to emerge only after puberty and may only be due in part to Mehta, P.D., Craft, S., 2012. Cognitive response to estradiol in postmenopausal
the activational effects of gonadal hormones on processes women is modified by high cortisol. Neurobiol. Aging 33 (4), 829.e820–829.e829.
Bakker, J., Baum, M.J., 2000. Neuroendocrine regulation of GnRH release in induced
such as fear extinction (Graham and Milad, 2013). Also, sex ovulators. Front. Neuroendocrinol. 21 (3), 220–262.
differences in depression incidence decrease after women go Bangasser, D.A., Shors, T.J., 2007. The hippocampus is necessary for enhancements
through menopause and their monthly hormonal changes and impairments of learning following stress. Nat. Neurosci. 10 (11), 1401–1403.
are no longer a factor (Sonnenberg et al., 2013). Barker, J.M., Galea, L.A.M., 2009. Sex and regional differences in estradiol content in
the prefrontal cortex, amygdala and hippocampus of adult male and female rats.
When assessing depression, it is most prevalent in women
General Comp. Endocrinol. 164 (1), 77–84.
during the reproductive years and periods of major hormonal Barros, L., Tufik, S., Andersen, M., 2015. The role of progesterone in memory: an
fluctuations, such as puberty, postchildbirth, and after meno- overview of three decades. Neurosci. Biobehav. Rev. 49, 193–204.
pause (Rapkin et al., 2002; Parker and Brotchie, 2010). Women Beck, K.D., McLaughlin, J., Bergen, M.T., Cominski, T.P., Moldow, R.L., Servatius, R.J.,
are also susceptible to postpartum depression (Shors and 2008. Facilitated acquisition of the classically conditioned eyeblink response in
women taking oral contraceptives. Behav. Pharmacol. 19 (8), 821–828.
Leuner, 2003), which begins after childbirth. During the time Berent-Spillson, A., Persad, C.C., Love, T., Tkaczyk, A., Wang, H., Reame, N.K.,
of childbirth, estrogen levels are extremely high and following Frey, K.A., Zubieta, J.-K., Smith, Y.R., 2010. Early menopausal hormone use
the birth of the child, they decline rapidly. This suggests that influences brain regions used for visual working memory. Menopause 1.
depression may be related to a decrease, or at least a dramatic Bimonte, H.A., Denenberg, V.H., 1999. Estradiol facilitates performance as working
memory load increases. Psychoneuroendocrinology 24 (2), 161–173.
change in ovarian steroid levels (Shors and Leuner, 2003);
Bingaman, E., Magnuson, D., Gray, T., Handa, R., 1994. Androgen inhibits the
therefore, estrogen levels may be necessary, but not sufficient increases in hypothalamic corticotropin-releasing hormone (CRH) and CRH-
for inducing depression in women. immunoreactivity following gonadectomy. Neuroendocrinology 59 (3), 228–234.
Sex Steroids, Learning and Memory 417

Blair, J.A., McGee, H., Bhatta, S., Palm, R., Casadesus, G., 2015. Hypothalamic– Cohen, S., Wills, T.A., 1985. Stress, social support, and the buffering hypothesis.
pituitary–gonadal axis involvement in learning and memory and Alzheimer’s Psychol. Bull. 98 (2), 310.
disease: more than “just” estrogen. Front. Endocrinol. 6. Conrad, C.D., Jackson, J.L., Wieczorek, L., Baran, S.E., Harman, J.S., Wright, R.L.,
Bloise, S.M., Johnson, M.K., 2007. Memory for emotional and neutral information: Korol, D.L., 2004. Acute stress impairs spatial memory in male but not female rats:
gender and individual differences in emotional sensitivity. Memory 15 (2), influence of estrous cycle. Pharmacol. Biochem. Behav. 78 (3), 569–579.
192–204. Cooper, C., Evans, A., Cook, S., Rawlings, N., 1995. Cortisol, progesterone and
Breier, A., Buchanan, R.W., 1992. The effects of metabolic stress on plasma b-endorphin response to stress in calves. Can. J. Anim. Sci. 75 (2), 197–201.
progesterone in healthy volunteers and schizophrenic patients. Life Sci. 51 (19), Couse, J.F., Yates, M.M., Walker, V.R., Korach, K.S., 2003. Characterization of the
1527–1534. hypothalamic-pituitary-gonadal axis in estrogen receptor (ER) null mice reveals
Brinton, R.D., Chen, S., Montoya, M., Hsieh, D., Minaya, J., Kim, J., Chu, H.-P., 2000. hypergonadism and endocrine sex reversal in females lacking ERa but not ERb.
The women’s health initiative estrogen replacement therapy is neurotrophic and Mol. Endocrinol. 17 (6), 1039–1053.
neuroprotective. Neurobiol. Aging 21, 475–496. Dalla, C., Whetstone, A.S., Hodes, G.E., Shors, T.J., 2009. Stressful experience has
Brinton, R.D., 2005. Investigative models for determining hormone therapy-induced opposite effects on dendritic spines in the hippocampus of cycling versus
outcomes in brain: evidence in support of a healthy cell bias of estrogen action. masculinized females. Neurosci. Lett. 449 (1), 52–56.
Ann. N. Y. Acad. Sci. 1052 (1), 57–74. Daniel, J.M., Fader, A.J., Spencer, A.L., Dohanich, G.P., 1997. Estrogen enhances
Brown, G., Courtney, G., Marotta, S., 1976. Progesterone secretion by adrenal glands performance of female rats during acquisition of a radial arm maze. Horm. Behav.
of hamsters and comparison of ACTH influence in rats and hamsters. Steroids 28 32, 217–225.
(2), 275–282. Davis, M., Emory, E., 1995. Sex differences in neonatal stress reactivity. Child. Dev.
Brown, S.L., Fredrickson, B.L., Wirth, M.M., Poulin, M.J., Meier, E.A., Heaphy, E.D., 66 (1), 14–27.
Cohen, M.D., Schultheiss, O.C., 2009. Social closeness increases salivary Davis, P.J., 1999. Gender differences in autobiographical memory for childhood
progesterone in humans. Horm. Behav. 56 (1), 108–111. emotional experiences. J. Pers. Soc. Psychol. 76 (3), 498.
Bullinger, M., Naber, D., Pickar, D., Cohen, R.M., Kalin, N.H., Pert, A., Deis, R., Leguizamon, E., Jahn, G., 1989. Feedback regulation by progesterone of
Bunney Jr., W.E., 1984. Endocrine effects of the cold pressor test: relationships to stress-induced prolactin release in rats. J. Endocrinol. 120 (1), 37–43.
subjective pain appraisal and coping. Psychiatry Res. 12, 227–233. Del Rio, G., Velardo, A., Zizzo, G., Avogaro, A., Cipolli, C., Della Casa, L., Marrama, P.,
Burgess, L.H., Handa, R.J., 1992. Chronic estrogen-induced alterations in adreno- Macdonald, I.A., 1994. Effect of estradiol on the sympathoadrenal response to
corticotropin and corticosterone secretion, and glucocorticoid receptor-mediated mental stress in normal men. J. Clin. Endocrinol. Metabolism 79 (3), 836–840.
functions in female rats. Endocrinology 131 (3), 1261–1269. Del Rio, G., Velardo, A., Menozzi, R., Zizzo, G., Tavernari, V., Venneri, M.G.,
Butcher, R., Collins, W., Fugo, N., 1974. Plasma concentration of LH, FSH, prolactin, Marrama, P., Petraglia, F., 1998. Acute estradiol and progesterone administration
progesterone and estradiol-17b throughout the 4-day estrous cycle of the rat. reduced cardiovascular and catecholamine responses to mental stress in meno-
Endocrinology 94 (6), 1704–1708. pausal women. Neuroendocrinology 67, 269–274.
Butts, K.A., Weinberg, J., Young, A.H., Phillips, A.G., 2011. Glucocorticoid receptors in Dennerstein, L., Spencer-Gardner, C., Gotts, G., Brown, J., Smith, M., Burrows, G.,
the prefrontal cortex regulate stress-evoked dopamine efflux and aspects of 1985. Progesterone and the premenstrual syndrome: a double blind crossover trial.
executive function. Proc. Natl. Acad. Sci. U.S.A. 108 (45), 18459–18464. BMJ 290 (6482), 1617–1621.
Cahill, L., van Stegeren, A., 2003. Sex-related impairment of memory for emotional Derntl, B., Windischberger, C., Robinson, S., Lamplmayr, E., Kryspin-Exner, I.,
events with b-adrenergic blockade. Neurobiol. Learn. Mem. 79 (1), 81–88. Gur, R.C., Moser, E., Habel, U., 2008. Facial emotion recognition and amygdala
Cahill, L., Haier, R.J., White, N.S., Fallon, J., Kilpatrick, L., Lawrence, C., Potkin, S.G., activation are associated with menstrual cycle phase. Psychoneuroendocrinology
Alkire, M.T., 2001. Sex-related difference in amygdala activity during emotionally 33 (8), 1031–1040.
influenced memory storage. Neurobiol. Learn. Mem. 75 (1), 1–9. Diamond, D.M., Fleshner, M., Ingersoll, N., Rose, G.M., 1996. Psychological stress
Cahill, L., Uncapher, M., Kilpatrick, L., Alkire, M.T., Turner, J., 2004. Sex-related impairs spatial working memory: relevance to electrophysiological studies of
hemispheric lateralization of amygdala function in emotionally influenced hippocampal function. Behav. Neurosci. 110 (4), 661–672.
memory: an FMRI investigation. Learn. Mem. 11 (3), 261–266. Diaz-Veliz, G., Soto, V., Dussaubat, N., Mora, S., 1989. Influence of the estrous cycle,
Campbell, S., Whitehead, M., 1977. Oestrogen therapy and the menopausal ovariectomy and estradiol replacement upon the acquisition of conditioned
syndrome. Clin. Obstet. Gynaecol. 4 (1), 31–47. avoidance responses in rats. Physiol. Behav. 46 (3), 397–401.
Canli, T., Desmond, J.E., Zhao, Z., Gabrieli, J.D., 2002. Sex differences in the Dixon, K.E., Thorn, B.E., Ward, L.C., 2004. An evaluation of sex differences in
neural basis of emotional memories. Proc. Natl. Acad. Sci. U.S.A. 99 (16), psychological and physiological responses to experimentally-induced pain: a path
10789–10794. analytic description. Pain 112 (1–2), 188–196.
Carlson, M., Zandi, P., Plassman, B., Tschanz, J., Welsh-Bohmer, K., Steffens, D., Dodson, J.D., 1915. The relation of strength of stimulus to rapidity of habit-formation
Bastian, L., Mehta, K., Breitner, J., 2001. Hormone replacement therapy in the kitten. J. Anim. Behav. 5 (4), 330–336.
and reduced cognitive decline in older women the Cache County Study. Duff, S.J., Hampson, E., 2000. A beneficial effect of estrogen on working memory
Neurology 57 (12), 2210–2216. in postmenopausal women taking hormone replacement therapy. Horm. Behav.
Carmeci, C., Thompson, D.A., Ring, H.Z., Francke, U., Weigel, R.J., 1997. Identifi- 38 (4), 262–276.
cation of a gene (GPR30) with homology to the G-protein-coupled receptor Duncan, G.E., Knapp, D.J., Carson, S.W., Breese, G.R., 1998. Differential effects of
superfamily associated with estrogen receptor expression in breast cancer. chronic antidepressant treatment on swim stress-and fluoxetine-induced secre-
Genomics 45 (3), 607–617. tion of corticosterone and progesterone. J. Pharmacol. Exp. Ther. 285 (2),
Ceresini, G., Freddi, M., Marganti, S., Rebecchi, I., Modena, A.B., Rinaldi, M., 579–587.
Manca, C., Amaducci, A., Del Rio, G., Valenti, G., 2000. The effects of transdermal Duncko, R., Johnson, L., Merikangas, K., Grillon, C., 2009. Working memory
estradiol on the response to mental stress in postmenopausal women: a random- performance after acute exposure to the cold pressor stress in healthy volunteers.
ized trial. Am. J. Med. 109, 463–468. Neurobiol. Learn. Mem. 91 (4), 377–381.
Cerqueira, J.J., Mailliet, F., Almeida, O.F., Jay, T.M., Sousa, N., 2007. The prefrontal Edelson, J.T., Robertson, G.L., 1986. The effect of the cold pressor test on vaso-
cortex as a key target of the maladaptive response to stress. J. Neurosci. 27 (11), pressin secretion in man. Psychoneuroendocrinology 11 (3), 307–316.
2781–2787. Egeland, J., Lund, A., Landrø, N.I., Rund, B.R., Sundet, K., Asbjørnsen, A.,
Chen, S., Nilsen, J., Brinton, R.D., 2006. Dose and temporal pattern of estrogen Mjellem, N., Roness, A., Stordal, K.I., 2005. Cortisol level predicts executive and
exposure determines neuroprotective outcome in hippocampal neurons: therapeutic memory function in depression, symptom level predicts psychomotor speed. Acta
implications. Endocrinology 147 (11), 5303–5313. Psychiatr. Scand. 112 (6), 434–441.
Chesler, E.J., Juraska, J.M., 2000. Acute administration of estrogen and progesterone Elman, I., Breier, A., 1997. Effects of acute metabolic stress on plasma progesterone
impairs the acquisition of the spatial Morris water maze in ovariectomized rats. and testosterone in male subjects: relationship to pituitary-adrenocortical axis
Horm. Behav. 38 (4), 234–242. activation. Life Sci. 61 (17), 1705–1712.
Childs, E., Dlugos, A., De Wit, H., 2010. Cardiovascular, hormonal, and emotional Elsabagh, S., Hartley, D., File, S., 2007. Cognitive function in late versus early
responses to the TSST in relation to sex and menstrual cycle phase. Psycho- postmenopausal stage. Maturitas 56 (1), 84–93.
physiology 47 (3), 550–559. Elzinga, B.M., Roelofs, K., 2005. Cortisol-induced impairments of working memory
Coburn-Litvak, P.S., Pothakos, K., Tata, D.A., McCloskey, D.P., Anderson, B.J., 2003. require acute sympathetic activation. Behav. Neurosci. 119 (1), 98–103.
Chronic administration of corticosterone impairs spatial reference memory before Ertman, N., Andreano, J.M., Cahill, L., 2011. Progesterone at encoding predicts
spatial working memory in rats. Neurobiol. Learn. Mem. 80 (1), 11–23. subsequent emotional memory. Learn. Mem. 18 (12), 759–763.
418 Sex Steroids, Learning and Memory

Espeland, M.A., Rapp, S.R., Shumaker, S.A., Brunner, R., Manson, J.E., Gerstner, B., Sifringer, M., Dzietko, M., Schüller, A., Lee, J., Simons, S., Obladen, M.,
Sherwin, B.B., Hsia, J., Margolis, K.L., Hogan, P.E., Wallace, R., Dailey, M., Volpe, J.J., Rosenberg, P.A., Felderhoff-Mueser, U., 2007. Estradiol attenuates
Freeman, R., Hays, J., 2004. Conjugated equine estrogens and global cognitive hyperoxia-induced cell death in the developing white matter. Ann. Neurol. 61 (6),
function in postmenopausal women. J. Am. Med. Assoc. 291 (24), 562–573.
2959–2968. Gibbs, R.B., Burke, A.M., Johnson, D.A., 1998. Estrogen replacement attenuates
Fader, A.J., Hendricson, A.W., Dohanich, G.P., 1998. Estrogen improves performance effects of scopolamine and lorazepam on memory acquisition and retention. Horm.
of reinforced T-maze alternation and prevents the amnestic effects of scopolamine Behav. 34 (2), 112–125.
administered systemically or intrahippocampally. Neurobiol. Learn. Mem. 69 (3), Gibbs, R.B., 1999. Estrogen replacement enhances acquisition of a spatial memory
225–240. task and reduces deficits associated with hippocampal muscarinic receptor inhi-
Fader, A.J., Johnson, P.E., Dohanich, G.P., 1999. Estrogen improves working but not bition. Horm. Behav. 36, 222–233.
reference memory and prevents amnestic effects of scopolamine on a radial-arm Goldstein, J.M., Jerram, M., Poldrack, R., Ahern, T., Kennedy, D.N., Seidman, L.J.,
maze. Pharmacol. Biochem. Behav. 62 (4), 711–717. Makris, N., 2005. Hormonal cycle modulates arousal circuitry in women using
Fajer, A., Holzbauer, M., Newport, H.M., 1971. The contribution of the adrenal gland to functional magnetic resonance imaging. J. Neurosci. 25 (40), 9309–9316.
the total amount of progesterone produced in the female rat. J. Physiol. 214 (1), Goldstein, J.M., Jerram, M., Abbs, B., Whitfield-Gabrieli, S., Makris, N., 2010. Sex
115–126. differences in stress response circuitry activation dependent on female hormonal
Faraday, M., Blakeman, K., Grunberg, N., 2005. Strain and sex alter effects of stress cycle. J. Neurosci. 30 (2), 431–438.
and nicotine on feeding, body weight, and HPA axis hormones. Pharmacol. Gordon, H.W., Corbin, E.D., Lee, P.A., 1986. Changes in specialized cognitive function
Biochem. Behav. 80 (4), 577–589. following changes in hormone levels. Cortex 22 (3), 399–415.
Farr, S.A., Flood, J.F., Scherrer, J.F., Kaiser, F.E., Taylor, G.T., Morley, J.E., 1995. Gould, E., Woolley, C.S., Frankfurt, M., McEwen, B.S., 1990. Gonadal steroids regulate
Effect of ovarian steroids on footshock avoidance learning and retention in female dendritic spine density in hippocampal pyramidal cells in adulthood. J. Neurosci. 10
mice. Physiol. Behav. 58 (4), 715–723. (4), 1286–1291.
Felmingham, K.L., Fong, W.C., Bryant, R.A., 2012a. The impact of progesterone on Graham, J.D., Clarke, C.L., 1997. Physiological action of progesterone in target tissues
memory consolidation of threatening images in women. Psychoneuroendocrinology 1. Endocr. Rev. 18 (4), 502–519.
37 (11), 1896–1900. Graham, B.M., Milad, M.R., 2013. Blockade of estrogen by hormonal contraceptives
Felmingham, K.L., Tran, T.P., Fong, W.C., Bryant, R.A., 2012b. Sex differences in impairs fear extinction in female rats and women. Biol. Psychiatry 73 (4),
emotional memory consolidation: the effect of stress-induced salivary alpha- 371–378.
amylase and cortisol. Biol. Psychol. 89 (3), 539–544. Gresack, J.E., Frick, K.M., 2003. Male mice exhibit better spatial working and
Fink, G.R., Halligan, P.W., Marshall, J.C., Frith, C.D., Frackowiak, R., Dolan, R.J., reference memory than females in a water-escape radial arm maze task. Brain
1996. Where in the brain does visual attention select the forest and the trees? Res. 982 (1), 98–107.
Nature 382. Grigorova, M., Sherwin, B.B., Tulandi, T., 2006. Effects of treatment with leuprolide
Fink, G.R., Marshall, J.C., Halligan, P.W., Frith, C.D., Driver, J., Frackowiak, R.S., acetate depot on working memory and executive functions in young premenopausal
Dolan, R.J., 1999. The neural consequences of conflict between intention and the women. Psychoneuroendocrinology 31 (8), 935–947.
senses. Brain 122 (3), 497–512. Gupta, R.R., Sen, S., Diepenhorst, L.L., Rudick, C.N., Maren, S., 2001. Estrogen
Foster, T.C., Sharrow, K.M., Kumar, A., Masse, J., 2003. Interaction of age and modulates sexually dimorphic contextual fear conditioning and hippocampal long-
chronic estradiol replacement on memory and markers of brain aging. Neurobiol. term potentiation (LTP) in rats. Brain Res. 888 (2), 356–365.
Aging 24 (6), 839–852. Ha, D.M., Xu, J., Janowsky, J.S., 2007. Preliminary evidence that long-term estrogen
Fowler, C.D., Liu, Y., Wang, Z., 2008. Estrogen and adult neurogenesis in the use reduces white matter loss in aging. Neurobiol. Aging 28 (12), 1936–1940.
amygdala and hypothalamus. Brain Res. Rev. 57 (2), 342–351. Halbreich, U., 1997. Role of estrogen in postmenopausal depression. Neurology 48
Freeman, E.W., Weinstock, L., Rickels, K., Sondheimer, S.J., Coutifaris, C., 1992. (5 Suppl. 7), 16S–20S.
A placebo-controlled study of effects of oral progesterone on performance and Hampson, E., Kimura, D., 1988. Reciprocal effects of hormonal fluctuations on human
mood. Br. J. Clin. Pharmacol. 33 (3), 293–298. motor and perceptual-spatial skills. Behav. Neurosci. 102 (3), 456–459.
Freeman, E., Purdy, R., Coutifaris, C., Rickels, K., Paul, S., 1993. Anxiolytic metab- Hampson, E., Moffat, S.D., 2004. The psychobiology of gender: cognitive effects of
olites of progesterone: correlation with mood and performance measures following reproductive hormones in the adult nervous system. In: The Psychology of Gender,
oral progesterone administration to healthy female volunteers. Neuroendocrinology pp. 38–64.
58 (4), 478–484. Hampson, E., Morley, E.E., 2013. Estradiol concentrations and working memory
Frye, C.A., 1995. Estrus-associated decrements in a water maze task are limited to performance in women of reproductive age. Psychoneuroendocrinology 38 (12),
acquisition. Physiol. Behav. 57 (1), 5–14. 2897–2904.
Fujita, F., Diener, E., Sandvik, E., 1991. Gender differences in negative affect and well- Hampson, E., 1990a. Estrogen-related variations in human spatial and articulatory-
being: the case for emotional intensity. J. Pers. Soc. Psychol. 61 (3), 427. motor skills. Psychoneuroendocrinology 15 (2), 97–111.
Funkoshi, T., Birsner, A., D’Amato, R., 2006. The effect of 2 Methoxyestradiol on Hampson, E., 1990b. Variations in sex-related cognitive abilities across the menstrual
acute respiratory distress syndrome. Anesthesiology 105, A222. cycle. Brain Cogn. 14 (1), 26–43.
Gaffey, A.E., Wirth, M.M., 2014. Stress, rejection, and hormones: cortisol and Handa, R.J., Burgess, L.H., Kerr, J.E., O’Keefe, J.A., 1994. Gonadal steroid hormone
progesterone reactivity to laboratory speech and rejection tasks in women and receptors and sex differences in the hypothalamo-pituitary-adrenal Axis. Horm.
men. F1000Research 3. Behav. 28 (4), 464–476.
Galea, L.A., Kimura, D., 1993. Sex differences in route-learning. Pers. Individ. Differ. Hao, J., Rapp, P.R., Janssen, W.G.M., Lou, W., Lasley, B.L., Hof, P.R., Morrison, J.H.,
14 (1), 53–65. 2007. Interactive effects of age and estrogen on cognition and pyramidal neurons in
Galea, L.A., Ossenkopp, K.-P., Kavaliers, M., 1994. Developmental changes in spatial monkey prefrontal cortex. Proc. Natl. Acad. Sci. U.S.A. 104 (27), 11465–11470.
learning in the Morris water-maze in young meadow voles, Microtus pennsylva- Henderson, V.W., Buckwalter, J.G., 1994. Cognitive deficits of men and women with
nicus. Behav. Brain Res. 60 (1), 43–50. Alzheimer’s disease. Neurology 44 (1), 90–96.
Galea, L.A., Kavaliers, M., Ossenkopp, K.-P., Hampson, E., 1995. Gonadal hormone Herrera, A.Y., Nielsen, S.E., Mather, M., 2016. Stress-induced increases in proges-
levels and spatial learning performance in the Morris water maze in male and terone and cortisol in naturally cycling women. Neurobiol. Stress 3.
female meadow voles, Microtus pennsylvanicus. Horm. Behav. 29 (1), Ho, H.-Z., Gilger, J.W., Brink, T.M., 1986. Effects of menstrual cycle on spatial
106–125. information-processes. Percept. Mot. Ski. 63 (2), 743–751.
Galea, L.A.M., Ormerod, B.K., Sampath, S., Kostaras, X., Wilkie, D.M., Phelps, M.T., Hodes, G.E., Shors, T.J., 2005. Distinctive stress effects on learning during puberty.
2000. Spatial working memory and hippocampal size across pregnancy in rats. Horm. Behav. 48 (2), 163–171.
Horm. Behav. 37 (1), 86–95. Hodges, H., 1996. Maze procedures: the radial-arm and water maze compared. Cogn.
Galea, L.A.M., Wide, J.K., Paine, T.A., Holmes, M.M., Ormerod, B.K., Floresco, S.B., Brain Res. 3 (3), 167–181.
2001. High levels of estradiol disrupt conditioned place preference learning, Hoffman, G., Moore, N., Fiksum, G., Murphy, A.Z., 2003. Ovarian steroid modulation of
stimulus response learning and reference memory but have limited effects on seizure severity and hippocampal cell death after kainic acid treatment. Exp.
working memory. Behav. Brain Res. 126, 115–126. Neurol. 182 (1), 124–134.
Garcia-Segura, L.M., Chowen, J., Parducz, A., Naftolin, F., 1994. Gonadal hormones Holloway, J.L., Beck, K.D., Servatius, R.J., 2011. Facilitated acquisition of the clas-
as promoters of structural synaptic plasticity: cellular mechanisms. Prog. Neurobiol. sically conditioned eyeblink response in females is augmented in those taking oral
44 (3), 279–307. contraceptives. Behav. Brain Res. 216 (1), 301–307.
Sex Steroids, Learning and Memory 419

Holmes, A., Wellman, C.L., 2009. Stress-induced prefrontal reorganization and Lupien, S.J., Gillin, C.J., Hauger, R.L., 1999. Working memory is more sensitive than
executive dysfunction in rodents. Neurosci. Biobehav. Rev. 33 (6), 773–783. declarative memory to the acute effects of corticosteroids: a dose-response study
Hosoda, T., Nakajima, H., Honjo, H., 2001. Estrogen protects neuronal cells from in humans. Behav. Neurosci. 113 (3), 420–430.
amyloid beta-induced apoptotic cell death. NeuroReport 12, 1965–1970. Mackiewicz, K.L., Sarinopoulos, I., Cleven, K.L., Nitschke, J.B., 2006. The effect
Janowsky, J.S., Chavez, B., Orwoll, E., 2000. Sex steroids modify working memory. of anticipation and the specificity of sex differences for amygdala and hippo-
J. Cogn. Neurosci. 12 (3), 407–414. campus function in emotional memory. Proc. Natl. Acad. Sci. U.S.A. 103 (38),
Jasnow, A.M., Schulkin, J., Pfaff, D.W., 2006. Estrogen facilitates fear conditioning 14200–14205.
and increases corticotropin-releasing hormone mRNA expression in the central Mahesh, V.B., Brann, D.W., Hendry, L.B., 1996. Diverse modes of action of
amygdala in female mice. Horm. Behav. 49 (2), 197–205. progesterone and its metabolites. J. Steroid Biochem. Mol. Biol. 56 (1), 209–219.
Kawas, C., Resnick, S., Morrison, A., Brookmeyer, R., Corrada, M., Zonderman, A., Maki, P.M., Zonderman, A.B., Resnick, S.M., 2001. Enhanced verbal memory in
Bacal, C., Lingle, D.D., Metter, E., 1997. A prospective study of estrogen nondemented elderly woman receiving hormone-replacement therapy. Am. J.
replacement therapy and the risk of developing Alzheimer’s disease the Baltimore Psychiatry 158, 227–233.
Longitudinal Study of Aging. Neurology 48 (6), 1517–1521. Maki, P.M., Dennerstein, L., Clark, M., Guthrie, J., LaMontagne, P., Fornelli, D.,
Kilpatrick, L., Zald, D., Pardo, J., Cahill, L., 2006. Sex-related differences in amygdala Little, D., Henderson, V.W., Resnick, S.M., 2011. Perimenopausal use of hormone
functional connectivity during resting conditions. Neuroimage 30 (2), 452–461. therapy is associated with enhanced memory and hippocampal function later in life.
Kirschbaum, C., Wust, S., Hellhammer, D.H., 1992. Consistent sex differences in Brain Res. 1379, 232–243.
cortisol responses to psychological stress. Psychosom. Med. 54, 648–657. Maki, P.M., 2013. Critical window hypothesis of hormone therapy and cognition:
Kirschbaum, C., Kudielka, B.M., Gaab, J., Schommer, N.C., Hellhammer, D.H., 1999. a scientific update on clinical studies. Menopause 20 (6), 695–709.
Impact of gender, menstrual cycle phase, and oral contraceptives on the activity of Malagutti, K.S., da Silva, A.P., Braga, H.C., Mitozo, P.A., Soares dos Santos, A.R.,
the hypothalamus-pituitary-adrenal axis. Psychosom. Med. 61 (2), 154–162. Dafre, A.L., de Bem, A.F., Farina, M., 2009. 17b-estradiol decreases
Kleen, J.K., Sitomer, M.T., Killeen, P.R., Conrad, C.D., 2006. Chronic stress impairs methylmercury-induced neurotoxicity in male mice. Environ. Toxicol. Pharmacol. 27
spatial memory and motivation for reward without disrupting motor ability and (2), 293–297.
motivation to explore. Behav. Neurosci. 120 (4), 842–851. Malenka, R.C., Nicoll, R.A., 1999. Long-term potentiation–a decade of progress?
Klinkenberg, I., Sambeth, A., Blokland, A., 2011. Acetylcholine and attention. Behav. Science 285 (5435), 1870–1874.
Brain Res. 221 (2), 430–442. Man, M., MacMillan, I., Scott, J., Young, A., 1999. Mood, neuropsychological function
Komesaroff, P.A., Elser, M.D., Sudhir, K., 1999. Estrogen supplementation attenuates and cognitions in premenstrual dysphoric disorder. Psychol. Med. 29 (03),
glucocorticoid and catecholamine responses to mental stress in perimenopausal 727–733.
women. J. Clin. Endocrinol. Metab. 84 (2), 606–610. Mannella, P., Sanchez, A.M., Giretti, M.S., Genazzani, A.R., Simoncini, T., 2009.
Krug, R., Born, J., Rasch, B., 2006. A 3-day estrogen treatment improves prefrontal Oestrogen and progestins differently prevent glutamate toxicity in cortical neurons
cortex-dependent cognitive function in postmenopausal women. Psychoneur- depending on prior hormonal exposure via the induction of neural nitric oxide
oendocrinology 31 (8), 965–975. synthase. Steroids 74 (8), 650–656.
Kudielka, B., Kirschbaum, C., 2005. Sex differences in HPA axis responses to stress: Markham, J., Pych, J., Juraska, J., 2002. Ovarian hormone replacement to aged
a review. Biol. Psychol. 69 (1), 113–132. ovariectomized female rats benefits acquisition of the morris water maze. Horm.
Kudielka, B., Hellhammer, J., Hellhammer, D.H., Wolf, O., Pirke, K., Varadi, E., Pilz, J., Behav. 42 (3), 284–293.
Kirschbaum, C., 1998. Sex differences in endocrine and psychological responses Mateo, J.M., 2008. Inverted-U shape relationship between cortisol and learning in
to psychosocial stress in healthy elderly subjects and the impact of a 2-Week ground squirrels. Neurobiol. Learn. Mem. 89 (4), 582–590.
dehydroepiandrosterone treatment. J. Clin. Endocrinol. Metab. 83, 1756–1761. Mather, M., Lighthall, N.R., Nga, L., Gorlick, M.A., 2010. Sex differences in how stress
Kudielka, B., Buske-Kirschbaum, A., Hellhammer, D.H., Kirschbaum, C., 2004. HPA affects brain activity during face viewing. NeuroReport 21 (14), 933–937.
axis responses to laboratory psychosocial stress in healthy elderly adults, younger McBurney, D.H., Gaulin, S.J., Devineni, T., Adams, C., 1997. Superior spatial memory
adults, and children: impact of age and gender. Psychoneuroendocrinology 29 (1), of women: stronger evidence for the gathering hypothesis. Evol. Hum. Behav. 18
83–98. (3), 165–174.
Kuhlmann, S., Wolf, O.T., 2005. Cortisol and memory retrieval in women: influence of McCormick, C.M., Linkroum, W., Sallinen, B.J., Miller, N.W., 2002. Peripheral and
menstrual cycle and oral contraceptives. Psychopharmacology 183 (1), 65–71. central sex steroids have differential effects on the HPA axis of male and female
Kuroki, Y., Fukushima, K., Kanda, Y., Mizuno, K., Watanabe, Y., 2001. Neuroprotection rats. Stress Int. J. Biol. Stress 5 (4), 235–247.
by estrogen via extracellular signal-regulated kinase against quinolinic acid-induced McEwen, B.S., Alves, S.E., 1999. Estrogen actions in the central nervous system 1.
cell death in the rat hippocampus. Eur. J. Neurosci. 13 (3), 472–476. Endocr. Rev. 20 (3), 279–307.
LaBuda, C.J., Mellgren, R.L., Hale, R.L., 2002. Sex differences in the acquisition of McEwen, B., 2002. Estrogen actions throughout the brain. Recent Prog. Horm. Res.
a radial maze task in the CD-1 mouse. Physiol. Behav. 76 (2), 213–217. 57, 357–384.
Lee, Y.-A., Goto, Y., 2015. Chronic stress effects on working memory: association with McGaugh, J.L., 2000. Memory–a century of consolidation. Science 287 (5451),
prefrontal cortical tyrosine hydroxylase. Behav. Brain Res. 286, 122–127. 248–251.
Leuner, B., Mendolia-Loffredo, S., Shors, T., 2004. High levels of estrogen enhance McMorris, T., Sproule, J., Turner, A., Hale, B.J., 2011. Acute, intermediate intensity
associative memory formation in ovariectomized females. Psychoneur- exercise, and speed and accuracy in working memory tasks: a meta-analytical
oendocrinology 29 (7), 883–890. comparison of effects. Physiol. Behav. 102 (3), 421–428.
Lighthall, N.R., Mather, M., Gorlick, M.A., 2009. Acute stress increases sex differ- Meethal, S.V., Atwood, C., 2005. The role of hypothalamic-pituitary-gonadal hormones
ences in risk seeking in the balloon analogue risk task. PLoS One 4 (7). in the normal structure and functioning of the brain. Cell Mol. Life Sci. 62 (3),
Lighthall, N.R., Sakaki, M., Vasunilashorn, S., Nga, L., Somayajula, S., Chen, E.Y., 257–270.
Samii, N., Mather, M., 2011. Gender differences in reward-related decision pro- Merz, C.J., Tabbert, K., Schweckendiek, J., Klucken, T., Vaitl, D., Stark, R., Wolf, O.T.,
cessing under stress. Soc. Cogn. Affect. Neurosci. 7 (4). 2012. Oral contraceptive usage alters the effects of cortisol on implicit fear
Lindheim, S.R., Legro, R.S., Bernstein, L., Stanczyk, F.Z., Vijod, M.A., Presser, S.C., learning. Horm. Behav. 62 (4), 531–538.
Lobo, R.A., 1992. Behavioral stress responses in premenopausal and post- Merz, C.J., Stark, R., Vaitl, D., Tabbert, K., Wolf, O.T., 2013a. Stress hormones are
menopausal women and the effects of estrogen. Am. J. Obstet. Gynecol. 167, associated with the neuronal correlates of instructed fear conditioning. Biol.
1831–1836. Psychol. 92 (1), 82–89.
Liston, C., McEwen, B., Casey, B., 2009. Psychosocial stress reversibly disrupts Merz, C.J., Wolf, O.T., Schweckendiek, J., Klucken, T., Vaitl, D., Stark, R., 2013b.
prefrontal processing and attentional control. Proc. Natl. Acad. Sci. U.S.A. 106 (3), Stress differentially affects fear conditioning in men and women. Psychoneur-
912–917. oendocrinology 38 (11), 2529–2541.
Lobo, R.A., Stanczyk, F.Z., 1994. New knowledge in the physiology of hormonal Micheau, J., Marighetto, A., 2011. Acetylcholine and memory: a long, complex and
contraceptives. Am. J. Obstet. Gynecol. 170 (5), 1499–1507. chaotic but still living relationship. Behav. Brain Res. 221 (2), 424–429.
Luethi, M., Meier, B., Sandi, C., 2009. Stress effects on working memory, explicit Miller, K.J., Conney, J.C., Rasgon, N.L., Fairbanks, L.A., Small, G.W., 2002. Mood
memory, and implicit memory for neutral and emotional stimuli in healthy men. symptoms and cognitive performance in women estrogen users and nonusers and
Front. Behav. Neurosci. 2, 1–9. men. J. Am. Geriatr. Soc. 50, 1826–1830.
Luine, V.N., Richards, S.T., Wu, V.Y., Beck, K.D., 1998. Estradiol enhances learning Moffat, S.D., Hampson, E., Hatzipantelis, M., 1998. Navigation in a “virtual” maze: sex
and memory in a spatial memory task and effects levels of monoaminergic differences and correlation with psychometric measures of spatial ability in
neurotransmitters. Horm. Behav. 34 (2), 149–162. humans. Evol. Hum. Behav. 19 (2), 73–87.
420 Sex Steroids, Learning and Memory

Montoya, D.C., Carrer, H., 1997. Estrogen facilitates induction of long term potentiation Pike, C.J., 1999. Estrogen modulates neuronal Bcl-xL expression and b-amyloid-
in the hippocampus of awake rats. Brain Res. 778 (2), 430–438. induced apoptosis: relevance to Alzheimer’s disease. J. Neurochem. 72,
Mora, S., Dussaubat, N., Díaz-Véliz, G., 1996. Effects of the estrous cycle and ovarian 1552–1563.
hormones on behavioral indices of anxiety in female rats. Psychoneur- Pohl, R.F., Bender, M., Lachmann, G., 2005. Autobiographical memory and social
oendocrinology 21 (7), 609–620. skills of men and women. Appl. Cogn. Psychol. 19 (6), 745–760.
Morgan, M., Pfaff, D., 2001. Effects of estrogen on activity and fear-related behaviors Postma, A., Winkel, J., Tuiten, A., van Honk, J., 1999. Sex differences and menstrual
in mice. Horm. Behav. 40 (4), 472–482. cycle effects in human spatial memory. Psychoneuroendocrinology 24 (2),
Mueller, S.C., Jackson, C.P., Skelton, R.W., 2008. Sex differences in a virtual water 175–192.
maze: an eye tracking and pupillometry study. Behav. Brain Res. 193 (2), Prossnitz, E.R., Arterburn, J.B., Smith, H.O., Oprea, T.I., Sklar, L.A., Hathaway, H.J.,
209–215. 2008. Estrogen signaling through the transmembrane G protein-coupled receptor
Mukai, H., Tsurugizawa, T., Ogiue-Ikeda, M., Murakami, G., Hojo, Y., Ishii, H., GPR30. Annu. Rev. Physiol. 70, 165–190.
Kimoto, T., Kawato, S., 2006. Local neurosteroid production in the Hippocampus: Puder, J.J., Freda, P.U., Goland, R.S., Wardlaw, S.L., 2001. Estrogen modulates the
influence on synaptic plasticity of memory. Neuroendocrinology 84 (4), 255–263. hypothalamic-pituitary-adrenal and inflammatory cytokine responses to endotoxin
Mulnard, R.A., Cotman, C.W., Kawas, C., van Dyck, C.H., Sano, M., Doody, R., in women. J. Clin. Endocrinol. Metab. 86 (6), 2403–2408.
Koss, E., Pfeiffer, E., Jin, S., Gamst, A., Grundman, M., Thomas, R., Thal, L.J., Rapkin, A.J., Mikacich, J.A., Moatakef-Imani, B., Rasgon, N., 2002. The clinical nature
2000. Estrogen replacement therapy for treatment of mild to moderate Alzheimer and formal diagnosis of premenstrual, postpartum, and perimenopausal affective
disease: a randomized controlled trial. J. Am. Med. Assoc. 283, 1007–1015. disorders. Curr. Psychiatry Rep. 4 (6), 419–428.
Nadel, L., Hardt, O., 2011. Update on memory systems and processes. Neuro- Rapkin, A.J., Biggio, G., Concas, A., 2006. Oral contraceptives and neuroactive
psychopharmacology 36 (1), 251–273. steroids. Pharmacol. Biochem. Behav. 84 (4), 628–634.
Nielsen, S.E., Ertman, N., Lakhani, Y.S., Cahill, L., 2011. Hormonal contraception Rapp, S.R., Espeland, M.A., Shumaker, S.A., Henderson, V.W., Brunner, R.L.,
usage is associated with altered memory for an emotional story. Neurobiol. Learn. Manson, J.E., Gass, M.L.S., Stefanick, M.L., Lane, D.S., Hays, J., Johnson, K.C.,
Mem. 96 (2), 378–384. Coker, L.H., Dailey, M., Bowen, D., 2003. Effect of estrogen plus progestin on global
Nielsen, S.E., Ahmed, I., Cahill, L., 2013a. Sex and menstrual cycle phase at encoding cognitive function in postmenopausal women the women’s health initiative memory
influence emotional memory for gist and detail. Neurobiol. Learn. Mem. 106, study: a randomized controlled trial. J. Am. Med. Assoc. 289 (20), 2663–2672.
56–65. Reilly, T., 2000. The menstrual cycle and human performance: an overview. Biol.
Nielsen, S.E., Segal, S.K., Worden, I.V., Yim, I.S., Cahill, L., 2013b. Hormonal Rhythm Res. 31 (1), 29–40.
contraception use alters stress responses and emotional memory. Biol. Psychol. 92 Resnick, S.M., Henderson, V.W., 2002. Hormone therapy and risk of Alzheimer
(2), 257–266. disease: a critical time. J. Am. Med. Assoc. 288 (17), 2170–2172.
Nielsen, S.E., Ahmed, I., Cahill, L., 2014. Postlearning stress differentially affects Resnick, S.M., Maki, P.M., Rapp, S.R., Espeland, M.A., Brunner, R., Coker, L.H.,
memory for emotional gist and detail in naturally cycling women and women on Granek, I.A., Hogan, P., Ockene, J.K., Shumaker, S.A., 2006. Effects of combi-
hormonal contraceptives. Behav. Neurosci. 128 (4), 482. nation estrogen plus progestin hormone treatment on cognition and affect. J. Clin.
Nolen-Hoeksema, S., 1987. Sex differences in unipolar depression: evidence and Endocrinol. Metab. 91 (5), 1802–1810.
theory. Psychol. Bull. 101 (2), 259. Revankar, C.M., Cimino, D.F., Sklar, L.A., Arterburn, J.B., Prossnitz, E.R., 2005.
Oei, N.Y.L., Everaerd, W.T.A.M., Elzinga, B.M., van Well, S., Bermond, B., 2006. A transmembrane intracellular estrogen receptor mediates rapid cell signaling.
Psychosocial stress impairs working memory at high loads: an association with Science 307 (5715), 1625–1630.
cortisol levels and memory retrieval. Stress Int. J. Biol. Stress 9 (3), 133–141. Risbrough, V.B., Stein, M.B., 2006. Role of corticotropin releasing factor in anxiety
Öhman, L., Nordin, S., Bergdahl, J., Birgander, L.S., Neely, A.S., 2007. Cognitive disorders: a translational research perspective. Horm. Behav. 50 (4), 550–561.
function in outpatients with perceived chronic stress. Scand. J. Work Environ. Rivas-Arancibia, S., Vazquez-Pereyra, F., 1994. Hormonal modulation of extinction
Health 223–232. responses induced by sexual steroid hormones in rats. Life Sci. 54 (21),
O’keefe, J., Nadel, L., 1978. The Hippocampus as a Cognitive Map. Clarendon Press PL363–PL367.
Oxford. Roca, C.A., Schmidt, P.J., Altemus, M., Deuster, P., Danaceau, M.A., Putman, K.,
Ormerod, B., Galea, L., 2001. Reproductive status influences cell proliferation and cell Rubinow, D.R., 2003. Differential menstrual cycle regulation of hypothalamic-
survival in the dentate gyrus of adult female meadow voles: a possible regulatory pituitary-adrenal axis in women with premenstrual syndrome and controls.
role for estradiol. Neuroscience 102 (2), 369–379. J. Clin. Endocrinol. Metab. 88 (7), 3057–3063.
Packard, M.G., Teather, L.A., 1997a. Intra-hippocampal estradiol infusion enhances Rollerova, E., Urbancikova, M., 2000. Intracellular estrogen receptors, their charac-
memory in ovariectomized rats. Neuroreport 8 (14), 3009–3013. terization and function (review). Endocr. Regul. 34, 203–218.
Packard, M.G., Teather, L.A., 1997b. Posttraining estradiol injections enhance memory Romeo, R.D., Lee, S.J., McEwen, B.S., 2004. Differential stress reactivity in intact and
in ovariectomized rats: cholinergic blockade and synergism. Neurobiol. Learn. ovariectomized prepubertal and adult female rats. Neuroendocrinology 80 (6),
Mem. 68 (2), 172–188. 387–393.
Paganini-Hill, A., Henderson, V.W., 1994. Estrogen deficiency and risk of Alzheimer’s Romeo, R.D., Karatsoreos, I.N., McEwen, B.S., 2006. Pubertal maturation and time of
disease in women. Am. J. Epidemiol. 140 (3), 256–261. day differentially affect behavioral and neuroendocrine responses following an
Paganini-Hill, A., Henderson, V.W., 1996. Estrogen replacement therapy and risk of acute stressor. Horm. Behav. 50 (3), 463–468.
Alzheimer disease. Archiv. Intern. Med. 156 (19), 2213–2217. Roozendaal, B., 2004. The basolateral amygdala interacts with the medial prefrontal
Park, C.R., Campbell, A.M., Diamond, D.M., 2001. Chronic psychosocial stress cortex in regulating glucocorticoid effects on working memory impairment.
impairs learning and memory and increases sensitivity to Yohimbine in adult rats. J. Neurosci. 24 (6), 1385–1392.
Biol. Psychiatry 50, 994–1004. Rosenberg, L., Park, S., 2002. Verbal and spatial functions across the menstrual cycle
Parker, G., Brotchie, H., 2010. Gender differences in depression. Int. Rev. Psychiatry in healthy young women. Psychoneuroendocrinology 27 (7), 835–841.
22 (5), 429–436. Rubinow, M.J., Arseneau, L.M., Beverly, J.L., Juraska, J.M., 2004. Effect of the
Patchev, V., Shoaib, M., Holsboer, F., Almeida, O., 1994. The neurosteroid tetrahy- estrous cycle on water maze acquisition depends on the temperature of the water.
droprogesterone counteracts corticotropin-releasing hormone-induced anxiety and Behav. Neurosci. 118 (4), 863.
alters the release and gene expression of corticotropin-releasing hormone in the rat Sakamoto, H., Matsuda, K.-I., Hosokawa, K., Nishi, M., Morris, J.F., Prossnitz, E.R.,
hypothalamus. Neuroscience 62 (1), 265–271. Kawata, M., 2007. Expression of G protein-coupled receptor-30, a G protein-
Pefanco, M.A., Kenny, A.M., Kaplan, R.F., Kuchel, G., Walsh, S., Kleppinger, A., coupled membrane estrogen receptor, in oxytocin neurons of the rat para-
Prestwood, K., 2007. The effect of 3-year treatment with 0.25 mg/day of ventricular and supraoptic nuclei. Endocrinology 148 (12), 5842–5850.
micronized 17b-estradiol on cognitive function in older postmenopausal women. Salehi, B., Cordero, M.I., Sandi, C., 2010. Learning under stress: the inverted-U-shape
J. Am. Geriatr. Soc. 55 (3), 426–431. function revisited. Learn. Mem. 17 (10), 522–530.
Phillips, K., Silverman, I., 1997. Differences in the relationship of menstrual cycle Sandstrom, N.J., Williams, C.L., 2001. Memory retention is modulated by acute
phase to spatial performance on two-and three-dimensional tasks. Horm. Behav. estradiol and progesterone replacement. Behav. Neurosci. 115 (2), 384–393.
32 (3), 167–175. Sandstrom, N.J., Kaufman, J., Huettel, S.A., 1998. Males and females use different distal
Pigott, T.A., 2003. Anxiety disorders in women. Psychiatr. Clin. N. Am. 26 (3), cues in a virtual environment navigation task. Cogn. Brain Res. 6 (4), 351–360.
621–672. Savic, I., Lindström, P., 2008. PET and MRI show differences in cerebral asymmetry
Pike, C.J., Carroll, J.C., Rosario, E.R., Barron, A.M., 2009. Protective actions of sex and functional connectivity between homo-and heterosexual subjects. Proc. Natl.
steroid hormones in Alzheimer’s disease. Front. Neuroendocrinol. 30 (2), 239–258. Acad. Sci. U.S.A. 105 (27), 9403–9408.
Sex Steroids, Learning and Memory 421

Schmitt, J.A., Jorissen, B.L., Dye, L., Markus, C.R., Deutz, N.E., Riedel, W.J., 2005. Suzuki, S., Lund, T.D., Price, R.H., Handa, R.J., 2001. Sex differences in the
Memory function in women with premenstrual complaints and the effect of hypothalamo-pituitary-adrenal axis: novel roles for androgen and estrogen recep-
serotonergic stimulation by acute administration of an alpha-lactalbumin protein. tors. Recent Res. Dev. Endocrinol. 69–86.
J. Psychopharmacol. 19 (4), 375–384. Tanapat, P., Hastings, N.B., Reeves, A.J., Gould, E., 1999. Estrogen stimulates
Scholz, U., La Marca, R., Nater, U.M., Aberle, I., Ehlert, U., Hornung, R., Martin, M., a transient increase in the number of new neurons in the dentate gyrus of the adult
Kliegel, M., 2009. Go no-go performance under psychosocial stress: beneficial female rat. J. Neurosci. 19 (14), 5792–5801.
effects of implementation intentions. Neurobiol. Learn. Mem. 91 (1), 89–92. Tang, M.-X., Jacobs, D., Stern, Y., Marder, K., Schofield, P., Gurland, B., Andrews, H.,
Schoofs, D., Preuß, D., Wolf, O.T., 2008. Psychosocial stress induces working memory Mayeux, R., 1996. Effect of oestrogen during menopause on risk and age at onset
impairments in an n-back paradigm. Psychoneuroendocrinology 33 (5), 643–653. of Alzheimer’s disease. Lancet 348 (9025), 429–432.
Schoofs, D., Wolf, O.T., Smeets, T., 2009. Cold pressor stress impairs performance on Toufexis, D.J., Myers, K.M., Davis, M., 2006. The effect of gonadal hormones and
working memory tasks requiring executive functions in healthy young men. Behav. gender on anxiety and emotional learning. Horm. Behav. 50 (4), 539–549.
Neurosci. 123 (5), 1066–1075. Van Wingen, G., Van Broekhoven, F., Verkes, R., Petersson, K., Bäckström, T.,
Seale, J., Wood, S., Atkinson, H., Bate, E., Lightman, S., Ingram, C., Jessop, D., Buitelaar, J., Fernandez, G., 2007. Progesterone selectively increases amygdala
Harbuz, M., 2004a. Gonadectomy reverses the sexually diergic patterns of reactivity in women. Mol. Psychiatry 13 (3), 325–333.
circadian and stress-induced hypothalamic-pituitary-adrenal axis activity in male Van Wingen, G., Van Broekhoven, F., Verkes, R., Petersson, K.M., Bäckström, T.,
and female rats. J. Neuroendocrinol. 16 (6), 516–524. Buitelaar, J., Fernandez, G., 2008. Progesterone selectively increases amygdala
Seale, J., Wood, S., Atkinson, H., Harbuz, M., Lightman, S., 2004b. Gonadal steroid reactivity in women. Mol. Psychiatry 13 (3), 325–333.
replacement reverses gonadectomy-induced changes in the corticosterone pulse Vaucher, E., Reymond, I., Najaffe, R., Kar, S., Quirion, R., Miller, M.M.,
profile and stress-induced hypothalamic-pituitary-adrenal axis activity of male and Franklin, K.B.J., 2002. Estrogen effects on object memory and cholinergic
female rats. J. Neuroendocrinol. 16 (12), 989–998. receptors in young and old female mice. Neurobiol. Aging 23, 87–95.
Seidlitz, L., Diener, E., 1998. Sex differences in the recall of affective experiences. Vierk, R., Brandt, N., Rune, G., 2014. Hippocampal estradiol synthesis and its
J. Pers. Soc. Psychol. 74 (1), 262. significance for hippocampal synaptic stability in male and female animals.
Sfikakis, A., Spyraki, C., Sitaras, N., Varonos, D., 1978. Implication of the estrous Neuroscience 274, 24–32.
cycle on conditioned avoidance behavior in the rat. Physiol. Behav. 21 (3), Vranic, A., Hromatko, I., 2008. Content-specific activational effects of estrogen on
441–446. working memory performance. J. Gen. Psychol. 135 (3), 323–336.
Shansky, R.M., Rubinow, K., Brennan, A., Arnsten, A., 2006. The effects of sex and Wang, P., Liao, S., Liu, R., Liu, C., Chao, H., Lu, S., Yu, H., Wang, S., Liu, H.-C.,
hormonal status on restraint-stress-induced working memory impairment. Behav. 2000. Effects of estrogen on cognition, mood, and cerebral blood flow in AD:
Brain Funct. 2 (8). a controlled study. Neurology 54 (11), 2061–2066.
Sherry, D.F., Schacter, D.L., 1987. The evolution of multiple memory systems. Psy- Warren, S.G., Juraska, J.M., 1997. Spatial and nonspatial learning across the rat
chol. Rev. 94 (4), 439. estrous cycle. Behav. Neurosci. 111 (2), 259.
Sherwin, B.B., 1998. Estrogen and cognitive functioning in women. Exp. Biol. Med. Warren, S.G., Humphreys, A.G., Juraska, J.M., Greenough, W.T., 1995. LTP varies
217 (1), 17–22. across the estrous cycle: enhanced synaptic plasticity in proestrus rats. Brain Res.
Shors, T.J., Leuner, B., 2003. Estrogen-mediated effects on depression and memory 703 (1), 26–30.
formation in females. J. Affect. Disord. 74 (1), 85–96. Warren, A.M., Gurvich, C., Worsley, R., Kulkarni, J., 2014. A systematic review of the
Shors, T.J., Lewczyk, C., Pacynski, M., Mathew, P.R., Pickett, J., 1998. Stages of impact of oral contraceptives on cognition. Contraception 90 (2), 111–116.
estrous mediate the stress-induced impairment of associative learning in the Waters, E.M., Thompson, L.I., Patel, P., Gonzales, A.D., Ye, H.Z., Filardo, E.J.,
female rat. Neuroreport 9 (3), 419–423. Clegg, D.J., Gorecka, J., Akama, K.T., McEwen, B.S., 2015. G-protein-coupled
Shors, T.J., Beylin, A.V., Wood, G.E., Gould, E., 2000. The modulation of Pavlovian estrogen receptor 1 is anatomically positioned to modulate synaptic plasticity in
memory. Behav. Brain Res. 110 (1), 39–52. the mouse hippocampus. J. Neurosci. 35 (6), 2384–2397.
Shors, T., 2001. Acute stress rapidly and persistently enhances memory formation in Weber, M.T., Rubin, L.H., Maki, P.M., 2013. Cognition in perimenopause: the effect of
the male rat. Neurobiol. Learn. Mem. 75 (1), 10–29. transition stage. Menopause (New York, NY) 20 (5).
Shors, T.J., 2004. Learning during stressful times. Learn. Mem. 11 (2), 137–144. Weiser, M.J., Handa, R.J., 2009. Estrogen impairs glucocorticoid dependent negative
Shumaker, S.A., Legault, C., Rapp, S.R., Thal, L., Wallace, R.B., Ockene, J.K., feedback on the hypothalamic–pituitary–adrenal axis via estrogen receptor alpha
Hendrix, S.L., Jones, B.N., Assaf, A.R., Jackson, R.D., Kotchen, J., Wassertheil- within the hypothalamus. Neuroscience 159 (2), 883–895.
Smoller, S., Wactawski-Wende, J., 2003. Estrogen plus progestin and the inci- Williams, C.L., Meck, W.H., 1991. The organizational effects of gonadal steroids
dence of dementia and mild cognitive impairment in postmenopausal women. The on sexually dimorphic spatial ability. Psychoneuroendocrinology 16 (1),
women’s health initiative memory study: a randomized controlled trial. J. Am. Med. 155–176.
Assoc. 289, 2651–2662. Williams, C.L., Barnett, A.M., Meck, W.H., 1990. Organizational effects of early
Silverman, I., Phillips, K., 1993. Effects of estrogen changes during the menstrual gonadal secretions on sexual differentiation in spatial memory. Behav. Neurosci.
cycle on spatial performance. Ethol. Sociobiol. 14 (4), 257–269. 104 (1), 84.
Singh, M., Meyer, E.M., Millard, W.J., Simpkins, J.W., 1994. Ovarian steroid depri- Wilson, I.A., Puolivali, J., Heikkinen, T., Riekkinen Jr., P., 1999. Estrogen and NMDA
vation results in a reversible learning impairment and compromised cholinergic receptor antagonism: effects upon reference and working memory. Eur. J. Phar-
function in female Sprague-Dawley rats. Brain Res. 644 (2), 305–312. macol. 381, 93–99.
Skorupskaite, K., George, J.T., Anderson, R.A., 2014. The kisspeptin-GnRH pathway Wirth, M., 2011. Beyond the HPA axis: progesterone-derived neuroactive steroids in
in human reproductive health and disease. Hum. Reprod. Update 20 (4), human stress and emotion. Front. Endocrinol. 2, 1–14.
485–500. Wolf, O.T., Kirschbaum, C., 2002. Endogenous estradiol and testosterone levels are
Smith, Y.R., Zubieta, J.-K., 2001. Neuroimaging of aging and estrogen effects on associated with cognitive performance in older women and men. Horm. Behav. 41
central nervous system physiology. Fertil. Steril. 76 (4), 651–659. (3), 259–266.
Smith, Y.R., Giordani, B., Lajiness-O’Neill, R., Zubieta, J.-K., 2001. Long-term Wolf, O.T., Kudielka, B., Hellhammer, D.H., Torber, S., McEwen, B.S., Kirschbaum, C.,
estrogen replacement is associated with improved nonverbal memory and atten- 1999. Two weeks of transdermal estradiol treatment in postmenopausal elderly
tional measures in postmenopausal women. Menopause 76 (6), 1101–1107. women and its effect on memory and mood: verbal memory changes are associated
Smith, Y.R., Bowen, L., Love, T.M., Berent-Spillson, A., Frey, K.A., Persad, C.C., with the treatment induced estradiol levels. Psychoneuroendocrinology 24, 727–741.
Reame, N.K., Koeppe, R.A., Zubieta, J.K., 2011. Early initiation of hormone therapy Wolf, O.T., Minnebusch, D., Daum, I., 2009. Stress impairs acquisition of delay eyeblink
in menopausal women is associated with increased hippocampal and posterior conditioning in men and women. Neurobiol. Learn. Mem. 91 (4), 431–436.
cingulate cholinergic activity. J. Clin. Endocrinol. Metab. 96 (11), E1761–E1770. Wood, G.E., Shors, T.J., 1998. Stress facilitates classical conditioning in males, but
Söderpalm, A.H., Lindsey, S., Purdy, R.H., Hauger, R., de Wit, H., 2004. Adminis- impairs classical conditioning in females through activation effects ovarian
tration of progesterone produces mild sedative-like effects in men and women. hormones. Proc. Natl. Acad. Sci. U.S.A. 95, 4066–4071.
Psychoneuroendocrinology 29 (3), 339–354. Woolley, C.S., McEwen, B.S., 1992. Estradiol mediates fluctuation in hippocampal
Sonnenberg, C., Deeg, D., Van Tilburg, T., Vink, D., Stek, M., Beekman, A., 2013. synapse density during the estrous cycle in the adult rat. J. Neurosci. 12 (7),
Gender differences in the relation between depression and social support in later 2549–2554.
life. Int. Psychogeriatr. 25 (01), 61–70. Woolley, C.S., McEwen, B.S., 1993. Roles of estradiol and progesterone in regulation
Starcke, K., Wolf, O.T., Markowitsch, H.J., Brand, M., 2008. Anticipatory stress influ- of hippocampal dendritic spine density during the estrous cycle in the rat. J. Comp.
ences decision making under explicit risk conditions. Behav. Neurosci. 122 (6), 1352. Neurol. 336, 293–306.
422 Sex Steroids, Learning and Memory

Woolley, C.S., McEwen, B.S., 1994. Estradiol regulates hippocampal dendritic spine Yerkes, R.M., Dodson, J.D., 1908. The relation of strength of stimulus to rapidity of
density via an N-Methyl-D-Aspartate receptor-dependent mechanism. J. Neurosci. habit-formation. J. Comp. Neurol. Psychol. 18 (5), 459–482.
14 (12), 7680–7687. Young, A.H., Sahakian, B.J., Robbins, T.W., Cowen, P.J., 1999. The effects of chronic
Woolley, C.S., Gould, E., McEwen, B.S., 1990. Exposure to excess glucocorticoids administration of hydrocortisone on cognitive function in normal male volunteers.
alters dendritic morphology of adult hippocampal pyramidal neurons. Brain Res. Psychopharmacology 145, 260–266.
531, 225–231. Zoladz, P.R., Peters, D.M., Cadle, C.E., Kalchik, A.E., Aufdenkampe, R.L., Dailey, A.M.,
Woolley, C.S., 1998. Estrogen-mediated structural and functional synaptic plasticity in Brown, C.M., Scharf, A.R., Earley, M.B., Knippen, C.L., 2015. Post-learning stress
the female rat hippocampus. Horm. Behav. 34 (2), 140–148. enhances long-term memory and differentially influences memory in females
Yaffe, K., Vittinghoff, E., Ensrud, K.E., Johnson, K.C., Diem, S., Hanes, V., Grady, D., depending on menstrual stage. Acta Psychol. 160, 127–133.
2006. Effects of ultra-low-dose transdermal estradiol on cognition and health-
related quality of life. Archiv. Neurol. 63, 945–950.
Yao, J., Chen, S., Cadenas, E., Brinton, R.D., 2011. Estrogen protection against
mitochondrial toxin-induced cell death in hippocampal neurons: antagonism by
progesterone. Brain Res. 1379, 2–10.
1.15 Adrenal Stress Hormone Effects on Memory
Benno Roozendaal and Erno J Hermans, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour,
Radboud University Nijmegen, Nijmegen, The Netherlands
Ó 2017 Elsevier Inc. All rights reserved.

1.15.1 Introduction 423


1.15.2 Effects of Adrenal Stress Hormones on Memory Formation 424
1.15.2.1 Epinephrine 424
1.15.2.2 Glucocorticoids 425
1.15.2.3 Adrenergic–Glucocorticoid Interactions 426
1.15.3 Role of Noradrenergic Activity within the Basolateral Amygdala in Mediating Adrenal Stress Hormone
Effects on Memory Consolidation 427
1.15.3.1 Noradrenergic Activity of the Basolateral Amygdala Mediates Epinephrine Effects on Memory
Consolidation 427
1.15.3.2 Glucocorticoids Interact with Noradrenergic Mechanisms within the Basolateral Amygdala 428
1.15.4 Adrenal Stress Hormone Effects on Other Memory Functions 431
1.15.4.1 Memory Retrieval 431
1.15.4.2 Working Memory 432
1.15.5 Concluding Remarks 433
References 433

1.15.1 Introduction arousing experiences typically leave lasting and vivid memo-
ries (Sandi and Pinelo-Nava, 2007) and, in most cases, it
Every day, we experience stress in varying forms and degrees. certainly seems highly adaptive to have such (neuro)biolog-
When we are exposed to potential threats (stressors), our ical processes that enable the significance of events to regulate
brain initiates a course of action that releases numerous trans- their remembrance. Extensive evidence indicates that
mitters, peptides, and hormones throughout our body epinephrine or glucocorticoids administered shortly after
(Joëls and Baram, 2009). In particular, two systems become a learning experience enhance the consolidation of memory
activated during stress: the fast-acting sympathetic nervous of such an experience (Roozendaal and McGaugh, 2011).
system and the slow hypothalamus–pituitary–adrenal Experimental studies in humans have shown that this
(HPA) axis. Sympathetic nervous system responses include memory retention benefit is observed also when stress is
the release of the catecholamines epinephrine (adrenaline) induced after learning (Abercrombie et al., 2006; Andreano
and norepinephrine (noradrenaline) from the adrenal and Cahill, 2006; Cahill et al., 2003; Preuss and Wolf, 2009;
medulla and presynaptic nerve terminals (McCarty and Smeets et al., 2008), demonstrating that the effects of stress
Gold, 1981; Smith and Vale, 2006), which induces, for and emotional arousal on memory cannot be explained
example, an increase in heart rate, energy metabolism, and simply by immediate effects on attention or memory encod-
enhanced blood flow to skeletal muscles to prepare the ing. In the first part of this chapter, we will summarize animal
organism for the ‘fight-or-flight’ response. Activation of the and human studies on the acute effects of epinephrine and
HPA axis triggers a cascade of events that induces the release glucocorticoids on the consolidation of memory processing.
of glucocorticoids (mainly cortisol in humans, corticosterone It is now well established that adrenal stress hormones prefer-
in rodents) from the adrenal cortex (Ulrich-Lai and Herman, entially strengthen the consolidation of memory of emotion-
2009). First, corticotropin-releasing factor (CRF) is released ally arousing, and not emotionally neutral, training
by the hypothalamic paraventricular nucleus into the portal experiences (de Quervain et al., 2009; Okuda et al., 2004).
system. CRF then induces the release of adrenocorticotropin In the second part, we will address how adrenal stress
from the adrenal pituitary gland, which subsequently stimu- hormones interact with arousal-induced noradrenergic
lates the release of glucocorticoids from the adrenal cortex activity within the basolateral complex of the amygdala
into the bloodstream (Axelrod and Reisine, 1984). In the (BLA), and several other brain regions, to selectively modulate
periphery, glucocorticoids exert, for example, immunosup- memory of emotionally arousing information or during
pressive actions and increases in blood glucose levels emotionally arousing test situations. Moreover, we will
(McEwen, 1998). In addition to preparing an individual for present some recent developments indicating that such gluco-
the acute consequences of dangerous or threatening situations corticoid interactions with the noradrenergic system do not
and the return to homeostasis, an important function of these appear to be mediated through the classic genomic action of
stress hormones and other stress mediators is to induce long- glucocorticoids but, rather, to involve rapid effects through
term adaptive responses (de Kloet et al., 2005; McEwen, an activation of membrane-associated steroid receptors and
1998). the recruitment of downstream endocannabinoid signaling
Here, we focus on how adrenal stress hormones affect (Atsak et al., 2012a; Tasker et al., 2006). Finally, we will
learning and memory processes. Stressful and emotionally briefly describe the modulatory influences of adrenal stress

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00015-8 423


424 Adrenal Stress Hormone Effects on Memory

hormones on other memory functions such as memory Introini-Collison and McGaugh, 1986; Izquierdo and Dias,
retrieval and working memory. 1985; Liang et al., 1986; Sternberg et al., 1985). Epinephrine
is most effective when given immediately after training, and
its memory-enhancing capacity rapidly decays with an
1.15.2 Effects of Adrenal Stress Hormones increasing delay between training and posttraining epinephrine
on Memory Formation administration (Gold and Van Buskirk, 1975). Moreover,
moderate doses produce greatest enhancement, whereas larger
1.15.2.1 Epinephrine
doses are less effective or even impair retention. Highly
Ralph Gerard (1961) may have been the first to suggest that the comparable to the findings of these studies in animals, epineph-
adrenomedullary stress hormone epinephrine might have rine administered to human subjects after they viewed emotion-
enhancing effects on memory consolidation. Memory consoli- ally arousing slides also enhanced the subjects’ long-term
dation is the process by which a fragile short-term memory memory of the slides (Cahill and Alkire, 2003). Systemic
trace is transferred into stable long-term memory. However, administration of the b-adrenoceptor antagonist propranolol
not all information is equally well transferred into long-term blocks epinephrine effects on memory consolidation
storage. In fact, it is well recognized that especially emotionally (Introini-Collison et al., 1992). As epinephrine does not readily
arousing experiences are well remembered, even after a long cross the blood–brain barrier (Weil-Malherbe et al., 1959), its
time (McGaugh, 2003). Gerard proposed that as, “. epineph- effects on memory consolidation appear to be initiated, at least
rine . is released in vivid emotional experiences, such an in part, by activation of b-adrenoceptors located in the
intense adventure should be highly memorable.” Early support periphery. This conclusion is supported by the finding that sota-
for this suggestion came from experiments using systemic lol, a b-adrenoceptor antagonist that does not readily enter the
injections of amphetamines. In addition to the well-known brain, also blocks the effects of systemically administered
effects of amphetamine on dopaminergic mechanisms, epinephrine on memory enhancement (Introini-Collison
amphetamine increases the release of adrenergic catechol- et al., 1992).
amines from peripheral and central storage sites (Rothman Epinephrine effects on memory consolidation are most
et al., 2001). Numerous studies have shown that amphetamine likely mediated by activation of b-adrenoceptors located on
enhances memory when administered peripherally shortly vagal afferents in the periphery that project to noradrenergic
after a training experience. This finding provides strong cell groups within the nucleus of the solitary tract (NTS) in
evidence that the drug enhanced memory by affecting time- the brainstem (Schreurs et al., 1986; Williams and Clayton,
dependent memory consolidation processes, as the subjects 2001). These noradrenergic cells directly innervate brain
are drug-free during both acquisition and retention testing regions involved in memory consolidation, including the
processes (McGaugh, 1973; McGaugh and Petrinovich, amygdala. The NTS may also influence noradrenergic activa-
1965). Enhancing effects of amphetamine in rats and mice tion via its projection to the locus coeruleus (LC), which has
have been observed in a variety of tasks such as inhibitory noradrenergic cells with more widespread projections to
avoidance, active avoidance, and appetitive discrimination forebrain regions, including the hippocampus, cortex, and
(McGaugh, 1989). Moreover, amphetamine administered to amygdala (Ricardo and Koh, 1978; Valentino and Aston-
human subjects either shortly before or immediately after Jones, 1995; Williams and Jensen, 1991; Williams and
learning lists of unrelated words gradually enhanced free recall McGaugh, 1993). The key role of the NTS in mediating
of this information (Soetens et al., 1995, 1993). As amphet- epinephrine effects on memory is indicated by the finding
amine readily passes the blood–brain barrier, other experi- that temporary inactivation of the NTS with lidocaine
ments investigated whether the memory-enhancing effects of prevents epinephrine-induced release of norepinephrine in
amphetamine involve influences on peripheral or central cate- other brain regions and blocks epinephrine influences on
cholamines. Posttraining systemic injections of 4-OH amphet- memory consolidation. Further, stimulating either the vagus
amine, a derivative of amphetamine that does not pass the (Clark et al., 1999) or the NTS immediately after training
blood–brain barrier, also enhanced inhibitory avoidance enhances memory consolidation (Williams and Jensen,
memory (Martinez et al., 1980). Moreover, adrenal demedulla- 1991). Thus, the NTS plays a critical role in mediating the
tion, i.e., elimination of peripheral epinephrine, blocked the transmission of adrenergic signals from the periphery to
effects of both amphetamine and 4-OH amphetamine on brain systems that process memory for emotionally signifi-
memory, whereas removal of peripheral norepinephrine by cant experiences.
sympathetic denervation did not attenuate the memory- Epinephrine may also influence memory consolidation by
enhancing effects of either drug (Martinez et al., 1980). These enhancing glycogenolysis in the liver (Gold, 1995; Messier
findings thus provide support for the view that amphetamine and White, 1984, 1987). Glucose administered peripherally
enhances memory consolidation by stimulating the release immediately after a training experience produces dose- and
of epinephrine from the adrenal medulla (Williams and time-dependent effects on memory comparable to those
McGaugh, 1994). produced by epinephrine (Gold, 1986). Further, doses of
Systemic administration of epinephrine after a training expe- epinephrine and glucose that are optimal for enhancing
rience also enhances memory consolidation in a wide variety of memory induce comparable levels of plasma glucose (Hall
tasks, including inhibitory avoidance, multitrial avoidance, and Gold, 1986). In contrast to their effects on epinephrine,
a one-trial appetitive task, an aversively motivated discrimina- b-adrenoceptor antagonists such as propranolol do not block
tion task, and object recognition (Costa-Miserachs et al., 1994; glucose effects on memory (Gold, 1986). Glucose readily
Dornelles et al., 2007; Gold and van Buskirk, 1976, 1975; enters the brain and, thus, can directly influence brain
Adrenal Stress Hormone Effects on Memory 425

mechanisms involved in memory consolidation (Oomura training impairs long-term memory (but see Atucha et al.,
et al., 1988). However, the finding that memory is also influ- 2015). Corticosterone or a GR agonist administered directly
enced by peripherally administered fructose, a sugar that has into a variety of brain regions enhances memory consolidation
little effect on the brain, suggests that this sugar, as well as of training on many different kinds of learning tasks, including
glucose, also acts, at least in part, at peripheral sites in influ- inhibitory avoidance, contextual and cued fear conditioning,
encing memory (Messier and White, 1987). In support of this water-maze spatial and cued training, object recognition,
view, Talley et al. (2002) reported that vagotomy blocks the conditioned taste aversion, and appetitive taste learning
memory-enhancing effects of peripherally administered (Roozendaal et al., 2006a). Glucocorticoids are known to act
L-glucose, an enantiomer of glucose that does not cross the through intracellular and intranuclear receptors and can affect
blood–brain barrier. gene transcription by direct binding of receptor homodimers
to glucocorticoid response elements on the DNA (Beato
et al., 1995; Datson et al., 2001) or via protein–protein interac-
1.15.2.2 Glucocorticoids
tions with other transcription factors such as Jun or Fos (Heck
There is compelling evidence from studies in both animals and et al., 1994). However, as discussed below, glucocorticoids may
humans that glucocorticoids are also involved in regulating also act more rapidly by interacting with membrane receptors
the consolidation of memory processes (de Kloet, 2000; de and/or potentiating the efficacy of the norepinephrine
Quervain et al., 2009; Flood et al., 1978; Het et al., 2005; signaling cascade via an interaction with G protein-mediated
McGaugh and Roozendaal, 2002; Roozendaal, 2000, 2002, actions and downstream molecular events, including activation
2009a; Sandi and Pinelo-Nava, 2007; Schwabe et al., 2012). of the transcription factor cAMP response-element binding
Blockade of glucocorticoid production with the synthesis (CREB) protein and associated epigenetic (e.g., histone acetyla-
inhibitor metyrapone impairs memory consolidation in both tion) mechanisms (Barsegyan et al., 2010; Dallman, 2005;
animals and humans (Cordero et al., 2002; Maheu et al., Joëls et al., 2011; Karst et al., 2010; Roozendaal, 2002, 2010;
2004). In contrast, acute systemic administration of glucocorti- Tasker et al., 2006). These genomic and nongenomic glucocor-
coids or synthetic glucocorticoid ligands such as dexametha- ticoid actions may ultimately, and collectively, result in
sone enhances long-term memory when given before neuroplasticity and structural changes, e.g., via modifications
(Abercrombie et al., 2003; Buchanan and Lovallo, 2001; Sandi of cell-adhesion molecules and strengthening of cell–cell inter-
and Rose, 1994) or immediately after a training experience actions (Sandi, 2011).
(Flood et al., 1978; Roozendaal and McGaugh, 1996; MR-induced effects have been primarily associated with
Roozendaal et al., 1999; Sandi and Rose, 1994; Wilhelm stress appraisal and responsiveness to stressful stimuli (Joëls
et al., 2011). Glucocorticoid manipulations applied before et al., 2008). Some studies, however, indicated that MR
learning may affect both encoding and consolidation function, either alone or in conjunction with GRs, might also
processes. Human work has shown that administration of be implicated in mnemonic functions, but in most cases
glucocorticoids affects sensory (Miller et al., 2015) and atten- evidence for a direct influence on consolidation processes is
tional processes (Hermans et al., 2014b; Putman and Roelofs, lacking. In healthy individuals it has been shown that oral
2011). Research on the effects of glucocorticoid administration administration of the MR agonist fludrocortisone improves
on medial temporal lobe activity during memory encoding performance in a wide range of neuropsychological tests,
remains inconclusive, with rapid deactivations (Lovallo et al., including visuospatial memory, working memory, and verbal
2010), delayed deactivations (Henckens et al., 2012), and memory (Hinkelmann et al., 2015), whereas blocking MR
null findings (van Stegeren et al., 2010). Glucocorticoids also function typically leads to impaired cognitive performance
play a role in the consolidation of memory of extinction (Cornelisse et al., 2011; Otte et al., 2007; Rimmele et al.,
training (Barrett and Gonzalez-Lima, 2004; Bohus and Lissák, 2012) and, for instance, prevents a stress-induced shift
1968; Cai et al., 2006; Yang et al., 2006). Like epinephrine away from hippocampal-dependent memory functions
effects, such glucocorticoid influences on memory consolida- (Vogel et al., 2015). There is also some evidence from animal
tion follow an inverted U-shaped dose–response relationship: studies that enhancing MR function improves memory
moderate doses enhance memory, whereas lower or higher performance (Harris et al., 2013) but mainly so in transgenic
doses are typically less effective or impair memory models (Ferguson and Sapolsky, 2007; Lai et al.,
consolidation (Lupien et al., 2007; Roozendaal, 2000). 2007). In one study, the MR antagonist spironolactone
Glucocorticoid hormones are highly lipophilic (McEwen administered systemically to rats after inhibitory avoidance
et al., 1979) and can bind directly to mineralocorticoid recep- training impaired later retention (Atucha et al., 2015), but
tors (MRs) and glucocorticoid receptors (GR) (de Kloet, earlier studies in mice found no effect of posttraining
2000; McEwen et al., 1968; Reul and de Kloet, 1985). MRs spironolactone treatment on context- and cue-dependent
have a high affinity for the natural steroids corticosterone, conditioning (Zhou et al., 2010). It is currently not known
cortisol, and aldosterone. GRs have an approximately 10 times whether the effects of peripherally administered MR agonists
lower affinity for corticosterone and cortisol but show a high and antagonists on cognitive performance in animals and
affinity for the synthetic ligands dexamethasone and RU humans are caused by a direct influence on MR function in
28362 (Reul et al., 1987; Sutanto and de Kloet, 1987). The the brain (Ferguson and Sapolsky, 2007; Karst et al., 2005) or
memory-enhancing effects of glucocorticoids appear to involve whether the effects are mediated via peripheral pathways by
the selective activation of the low-affinity GR (Oitzl and de modifying MR-induced influences on, for example, HPA-axis
Kloet, 1992; Roozendaal and McGaugh, 1997) as a blockade activity (Lembke et al., 2013) or autonomic outflow (Borrell
of GRs, but not MRs, shortly before or immediately after et al., 1984; Rahmouni et al., 2001). The cognitive changes
426 Adrenal Stress Hormone Effects on Memory

may also involve an MR-dependent influence on GR function evidence that training-associated emotional arousal is essential
(Liu et al., 1995). for enabling glucocorticoid effects on memory consolidation.
Other findings indicated that training-induced (nor)adren-
ergic activation is a critical component of emotional arousal
1.15.2.3 Adrenergic–Glucocorticoid Interactions
in enabling glucocorticoid effects on memory consolidation.
Evidence from several kinds of studies indicates that although As shown in Figure 1(a), the b-adrenoceptor antagonist
epinephrine and glucocorticoid effects are initiated by different propranolol administered systemically together with the corti-
brain mechanisms, these adrenal stress hormones interact to costerone immediately after object recognition training
influence memory consolidation. For example, glucocorticoid blocked the corticosterone-induced memory enhancement of
administration alters the efficacy of epinephrine for influencing rats without prior habituation to the training context
memory consolidation in adrenalectomized rats (Borrell et al., (Roozendaal et al., 2006b). Additionally, and shown in
1984, 1983). Moreover, the corticosterone-synthesis inhibitor Figure 1(b), in habituated rats, pharmacological reinstatement
metyrapone attenuates the memory-enhancing effects of of adrenergic activity by the administration of the noradren-
amphetamine and epinephrine (Roozendaal et al., 1996). ergic stimulant yohimbine (an a2-adrenoceptor antagonist)
Thus, synergistic actions of epinephrine and corticosterone after object recognition training enabled corticosterone to
may be essential in mediating stress effects on memory induce dose-dependent enhancement of memory (Roozendaal
enhancement (Joëls et al., 2011). et al., 2006b). These findings strongly suggest that emotional
Animal experiments investigating stress hormone effects on arousal-induced adrenergic activation is essential for enabling
memory consolidation typically use highly arousing training glucocorticoid enhancement of memory consolidation.
conditions, such as inhibitory avoidance or contextual and Human studies generally support the conclusion of animal
cued fear conditioning, that are known to induce the release experiments that glucocorticoids can enhance memory consol-
of both corticosterone and epinephrine. Studies using an object idation only when their activity is paralleled by emotional
recognition task investigated whether adrenergic activation arousal (i.e., noradrenergic activity). Cortisol administered
induced by emotional arousal is essential in enabling cortico- shortly before or after training selectively enhances long-term
sterone effects on memory consolidation (Okuda et al., memory of emotionally arousing, but not of emotionally
2004). Rats were given either extensive habituation to an appa- neutral, items (Buchanan and Lovallo, 2001; Kuhlmann and
ratus, or no prior habituation, and were then allowed to Wolf, 2006). Moreover, a cold pressor stress in humans (i.e.,
explore objects in the apparatus. Placing rats in a novel testing placing the arm in ice water), a procedure that significantly
apparatus evokes novelty-induced arousal and habituation of elevates endogenous cortisol levels, enhances memory of
rats to the apparatus is known to reduce this arousal response emotionally arousing slides but does not affect memory of
(De Boer et al., 1990). Corticosterone administered immedi- emotionally neutral slides (Cahill et al., 2003; but see Preuss
ately posttraining to nonhabituated (i.e., emotionally aroused) and Wolf, 2009). Consistent with these findings, it has been re-
rats enhanced their 24-h retention performance. In contrast, ported that levels of endogenous cortisol at the time of learning
posttraining corticosterone did not enhance retention of object correlated with enhanced memory consolidation only in indi-
recognition in habituated rats (Okuda et al., 2004), providing viduals who were emotionally aroused (Abercrombie et al.,

(a) (b)

Figure 1 Glucocorticoid effects on memory consolidation for object recognition training require arousal-induced noradrenergic activation. Rats were
either habituated to the training context for 7 days (prior habituation) or not habituated (no prior habituation). On day 8, they were given a 3-min
training trial during which they could freely explore two identical objects, followed by systemic drug administration. Retention was tested 24 h later
by placing the rats back into the apparatus for 3 min. On the retention trial, one object was similar to the training objects, whereas the other was
novel. Data represent discrimination index (%) on the 24-h retention trial, expressed as mean  SEM. The discrimination index was calculated as the
difference in time spent exploring the novel and the familiar object, expressed as the ratio of the total time spent exploring both objects. (a) Effect of
immediate posttraining administration of the b-adrenoceptor antagonist propranolol (3.0 mg kg1, s.c.) on corticosterone-induced enhancement of
object recognition memory in nonhabituated (emotionally aroused) rats. (b) Effect of coadministration of the a2-adrenoceptor antagonist yohimbine
(0.3 mg kg1, s.c.) with corticosterone on object recognition memory in habituated (emotionally nonaroused) rats. **, p < 0.0001 vs vehicle. Adapt-
ed from Roozendaal, B., Okuda, S., van der Zee, E.A., McGaugh, J.L., 2006. Glucocorticoid enhancement of memory requires arousal-induced norad-
renergic activation in the basolateral amygdala. Proc. Natl. Acad. Sci. U.S.A. 103 (17), 6741–6746.
Adrenal Stress Hormone Effects on Memory 427

2006). Thus, these findings from animal and human studies Extensive evidence now indicates that the BLA is the critical
indicate that training-associated endogenous emotional region of the amygdala involved in mediating noradrenergic
arousal is essential for enabling glucocorticoid effects on influences on memory consolidation. The BLA is a cortical-
memory consolidation. like structure that has many reciprocal connections with other
higher brain regions, including the hippocampus and
prefrontal cortex (Petrovich et al., 2001; Pitkänen, 2000).
Many studies found that posttraining infusions of norepineph-
1.15.3 Role of Noradrenergic Activity within
rine or the b-adrenoceptor agonist clenbuterol into the amyg-
the Basolateral Amygdala in Mediating Adrenal
dala (or selectively into the BLA) induce a dose-dependent
Stress Hormone Effects on Memory Consolidation
enhancement of retention (Barsegyan et al., 2014; Bianchin
et al., 1999; Hatfield and McGaugh, 1999; Huff et al., 2005;
As summarized up to this point, both epinephrine and gluco-
Izquierdo et al., 1992; Liang et al., 1986, 1995). In contrast,
corticoids enhance memory consolidation. The memory-
b-adrenoceptor agonists or antagonists infused into other
enhancing effects of epinephrine are initiated by activating
amygdala regions such as the central amygdala are ineffective
b-adrenoceptors on vagal afferents in the periphery, which
(Hatfield and McGaugh, 1999). In addition to b-adrenoceptor
will then activate noradrenergic mechanisms in the brain.
effects, a-adrenoceptor influences in the BLA also modulate
Glucocorticoid hormones readily enter the brain, but also their
memory consolidation. Intra-BLA administration of the
effects on memory consolidation depend critically on interac-
a1-adrenoceptor antagonist prazosin impairs inhibitory avoid-
tions with emotional arousal-induced noradrenergic activity.
ance memory, whereas an activation of a1-adrenoceptors
In this section we will go deeper into the neurobiological mech-
enhances retention (Ferry et al., 1999a). The a1-adrenoceptor-
anisms of how epinephrine and glucocorticoids interact with
induced memory enhancement most likely involves an interac-
central noradrenergic activation in enhancing memory consol-
tion with b-adrenoceptors, as posttraining intra-BLA infusions
idation of emotionally arousing experiences. Furthermore, we
of the b-adrenoceptor antagonist atenolol block the memory
will present some recent evidence indicating that glucocorti-
enhancement produced by activation of a1-adrenoceptors
coid interactions with the noradrenergic arousal system involve
(Ferry et al., 1999b). Considered together, these findings
fast, nongenomically mediated actions via a membrane-
strongly suggest that noradrenergic activation in the BLA plays
associated GR and downstream endocannabinoid signaling.
a critical role in enhancing memory consolidation and medi-
ating the effects of peripheral epinephrine on memory
consolidation.
1.15.3.1 Noradrenergic Activity of the Basolateral Amygdala
Gold and van Buskirk (1978) reported evidence suggesting
Mediates Epinephrine Effects on Memory Consolidation
that noradrenergic activity in the brain is increased in rats
Extensive evidence indicates that norepinephrine, originating trained on an inhibitory avoidance task or given memory-
from cell groups in the NTS and LC, affects memory consolida- enhancing doses of epinephrine immediately after training.
tion by influences involving the amygdala and that the effects Subsequent studies using in vivo microdialysis and high-
of peripheral epinephrine on memory involve noradrenergic performance liquid chromatography (HPLC) to measure
activation of the amygdala. Early findings have shown that ongoing changes in norepinephrine levels indicated that
adrenal demedullation or posttraining administration of norepinephrine is normally released within the amygdala
epinephrine alters the memory-modulating effects of electrical during training on aversively motivated tasks. A single, brief
stimulation of the amygdala (Liang et al., 1985) and that footshock stimulation of the kind typically used in fear-based
lesions of either the amygdala or stria terminalis, an important training induced norepinephrine release within the amygdala
amygdala input–output pathway, block epinephrine effects on that varied directly with the stimulus intensity (Galvez et al.,
memory consolidation (Cahill and McGaugh, 1991; Liang and 1996; Quirarte et al., 1998). Moreover, epinephrine adminis-
McGaugh, 1983). Experiments using the expression of imme- tered systemically in a dose known to enhance memory
diate early genes as markers for activated neurons reported consolidation potentiated footshock-induced increases in
that stimulation of the vagus nerve, the pathway that mediates norepinephrine levels in the amygdala (Williams et al.,
epinephrine effects on the brain, increases the number of amyg- 1998). Further, stimulation of the vagus nerve or the NTS
dala neurons expressing c-fos. Electrophysiological findings increases norepinephrine levels in the amygdala and enhances
indicate that the firing rate of amygdala neurons is increased memory consolidation (Clayton and Williams, 2000; Hassert
following electrical stimulation of either the vagus nerve et al., 2004). McIntyre et al. (2002) found that norepinephrine
(Radna and MacLean, 1981) or the NTS (Rogers and Fryman, levels in the amygdala increased significantly after inhibitory
1988). In contrast, depletion of amygdala norepinephrine avoidance training. Additionally, and importantly, the magni-
with local injections of the norepinephrine-specific neurotoxin tude of training-induced increases in amygdala norepinephrine
DSP-4 blocks systemic epinephrine effects on memory consol- levels assessed shortly after training correlated highly with the
idation (Liang, 1998). Posttraining intra-amygdala infusions of rats’ subsequent long-term retention performance, providing
the b-adrenoceptor antagonist propranolol also block the additional evidence suggesting that noradrenergic activity of
memory enhancement induced by systemic administration of the amygdala regulates memory consolidation.
epinephrine (Liang et al., 1986, 1995), whereas posttraining Human research corroborates these findings: administra-
infusions of norepinephrine into the amygdala attenuate reten- tion of yohimbine either before (O’Carroll et al., 1999) or after
tion impairment induced by adrenal demedullation (Liang (Southwick et al., 2002) learning has been shown to enhance
et al., 1986). memory. Carriers of a common functional deletion in the
428 Adrenal Stress Hormone Effects on Memory

gene coding for the presynaptic a2b-adrenoceptor (ADRA2B), (a)


exhibit a stronger enhancement of emotional memory (de
Quervain et al., 2007b). Conversely, blocking the effects of
norepinephrine using the b-adrenoceptor antagonist propran-
olol results in impaired memory for emotionally arousing
events (Cahill et al., 1994). Such noradrenergic effects are cen-
trally mediated: nadolol, a b-adrenoceptor blocker that does
not readily cross the blood–brain barrier, does not mimic the
effects of propranolol (van Stegeren et al., 1998). Human neu-
roimaging work furthermore supports the notion that such
noradrenergic effects are mediated by the amygdala. For
instance, increased activity in the amygdala was observed
shortly after experimental induction of stress in healthy volun-
teers (Cousijn et al., 2010; Oei et al., 2012; van Marle et al., (b)
2009) or after symptom provocation in anxiety-disordered
patients (Osuch et al., 2008). Pharmacological elevation of
levels of norepinephrine using reboxetine yields similar results
(Onur et al., 2009). Furthermore, carriers of the functional
deletion in the ADRA2B gene mentioned above show stronger
amygdala responses to aversive stimuli (Rasch et al., 2009) and
stronger stress-induced enhancements of amygdala activation
(Cousijn et al., 2010).
There is now compelling evidence that this amygdala (or
more specifically, BLA) activation induced by stress hormones
and norepinephrine during emotionally arousing stimulation
acts via efferent projections to enhance memory storage and
neuroplasticity mechanisms in other brain regions (McGaugh, Figure 2 Corticosterone treatment after inhibitory avoidance training
2004; McIntyre et al., 2005). These regions include the hippo- increases norepinephrine levels in the BLA. Microdialysis samples were
campus, entorhinal cortex, striatum, medial prefrontal cortex, collected every 15 min. Norepinephrine levels (mean  SEM) are
anterior cingulate cortex, and insula, and this list is likely to expressed as a percentage change from average baseline levels.
expand in the future (Hermans et al., 2014a). Notably, the (a) Corticosterone treatment (3.0 mg kg1, i.p.) significantly increased
norepinephrine release in the amygdala of animals trained on an inhibi-
same regions have been shown to exhibit strong intrinsic func-
tory avoidance task compared with vehicle-injected animals. *, p < 0.05
tional connectivity within distinct large-scale neural systems in
vs vehicle. (b) No changes in norepinephrine levels were observed in
the human brain. For instance, the hippocampal complex and corticosterone- or vehicle-injected animals that were not trained on the
medial prefrontal cortex are part of the midline-centered task. Adapted from McReynolds, J.R., Donowho, K., Abdi, A.,
‘default mode network,’ while insular, anterior cingulate, and McGaugh, J.L., Roozendaal, B., McIntyre, C.K., 2010. Memory-
striatal structures are part of the cingulo-opercular ‘salience’ enhancing corticosterone treatment increases amygdala norepinephrine
network (Hermans et al., 2014b; Menon, 2011). Functional and Arc protein expression in hippocampal synaptic fractions. Neuro-
connectivity, in particular within the latter network, is regulated biol. Learn. Mem. 93 (3), 312–321.
by norepinephrine (Hermans et al., 2011). Our understanding
of these network interactions and their contributions to
memory functions is likely to develop rapidly in the coming the b-adrenoceptor antagonists propranolol or atenolol into
years due to novel techniques such as multiunit recording the BLA, but not into the neighboring central amygdala,
and optogenetics. blocked the memory enhancement induced by a glucocorticoid
administered systemically (Quirarte et al., 1997). Atenolol or
the protein kinase A (PKA) inhibitor Rp-cAMPS administered
1.15.3.2 Glucocorticoids Interact with Noradrenergic
into the BLA also blocks memory enhancement induced by
Mechanisms within the Basolateral Amygdala
infusions of a GR agonist into the BLA (Roozendaal et al.,
Considerable evidence indicates that glucocorticoids also 2002) or dorsal hippocampus (Roozendaal et al., 1999). These
interact with training-associated noradrenergic activation findings, indicating that glucocorticoid effects on memory
within the BLA in enhancing the consolidation of memory of consolidation require concurrent activation of the b-adreno-
emotionally arousing training experiences (Roozendaal et al., ceptor-cAMP/PKA pathway in the BLA, are thus consistent
2009a). As shown in Figure 2(a), an in vivo microdialysis study with the notion that glucocorticoid administration selectively
reported that systemic administration of a memory-enhancing modulates memory consolidation of emotionally arousing
dose of corticosterone after inhibitory avoidance training information that induces the release of norepinephrine within
rapidly augmented norepinephrine levels within the amygdala the BLA. Other findings suggested that glucocorticoids
(McReynolds et al., 2010). In contrast, the same dose of corti- enhance memory consolidation, in a permissive fashion, by
costerone administered to nontrained control rats did not potentiating b-adrenoceptor-cAMP/PKA efficacy in the BLA
increase amygdala norepinephrine levels (Figure 2(b)). More- (Roozendaal et al., 2002). Posttraining intra-BLA infusions of
over, attenuation of noradrenergic signaling with infusions of the b-adrenoceptor agonist clenbuterol or the cAMP analog
Adrenal Stress Hormone Effects on Memory 429

8-bromo-cAMP are known to dose-dependently enhance neural plasticity and cellular activity in BLA neurons (Joëls
memory consolidation. Importantly, suppression of glucocorti- et al., 2011). Corticosterone applied simultaneously with
coid signaling with a GR antagonist infused into the BLA a b-adrenoceptor agonist rapidly facilitates AMPA receptor-
shortly before training reduced the sensitivity of BLA neurons mediated responses in BLA neurons (Liebmann et al., 2009).
to the memory-enhancing effects of clenbuterol such that Nongenomic glucocorticoid actions likely involve the activa-
a much higher dose of clenbuterol was required to induce tion of a membrane-associated variant(s) of the steroid
memory enhancement (Roozendaal et al., 2002). In contrast, receptor (Dallman, 2005; Johnson et al., 2005; Losel et al.,
the GR antagonist did not modify the dose–response effects 2003; Riedemann et al., 2010). In support of this view, recent
of 8-bromo-cAMP, indicating that cAMP acts in the BLA down- findings indicate that the administration of the membrane-
stream from the locus of interaction of glucocorticoids with the impermeable glucocorticoid ligand cort:BSA, corticosterone
b-adrenoceptor-cAMP/PKA pathway. In subsequent studies we conjugated to a large bovine-serum albumin molecule, into
reported that such glucocorticoid effects on potentiating a variety of brain regions of the rat is sufficient to enhance
b-adrenoceptor-cAMP activity results in increased phosphoryla- the consolidation of long-term memory of emotionally
tion of CREB protein (Roozendaal et al., 2006b, 2010). Collec- arousing training experiences (Lee et al., 2011; Roozendaal
tively, these findings indicate that glucocorticoids interact with et al., 2010). As these cort:BSA effects are blocked by coadmin-
the noradrenergic system in modulating the molecular istration of a GR antagonist (Barsegyan et al., 2010;
machinery involved in memory consolidation to selectively Roozendaal et al., 2010), these findings suggest a role for
strengthen the consolidation of long-term memory of emotion- a membrane-associated GR in mediating rapid glucocorticoid
ally significant events. effects on memory.
An interaction between glucocorticoids and noradrenergic Recent findings indicate that such rapid actions of glucocorti-
activity within the amygdala in emotionally influenced coid hormones on memory consolidation involve the endocan-
memory has also been investigated in studies using functional nabinoid system. The endocannabinoid system is a fast lipid
MRI (fMRI) in healthy humans. van Stegeren et al. (2007) re- system in the brain and recently emerged as an important
ported that the relationship between amygdala activity during stress–response system (Atsak et al., 2012b; Campolongo
encoding and subsequent long-term memory was greatest for et al., 2009; Evanson et al., 2010; Hill and McEwen, 2009; Hill
the most emotionally arousing stimuli and for participants and Tasker, 2012; Morena et al., 2016). The first evidence for
with higher endogenous levels of cortisol. Importantly, b-adre- a role of the endocannabinoid system in regulating glucocorti-
noceptor antagonists blocked both the increase in amygdala coid effects on the brain originated from an elegant series of
activity and the enhanced retention induced by emotional in vitro studies by Tasker and colleagues. They demonstrated
stimuli. Kukolja et al. (2008) furthermore showed increased that corticosterone rapidly induced the release of endocannabi-
activity in the amygdala in response to negatively arousing noids in the hypothalamus. Endocannabinoids then act in
images after combined administration of reboxetine and a retrograde fashion to inhibit the release of glutamate in the par-
hydrocortisone but not with either drug alone. Highlighting aventricular nucleus and suppress HPA-axis activity (Di et al.,
again the fact that the effects of hydrocortisone depend on 2003, 2005). More recently, an in vivo study by Hill et al.
concurrent noradrenergic activity, a study with administration (2010) corroborated these findings and showed that
of only hydrocortisone showed suppression of amygdala a single injection of corticosterone rapidly (within 10 min)
activity (Henckens et al., 2010). elevated levels of the endocannabinoid anandamide in the
Importantly, glucocorticoid interactions with the noradren- hypothalamus but also in the amygdala and hippocampus.
ergic system are not limited to the BLA. For example, Endocannabinoid transmission appears to be essentially
a b-adrenoceptor antagonist administered into the nucleus involved in mediating glucocorticoid effects on memory
accumbens shell prevented glucocorticoid-induced memory consolidation as the cannabinoid type 1 (CB1) receptor
enhancement on both an appetitive and an aversive version antagonist AM251 administered into the BLA blocked the
of taste learning (Wichmann et al., 2012). Posttraining infusion ability of posttraining systemic corticosterone to facilitate
of the GR agonist RU 28362 into the medial prefrontal cortex memory consolidation of inhibitory avoidance training
also enhances memory consolidation of inhibitory avoidance (Campolongo et al., 2009). Similarly, a CB1 receptor
training (Roozendaal et al., 2009b), and a b-adrenoceptor antagonist infused into the hippocampus blocked memory
antagonist or PKA inhibitor coinfused into the medial enhancement induced by the synthetic glucocorticoid
prefrontal cortex prevented this memory enhancement dexamethasone (de Oliveira Alvares et al., 2010). As shown in
(Barsegyan et al., 2010). Moreover, corticosterone adminis- Figure 3(a), training rats on an emotionally arousing
tered systemically immediately after inhibitory avoidance inhibitory avoidance task with high footshock intensity rapidly
training increased PKA activity in the medial prefrontal cortex increased anandamide levels in the BLA, whereas training rats
within 30 min. on this task with a lower footshock intensity did not increase
Several experimental findings further suggested that gluco- anandamide levels (Morena et al., 2014). These findings thus
corticoid effects on increasing noradrenergic signaling might indicate that endocannabinoids are released within the BLA
have an onset that is too fast to be mediated via transcriptional during emotionally arousing training conditions that are
regulation in the nucleus and likely involve a rapid, nonge- associated with glucocorticoid release. To investigate whether
nomic mode of action (Dallman, 2005; de Kloet, 2000; Popoli glucocorticoid–endocannabinoid interactions on memory
et al., 2012). Findings of electrophysiological experiments in consolidation are dependent upon an adrenal steroid receptor
brain slices are consistent with the view that glucocorticoids on the cell surface, we performed an additional experiment. As
rapidly interact with the noradrenergic system to influence shown in Figure 3(b) and 3(c), the CB1 receptor antagonist
430 Adrenal Stress Hormone Effects on Memory

(a) (b)

(c) (d)

Figure 3 Glucocorticoids interact with the endocannabinoid system of the BLA in enhancing memory consolidation of inhibitory avoidance training.
(a) Training rats on an inhibitory avoidance task with a high footshock (high FS; 0.45 mA, 1 s), but not low footshock (Low FS; 0.35 mA, 1 s), inten-
sity increased anandamide (AEA) levels in the amygdala 10, 30, and 60 min after the training. *, p < 0.05; **, p < 0.01 vs the no footshock (No FS)
group. Results represent mean  SEM. (b) Forty-eight-hour retention latencies (in seconds, mean  SEM) of rats given immediate posttraining intra-
BLA infusions of the GR agonist RU 28362 (1, 3, or 10 ng in 0.2 mL) alone or together with the CB1 receptor antagonist AM251 (0.14 ng). **,
p < 0.01 vs vehicle; , p < 0.01 vs RU 28362 alone. (c) Forty-eight-hour retention latencies of rats given immediate posttraining intra-BLA infu-
sions of the membrane-impermeable glucocorticoid cort:BSA (1, 3, or 10 ng in 0.2 mL) alone or together with AM251 (0.14 ng). *, p < 0.05; **,
p < 0.01 vs vehicle; , p < 0.05 vs cort:BSA alone. (d) Forty-eight-hour retention latencies of rats given immediate posttraining intra-BLA infusions
of the cannabinoid agonist WIN55,212-2 (10, 30, or 100 ng in 0.2 mL) alone or together with the GR antagonist RU 38486 (1 ng). **, p < 0.01 vs
vehicle. Adapted from Morena, M., Roozendaal, B., Trezza, V., Ratano, P., Peloso, A., Hauer, D., Atsak, P., et al., 2014. Endogenous cannabinoid
release within prefrontal-limbic pathways affects memory consolidation of emotional training. Proc. Natl. Acad. Sci. U.S.A. 111 (51), 18333–18338;
Atsak, P., Hauer, D., Campolongo, P., Schelling, G., Fornari, R.V., Roozendaal, B., 2015. Endocannabinoid signaling within the basolateral amygdala
integrates multiple stress hormone effects on memory consolidation. Neuropsychopharmacology 40 (6), 1485–1494.

AM251 infused into the BLA blocked the memory-enhancing activity or, alternatively, alter noradrenergic function indi-
effects induced by concurrent infusions of either a specific GR rectly via a modulation of GABAergic or glutamatergic
agonist or the membrane-impermeable ligand cort:BSA activity. Within the BLA, CB1 receptors are in particular
(Atsak et al., 2015). In contrast, the GR antagonist RU 38486 abundantly expressed on GABAergic interneurons (Katona
infused into the BLA did not alter the memory-enhancing et al., 2001) and activation of CB1 receptors has consistently
effects of a CB1 receptor agonist (Figure 3(d)). Therefore, these been shown to suppress the release of GABA (Katona et al.,
findings indicate that endocannabinoid transmission is 2001, 1999; Ohno-Shosaku et al., 2001) via a rapid inhibi-
required for mediating glucocorticoid effects on memory tion of calcium entry into the terminals (Hoffman and Lup-
consolidation, presumably involving the activation of a GR on ica, 2000; Wilson and Nicoll, 2001). Interestingly, inhibition
the cell surface. Subsequent experiments indicated that such of GABAergic transmission with GABA receptor antagonists
glucocorticoid interactions with the endocannabinoid system is known to enhance memory consolidation by stimulating
also mediate the rapid effects of glucocorticoids onto the norepinephrine release (Hatfield and McGaugh, 1999). As
noradrenergic system (Atsak et al., 2015). These findings are in illustrated in Figure 4, these findings thus suggest that corti-
line with prior evidence that systemic or local administration costerone might bind to a GR on the cell surface and rapidly
of a CB1 receptor agonist increases norepinephrine levels in induce the release of endocannabinoids. The released endo-
limbic and cortical brain regions (Oropeza et al., 2005; Page cannabinoids then bind to CB1 receptors on GABAergic
et al., 2007). interneurons and inhibit the release of GABA that can then
Such glucocorticoid-induced increases in endocannabi- result in an increased noradrenergic transmission in BLA
noid signaling might either directly influence noradrenergic neurons.
Adrenal Stress Hormone Effects on Memory 431

before memory consolidation took place and when glucocorti-


coid levels are still elevated (Diamond et al., 1996; Kirschbaum
et al., 1996), provided the first indication that glucocorticoids
might influence memory retrieval processes. Subsequent
studies specifically investigated this issue by exposing rats to
stress or by administering glucocorticoids shortly before reten-
tion testing. Stress exposure or systemic corticosterone admin-
istration to rats shortly before testing for memory of training on
inhibitory avoidance, contextual fear conditioning, or water-
maze spatial tasks (24 h earlier) produces temporary impair-
ment of retention performance (Bohus, 1973; Cai et al.,
2006; de Quervain et al., 1998; Pakdel and Rashidy-Pour,
2006; Roozendaal et al., 2004a; Sajadi et al., 2006; Yang
et al., 2003). As the same treatments administered shortly
before training do not affect either acquisition or retention
performance assessed immediately after acquisition, such find-
ings indicate that glucocorticoids impair retention by influ-
encing memory retrieval. Several further studies from
different laboratories have indicated that glucocorticoids
impair memory retrieval of spatial or contextual memory in
rats and declarative (mostly episodic) memory in humans
(Buss et al., 2004; Coluccia et al., 2008; de Quervain et al.,
Figure 4 A model illustrating the role of the endocannabinoid system 2000, 2003; Het et al., 2005; Kuhlmann and Wolf, 2005;
in integrating the effects of glucocorticoids and norepinephrine within Rashidy-Pour et al., 2004; Roozendaal et al., 2003, 2004b;
the BLA on memory consolidation. Glucocorticoids, released during Sajadi et al., 2007; Wolf, 2008; Wolf et al., 2001). Moreover,
emotionally arousing situations, bind to a membrane-bound GR and increased cortisol levels due to psychological stress have also
activate the intracellular cAMP/PKA signaling cascade. This triggers the been shown to impair declarative memory retrieval (Buchanan
release of endocannabinoids, particularly anandamide (AEA). Ananda- et al., 2006; Domes et al., 2004; Kuhlmann et al., 2005b).
mide then activates CB1 receptors on GABAergic interneurons and Although the vast majority of studies have investigated the
thereby inhibits GABA release. This subsequently disinhibits norepi- effects of stress and glucocorticoids on the retrieval of
nephrine (NE) release and increases the excitability of pyramidal
hippocampus-dependent forms of memory, some animal and
neurons within the BLA. This overall increases the sensitivity of BLA
human studies have shown that stress exposure or glucocorti-
neurons to the effects of norepinephrine and results in an increased
activation of the cAMP/PKA pathway and phosphorylation of CREB coid administration before retention testing also impairs the
protein. These stress hormone effects in the BLA are required for the retrieval of cortex-dependent recognition memory (Barsegyan
optimal enhancement of memory for emotionally arousing experiences et al., 2015) and striatal-dependent stimulus–response associa-
by influencing information storage processes in other brain regions. tions (Atsak et al., 2016; Guenzel et al., 2013).
Adapted from Atsak, P., Hauer, D., Campolongo, P., Schelling, G., Glucocorticoid effects on memory retrieval are highly
Fornari, R.V., Roozendaal, B., 2015. Endocannabinoid signaling within comparable to those seen in studies investigating memory
the basolateral amygdala integrates multiple stress hormone effects on consolidation in that the effects depend on emotional arousal.
memory consolidation. Neuropsychopharmacology 40 (6), 1485–1494. Specifically, it has been shown in studies in humans that
emotionally arousing information is especially sensitive to
1.15.4 Adrenal Stress Hormone Effects on Other the retrieval-impairing effects of glucocorticoids (Buchanan
Memory Functions et al., 2006; Buchanan and Adolphs, 2004; Buss et al., 2004;
de Quervain et al., 2000, 2007a; Het et al., 2005; Kuhlmann
Most studies investigating the effects of adrenal stress hormones et al., 2005a,b; Kuhlmann and Wolf, 2006; Schwabe and
on memory have focused on the neurobiological processes Wolf, 2009; Smeets et al., 2008, 2009; Tollenaar et al., 2009;
underlying the consolidation of recent experiences. There is, Wolf et al., 2001). Studies of rats investigating the neural mech-
however, considerable evidence that adrenal stress hormones anisms underlying this selectivity have indicated that glucocor-
also influence memory retrieval and working memory. In ticoid effects on memory retrieval depend critically on
contrast to the enhancing effects of stress hormones on memory noradrenergic activity within the brain. The b-adrenoceptor
consolidation, adrenal stress hormones, and in particular gluco- antagonist propranolol administered systemically 30 min
corticoids, have been shown to impair memory retrieval and before inhibitory avoidance retention testing blocks the
working memory. As will be discussed below, these stress memory retrieval impairment induced by concurrent injections
hormone effects also depend on rapidly mediated interactions of corticosterone (Roozendaal et al., 2004a). A b-adrenoceptor
with both the noradrenergic and endocannabinoid systems. antagonist infused into the hippocampus also prevents the
retrieval-impairing effect of a GR agonist administered concur-
rently (Roozendaal et al., 2004b). Together with the finding
1.15.4.1 Memory Retrieval
that norepinephrine is activated by emotional arousal, this
Studies showing that stress induces subsequent memory could explain why emotionally arousing information or an
impairments when retention is tested soon after learning, emotionally arousing test situation is a prerequisite for
432 Adrenal Stress Hormone Effects on Memory

enabling glucocorticoid effects on memory retrieval. In line contextual fear memory were examined. The CB receptor
with this idea and illustrated in Figure 5, it has been reported agonist WIN55,212-2 infused into the hippocampus 1 h before
that the b-adrenoceptor antagonist propranolol blocked the retention testing impaired the retrieval of contextual fear
impairing effect of cortisone administration on the retrieval memory; however, the b-adrenoceptor antagonist propranolol
of emotionally arousing material in healthy humans (de blocked the impairing effect of WIN55,212-2 on memory
Quervain et al., 2007a). Another study indicated that propran- retrieval. Conversely, the CB1 receptor antagonist AM251
olol also blocked the impairing effect of stress induction on infused into hippocampus together with an impairing dose
memory retrieval (Schwabe et al., 2009). Further studies have of norepinephrine failed to abolish the impairing effect of
indicated that arousal-induced BLA activation interacts with norepinephrine on memory retrieval.
the hippocampus in mediating glucocorticoid effects on the
retrieval of emotionally arousing information. Lesions of the
1.15.4.2 Working Memory
BLA or infusions of a b-adrenoceptor antagonist into the BLA
block the impairing effect of a GR agonist infused into the Stress exposure is also known to impair working memory
hippocampus on memory retrieval of spatial information (Arnsten and Goldman-Rakic, 1998; Schoofs et al., 2008).
(Roozendaal et al., 2003, 2004b). In support of the importance Working memory is a dynamic process whereby information
of an interaction between the amygdala and the hippocampus is updated continuously, providing a temporary storage of
during retrieval of emotionally arousing information, human information (Baddeley, 1992; Jones, 2002). Evidence from
imaging studies have indicated that the degree of interaction lesion, pharmacological, imaging, and clinical studies indicates
between these two brain regions is greater during the retrieval that working memory depends on the integrity of the medial
of emotionally arousing declarative information as compared prefrontal cortex (Brito et al., 1982; Fuster, 1991; Lee and
to neutral information (Dolcos et al., 2005; Smith et al., 2006). Kesner, 2003; Levy and Farrow, 2001; Rowe et al., 2000; Stern
The temporary effects of glucocorticoids on memory et al., 2001; Taylor et al., 1999). Basal levels of endogenous
retrieval impairment are fast and do not seem to depend on glucocorticoids are required to maintain prefrontal cortical
gene transcription (Sajadi et al., 2006). Recent findings indicate function (Mizoguchi et al., 2004), but systemic injections of
that endocannabinoid signaling is also involved in mediating stress doses of corticosterone or a GR agonist administered
glucocorticoid-induced impairment of memory retrieval. For into the prelimbic region of the medial prefrontal cortex
this experiment, rats were trained on a contextual fear condi- impair delayed alternation performance in rats, a task
tioning task and tested 24 h later for fear memory retention commonly used to assess working memory performance in
(Atsak et al., 2012a). Blockade of hippocampal CB1 receptors rodents (Roozendaal et al., 2004c). As similar GR agonist infu-
by local infusions of AM251, 1 h before retention testing pre- sions into the medial prefrontal cortex do not impair delayed
vented the impairing effects of systemically coadministered alternation performance on nonmnemonic control tasks that
glucocorticoids on retrieval of contextual fear memory. More- have similar motivational and motor demands (Barsegyan
over, a retrieval-impairing dose of corticosterone elevated et al., 2010), these findings indicate that stress levels of gluco-
hippocampal levels of 2-arachidonoyl glycerol (2-AG), but corticoids, via GR activation, impair working memory. Addi-
not anandamide. As mentioned before, glucocorticoid effects tionally, stress-level cortisol treatment impairs prefrontal
on memory retrieval highly depend on noradrenergic activity. cortex-dependent inhibitory control of behaviors in squirrel
Thus, in order to determine whether endocannabinoids monkeys (Lyons et al., 2000), as well as working memory
mediate the effects of glucocorticoids on the noradrenergic performance in human subjects during demanding tasks which
system, possible interactions between the endocannabinoid require a high level of arousal (Lupien et al., 1999; Wolf et al.,
and noradrenergic systems during retrieval processing of 2001; Young et al., 1999).

(a) (b)

Figure 5 Glucocorticoid effects on memory retrieval depend on emotional arousal and noradrenergic activity. (a) Cortisone (25 mg) administered to
humans 1 h before recall testing impaired retrieval of high-arousal words, but not of low-arousal words. (b) Concurrent administration of propranolol
(40 mg) prevented the impairing effect of cortisone on the recall of high-arousal words. Data are presented as mean þ SEM. **, p < 0.0001 vs the
corresponding placebo condition. Adapted from de Quervain, D.J.-F., Aerni, A., Roozendaal, B., 2007. Preventive effect of beta-adrenoceptor blockade
on glucocorticoid-induced memory retrieval deficits. Am. J. Psychiatry 164 (6), 967–969.
Adrenal Stress Hormone Effects on Memory 433

Importantly, glucocorticoid effects on working memory Arnsten, A.F., Goldman-Rakic, P.S., 1998. Noise stress impairs prefrontal cortical
impairment also depend on interactions with arousal- cognitive function in monkeys: evidence for a hyperdopaminergic mechanism.
Arch. Gen. Psychiatry 55 (4), 362–368.
associated noradrenergic mechanisms. In humans, negative
Atsak, P., Hauer, D., Campolongo, P., Schelling, G., McGaugh, J.L., Roozendaal, B.,
effects of stress induction on working memory (Elzinga and 2012a. Glucocorticoids interact with the hippocampal endocannabinoid system in
Roelofs, 2005; Luethi et al., 2008; Oei et al., 2006; Taverniers impairing retrieval of contextual fear memory. Proc. Natl. Acad. Sci. U.S.A. 109 (9),
et al., 2010; Qin et al., 2012; Schoofs et al., 2008, 2009) and 3504–3509.
prefrontal cortex activity (Ossewaarde et al., 2011; Qin et al., Atsak, P., Roozendaal, B., Campolongo, P., 2012b. Role of the endocannabinoid
system in regulating glucocorticoid effects on memory for emotional experiences.
2009) are usually observed within the time window in Neuroscience 204, 104–116.
which both glucocorticoid and noradrenergic activity are Atsak, P., Hauer, D., Campolongo, P., Schelling, G., Fornari, R.V., Roozendaal, B.,
elevated. In rats, a b-adrenoceptor antagonist administered 2015. Endocannabinoid signaling within the basolateral amygdala integrates
systemically blocks the impairing effect of corticosterone on multiple stress hormone effects on memory consolidation. Neuro-
psychopharmacology 40 (6), 1485–1494.
working memory (Roozendaal et al., 2004c). Furthermore,
Atsak, P., Guenzel, F.M., Kantar-Gok, D., Zalachoras, I., Yargicoglu, P.,
a b-adrenoceptor antagonist or cAMP blocker infused into the Meijer, O.C., Quirarte, G.L., et al., 2016. Glucocorticoids mediate stress-
medial prefrontal cortex blocks working memory impairment induced impairment of retrieval of stimulus - response memory. Psychoneur-
induced by a GR agonist administered concurrently (Barsegyan oendocrinology 67.
et al., 2010). Animal studies have shown that glucocorticoid Atucha, E., Zalachoras, I., van den Heuvel, J.K., van Weert, L.T.C.M., Melchers, D.,
Mol, I.M., Belanoff, J.K., et al., 2015. A mixed glucocorticoid/mineralocorticoid
effects on working memory also depend on functional interac- selective modulator with dominant antagonism in the male rat brain. Endocrinology
tions between the BLA and the medial prefrontal cortex. The 156 (11), 4105–4114.
BLA both sends projections to and receives projections from Axelrod, J., Reisine, T.D., 1984. Stress hormones: their interaction and regulation.
the medial prefrontal cortex (Likhtik et al., 2005; McDonald, Science 224 (4648), 452–459.
Baddeley, A., 1992. Working memory. Science 255 (5044), 556–559.
1991; Pérez-Jaranay and Vives, 1991; Rosenkranz and Grace,
Barrett, D., Gonzalez-Lima, F., 2004. Behavioral effects of metyrapone on Pavlovian
2002). Drug administration into the BLA does not appear to extinction. Neurosci. Lett. 371 (2–3), 91–96.
affect working memory (Bianchin et al., 1999; Wan et al., Barsegyan, A., Mackenzie, S.M., Kurose, B.D., McGaugh, J.L., Roozendaal, B., 2010.
1994), but disruption of BLA activity blocks the impairment Glucocorticoids in the prefrontal cortex enhance memory consolidation and impair
induced by either systemic administration of corticosterone working memory by a common neural mechanism. Proc. Natl. Acad. Sci. U.S.A.
107 (38), 16655–16660.
or infusions of a GR agonist into the medial prefrontal cortex Barsegyan, A., McGaugh, J.L., Roozendaal, B., 2014. Noradrenergic activation of the
(Roozendaal et al., 2004b). basolateral amygdala modulates the consolidation of object-in-context recognition
memory. Front. Behav. Neurosci. 8, 160.
Barsegyan, A., Atsak, P., Hornberger, W.B., Jacobson, P.B., van Gaalen, M.M.,
Roozendaal, B., 2015. The vasopressin 1b receptor antagonist A-988315 blocks
1.15.5 Concluding Remarks stress effects on the retrieval of object-recognition memory. Neuro-
psychopharmacology 40 (8), 1979–1989.
The past decades have witnessed a steady increase in our under- Beato, M., Herrlich, P., Schütz, G., 1995. Steroid hormone receptors: many actors in
standing of how adrenal stress hormones affect different search of a plot. Cell 83 (6), 851–857.
Bianchin, M., Mello e Souza, T., Medina, J.H., Izquierdo, I., 1999. The amygdala is
aspects of memory function. This knowledge is increasingly
involved in the modulation of long-term memory, but not in working or short-term
used to gain a mechanistic understanding of the etiology and memory. Neurobiol. Learn. Mem. 71 (2), 127–131.
treatment of intense emotionally arousing or traumatic Bohus, B., Lissák, K., 1968. Adrenocortical hormones and avoidance behaviour of rats.
memory in psychopathology. For instance, the finding that Int. J. Neuropharmacol. 7 (4), 301–306.
glucocorticoids impair memory retrieval of emotionally Bohus, B., 1973. Pituitary-adrenal influences on avoidance and approach behavior of
the rat. In: Zimmerman, E., Gispen, W.H., Marks, B.H., de Wied, D. (Eds.),
arousing experiences has been successfully translated to patient Progress in Brain Research: Drug Effects on Neuroendocrine Regulations. Elsevier,
studies to alleviate symptoms of chronic stress after a highly Amsterdam, the Netherlands, pp. 407–430.
traumatic experience (Schelling et al., 2001) and in patients Borrell, J., de Kloet, E.R., Versteeg, D.H., Bohus, B., 1983. Inhibitory avoidance deficit
with phobic fear (Soravia et al., 2006). A recent study further following short-term adrenalectomy in the rat: the role of adrenal catecholamines.
Behav. Neural Biol. 39 (2), 241–258.
showed that administration of hydrocortisone can enhance
Borrell, J., de Kloet, E.R., Bohus, B., 1984. Corticosterone decreases the efficacy of
the efficacy of extinction-based psychotherapy such as exposure adrenaline to affect passive avoidance retention of adrenalectomized rats. Life Sci.
therapy in phobic patients (de Quervain et al., 2011). A critical 34 (1), 99–104.
future challenge for this field is therefore to apply the detailed Brito, G.N., Thomas, G.J., Davis, B.J., Gingold, S.I., 1982. Prelimbic cortex, medi-
knowledge acquired in experimental work to improve treat- odorsal thalamus, septum, and delayed alternation in rats. Exp. Brain Res. 46 (1),
52–58.
ment and prevention of memory-related and stress-related Buchanan, T.W., Adolphs, R., 2004. The neuroanatomy of emotional memory in
disorders such as posttraumatic stress disorder and phobias. humans. In: Reisberg, D., Hertel, P. (Eds.), Memory and Emotion. Oxford University
Press, New York, NY.
Buchanan, T.W., Lovallo, W.R., 2001. Enhanced memory for emotional material
following stress-level cortisol treatment in humans. Psychoneuroendocrinology 26
References (3), 307–317.
Buchanan, T.W., Tranel, D., Adolphs, R., 2006. Impaired memory retrieval correlates
Abercrombie, H.C., Kalin, N.H., Thurow, M.E., Rosenkranz, M.A., Davidson, R.J., with individual differences in cortisol response but not autonomic response. Learn.
2003. Cortisol variation in humans affects memory for emotionally laden and Mem. 13 (3), 382–387.
neutral information. Behav. Neurosci. 117 (3), 505–516. Buss, C., Wolf, O.T., Witt, J., Hellhammer, D.H., 2004. Autobiographic memory
Abercrombie, H.C., Speck, N.S., Monticelli, R.M., 2006. Endogenous cortisol eleva- impairment following acute cortisol administration. Psychoneuroendocrinology 29
tions are related to memory facilitation only in individuals who are emotionally (8), 1093–1096.
aroused. Psychoneuroendocrinology 31 (2), 187–196. Cahill, L., Alkire, M.T., 2003. Epinephrine enhancement of human memory consoli-
Andreano, J.M.J., Cahill, L.L., 2006. Glucocorticoid release and memory consolidation dation: interaction with arousal at encoding. Neurobiol. Learn. Mem. 79 (2),
in men and women. Psychol. Sci. 17 (6), 466–470. 194–198.
434 Adrenal Stress Hormone Effects on Memory

Cahill, L., McGaugh, J.L., 1991. NMDA-induced lesions of the amygdaloid complex Ferry, B., Roozendaal, B., McGaugh, J.L., 1999a. Involvement of alpha1-
block the retention-enhancing effect of posttraining epinephrine. Psychobiology adrenoceptors in the basolateral amygdala in modulation of memory storage.
19 (3), 206–210. Eur. J. Pharmacol. 372 (1), 9–16.
Cahill, L., Prins, B., Weber, M., McGaugh, J.L., 1994. Beta-adrenergic activation and Ferry, B., Roozendaal, B., McGaugh, J.L., 1999b. Basolateral amygdala noradrenergic
memory for emotional events. Nature 371 (6499), 702–704. influences on memory storage are mediated by an interaction between beta- and
Cahill, L., Gorski, L., Le, K., 2003. Enhanced human memory consolidation with post- alpha1-adrenoceptors. J. Neurosci. 19 (12), 5119–5123.
learning stress: interaction with the degree of arousal at encoding. Learn. Mem. 10 Flood, J.F., Vidal, D., Bennett, E.L., Orme, A.E., Vasquez, S., Jarvik, M.E., 1978.
(4), 270–274. Memory facilitating and anti-amnesic effects of corticosteroids. Pharmacol. Bio-
Cai, W.-H., Blundell, J., Han, J., Greene, R.W., Powell, C.M., 2006. Postreactivation chem. Behav. 8 (1), 81–87.
glucocorticoids impair recall of established fear memory. J. Neurosci. 26 (37), Fuster, J.M., 1991. The prefrontal cortex and its relation to behavior. Prog. Brain Res.
9560–9566. 87, 201–211.
Campolongo, P., Roozendaal, B., Trezza, V., Hauer, D., Schelling, G., McGaugh, J.L., Galvez, R., Mesches, M.H., McGaugh, J.L., 1996. Norepinephrine release in the
Cuomo, V., 2009. Endocannabinoids in the rat basolateral amygdala enhance amygdala in response to footshock stimulation. Neurobiol. Learn. Mem. 66 (3),
memory consolidation and enable glucocorticoid modulation of memory. Proc. Natl. 253–257.
Acad. Sci. U.S.A. 106 (12), 4888–4893. Gerard, R.W., 1961. The fixation of experience. In: Fessard, A., Gerard, R.W.,
Clark, K.B., Naritoku, D.K., Smith, D.C., Browning, R.A., Jensen, R.A., 1999. Konorski, J. (Eds.), Brain Mechanisms and Learning. Charles C. Thomas,
Enhanced recognition memory following vagus nerve stimulation in human Springfield, IL, pp. 21–35.
subjects. Nat. Neurosci. 2 (1), 94–98. Gold, P.E., Van Buskirk, R.B., 1975. Facilitation of time-dependent memory processes
Clayton, E.C., Williams, C.L., 2000. Adrenergic activation of the nucleus tractus sol- with posttrial epinephrine injections. Behav. Biol. 13 (2), 145–153.
itarius potentiates amygdala norepinephrine release and enhances retention Gold, P.E., van Buskirk, R., 1976. Effects of posttrial hormone injections on memory
performance in emotionally arousing and spatial memory tasks. Behav. Brain Res. processes. Horm. Behav. 7 (4), 509–517.
112 (1–2), 151–158. Gold, P.E., van Buskirk, R., 1978. Effects of alpha- and beta-adrenergic receptor
Coluccia, D., Wolf, O.T., Kollias, S., Roozendaal, B., Forster, A., de Quervain, D.J.-F., antagonists on post-trial epinephrine modulation of memory: relationship to post-
2008. Glucocorticoid therapy-induced memory deficits: acute versus chronic training brain norepinephrine concentrations. Behav. Biol. 24 (2), 168–184.
effects. J. Neurosci. 28 (13), 3474–3478. Gold, P.E., 1986. Glucose modulation of memory storage processing. Behav. Neural
Cordero, M.I., Kruyt, N.D., Merino, J.J., Sandi, C., 2002. Glucocorticoid Biol. 45 (3), 342–349.
involvement in memory formation in a rat model for traumatic memory. Stress Gold, P.E., 1995. Role of glucose in regulating the brain and cognition. Am. J. Clin.
5 (1), 73–79. Nutr. 61 (4 Suppl.), 987S–995S.
Cornelisse, S., Joëls, M., Smeets, T., 2011. A randomized trial on mineralocorticoid Guenzel, F.M., Wolf, O.T., Schwabe, L., 2013. Stress disrupts response memory
receptor blockade in men: effects on stress responses, selective attention, and retrieval. Psychoneuroendocrinology 38 (8), 1460–1465.
memory. Neuropsychopharmacology 36 (13), 2720–2728. Hall, J.L., Gold, P.E., 1986. The effects of training, epinephrine, and glucose injections
Costa-Miserachs, D., Portell-Cortés, I., Aldavert-Vera, L., Torras-García, M., Morgado- on plasma glucose levels in rats. Behav. Neural Biol. 46 (2), 156–167.
Bernal, I., 1994. Long-term memory facilitation in rats by posttraining epinephrine. Harris, A.P., Holmes, M.C., de Kloet, E.R., Chapman, K.E., Seckl, J.R., 2013.
Behav. Neurosci. 108 (3), 469–474. Mineralocorticoid and glucocorticoid receptor balance in control of HPA axis and
Cousijn, H., Rijpkema, M., Qin, S., van Marle, H.J.F., Franke, B., Hermans, E.J., behaviour. Psychoneuroendocrinology 38 (5), 648–658.
van Wingen, G., et al., 2010. Acute stress modulates genotype effects on Hassert, D.L., Miyashita, T., Williams, C.L., 2004. The effects of peripheral vagal nerve
amygdala processing in humans. Proc. Natl. Acad. Sci. U.S.A. 107 (21), stimulation at a memory-modulating intensity on norepinephrine output in the
9867–9872. basolateral amygdala. Behav. Neurosci. 118 (1), 79–88.
Dallman, M.F., 2005. Fast glucocorticoid actions on brain: back to the future. Front. Hatfield, T., McGaugh, J.L., 1999. Norepinephrine infused into the basolateral
Neuroendocrinol. 26 (3–4), 103–108. amygdala posttraining enhances retention in a spatial water maze task. Neurobiol.
Datson, N.A., van der Perk, J., de Kloet, E.R., Vreugdenhil, E., 2001. Identification of Learn. Mem. 71 (2), 232–239.
corticosteroid-responsive genes in rat hippocampus using serial analysis of gene Heck, S., Kullmann, M., Gast, A., Ponta, H., Rahmsdorf, H.J., Herrlich, P., Cato, A.C.,
expression. Eur. J. Neurosci. 14 (4), 675–689. 1994. A distinct modulating domain in glucocorticoid receptor monomers in the
De Boer, S.F., Slangen, J.L., Van der Gugten, J., 1990. Plasma catecholamine and repression of activity of the transcription factor AP-1. EMBO J. 13 (17),
corticosterone levels during active and passive shock-prod avoidance behavior in 4087–4095.
rats: effects of chlordiazepoxide. Physiol. Behav. 47 (6), 1089–1098. Henckens, M.J.A.G., van Wingen, G.A., Joëls, M., Fernández, G., 2010. Time-
Di, S., Malcher-Lopes, R., Halmos, K.C., Tasker, J.G., 2003. Nongenomic glucocor- dependent effects of corticosteroids on human amygdala processing.
ticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback J. Neurosci. 30 (38), 12725–12732.
mechanism. J. Neurosci. 23 (12), 4850–4857. Henckens, M.J.A.G., Pu, Z., Hermans, E.J., van Wingen, G.A., Joëls, M.,
Di, S., Malcher-Lopes, R., Marcheselli, V.L., Bazan, N.G., Tasker, J.G., 2005. Rapid Fernández, G., 2012. Dynamically changing effects of corticosteroids on
glucocorticoid-mediated endocannabinoid release and opposing regulation of human hippocampal and prefrontal processing. Hum. Brain Mapp. 33 (12),
glutamate and gamma-aminobutyric acid inputs to hypothalamic magnocellular 2885–2897.
neurons. Endocrinology 146 (10), 4292–4301. Hermans, E.J., van Marle, H.J.F., Ossewaarde, L., Henckens, M.J.A.G., Qin, S., van
Diamond, D.M., Fleshner, M., Ingersoll, N., Rose, G.M., 1996. Psychological stress Kesteren, M.T.R., Schoots, V.C., et al., 2011. Stress-related noradrenergic activity
impairs spatial working memory: relevance to electrophysiological studies of prompts large-scale neural network reconfiguration. Science 334 (6059),
hippocampal function. Behav. Neurosci. 110 (4), 661–672. 1151–1153.
Dolcos, F., Labar, K.S., Cabeza, R., 2005. Remembering one year later: role of the Hermans, E.J., Battaglia, F.P., Atsak, P., de Voogd, L.D., Fernández, G.,
amygdala and the medial temporal lobe memory system in retrieving emotional Roozendaal, B., 2014a. How the amygdala affects emotional memory by altering
memories. Proc. Natl. Acad. Sci. U.S.A. 102 (7), 2626–2631. brain network properties. Neurobiol. Learn. Mem. 112, 2–16.
Domes, G., Heinrichs, M., Rimmele, U., Reichwald, U., Hautzinger, M., 2004. Acute Hermans, E.J., Henckens, M.J.A.G., Joëls, M., Fernández, G., 2014b. Dynamic
stress impairs recognition for positive words–association with stress-induced adaptation of large-scale brain networks in response to acute stressors. Trends
cortisol secretion. Stress 7 (3), 173–181. Neurosci. 37 (6), 304–314.
Dornelles, A., de Lima, M.N.M., Grazziotin, M., Presti-Torres, J., Garcia, V.A., Het, S., Ramlow, G., Wolf, O.T., 2005. A meta-analytic review of the effects of acute
Scalco, F.S., Roesler, R., et al., 2007. Adrenergic enhancement of consolidation of cortisol administration on human memory. Psychoneuroendocrinology 30 (8),
object recognition memory. Neurobiol. Learn. Mem. 88 (1), 137–142. 771–784.
Elzinga, B.M., Roelofs, K., 2005. Cortisol-induced impairments of working memory Hill, M.N., McEwen, B.S., 2009. Endocannabinoids: the silent partner of glucocorti-
require acute sympathetic activation. Behav. Neurosci. 119 (1), 98–103. coids in the synapse. Proc. Natl. Acad. Sci. U.S.A. 106 (12), 4579–4580.
Evanson, N.K., Tasker, J.G., Hill, M.N., Hillard, C.J., 2010. Fast feedback inhibition of Hill, M.N., Tasker, J.G., 2012. Endocannabinoid signaling, glucocorticoid-mediated
the HPA axis by glucocorticoids is mediated by endocannabinoid signaling. negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis.
Endocrinology 151 (10), 4811–4819. Neuroscience 204, 5–16.
Ferguson, D., Sapolsky, R., 2007. Mineralocorticoid receptor overexpression differ- Hill, M.N., McLaughlin, R.J., Bingham, B., Shrestha, L., Lee, T.T.Y., Gray, J.M.,
entially modulates specific phases of spatial and nonspatial memory. J. Neurosci. Hillard, C.J., et al., 2010. Endogenous cannabinoid signaling is essential for stress
27 (30), 8046–8052. adaptation. Proc. Natl. Acad. Sci. U.S.A. 107 (20), 9406–9411.
Adrenal Stress Hormone Effects on Memory 435

Hinkelmann, K., Wingenfeld, K., Kuehl, L.K., Fleischer, J., Heuser, I., Wiedemann, K., Lee, E.J., Son, G.H., Chung, S., Lee, S., Kim, J., 2011. Impairment of fear memory
Otte, C., 2015. Stimulation of the mineralocorticoid receptor improves memory in consolidation in maternally stressed male mouse offspring: evidence for non-
young and elderly healthy individuals. Neurobiol. Aging 36 (2), 919–924. genomic glucocorticoid action on the amygdala. J. Neurosci. 31 (19), 7131–7140.
Hoffman, A.F., Lupica, C.R., 2000. Mechanisms of cannabinoid inhibition of GABA(A) Lembke, A., Gomez, R., Tenakoon, L., Keller, J., Cohen, G., Williams, G.H.,
synaptic transmission in the hippocampus. J. Neurosci. 20 (7), 2470–2479. Kraemer, F.B., et al., 2013. The mineralocorticoid receptor agonist, flu-
Huff, N.C., Wright-Hardesty, K.J., Higgins, E.A., Matus-Amat, P., Rudy, J.W., 2005. drocortisone, differentially inhibits pituitary-adrenal activity in humans with
Context pre-exposure obscures amygdala modulation of contextual-fear condi- psychotic major depression. Psychoneuroendocrinology 38 (1), 115–121.
tioning. Learn. Mem. 12 (5), 456–460. Levy, F., Farrow, M., 2001. Working memory in ADHD: prefrontal/parietal connections.
Introini-Collison, I.B., McGaugh, J.L., 1986. Epinephrine modulates long-term Curr. Drug Targets 2 (4), 347–352.
retention of an aversively motivated discrimination. Behav. Neural Biol. 45 (3), Liang, K.C., McGaugh, J.L., 1983. Lesions of the stria terminalis attenuate the
358–365. enhancing effect of post-training epinephrine on retention of an inhibitory avoid-
Introini-Collison, I., Saghafi, D., Novack, G.D., McGaugh, J.L., 1992. Memory- ance response. Behav. Brain Res. 9 (1), 49–58.
enhancing effects of post-training dipivefrin and epinephrine: involvement of Liang, K.C., Bennett, C., McGaugh, J.L., 1985. Peripheral epinephrine modulates the
peripheral and central adrenergic receptors. Brain Res. 572 (1–2), 81–86. effects of post-training amygdala stimulation on memory. Behav. Brain Res. 15 (2),
Izquierdo, I., Dias, R.D., 1985. Influence on memory of posttraining or pre-test 93–100.
injections of ACTH, vasopressin, epinephrine, and beta-endorphin, and their Liang, K.C., Juler, R.G., McGaugh, J.L., 1986. Modulating effects of posttraining
interaction with naloxone. Psychoneuroendocrinology 10 (2), 165–172. epinephrine on memory: involvement of the amygdala noradrenergic system. Brain
Izquierdo, I., Medina, J.H., Jeriisalinsky, D., Da Cunha, C., 1992. Post-training memory Res. 368 (1), 125–133.
processing in amygdala, septum and hippocampus: role of benzodiazepine/GABAA Liang, K.C., Chen, L.L., Huang, T.E., 1995. The role of amygdala norepinephrine in
receptors, and their interaction with other neurotransmitter systems. Rev. Neurosci. memory formation: involvement in the memory enhancing effect of peripheral
3 (1), 11–24. epinephrine. Chin. J. Physiol. 38 (2), 81–91.
Joëls, M., Baram, T.Z., 2009. The neuro-symphony of stress. Nat. Rev. Neurosci. 10 Liang, K.C., 1998. Pretraining infusion of DSP-4 into the amygdala impaired retention
(6), 459–466. in the inhibitory avoidance task: involvement of norepinephrine but not serotonin in
Joëls, M., Karst, H., DeRijk, R., de Kloet, E.R., 2008. The coming out of the brain memory facilitation. Chin. J. Physiol. 41 (4), 223–233.
mineralocorticoid receptor. Trends Neurosci. 31 (1), 1–7. Liebmann, L., Karst, H., Joëls, M., 2009. Effects of corticosterone and the beta-
Joëls, M., Fernández, G., Roozendaal, B., 2011. Stress and emotional memory: agonist isoproterenol on glutamate receptor-mediated synaptic currents in the
a matter of timing. Trends Cogn. Sci. 15 (6), 280–288. rat basolateral amygdala. Eur. J. Neurosci. 30 (5), 800–807.
Johnson, L.R., Farb, C., Morrison, J.H., McEwen, B.S., LeDoux, J.E., 2005. Locali- Likhtik, E., Pelletier, J.G., Paz, R., Paré, D., 2005. Prefrontal control of the amygdala.
zation of glucocorticoid receptors at postsynaptic membranes in the lateral J. Neurosci. 25 (32), 7429–7437.
amygdala. Neuroscience 136 (1), 289–299. Liu, W., Wang, J., Sauter, N.K., Pearce, D., 1995. Steroid receptor hetero-
Jones, M.W., 2002. A comparative review of rodent prefrontal cortex and working dimerization demonstrated in vitro and in vivo. Proc. Natl. Acad. Sci. U.S.A. 92
memory. Curr. Mol. Med. 2 (7), 639–647. (26), 12480–12484.
de Kloet, E.R., Joëls, M., Holsboer, F., 2005. Stress and the brain: from adaptation to Losel, R.M., Falkenstein, E., Feuring, M., Schultz, A., Tillmann, H.-C., Rossol-
disease. Nat. Rev. Neurosci. 6 (6), 463–475. Haseroth, K., Wehling, M., 2003. Nongenomic steroid action: controversies,
de Kloet, E.R., 2000. Stress in the brain. Eur. J. Pharmacol. 405 (1–3), 187–198. questions, and answers. Physiol. Rev. 83 (3), 965–1016.
Karst, H., Berger, S., Turiault, M., Tronche, F., Schütz, G., Joëls, M., 2005. Miner- Lovallo, W.R., Robinson, J.L., Glahn, D.C., Fox, P.T., 2010. Acute effects of hydro-
alocorticoid receptors are indispensable for nongenomic modulation of hippo- cortisone on the human brain: an fMRI study. Psychoneuroendocrinology 35 (1),
campal glutamate transmission by corticosterone. Proc. Natl. Acad. Sci. U.S.A. 15–20.
102 (52), 19204–19207. Luethi, M., Meier, B., Sandi, C., 2008. Stress effects on working memory, explicit
Karst, H., Berger, S., Erdmann, G., Schütz, G., Joëls, M., 2010. Metaplasticity of memory, and implicit memory for neutral and emotional stimuli in healthy men.
amygdalar responses to the stress hormone corticosterone. Proc. Natl. Acad. Sci. Front. Behav. Neurosci. 2, 5.
U.S.A. 107 (32), 14449–14454. Lupien, S.J., Gillin, C.J., Hauger, R.L., 1999. Working memory is more sensitive than
Katona, I., Sperlágh, B., Sík, A., Käfalvi, A., Vizi, E.S., Mackie, K., Freund, T.F., 1999. declarative memory to the acute effects of corticosteroids: a dose-response study
Presynaptically located CB1 cannabinoid receptors regulate GABA release from in humans. Behav. Neurosci. 113 (3), 420–430.
axon terminals of specific hippocampal interneurons. J. Neurosci. 19 (11), Lupien, S., Maheu, F., Tu, M., Fiocco, A., Schramek, T., 2007. The effects of stress
4544–4558. and stress hormones on human cognition: implications for the field of brain and
Katona, I., Rancz, E.A., Acsady, L., Ledent, C., Mackie, K., Hajos, N., cognition. Brain Cogn. 65 (3), 209–237.
Freund, T.F., 2001. Distribution of CB1 cannabinoid receptors in the Lyons, D.M., Lopez, J.M., Yang, C., Schatzberg, A.F., 2000. Stress-level cortisol
amygdala and their role in the control of GABAergic transmission. treatment impairs inhibitory control of behavior in monkeys. J. Neurosci. 20 (20),
J. Neurosci. 21 (23), 9506–9518. 7816–7821.
Kirschbaum, C., Wolf, O.T., May, M., Wippich, W., Hellhammer, D.H., 1996. Stress- Maheu, F.S., Joober, R., Beaulieu, S., Lupien, S.J., 2004. Differential effects of
and treatment-induced elevations of cortisol levels associated with impaired adrenergic and corticosteroid hormonal systems on human short- and long-term
declarative memory in healthy adults. Life Sci. 58 (17), 1475–1483. declarative memory for emotionally arousing material. Behav. Neurosci. 118 (2),
Kuhlmann, S., Wolf, O.T., 2005. Cortisol and memory retrieval in women: influence of 420–428.
menstrual cycle and oral contraceptives. Psychopharmacology 183 (1), 65–71. Martinez, J.L., Jensen, R.A., Messing, R.B., Vasquez, B.J., Soumireu-Mourat, B.,
Kuhlmann, S., Wolf, O.T., 2006. Arousal and cortisol interact in modulating memory Geddes, D., Liang, K.C., et al., 1980. Central and peripheral actions of amphet-
consolidation in healthy young men. Behav. Neurosci. 120 (1), 217–223. amine on memory storage. Brain Res. 182 (1), 157–166.
Kuhlmann, S., Kirschbaum, C., Wolf, O.T., 2005a. Effects of oral cortisol treatment in McCarty, R., Gold, P.E., 1981. Plasma catecholamines: effects of footshock level and
healthy young women on memory retrieval of negative and neutral words. Neu- hormonal modulators of memory storage. Horm. Behav. 15 (2), 168–182.
robiol. Learn. Mem. 83 (2), 158–162. McDonald, A.J., 1991. Organization of amygdaloid projections to the prefrontal cortex
Kuhlmann, S., Piel, M., Wolf, O.T., 2005b. Impaired memory retrieval after psycho- and associated striatum in the rat. Neuroscience 44 (1), 1–14.
social stress in healthy young men. J. Neurosci. 25 (11), 2977–2982. McEwen, B.S., Weiss, J.M., Schwartz, L.S., 1968. Selective retention of corticosterone
Kukolja, J., Schläpfer, T.E., Keysers, C., Klingmüller, D., Maier, W., Fink, G.R., by limbic structures in rat brain. Nature 220 (5170), 911–912.
Hurlemann, R., 2008. Modeling a negative response bias in the human McEwen, B.S., Davis, P.G., Parsons, B., Pfaff, D.W., 1979. The brain as a target for
amygdala by noradrenergic-glucocorticoid interactions. J. Neurosci. 28 (48), steroid hormone action. Annu. Rev. Neurosci. 2 (1), 65–112.
12868–12876. McEwen, B.S., 1998. Stress, adaptation, and disease. Allostasis and allostatic load.
Lai, M., Horsburgh, K., Bae, S.-E., Carter, R.N., Stenvers, D.J., Fowler, J.H., Yau, J.L., Ann. N.Y. Acad. Sci. 840, 33–44.
et al., 2007. Forebrain mineralocorticoid receptor overexpression enhances McGaugh, J.L., Petrinovich, L.F., 1965. Effects of drugs on learning and memory. Int.
memory, reduces anxiety and attenuates neuronal loss in cerebral ischaemia. Eur. Rev. Neurobiol. 8, 139.
J. Neurosci. 25 (6), 1832–1842. McGaugh, J.L., Roozendaal, B., 2002. Role of adrenal stress hormones in forming
Lee, I., Kesner, R.P., 2003. Time-dependent relationship between the dorsal hippo- lasting memories in the brain. Curr. Opin. Neurobiol. 12 (2), 205–210.
campus and the prefrontal cortex in spatial memory. J. Neurosci. 23 (4), McGaugh, J.L., 1973. Drug facilitation of learning and memory. Annu. Rev. Pharmacol.
1517–1523. 13, 229–241.
436 Adrenal Stress Hormone Effects on Memory

McGaugh, J.L., 1989. Involvement of hormonal and neuromodulatory systems in the Ossewaarde, L., Qin, S., van Marle, H.J.F., van Wingen, G.A., Fernández, G.,
regulation of memory storage. Annu. Rev. Neurosci. 12 (1), 255–287. Hermans, E.J., 2011. Stress-induced reduction in reward-related prefrontal cortex
McGaugh, J.L., 2003. Memory and Emotion: The Making of Lasting Memories. function. NeuroImage 55 (1), 345–352.
Weidenfeld and Nicolson, London. Osuch, E.A., Willis, M.W., Bluhm, R., CSTS Neuroimaging Study Group, Ursano, R.J.,
McGaugh, J.L., 2004. The amygdala modulates the consolidation of memories of Drevets, W.C., 2008. Neurophysiological responses to traumatic reminders in the
emotionally arousing experiences. Annu. Rev. Neurosci. 27 (1), 1–28. acute aftermath of serious motor vehicle collisions using [15O]-H2O positron
McIntyre, C.K., Hatfield, T., McGaugh, J.L., 2002. Amygdala norepinephrine levels emission tomography. Biol. Psychiatry 64 (4), 327–335.
after training predict inhibitory avoidance retention performance in rats. Eur. J. Otte, C., Moritz, S., Yassouridis, A., Koop, M., Madrischewski, A.M., Wiedemann, K.,
Neurosci. 16 (7), 1223–1226. Kellner, M., 2007. Blockade of the mineralocorticoid receptor in healthy men:
McIntyre, C.K., Miyashita, T., Setlow, B., Marjon, K.D., Steward, O., Guzowski, J.F., effects on experimentally induced panic symptoms, stress hormones, and cogni-
McGaugh, J.L., 2005. Memory-influencing intra-basolateral amygdala drug infu- tion. Neuropsychopharmacology 32 (1), 232–238.
sions modulate expression of Arc protein in the hippocampus. Proc. Natl. Acad. Page, M.E., Oropeza, V.C., Sparks, S.E., Qian, Y., Menko, A.S., Van Bockstaele, E.J.,
Sci. U.S.A. 102 (30), 10718–10723. 2007. Repeated cannabinoid administration increases indices of noradrenergic
McReynolds, J.R., Donowho, K., Abdi, A., McGaugh, J.L., Roozendaal, B., activity in rats. Pharmacol. Biochem. Behav. 86 (1), 162–168.
McIntyre, C.K., 2010. Memory-enhancing corticosterone treatment increases Pakdel, R., Rashidy-Pour, A., 2006. Glucocorticoid-induced impairment of long-term
amygdala norepinephrine and Arc protein expression in hippocampal synaptic memory retrieval in rats: an interaction with dopamine D2 receptors. Neurobiol.
fractions. Neurobiol. Learn. Mem. 93 (3), 312–321. Learn. Mem. 85 (3), 300–306.
Menon, V., 2011. Large-scale brain networks and psychopathology: a unifying triple Pérez-Jaranay, J.M., Vives, F., 1991. Electrophysiological study of the response of
network model. Trends Cogn. Sci. 15 (10), 483–506. medial prefrontal cortex neurons to stimulation of the basolateral nucleus of the
Messier, C., White, N.M., 1984. Contingent and non-contingent actions of sucrose and amygdala in the rat. Brain Res. 564 (1), 97–101.
saccharin reinforcers: effects on taste preference and memory. Physiol. Behav. 32 Petrovich, G.D., Canteras, N.S., Swanson, L.W., 2001. Combinatorial amygdalar inputs
(2), 195–203. to hippocampal domains and hypothalamic behavior systems. Brain Res. Rev. 38
Messier, C., White, N.M., 1987. Memory improvement by glucose, fructose, and two (1–2), 247–289.
glucose analogs: a possible effect on peripheral glucose transport. Behav. Neural Pitkänen, A., 2000. Connectivity of the rat amygdaloid complex. In:
Biol. 48 (1), 104–127. Aggleton, J.P. (Ed.), The Amygdala. Oxford University Press, London, England,
Miller, R., Weckesser, L.J., Smolka, M.N., Kirschbaum, C., Plessow, F., 2015. pp. 31–117.
Hydrocortisone accelerates the decay of iconic memory traces: on the modulation Popoli, M., Yan, Z., McEwen, B.S., Sanacora, G., 2012. The stressed synapse: the
of executive and stimulus-driven constituents of sensory information maintenance. impact of stress and glucocorticoids on glutamate transmission. Nat. Rev. Neu-
Psychoneuroendocrinology 53, 148–158. rosci. 13 (1), 22–37.
Mizoguchi, K., Ishige, A., Takeda, S., Aburada, M., Tabira, T., 2004. Endogenous Preuss, D., Wolf, O.T., 2009. Post-learning psychosocial stress enhances consolida-
glucocorticoids are essential for maintaining prefrontal cortical cognitive function. tion of neutral stimuli. Neurobiol. Learn. Mem. 92 (3), 318–326.
J. Neurosci. 24 (24), 5492–5499. Putman, P., Roelofs, K., 2011. Effects of single cortisol administrations on human
Morena, M., Roozendaal, B., Trezza, V., Ratano, P., Peloso, A., Hauer, D., Atsak, P., affect reviewed: coping with stress through adaptive regulation of automatic
et al., 2014. Endogenous cannabinoid release within prefrontal-limbic pathways cognitive processing. Psychoneuroendocrinology 36 (4), 439–448.
affects memory consolidation of emotional training. Proc. Natl. Acad. Sci. U.S.A. de Quervain, D.J., Roozendaal, B., McGaugh, J.L., 1998. Stress and glucocor-
111 (51), 18333–18338. ticoids impair retrieval of long-term spatial memory. Nature 394 (6695),
Morena, M., Patel, S., Bains, J.S., Hill, M.N., 2016. Neurobiological interactions 787–790.
between stress and the endocannabinoid system. Neuropsychopharmacology 41 de Quervain, D.J., Roozendaal, B., Nitsch, R.M., McGaugh, J.L., Hock, C., 2000.
(1), 80–102. Acute cortisone administration impairs retrieval of long-term declarative memory in
van Marle, H.J.F., Hermans, E.J., Qin, S., Fernández, G., 2009. From specificity to humans. Nat. Neurosci. 3 (4), 313–314.
sensitivity: how acute stress affects amygdala processing of biologically salient de Quervain, D.J.-F., Henke, K., Aerni, A., Treyer, V., McGaugh, J.L., Berthold, T.,
stimuli. Biol. Psychiatry 66 (7), 649–655. Nitsch, R.M., et al., 2003. Glucocorticoid-induced impairment of declarative
de Oliveira Alvares, L., Engelke, D.S., Diehl, F., Scheffer-Teixeira, R., Haubrich, J., de memory retrieval is associated with reduced blood flow in the medial temporal lobe.
Freitas Cassini, L., Molina, V.A., et al., 2010. Stress response recruits the Eur. J. Neurosci. 17 (6), 1296–1302.
hippocampal endocannabinoid system for the modulation of fear memory. Learn. de Quervain, D.J.-F., Aerni, A., Roozendaal, B., 2007a. Preventive effect of beta-
Mem. 17 (4), 202–209. adrenoceptor blockade on glucocorticoid-induced memory retrieval deficits. Am.
O’Carroll, R.E., Drysdale, E., Cahill, L., Shajahan, P., Ebmeier, K.P., 1999. Stimulation J. Psychiatry 164 (6), 967–969.
of the noradrenergic system enhances and blockade reduces memory for emotional de Quervain, D.J.-F., Kolassa, I.-T., Ertl, V., Onyut, P.L., Neuner, F., Elbert, T.,
material in man. Psychol. Med. 29 (5), 1083–1088. Papassotiropoulos, A., 2007b. A deletion variant of the alpha2b-adrenoceptor is
Oei, N.Y.L., Everaerd, W.T.A.M., Elzinga, B.M., van Well, S., Bermond, B., 2006. related to emotional memory in Europeans and Africans. Nat. Neurosci. 10 (9),
Psychosocial stress impairs working memory at high loads: an association with 1137–1139.
cortisol levels and memory retrieval. Stress 9 (3), 133–141. de Quervain, D.J.-F., Aerni, A., Schelling, G., Roozendaal, B., 2009. Glucocorticoids
Oei, N.Y.L., Veer, I.M., Wolf, O.T., Spinhoven, P., Rombouts, S.A.R.B., Elzinga, B.M., and the regulation of memory in health and disease. Front. Neuroendocrinol. 30
2012. Stress shifts brain activation towards ventral “affective” areas during (3), 358–370.
emotional distraction. Soc. Cogn. Affect. Neurosci. 7 (4), 403–412. de Quervain, D.J.-F., Bentz, D., Michael, T., Bolt, O.C., Wiederhold, B.K., Margraf, J.,
Ohno-Shosaku, T., Maejima, T., Kano, M., 2001. Endogenous cannabinoids mediate Wilhelm, F.H., 2011. Glucocorticoids enhance extinction-based psychotherapy.
retrograde signals from depolarized postsynaptic neurons to presynaptic terminals. Proc. Natl. Acad. Sci. U.S.A. 108 (16), 6621–6625.
Neuron 29 (3), 729–738. Qin, S., Hermans, E.J., van Marle, H.J.F., Luo, J., Fernández, G., 2009. Acute
Oitzl, M.S., de Kloet, E.R., 1992. Selective corticosteroid antagonists modulate specific psychological stress reduces working memory-related activity in the dorsolateral
aspects of spatial orientation learning. Behav. Neurosci. 106 (1), 62–71. prefrontal cortex. Biol. Psychiatry 66 (1), 25–32.
Okuda, S., Roozendaal, B., McGaugh, J.L., 2004. Glucocorticoid effects on object Qin, S., Cousijn, H., Rijpkema, M., Luo, J., Franke, B., Hermans, E.J., Fernández, G.,
recognition memory require training-associated emotional arousal. Proc. Natl. 2012. The effect of moderate acute psychological stress on working memory-
Acad. Sci. U.S.A. 101 (3), 853–858. related neural activity is modulated by a genetic variation in catecholaminergic
Onur, O.A., Walter, H., Schlaepfer, T.E., Rehme, A.K., Schmidt, C., Keysers, C., function in humans. Front. Integr. Neurosci. 6, 16.
Maier, W., et al., 2009. Noradrenergic enhancement of amygdala responses to Quirarte, G.L., Roozendaal, B., McGaugh, J.L., 1997. Glucocorticoid enhancement of
fear. Soc. Cogn. Affect. Neurosci. 4 (2), 119–126. memory storage involves noradrenergic activation in the basolateral amygdala.
Oomura, Y., Nakano, Y., Lénárd, L., Nishino, H., 1988. Catecholaminergic and opioid Proc. Natl. Acad. Sci. U.S.A. 94 (25), 14048–14053.
mechanisms in conditioned food intake behavior of the monkey amygdala. In: Quirarte, G.L., Galvez, R., Roozendaal, B., McGaugh, J.L., 1998. Norepinephrine
Woody, C.D., Alkon, D.L., McGaugh, J.L. (Eds.), Cellular Mechanisms of Condi- release in the amygdala in response to footshock and opioid peptidergic drugs.
tioning and Behavioral Plasticity. Plenum, New York, pp. 109–118. Brain Res. 808 (2), 134–140.
Oropeza, V.C., Page, M.E., Van Bockstaele, E.J., 2005. Systemic administration of WIN Radna, R.J., MacLean, P.D., 1981. Vagal elicitation of respiratory-type and other unit
55,212-2 increases norepinephrine release in the rat frontal cortex. Brain Res. responses in basal limbic structures of squirrel monkeys. Brain Res. 213 (1),
1046 (1–2), 45–54. 45–61.
Adrenal Stress Hormone Effects on Memory 437

Rahmouni, K., Barthelmebs, M., Grima, M., Imbs, J.L., De Jong, W., 2001. Involve- Roozendaal, B., Hernandez, A., Cabrera, S.M., Hagewoud, R., Malvaez, M.,
ment of brain mineralocorticoid receptor in salt-enhanced hypertension in spon- Stefanko, D.P., Haettig, J., et al., 2010. Membrane-associated glucocorticoid
taneously hypertensive rats. Hypertension 38 (4), 902–906. activity is necessary for modulation of long-term memory via chromatin modifi-
Rasch, B., Spalek, K., Buholzer, S., Luechinger, R., Boesiger, P., cation. J. Neurosci. 30 (14), 5037–5046.
Papassotiropoulos, A., de Quervain, D.J.-F., 2009. A genetic variation of the Roozendaal, B., 2000. 1999 Curt P. Richter award. Glucocorticoids and the regulation
noradrenergic system is related to differential amygdala activation during encoding of memory consolidation. Psychoneuroendocrinology 25 (3), 213–238.
of emotional memories. Proc. Natl. Acad. Sci. U.S.A. 106 (45), 19191–19196. Roozendaal, B., 2002. Stress and memory: opposing effects of glucocorticoids on
Rashidy-Pour, A., Sadeghi, H., Taherain, A.A., Vafaei, A.A., Fathollahi, Y., 2004. The memory consolidation and memory retrieval. Neurobiol. Learn. Mem. 78 (3),
effects of acute restraint stress and dexamethasone on retrieval of long-term 578–595.
memory in rats: an interaction with opiate system. Behav. Brain Res. 154 (1), Rosenkranz, J.A., Grace, A.A., 2002. Cellular mechanisms of infralimbic and prelimbic
193–198. prefrontal cortical inhibition and dopaminergic modulation of basolateral amygdala
Reul, J.M., de Kloet, E.R., 1985. Two receptor systems for corticosterone in rat neurons in vivo. J. Neurosci. 22 (1), 324–337.
brain: microdistribution and differential occupation. Endocrinology 117 (6), Rothman, R.B., Baumann, M.H., Dersch, C.M., Romero, D.V., Rice, K.C., Carroll, F.I.,
2505–2511. Partilla, J.S., 2001. Amphetamine-type central nervous system stimulants release
Reul, J.M., van den Bosch, F.R., de Kloet, E.R., 1987. Relative occupation of type-I norepinephrine more potently than they release dopamine and serotonin. Synapse
and type-II corticosteroid receptors in rat brain following stress and dexametha- 39 (1), 32–41.
sone treatment: functional implications. J. Endocrinol. 115 (3), 459–467. Rowe, J.B., Toni, I., Josephs, O., Frackowiak, R.S., Passingham, R.E., 2000. The
Ricardo, J.A., Koh, E.T., 1978. Anatomical evidence of direct projections from the prefrontal cortex: response selection or maintenance within working memory?
nucleus of the solitary tract to the hypothalamus, amygdala, and other forebrain Science 288 (5471), 1656–1660.
structures in the rat. Brain Res. 153 (1), 1–26. Sajadi, A.A., Samaei, S.A., Rashidy-Pour, A., 2006. Intra-hippocampal microinjections
Riedemann, T., Patchev, A.V., Cho, K., Almeida, O.F.X., 2010. Corticosteroids: way of anisomycin did not block glucocorticoid-induced impairment of memory retrieval
upstream. Mol. Brain 3, 2. in rats: an evidence for non-genomic effects of glucocorticoids. Behav. Brain Res.
Rimmele, U., Besedovsky, L., Lange, T., Born, J., 2012. Blocking mineralocorticoid 173 (1), 158–162.
receptors impairs, blocking glucocorticoid receptors enhances memory retrieval in Sajadi, A.A., Samaei, S.A., Rashidy-Pour, A., 2007. Blocking effects of intra-
humans. Neuropsychopharmacology 38 (5), 884–894. hippocampal naltrexone microinjections on glucocorticoid-induced impairment of
Rogers, R.C., Fryman, D.L., 1988. Direct connections between the central nucleus of spatial memory retrieval in rats. Neuropharmacology 52 (2), 347–354.
the amygdala and the nucleus of the solitary tract: an electrophysiological study in Sandi, C., Pinelo-Nava, M.T., 2007. Stress and memory: behavioral effects and
the rat. J. Aut. Nerv. Syst. 22 (1), 83–87. neurobiological mechanisms. Neural Plast. 2007, 1–21.
Roozendaal, B., McGaugh, J.L., 1996. Amygdaloid nuclei lesions differentially affect Sandi, C., Rose, S.P., 1994. Corticosterone enhances long-term retention in one-day-
glucocorticoid-induced memory enhancement in an inhibitory avoidance task. old chicks trained in a weak passive avoidance learning paradigm. Brain Res. 647
Neurobiol. Learn. Mem. 65 (1), 1–8. (1), 106–112.
Roozendaal, B., McGaugh, J.L., 1997. Glucocorticoid receptor agonist and antagonist Sandi, C., 2011. Glucocorticoids act on glutamatergic pathways to affect memory
administration into the basolateral but not central amygdala modulates memory processes. Trends Neurosci. 34 (4), 165–176.
storage. Neurobiol. Learn. Mem. 67 (2), 176–179. Schelling, G., Briegel, J., Roozendaal, B., Stoll, C., Rothenhäusler, H.B.,
Roozendaal, B., McGaugh, J.L., 2011. Memory modulation. Behav. Neurosci. 125 (6), Kapfhammer, H.P., 2001. The effect of stress doses of hydrocortisone during
797–824. septic shock on posttraumatic stress disorder in survivors. Biol. Psychiatry 50 (12),
Roozendaal, B., Carmi, O., McGaugh, J.L., 1996. Adrenocortical suppression blocks 978–985.
the memory-enhancing effects of amphetamine and epinephrine. Proc. Natl. Acad. Schoofs, D., Preuss, D., Wolf, O.T., 2008. Psychosocial stress induces working
Sci. U.S.A. 93 (4), 1429–1433. memory impairments in an n-back paradigm. Psychoneuroendocrinology 33 (5),
Roozendaal, B., Nguyen, B.T., Power, A.E., McGaugh, J.L., 1999. Basolateral 643–653.
amygdala noradrenergic influence enables enhancement of memory consolidation Schoofs, D., Wolf, O.T., Smeets, T., 2009. Cold pressor stress impairs performance on
induced by hippocampal glucocorticoid receptor activation. Proc. Natl. Acad. Sci. working memory tasks requiring executive functions in healthy young men. Behav.
U.S.A. 96 (20), 11642–11647. Neurosci. 123 (5), 1066–1075.
Roozendaal, B., Quirarte, G.L., McGaugh, J.L., 2002. Glucocorticoids interact with the Schreurs, J., Seelig, T., Schulman, H., 1986. Beta 2-adrenergic receptors on
basolateral amygdala beta-adrenoceptor–cAMP/PKA system in influencing memory peripheral nerves. J. Neurochem. 46 (1), 294–296.
consolidation. Eur. J. Neurosci. 15 (3), 553–560. Schwabe, L., Wolf, O.T., 2009. The context counts: congruent learning and testing
Roozendaal, B., Griffith, Q.K., Buranday, J., de Quervain, D.J.-F., McGaugh, J.L., environments prevent memory retrieval impairment following stress. Cogn. Affect.
2003. The hippocampus mediates glucocorticoid-induced impairment of spatial Behav. Neurosci. 9 (3), 229–236.
memory retrieval: dependence on the basolateral amygdala. Proc. Natl. Acad. Sci. Schwabe, L., Römer, S., Richter, S., Dockendorf, S., Bilak, B., Schachinger, H., 2009.
U.S.A. 100 (3), 1328–1333. Stress effects on declarative memory retrieval are blocked by a beta-adrenoceptor
Roozendaal, B., de Quervain, D.J.-F., Schelling, G., McGaugh, J.L., 2004a. antagonist in humans. Psychoneuroendocrinology 34 (3), 446–454.
A systemically administered beta-adrenoceptor antagonist blocks corticosterone- Schwabe, L., Joëls, M., Roozendaal, B., Wolf, O.T., Oitzl, M.S., 2012. Stress effects
induced impairment of contextual memory retrieval in rats. Neurobiol. Learn. on memory: an update and integration. Neurosci. Biobehav. Rev. 36 (7),
Mem. 81 (2), 150–154. 1740–1749.
Roozendaal, B., Hahn, E.L., Nathan, S.V., de Quervain, D.J.-F., McGaugh, J.L., Smeets, T., Otgaar, H., Candel, I., Wolf, O.T., 2008. True or false? Memory is
2004b. Glucocorticoid effects on memory retrieval require concurrent norad- differentially affected by stress-induced cortisol elevations and sympathetic activity
renergic activity in the hippocampus and basolateral amygdala. J. Neurosci. 24 at consolidation and retrieval. Psychoneuroendocrinology 33 (10), 1378–1386.
(37), 8161–8169. Smeets, T., Wolf, O.T., Giesbrecht, T., Sijstermans, K., Telgen, S., Joëls, M., 2009.
Roozendaal, B., McReynolds, J.R., McGaugh, J.L., 2004c. The basolateral amygdala Stress selectively and lastingly promotes learning of context-related high arousing
interacts with the medial prefrontal cortex in regulating glucocorticoid effects on information. Psychoneuroendocrinology 34 (8), 1152–1161.
working memory impairment. J. Neurosci. 24 (6), 1385–1392. Smith, S.M., Vale, W.W., 2006. The role of the hypothalamic-pituitary-adrenal axis in
Roozendaal, B., Okuda, S., de Quervain, D.J.-F., McGaugh, J.L., 2006a. Glucocorti- neuroendocrine responses to stress. Dialogues Clin. Neurosci. 8 (4), 383–395.
coids interact with emotion-induced noradrenergic activation in influencing different Smith, A.P.R., Stephan, K.E., Rugg, M.D., Dolan, R.J., 2006. Task and content
memory functions. Neuroscience 138 (3), 901–910. modulate amygdala-hippocampal connectivity in emotional retrieval. Neuron 49 (4),
Roozendaal, B., Okuda, S., van der Zee, E.A., McGaugh, J.L., 2006b. Glucocorticoid 631–638.
enhancement of memory requires arousal-induced noradrenergic activation in the Soetens, E., D’Hooge, R., Hueting, J.E., 1993. Amphetamine enhances human-
basolateral amygdala. Proc. Natl. Acad. Sci. U.S.A. 103 (17), 6741–6746. memory consolidation. Neurosci. Lett. 161 (1), 9–12.
Roozendaal, B., McEwen, B.S., Chattarji, S., 2009a. Stress, memory and the amyg- Soetens, E.E., Casaer, S.S., D’Hooge, R.R., Hueting, J.E.J., 1995. Effect of
dala. Nat. Rev. Neurosci. 10 (6), 423–433. amphetamine on long-term retention of verbal material. Psychopharmacology 119
Roozendaal, B., McReynolds, J.R., van der Zee, E.A., Lee, S., McGaugh, J.L., (2), 155–162.
McIntyre, C.K., 2009b. Glucocorticoid effects on memory consolidation depend on Soravia, L.M., Heinrichs, M., Aerni, A., Maroni, C., Schelling, G., Ehlert, U.,
functional interactions between the medial prefrontal cortex and basolateral Roozendaal, B., et al., 2006. Glucocorticoids reduce phobic fear in humans. Proc.
amygdala. J. Neurosci. 29 (45), 14299–14308. Natl. Acad. Sci. U.S.A. 103 (14), 5585–5590.
438 Adrenal Stress Hormone Effects on Memory

Southwick, S.M., Davis, M., Horner, B., Cahill, L., Morgan, C.A., Gold, P.E., Weil-Malherbe, H., Axelrod, J., Tomchick, R., 1959. Blood-brain barrier for adrenaline.
Bremner, J.D., et al., 2002. Relationship of enhanced norepinephrine activity Science 129 (3357), 1226–1227.
during memory consolidation to enhanced long-term memory in humans. Am. J. Wichmann, R., Fornari, R.V., Roozendaal, B., 2012. Glucocorticoids interact with the
Psychiatry 159 (8), 1420–1422. noradrenergic arousal system in the nucleus accumbens shell to enhance memory
Stern, C.E., Sherman, S.J., Kirchhoff, B.A., Hasselmo, M.E., 2001. Medial temporal consolidation of both appetitive and aversive taste learning. Neurobiol. Learn. Mem.
and prefrontal contributions to working memory tasks with novel and familiar 98 (2), 197–205.
stimuli. Hippocampus 11 (4), 337–346. Wilhelm, I., Wagner, U., Born, J., 2011. Opposite effects of cortisol on consolidation of
Sternberg, D.B., Isaacs, K.R., Gold, P.E., McGaugh, J.L., 1985. Epinephrine facilitation temporal sequence memory during waking and sleep. J. Cogn. Neurosci. 23 (12),
of appetitive learning: attenuation with adrenergic receptor antagonists. Behav. 3703–3712.
Neural Biol. 44 (3), 447–453. Williams, C.L., Clayton, E.C., 2001. Contribution of brainstem structures in modulating
Sutanto, W., de Kloet, E.R., 1987. Species-specificity of corticosteroid receptors in memory storage processes. In: Gold, P.E., Greenough, W.T. (Eds.), Memory
hamster and rat brains. Endocrinology 121 (4), 1405–1411. Consolidation: Essays in Honor of James L. Mcgaugh. American Psychological
van Stegeren, A.H., Everaerd, W., Cahill, L., McGaugh, J.L., Gooren, L.J., 1998. Association, Washington, DC, pp. 141–163.
Memory for emotional events: differential effects of centrally versus peripherally Williams, C.L., Jensen, R.A., 1991. Vagal afferents: a possible mechanism for the
acting beta-blocking agents. Psychopharmacology 138 (3–4), 305–310. modulation of memory by peripherally acting agents. In: Fredrickson, R.C.A.,
van Stegeren, A.H., Wolf, O.T., Everaerd, W., Scheltens, P., Barkhof, F., McGaugh, J.L., Felten, D.L. (Eds.), Peripheral Signaling of the Brain. Hogrefe &
Rombouts, S.A.R.B., 2007. Endogenous cortisol level interacts with noradrenergic Huber, New York, NY, pp. 467–472.
activation in the human amygdala. Neurobiol. Learn. Mem. 87 (1), 57–66. Williams, C.L., McGaugh, J.L., 1993. Reversible lesions of the nucleus of the solitary
van Stegeren, A.H., Roozendaal, B., Kindt, M., Wolf, O.T., Joëls, M., 2010. Interacting tract attenuate the memory-modulating effects of posttraining epinephrine. Behav.
noradrenergic and corticosteroid systems shift human brain activation patterns Neurosci. 107 (6), 955–962.
during encoding. Neurobiol. Learn. Mem. 93 (1), 56–65. Williams, C.L., McGaugh, J.L., 1994. Enhancement of memory processing in an
Talley, C.P., Clayborn, H., Jewel, E., McCarty, R., Gold, P.E., 2002. Vagotomy inhibitory avoidance and radial maze task by post-training infusion of bombesin into
attenuates effects of L-glucose but not of D-glucose on spontaneous alternation the nucleus tractus solitarius. Brain Res. 654 (2), 251–256.
performance. Physiol. Behav. 77 (2–3), 243–249. Williams, C.L., Men, D., Clayton, E.C., Gold, P.E., 1998. Norepinephrine release in
Tasker, J.G., Di, S., Malcher-Lopes, R., 2006. Minireview: rapid glucocorticoid signaling the amygdala after systemic injection of epinephrine or escapable footshock:
via membrane-associated receptors. Endocrinology 147 (12), 5549–5556. contribution of the nucleus of the solitary tract. Behav. Neurosci. 112 (6),
Taverniers, J., Van Ruysseveldt, J., Smeets, T., von Grumbkow, J., 2010. High- 1414–1422.
intensity stress elicits robust cortisol increases, and impairs working memory Wilson, R.I., Nicoll, R.A., 2001. Endogenous cannabinoids mediate retrograde sig-
and visuo-spatial declarative memory in special forces candidates: a field experi- nalling at hippocampal synapses. Nature 410 (6828), 588–592.
ment. Stress 13 (4), 323–333. Wolf, O.T., Convit, A., McHugh, P.F., Kandil, E., Thorn, E.L., De Santi, S.,
Taylor, J.R., Birnbaum, S., Ubriani, R., Arnsten, A.F., 1999. Activation of cAMP- McEwen, B.S., et al., 2001. Cortisol differentially affects memory in young and
dependent protein kinase A in prefrontal cortex impairs working memory elderly men. Behav. Neurosci. 115 (5), 1002–1011.
performance. J. Neurosci. 19 (18), RC23. Wolf, O.T., 2008. The influence of stress hormones on emotional memory: relevance
Tollenaar, M.S., Elzinga, B.M., Spinhoven, P., Everaerd, W., 2009. Autobiographical for psychopathology. Acta Psychol. 127 (3), 513–531.
memory after acute stress in healthy young men. Memory 17 (3), 301–310. Yang, Y., Cao, J., Xiong, W., Zhang, J., Zhou, Q., Wei, H., Liang, C., et al.,
Ulrich-Lai, Y.M., Herman, J.P., 2009. Neural regulation of endocrine and autonomic 2003. Both stress experience and age determine the impairment or
stress responses. Nat. Rev. Neurosci. 10 (6), 397–409. enhancement effect of stress on spatial memory retrieval. J. Endocrinol. 178
Valentino, R.J., Aston-Jones, G., 1995. Physiological and anatomic determinants of (1), 45–54.
locus coeruleus discharge: behavioral and clinical implications. In: Bloom, F.E., Yang, Y.-L., Chao, P.-K., Lu, K.-T., 2006. Systemic and intra-amygdala administration
Kupfer, D.J. (Eds.), Psychopharmacology: The Fourth Generation of Progress. of glucocorticoid agonist and antagonist modulate extinction of conditioned fear.
Raven Press, New York, NY, pp. 373–386. Neuropsychopharmacology 31 (5), 912–924.
Vogel, S., Klumpers, F., Kroes, M.C.W., Oplaat, K.T., Krugers, H.J., Oitzl, M.S., Young, A.H., Sahakian, B.J., Robbins, T.W., Cowen, P.J., 1999. The effects of chronic
Joëls, M., et al., 2015. A stress-induced shift from trace to delay conditioning administration of hydrocortisone on cognitive function in normal male volunteers.
depends on the mineralocorticoid receptor. Biol. Psychiatry 78 (12), 830–839. Psychopharmacology 145 (3), 260–266.
Wan, R.Q., Pang, K., Olton, D.S., 1994. Hippocampal and amygdaloid involvement in Zhou, M., Bakker, E.H.M., Velzing, E.H., Berger, S., Oitzl, M., Joëls, M., Krugers, H.J.,
nonspatial and spatial working memory in rats: effects of delay and interference. 2010. Both mineralocorticoid and glucocorticoid receptors regulate emotional
Behav. Neurosci. 108 (5), 866–882. memory in mice. Neurobiol. Learn. Mem. 94 (4), 530–537.
1.16 Ovarian Hormones and Prefrontal Cortex-Related Cognition
Nicole J Gervais, University of Massachusetts, Amherst, MA, USA
Wayne G Brake, Center for Studies in Behavioral Neurobiology (CSBN), Concordia University, Montreal, QC, Canada
Agnès Lacreuse, University of Massachusetts, Amherst, MA, USA
Ó 2017 Elsevier Inc. All rights reserved.

1.16.1 Introduction 439


1.16.2 Anatomy of the PFC 439
1.16.3 Effects of Ovarian Hormones on PFC-Mediated Cognition 440
1.16.3.1 Working Memory 440
1.16.3.1.1 Humans 440
1.16.3.1.2 Nonhuman Primates 440
1.16.3.1.3 Rodents 441
1.16.3.1.4 Summary of E2 Effects on WM 442
1.16.3.2 Executive Function 442
1.16.3.2.1 Updating 442
1.16.3.2.2 Selective Attention 443
1.16.3.2.3 Response Inhibition 444
1.16.3.2.4 Task Switching 444
1.16.3.2.5 Decision Making 445
1.16.3.3 Multiple Memory System Bias 446
1.16.4 Effects of Ovarian Hormones on the PFC 447
1.16.4.1 Receptor Expression 447
1.16.4.2 Local Synthesis 447
1.16.4.3 Effects on Synaptic Plasticity and Signaling in the PFC 447
1.16.4.4 Correlations between Changes in Neural Substrates and Cognitive Performance 447
1.16.5 Conclusion 448
References 448

1.16.1 Introduction Finally, we will offer some suggestions for future studies
on this topic.
This chapter focuses on the effects of ovarian hormones on
prefrontal cortex (PFC)-mediated cognitive function. These
cognitive processes encompass several distinct, yet overlap- 1.16.2 Anatomy of the PFC
ping, constructs that together form executive function.
Executive function includes working memory (WM) and The PFC in primates is located in the anterior portion of the
processes that manipulate WM, such as updating, selective frontal lobe in both humans and NHPs. Although the subdivi-
attention, response inhibition, task switching, planning, sions are not entirely clear, and vary across studies (Wilson
and decision making (Chudasama and Robbins, 2006). et al., 2010), there is general agreement that the primate PFC
Although there is some disagreement as to whether executive is composed of three main subregions, the dorsolateral
function is a functionally homogeneous or heterogeneous (DLPFC, Brodmann’s areas 9, 46, and 9/46; Petrides et al.,
ability (Dalley et al., 2004), for the purpose of the present 2012), ventrolateral (VLPFC, areas 45 and 47/12; Petrides
chapter, each construct will be described separately. We and Pandya, 2002), and orbitofrontal cortices (OFC, areas
will address how ovarian hormones modulate each of these 10, 11, 14, and 25, Elliott et al., 2000).
processes, by reviewing data from the human, nonhuman The DLPFC (area 9, 46, and 9/46 in primates) receives input
primate (NHP), and rodent literature. Much work has been from the multimodal superior temporal sulcal cortex, the
conducted on the effects of estrogens on PFC-dependent rostral superior temporal gyrus, the anterior and posterior
function in females, primarily in the context of hormone cingulate cortex, and the retrosplenial cortex (Petrides and Pan-
replacement therapy (HRT) in postmenopausal women and dya, 1999; Petrides et al., 2005). This connectivity between
related animal models. Less is known about the effects of multimodal temporal areas and paralimbic cortical areas
estrogens in younger women, and there is a remarkable (cingulate, retrosplenial, and rostral temporal cortex) reflects
paucity of studies devoted to the effects of progestins. The its role in cognitive control and WM (Petrides et al., 1999).
later sections of the chapter will summarize what is The majority of the VLPFC of primates lies on the inferior
currently known about the neural circuitry underlying the frontal gyrus. Area 45 receives input from the superior temporal
effects of ovarian hormones on PFC-mediated function. gyrus (auditory system) and multimodal areas of the superior

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00016-X 439


440 Ovarian Hormones and Prefrontal Cortex-Related Cognition

temporal sulcus (Petrides et al., 2005). Area 47/12 has strong the effects of estrogens on WM per se, as opposed to more
connections with the rostral inferotemporal visual association complex tasks of executive function. Estrogen replacement
cortex and ventral limbic areas (i.e., perirhinal cortex, rostral therapy (ERT) administered cyclically or continuously to post-
parahippocampal gyrus; Petrides and Pandya, 2002). The menopausal women has no effect on the Digit Span-forward
OFC lies on the orbital surface of the frontal lobe (Elliott subtest (Duff and Hampson, 2000; Keenan et al., 2001), but
et al., 2000) and receives input from visual association areas, improves performance on the backward subtest relative to the
somatosensory, inferior temporal cortex and temporal pole, non-ERT group (Duff and Hampson, 2000). Since the Digit
amygdala, and mediodorsal thalamus (Elliott et al., 2000). Span-backward subtest involves some manipulation of infor-
The OFC projects to the inferior temporal and entorhinal mation in WM, the observed effect of ERT on this subtest, in
cortices, anterior cingulate, hypothalamus, ventral tegmental the absence of an effect on the Digit Span-forward subtest,
area, and caudate nucleus (Elliott et al., 2000). suggests a potential effect of estrogens on the manipulation
There is some disagreement as to whether rodents are suit- of WM.
able models for understanding the functions of the PFC (see
Wise, 2009). While rats do not have a frontal lobe, they do 1.16.3.1.2 Nonhuman Primates
possess a PFC (Uylings et al., 2003) with three subregions, One commonly used WM test in NHPs is the delayed response
including the medial PFC (mPFC). The mPFC can be further (DR) task, initially implemented by Jacobsen (1936). In this
subdivided into the prelimbic cortex (PL), infralimbic cortex task, the monkey watches as food reward is placed into one
(IL), frontal cortical area (area Fr2), and dorsal anterior cingu- of two food wells. Two identical tokens are then placed over
late area (Heidbreder and Groenewegen, 2003; Uylings et al., the wells, and then an opaque door is closed for a brief reten-
2003). The PL is located in the dorsal portion of the mPFC, tion delay blocking the subject’s view of the wells. Following
while the IL is located below it. These two subregions show the delay, the door is lifted and the monkey is allowed to
functional similarities with the DLPFC of primates (Uylings displace one of the two tokens. The monkey must remember
et al., 2003). While the IL/PL are not as anatomically special- the location of the food reward during the retention delay in
ized as the primate DLPFC, as will be discussed below, these order to perform well on this task. Several lines of research
regions are associated with abilities in rodents that overlap demonstrate a role of the DLPFC in DR performance. For
with PFC-dependent abilities in primates. Therefore, while example, single unit recordings in monkeys demonstrate
rodents may not demonstrate all the PFC-dependent abilities persistent firing of DLPFC neurons during the delay phase of
observed in primates, they are useful models to study the the DR task (Fuster and Alexander, 1971). An imaging study
functional role of the PFC and neuronal mechanisms in less conducted in humans reported sustained activation of the
complex cognitive abilities. lateral PFC during the delay phase of a spatial WM test
(Courtney et al., 1998). It is thought that this persistent firing
in the DLPFC is necessary to maintain task-relevant informa-
1.16.3 Effects of Ovarian Hormones tion in WM (Goldman-Rakic, 1996). Several studies demon-
on PFC-Mediated Cognition strate that damage to the DLPFC results in impaired
performance on the DR task (for a review, see Chudasama
1.16.3.1 Working Memory
and Robbins, 2006). Altogether, these studies convincingly
WM in humans is defined as “a system for temporary storage show a crucial role of the DLPFC for DR task performance.
and a mechanism for online manipulation of information Another commonly used test of WM in NHP is the delayed
that occurs during a wide variety of cognitive activities” matching/nonmatching-to-sample (DMS/DNMS) task, first
(Baddeley and Hitch, 1974). For nonhuman animals, WM is described by Mishkin and Delacour (1975). Similar to the
defined instead as “short-term memory for an object, stimulus, DR task, the DMS/DNMS task involves two phases, separated
or location that is used within a trial, but not typically across by a retention delay that varies from a few seconds to minutes.
trials” (Dudchenko, 2004). This type of memory is often distin- During the first phase, the monkey is presented with a sample
guished from reference memory, which is memory for informa- object and is required to displace it in order to obtain a food
tion that is relevant across trials, such as rules of a particular reward hidden underneath it. An opaque door is then closed
task (Dudchenko, 2004). In contrast, WM involves a representa- and following a delay, the door is lifted, and the monkey
tion of stimuli that is delay-dependent, subject to decay over must now choose between the now familiar (i.e., sample
time, and sensitive to attentional processes. object), or a novel object. For the DMS task, the sample object
is paired with the reward, and during the DNMS task, it is the
1.16.3.1.1 Humans novel object that is paired with reward. While this task, like
Many tests are available for assessing WM in humans, most of many other WM tasks, requires maintaining information
which involve the manipulation of WM through executive acquired during the first phase of each trial, it also requires
control, rather than simple maintenance of information in recognition of the sample object, which is an ability associated
memory. The Digit Span subtest of the Wechsler Adult Intelli- with medial temporal lobe structures, such as the perirhinal
gence Scale (Wechsler, 2008), which assesses verbal WM, is cortex (as reviewed by Winters et al., 2008; Warburton and
one exception. In this test, participants are required to recall Brown, 2010). There is also evidence that the DLPFC (Moore
the order of digits either in the same (Digit Span forward) or et al., 2012) and the VMPFC play a role, as damage to these
opposite order (Digit Span backward) in which they are pre- regions impairs performance on this task (Bachevalier and
sented. The total number of correctly listed items is taken to Mishkin, 1986; Meunier et al., 1997). However, Meunier
reflect the strength of WM. Few studies in humans examined et al. (1997) suggest that the role of the PFC in recognition
Ovarian Hormones and Prefrontal Cortex-Related Cognition 441

tasks is likely to maintain associative memory, rather than arms are accessible, and the rodent must enter the previously
recognition per se. unentered goal arm to retrieve the reward. Like WM tests for
Studies focusing on DR and DNMS performance in NHPs NHPs, longer delays can be introduced between the two
have provided mixed evidence for beneficial effects of E2. phases of each trial to increase WM load. Lesion studies
Roberts et al. (1997) compared performance of peri-/postmen- demonstrate that the mPFC, and in particular the PL, is
opausal macaques to age-matched premenopausal females, necessary for accurate performance on this test (Larsen et al.,
and young control monkeys on the DR task. While the two 1978; Thomas and Brito, 1980; Wolf et al., 1987).
older groups performed worse than the young control Studies in aged rats provide support for the idea that
group, the greatest WM deficit was observed in the peri-/ estrogen loss during midlife accelerates age-related cognitive
postmenopausal group. In addition, accuracy scores were posi- decline. For example, Markowska and Savonenko (2002)
tively correlated with urinary levels of estrogen metabolites tested the effects of long-term estrogen loss on the DSA in
(i.e., estrone conjugate). These data are consistent with the middle-aged-to-old rats (13–23 months). Performance deficits
idea that estrogen loss during menopause accelerates the effect emerged beginning 4 months post-OVX. WM ability was
of normal aging on WM. A subsequent study (Rapp et al., restored among females given a cyclic regimen of E2 replace-
2003) tested aged OVX macaques (22 years) on the DR and ment involving continuous low E2 in addition to three daily
DNMS, following replacement with low cyclic E2 (100 mg estra- subcutaneous injections of E2 (25 pg ml1, serum) followed
diol cypionate, i.m.) or vehicle every 3 weeks. Relative to the by 1 day of no injection. A similar pattern of results was
vehicle-treated group, E2 improved performance on the DR observed in a study by Gibbs (2000) using the delayed
and on two delays of the DNMS. While these findings suggest matching-to-place (DMP) task, a task similar to the DSA test
that E2 can reverse age-related cognitive decline on WM, not except that the reward appears in the same arm during both
all studies report consistent effects. For example, Voytko et al. phases of each trial: continuous low E2 (15–25 pg ml1,
(2008) observed modest improvements on the DMS and no serum, via silastic implants) or cyclic E2 (10 mg, s.c., once
effect on DR performance in OVX aged macaques a week) and progesterone (500 mg, s.c. 2 days after E2) replace-
(19.7  0.5 years) given a cyclic hormonal regimen that ment prevented impairments in the middle-aged rats when
included continuous low E2 replacement (40–80 pg ml1, administered within 3 months of OVX.
serum, via silastic implants) and acute injections of E2 valerate Several lines of evidence suggest that DSA and DMP
(0.10–0.20 mg ml1). Another study comparing long-term performance is also improved by estrogens in young rats.
OVX (>12 years) monkeys to gonadally intact age-matched Impaired DSA performance was observed during estrous (low
only revealed small impairments on the DNMS task in OVX E2) relative to the other phases of the estrous cycle in one study
animals (Lacreuse et al., 2000). Furthermore, administration (Korol et al., 2004). In another study, DMP performance was
of continuous ethinyl estradiol (EE2, 450 ng kg1 per day, improved by acute E2 replacement. Velázquez-Zamora et al.
p.o.) or vehicle for a month did not affect performance on (2012) showed that young rats (75–90 days) given estradiol
the DR or DNMS task (Lacreuse et al., 2002). So, while ERT benzoate (EB, 10 mg, s.c.) or vehicle 6 days post-OVX
appears to attenuate age-related cognitive decline in WM, this performed better on the DMP than vehicle-treated rats. Interest-
appears to be influenced by both the hormone regimen (cyclic ingly, performance was also improved following administra-
versus continuous) and the duration of estrogen deprivation. tion of two selective estrogen receptor modulators, tamoxifen
While studies using aged macaques (either peri-/postmeno- (1 mg kg1) and raloxifene (1 mg kg1). Unlike acute adminis-
pausal, or OVX with ERT within a critical window) suggest that tration, continuous E2 replacement does not necessarily
estrogens improve WM, this is not observed in younger improve WM in young rats. For example, continuous E2
monkeys. For example, two studies found that endogenous replacement (86.2  8.2 pg ml1 serum, via silastic implants)
fluctuations in ovarian hormones do not influence WM perfor- given immediately following OVX was found to reduce
mance, as assessed by the DMS (Lacreuse et al., 2001; Kromney, accuracy on the DSA test (Wang et al., 2008, 2009). While these
2015). In addition, OVX and various regimens of E2 replace- data suggest that acute and continuous E2 regimens have
ment have no effect on DR performance (Voytko, 2000; Hao differential effects on WM, they might also reflect dose-
et al., 2007) or DMS tasks in young OVX macaques (Lacreuse dependent effects, as opposing effects of low and high
and Herndon, 2003, 2009). E2 replacement are observed on the DSA task when reliance
on extramaze cues is minimized (Wide et al., 2004).
1.16.3.1.3 Rodents In that study, low continuous E2 replacement
The most basic WM test used with rodents is the delayed spatial (27.6  6.3 pg ml1, serum) improved, whereas higher doses
alternation (DSA) test (Tolman, 1925). This test is often (medium: 103.3  8.5 pg ml1; high: 197.0  28.1 pg ml1,
administered in a T-shaped maze. The starting location is the serum) impaired, DSA performance in adult rats. These studies
end of one arm. At the end of the starting arm is a junction, provide evidence that low E2 replacement given within
with a goal arm to the left and right. Food rewards are placed 3–4 months of OVX enhances performance on WM tasks in
in wells at the end of each goal arm. Like WM tasks in NHPs, both young and older rats. It is important to note that the tests
the DSA test involves two phases, which are separated by used (DMP and DSA) rely on the integrity of both the mPFC
a retention delay. During the first phase, one of the goal arms and HPC (Dudchenko, 2004; Yoon et al., 2008). With the
is closed and so the animal is forced to turn either left or exception of the study by Wide et al. (2004), which reduced
right to retrieve a reward from the accessible arm. The animal reliance on the HPC by limiting the availability of extramaze
is returned to the start arm for a retention delay, after which cues, the reported improvement of WM performance by E2 in
the test phase of the trial begins. During this phase, both goal rats may reflect enhanced function of either brain region.
442 Ovarian Hormones and Prefrontal Cortex-Related Cognition

1.16.3.1.4 Summary of E2 Effects on WM Ordered Search test in older hysterectomized women


Limited support exists for the enhancement of WM by estro- (61–74 years). Similarly, 12 weeks of ERT (transdermal E2,
gens in postmenopausal women. Stronger evidence for the 50 mg day1) had no effect on spatial WM in older postmeno-
enhancement of WM following estrogen administration is pausal sample (mean age 71; Schiff et al., 2005). These results
provided by studies in NHP and rats. However, a few factors are consistent with the critical period hypothesis in that estro-
influence whether the effects are detected. Age, in particular, gens may improve performance in WM tasks that require
appears to be an important factor in NHP studies. In aged updating, only if given within a short period of time following
macaques, OVX impairs while E2 replacement enhances WM estrogen loss.
ability, but the effects are more reliable following cyclic than In women of reproductive age, elevated circulating levels of
continuous replacement, and only occurs within a critical E2 are associated with better performance on the SPWM task
window following OVX. While E2 is shown to enhance WM (Hampson and Morley, 2013).
performance in both young and older rats, the effect appears
to be dependent on the dose of E2 replacement, with low 1.16.3.2.1.2 Nonhuman Primates
replacement improving while moderate to high replacement In NHPs, several tests have been developed to closely model
impairing WM ability. However, the majority of studies in the updating of WM, such as the monkey version of the
rats use tests that rely on both the PFC and HPC, and so these SOPT (Petrides, 2000), the Self-Ordered Spatial Search task
effects might not solely reflect an influence of E2 on PFC func- (Weed, 1999), or the Delayed Recognition Span tests (DRST;
tion. Studies incorporating intra-PFC infusions of E2 are Moss et al., 1997). All require the animals to monitor their
needed to confirm whether E2 influences performance on these selection in order to avoid retouching a previously selected
tests via effects on this structure. stimulus. The PFC has been shown to be involved in the perfor-
mance of these tasks (Beason-Held et al., 1999; Lamar and
Driscoll, 2007; Collins et al., 1998).
1.16.3.2 Executive Function
Seemingly contrasting the results in human participants,
WM involves maintaining information during a delay often to Lacreuse et al. (2000) reported positive effects of long-term
perform higher-order cognitive operations, such as updating, ovariectomy (12 years) on the spatial-DRST in aged macaques,
selective attention, response inhibition, task switching, plan- when compared to gonadally intact age-matched controls.
ning, and decision making. Together, these abilities are referred However, a subsequent study (Lacreuse et al., 2002) found
to as executive function. beneficial effects of EE2 (450 ng kg1 per day, p.o. for
1 month) on the same task. Later work in rodents provided
1.16.3.2.1 Updating convincing evidence that the loss of progesterone in long-
1.16.3.2.1.1 Humans term OVX animals benefits spatial WM (assessed with a water
This ability is typically assessed in humans using the N-back radial-arm maze task (RAM); Bimonte-Nelson et al., 2003,
test (Cohen et al., 1997). In this test, participants are given 2004). Thus, both the lack of progesterone and the presence
a random list of items, usually numbers, and when a target of E2 can benefit WM tasks requiring updating in aged NHP.
item is identified, he/she must identify the item that appeared In young, gonadally intact macaques (5–7 years old),
N items previously. For example, in the two-back test, partici- impaired performance on the spatial-DRST was observed
pants must identify the number that preceded the target by during the periovulatory period (high E2; Lacreuse et al.,
two items. To perform well on this task, participants must 2001). No effect was found on DMS, suggesting that the
keep track of the last two numbers presented to them and impairing effect of E2 is specific to updating, without affecting
update these items as new numbers are presented. Both the the simple maintenance of information in WM. In OVX young
DLPFC and VLPFC are activated during this test (Owen et al., macaques, E2 administration (EB, 2 mg kg1, s.c. given for
2005). Similar tests include the Digit Ordering task (Petrides 2 weeks) had negligible effects on three versions of the DRST,
et al., 1993), the Self-Ordered Pointing test (SOPT; Petrides the spatial-, object-, and face-DRST (Lacreuse et al., 2009).
and Milner, 1982; Petrides, 2000), and the spatial WM task However, a previous study with continuous EE2
(SPWM; Duff and Hampson, 2000), which involve selecting (450 ng kg1 day, p.o.) given for 1 month reported negative
items among an array without repeating any selections. effects of EE2 on the face version of the DRST (Lacreuse and
Positive effects of estrogens have often been reported in Herndon, 2003), possibly due to the interactions between
tasks that require updating WM. For example, menopausal estrogens and the processing of emotional material (i.e.,
women (52 years) on ERT (either oral CEE, transdermal E2, conspecific faces).
or vaginal CEE cream) had stronger performance on the
N-back test relative to menopausal women not on ERT (Keenan 1.16.3.2.1.3 Rodents
et al., 2001). ERT given briefly (100 mg, transdermally over A rodent task that requires the updating of WM is the
3 days; Krug et al., 2006), either alone or combined with RAM (Olton and Samuelson, 1976). The more common
progesterone for at least 70 months (Duff and Hampson, configuration of the maze involves eight goal arms radiating
2000), is also associated with improved performance on the from a center platform. A food well is located at the end of
Digit Ordering test. In addition to the Digit Ordering test, each arm, and doors can be lifted or lowered by the experi-
HRT also improved performance on the SPWM (Duff and menter to control access to each arm. During the first phase
Hampson, 2000). ERT may not have the same beneficial effects of each trial, doors to a number of arms (e.g., three or four)
in older women, however. Janowsky et al. (2000) reported no are lifted allowing the rodent to enter each arm to retrieve
effect of CEE (0.625 mg day1 for 1 month) on the Self- the rewards. Once rewards from these arms are consumed,
Ovarian Hormones and Prefrontal Cortex-Related Cognition 443

the rodent reenters the center compartment for a retention on a win-shift version of the RAM in young rats. The extent
delay, whereby access to all arms is blocked. All doors are to which the tests used involve the HPC may contribute to
then lifted allowing the rodent to collect remaining rewards. the inconsistent findings.
WM is inferred from the number of errors, i.e., the number of
times the rodent reenters each arm. In addition to maintaining 1.16.3.2.2 Selective Attention
spatial information about the previously entered arms, this test The PFC, and in particular, the DLPFC, is also implicated in
also requires monitoring arm entries to avoid reentering an visual attention, which is the ability to process important
arm. The PL (Seamans et al., 1995) and the HPC (Dudchenko, sensory information while ignoring irrelevant information
2004) appear to play a role in this task. (Squire et al., 2013). Damage to this region results in impaired
Similar to effects seen on WM tests, E2 replacement performance on tasks that require selective attention in both
enhances RAM performance in middle-aged and older rats. In macaques (Wardak et al., 2006) and humans (Knight et al.,
one study (Daniel et al., 2006), 12- and 17-month-old female 1995; Rueckert and Grafman, 1996). Impaired attention is
rats were OVX, then given continuous low E2 replacement (20– also observed in both rats (Muir et al., 1996; Chudasama and
25 pg ml1, serum) or vehicle. E2 replacement began either Muir, 2001; Passetti et al., 2002) and mice (Kahn et al.,
immediately following OVX, or 5 months later. While E2 2012) following damage to the mPFC. In addition, enhanced
replacement was associated with fewer WM errors (arm reen- neuronal responses have been observed in the DLPFC when
tries), this was only true for those that received replacement attention is directed at stimuli within the visual receptive field
within 5 months of OVX. These data provide support for the (Lebedev et al., 2004; Buschman and Miller, 2007; Armstrong
critical period hypothesis in that higher E2 levels benefit updat- et al., 2009; Gregoriou et al., 2009).
ing only if administered around the time of estrogen loss.
RAM performance is also influenced by E2 in younger rats. 1.16.3.2.2.1 Humans
During proestrus, fewer WM errors are observed relative to the A test of selective visual attention is the continuous perfor-
estrous phase in gonadally intact female rats (Pompili et al., mance test (Rosvold et al., 1956). Although there are several
2010). Similar to the DSA test, there appears to be a dose- versions of this test, they all involve presentation of stimuli
dependent effect of E2 replacement on RAM in young OVX on a screen, and the participant must respond by pressing
rats, as low EB replacement (0.3 mg day1, s.c.) is associated a key when the target stimulus is presented. Other tests include
with more WM errors, whereas high EB replacement the Digit Symbol subtest of the WAIS. This test involves pre-
(5 mg day1, s.c.) is associated with fewer WM errors relative senting a set of digit–symbol pairs, and participants are shown
to controls (Sinopoli et al., 2006). Reduced WM errors is also a list of digits and are asked to identify the symbols that are
observed when E2 (0.9 mg/0.5 ml, but not 0.1 mg/0.5 ml) is paired with each symbol as quickly as possible. Studies report
infused directly into the PL prior to testing on the RAM. inconsistent results, with some studies reporting no effect of
Another study (Holmes et al., 2002) reported opposite dose- HRT on attention (Keenan et al., 2001; Wolf et al., 2005), while
dependent effects of EB replacement on RAM performance, other studies report improved attention following HRT (Smith
where low replacement (0.3 mg day1, s.c) enhanced perfor- et al., 2001; Schmidt et al., 1996).
mance, whereas medium (1 mg day1, s.c.) and high
(5 mg day1, s.c.) replacement impaired performance. Whereas 1.16.3.2.2.2 Nonhuman Primates
Sinopoli et al. (2006) used the win-shift version of this task, Visual attention can be assessed in NHPs using a variety of cued
which is dependent on both PFC and HPC, Holmes et al. search tasks. These tests require monkeys to identify a target
(2002) kept the same pattern of baited and unbaited arms stimulus among an array of distractors or to attend to a cue
across sessions, decreasing reliance on the PFC. Therefore, the that indicates the probable location of a target (Baxter and Voy-
majority of the results suggest that E2 enhances performance tko, 1996). Similar to the results in humans, studies using
on the RAM, with some evidence that it acts directly in the PL NHPs report inconsistent effects of E2 replacement on atten-
to promote the updating of WM. tion. Voytko et al. (2009) trained aged macaques (20 years
old) to criterion before ovariectomies were performed.
1.16.3.2.1.4 Summary of E2 Effects on Updating WM Some animals received immediate cyclic E2 replacement
There is reasonable evidence that updating information in WM (E2 70 pg ml1 via silastic implants) plus acute E2
is improved by HRT or ERT in women. This is observed in (0.10–0.20 mg ml1, s.c. given for 12 days every 4 weeks).
younger women and in postmenopausal women who begin Other monkeys received continuous E2 replacement (via
therapy during a critical window. In NHPs, the effect of E2 implants) and cyclic P replacement (0.2 mg kg1 given for
on updating is less clear, as higher levels are associated with 12 days month1). A third group received no replacement.
improved updating in older, but impaired updating in All macaques were tested on the cued search task 2, 12, and
younger, monkeys. A study reporting contrasting results 24 weeks post-OVX. While hormone replacement had no effect
(Lacreuse and Herndon, 2003) used emotional stimuli on overall performance, it increased release times on valid
(conspecific faces) that may have reflected an effect of E2 on (when cue is presented on same side as target) and invalid trials
emotional reactivity rather than cognitive performance per se. (when cue is presented on opposite side as target), suggesting
Studies in rats used the RAM, which is a task that involves a specific impairment in the processing of cues. The opposite
both the PFC and HPC. Low continuous E2 replacement is pattern of results was reported in a study with young monkeys
associated with improved RAM performance in older rats, but (6–11 years old). Decreased release times were observed
impaired RAM performance in younger rats. Higher EB following E2 replacement (130 pg ml1 via silastic implants)
replacement or intra-PL infusion of E2 enhances performance relative to a no replacement group (Voytko, 2002). In another
444 Ovarian Hormones and Prefrontal Cortex-Related Cognition

study, no effect of EB replacement (2 mg kg1, s.c. given for or combined with P (daily E2: 2 mg day1; P: 100 mg day1,
2 weeks) in young monkeys (7–16 years old) was observed p.o.), or placebo (Wolf et al., 2005). These contrasting results
(Lacreuse et al., 2009). are consistent with the critical period hypothesis. Findings in
younger women suggest a different pattern of results. For
1.16.3.2.2.3 Rodents example, high E2 during the menstrual cycle impairs response
In rodents, selective attention is typically assessed by using the inhibition, as assessed by a Stop-Signal task (Colzato et al.,
5-choice serial reaction time test (5-CSRT; Robbins, 2002). The 2010) and a Go/No-Go task (Reimers et al., 2014).
rodent responds to a brief visual stimulus presented randomly There is a paucity of studies investigating the effects of
in one of five locations in order to obtain a food reward. The ovarian hormones on response inhibition in NHPs. A recent
number of correct responses over incorrect ones is taken as study in baboons (Lacreuse et al., 2016) found that Stop-
a measure of attention. Performance on the 5-CSRT is Signal performance was unaffected by the menstrual cycle in
enhanced by continuous E2 replacement (37 pg ml1, females, although large sex differences in favor of females
serum) relative to a no replacement condition (Bohacek and were observed in response inhibition and overall response
Daniel, 2010) in both young and older rats. In older rats, the times. Additional studies are needed to understand how
enhanced effect on selective attention is only observed when gonadal hormones may influence these sexually differentiated
given immediately, but not 5 months following OVX. patterns of performance.
Latent inhibition (LI) is another way to assess selective The effect of E2 replacement on response inhibition has also
attention (Escobar et al., 2002). LI occurs when preexposure been assessed in young, middle-aged, and older OVX rats
to the conditioned stimulus results in a reduction in the (Wang et al., 2011). OVX rats receiving continuous E2 replace-
subsequent association with the unconditioned stimulus ment were trained on the differential reinforcement of low rates
(Cassaday et al., 2014). In contrast to the 5-CSRT test, LI is of responding task, which requires the rats to suppress a lever-
impaired by both continuous (17–37 pg ml1, serum via pressing response for a fixed period of time in order to obtain
silastic implant) and cyclic (implant: 28 pg ml1, serum, a food reward. In the young and middle-aged groups, contin-
and acute E2:10 mg kg1, s.c. every fourth day) E2 replacement uous E2 replacement (20 and 40 pg ml1, serum) was associ-
(Almey et al., 2013). LI is also reduced when conditioning ated with reduced ratio of reinforced to nonreinforced
occurs during proestrus, when circulating levels of E2 are responses, suggesting impaired response inhibition in these
highest (Quinlan et al., 2010). These data provide paradoxical animals.
effects on two different measures of selective attention in
OVX rats. Research is needed to identify potential mecha- 1.16.3.2.3.1 Summary of E2 Effects on Response Inhibition
nisms by which E2 impairs LI while enhancing attention on Few studies have investigated the effects of ovarian hormones
the 5-CRST test. on response inhibition. Studies in postmenopausal women
suggest improved response inhibition following ERT. However,
1.16.3.2.2.4 Summary of E2 Effects on Selective Attention higher E2 levels appear to impair response inhibition in young
Studies on selective attention have provided mixed evidence for cycling women and in rats regardless of age. More research is
an effect of E2, with several studies in humans and NHPs necessary to better understand the role of E2 in response
reporting no effect. Among those that do report an effect, inhibition.
some find enhanced attention following E2 replacement, while
others report impaired attention. In rats, E2 replacement 1.16.3.2.4 Task Switching
appears to enhance performance on the 5-CSRT test, while Task switching is defined as the ability to switch attention from
impairing LI. More research is needed to clarify the role of an initially important feature to another that subsequently
ovarian hormones in attention. conveys information about where to respond correctly to
obtain a reward (Butts et al., 2013). Task switching ability
1.16.3.2.3 Response Inhibition involves the DLPFC in humans and NHPs (Heekeren et al.,
Response inhibition in humans is typically assessed using the 2006; Seo et al., 2007; Nakahara, 2002) and the mPFC in
Stroop test (Stroop, 1935), the Go/No-Go test (Donders, rodents (Floresco et al., 2009; Ragozzino et al., 1999). One
1969), or the Stop-Signal task (Logan et al., 1984). These tasks basic form of task switching is reversal learning, which is the
involve inhibiting a prepotent response. For example, the ability to shift responses to stimuli following a shift in stim-
Stroop test involves presenting a list of words describing colors. ulus/reward contingencies (Butts et al., 2013). This ability is
The words themselves are written in different colors. Partici- dependent on the OFC (Butts et al., 2013; Kehagia et al.,
pants are required to report the color each word is written in, 2010). Damage to the OFC is often associated with persevera-
while avoiding reading the word. The Go/No-Go test and tive responding, the selection of the previously rewarded stim-
Stop-Signal task require participants to make a response ulus, despite a change in stimulus/reward contingency (Jones
when certain stimuli are presented and withhold a response and Mishkin, 1972; Rolls et al., 2004; Dias et al., 1996a,b;
when other stimuli are presented. Fellows and Farah, 2003).
One study (Krug et al., 2006) reported beneficial effects of
ERT (100 mg, transdermally over 3 days) in postmenopausal 1.16.3.2.4.1 Humans
women (50–65 years old) on the incongruent component of Task switching is typically assessed in humans using the Wis-
the Stroop test. This effect was not replicated in an older sample consin Card Sorting Test (WCST, Heaton, 1981). In this test,
(58–75 years old) of women who were hysterectomized participants are presented with cards displaying a varying
20 years prior, then given either E2 alone (2 mg day1, p.o.), number of shapes of different colors. The participant is
Ovarian Hormones and Prefrontal Cortex-Related Cognition 445

required to figure out, by using the experimenter’s feedback, one possibility is that E2 has opposing effects on the OFC
whether to sort the cards by shape, number, or color. Once and PFC. More research is needed to address this potential
the rule is acquired, the experimenter switches the rule and explanation.
records the number of trials required to acquire the new
rule. There is conflicting evidence that ERT improves this
1.16.3.2.5 Decision Making
ability. For example, Schmidt et al. (1996) compared WCST
Decision making is often assessed in humans via the Iowa
performance between postmenopausal women (60 years
Gambling task (IGT; Bechara et al., 1994). The IGT is a comput-
old) taking ERT and those who never took ERT. While the
erized task that involves presenting four deck of cards on
number of category shifts was higher for the ERT group, the
a screen. Decks vary in the number of cards that result in lost
overall number of errors, and number of perseverative errors,
money, with two decks being considered advantageous, and
was equal across the two groups. Using a cross-over design,
two decks disadvantageous. Performing well on the IGT
Berman et al. (1997) reported no effect of ERT or P on
involves learning from as few trials as possible the payoffs asso-
WCST performance in younger women (age: 27–49 years
ciated with each deck and selecting cards from the advanta-
old) whose ovarian function was suppressed via injection of
geous decks, while minimizing selections from the
a gonadotropin-releasing hormone agonist (GnRH-a).
disadvantageous ones. The task involves the VMPFC/OFC
However, the activity patterns in the PFC were clearly
and the DLPFC (Bechara et al., 1998; Christakou et al., 2009;
reduced by the GnRH-a and restored following treatment
Lawrence et al., 2009). Another decision-making task for use
with E2 or P.
in humans is the Delayed Discounting (DD) task (Altamirano
et al., 2011; Smith and Boettiger, 2012). This task requires
1.16.3.2.4.2 Nonhuman Primates
participants to make choices between smaller, immediate
NHP studies provide evidence for a modulatory effect of E2 on
rewards and larger, eventual ones. This task taps into the ability
task switching. Voytko et al. (2009) reported that aged
to choose delayed larger rewards over smaller immediate ones.
macaques (20 years old) treated with cyclic E2 or E þ P
A related decision-making task used in rodents is called the
performed better than the no replacement group in a monkey
Effort Discounting (ED) task. Subjects are trained in an operant
version of the WCST, as indicated by fewer trials to learn the
chamber containing two levers. One lever is paired with the
set-shift and fewer perseverative errors. However, E2 replace-
immediate delivery of a small reward (two pellets), and the
ment (EE2, 450 ng kg1 per day) failed to enhance perfor-
second reward is paired with a delayed delivery of a large
mance on the primate-WCST in aged (23–26 years) long-term
reward (four pellets). The rodent must choose between the
OVX macaques (Lacreuse et al., 2004).
two levers. Upon choosing the lever paired with the large
E2 replacement also modulates reversal learning in marmo-
reward, the subject must press that lever an additional 5–20
sets. Adult OVX marmosets (2.5–5 years) given immediate
times, with the number of additional level presses increasing
continuous E2 replacement (360  24.21 pg ml1, serum) per-
as the test sessions progress. Therefore, choosing the lever
formed worse on one of three reversals and showed an increase
paired with a large reward becomes increasingly costly as the
in perseverative responding across the three reversals compared
session progresses.
to the control group (Lacreuse et al., 2014).
E2 does not appear to influence performance on the IGT
in both young and postmenopausal women (Reavis and
1.16.3.2.4.3 Rodents
Overman, 2001). However, there is some evidence that perfor-
Impaired reversal learning was also found in aged OVX rats
mance on the DD task varies across the menstrual cycle. Smith
given continuous E2 replacement (39 pg ml1; Gibbs et al.,
et al. (2014) tested naturally cycling women (age 18–40 years)
2011). Another study in young OVX rats used the extradimen-
and reported a decrease in the tendency to prefer smaller
sional set-shifting task to assess task switching (ED task;
immediate rewards from the menstrual (low E2) to the follic-
Lipatova et al., 2016). This task was administered in a plus
ular phase (high E2). In addition, this decrease was negatively
maze, with arms that differed in terms of brightness (light or
correlated with E2 levels, suggesting that higher levels of E2 are
dark) and texture (smooth or rough), and the rat had to attend
associated with a decreased bias for immediate reward (Smith
one or the other feature. Three groups were tested, an acute E2
et al., 2014).
group (two injections, 10 mg kg1, s.c.; 48 h and 24 h before
The effects of E2 on decision making has also been assessed
training), a continuous E2 (40 pg ml1, serum via silastic
in rats (Uban et al., 2012) but yielded opposite results, as high
implants), and a no replacement group. Both E2 groups
EB replacement (10 mg, s.c.) resulted in reduced preference for
required less trials than the no replacement group to acquire
the more costly/higher reward lever on the ED task.
the ED shift.

1.16.3.2.4.4 Summary of E2 Effects on Task Switching 1.16.3.2.5.1 Summary of E2 Effects on Decision Making
Mixed findings are reported across species, and the direction Few studies have investigated the effects of ovarian
of the effect appears to depend on the task. E2 impairs hormones on decision making. While ovarian hormones
reversal learning in both NHPs and aged rats and enhances do not appear to influence performance on the IGT,
set-shifting in postmenopausal women, aged monkeys, and elevated E2 levels during the menstrual cycle in young
young rats. The enhanced effect of E2 on set-shifting is women are associated with improved performance on the
dependent on the timing of replacement, as it has no effect DD task. However, high EB replacement in young rats is
when administered following a critical period. Since associated with impaired ED task performance. While these
different PFC structures are implicated in these two tasks, results provide some support for the idea that E2 modulates
446 Ovarian Hormones and Prefrontal Cortex-Related Cognition

cost/benefit analysis, more research is necessary to clarify not only biases animals to use response memory, but it
the direction of the effect. also enhances response learning and memory and vice versa.
Under normal circumstances, male rats will begin to solve
a maze by using place memory, but as they become highly
1.16.3.3 Multiple Memory System Bias
trained on the task they switch to using response memory
Rats learning to complete a maze can use one of several (Chang and Gold, 2003).
memory systems (Tolman et al., 1946). Firstly, they may use A seminal paper by Donna Korol et al. (2004) showed
several cues from the environment, or allocentric cues, to that when naturally cycling female rats were allowed to
develop a cognitive map and use this approach to find the solve a modified plus maze using any learning and memory
reward. This approach is referred to as ‘place’ learning and system, the majority of proestrous rats used place memory
memory and is mediated predominantly by the hippocampus while most estrous rats used response memory. This finding
(Packard and McGaugh, 1996). Secondly, rats may use egocen- has been repeatedly confirmed (for review see Hussain
tric cues to learn where to go to find the reward. In this case, the et al., 2014). Female rats in the modified plus maze para-
rats would learn a series of habitual motor responses such as digm are considered overtrained; that is, upon reaching
turn left or turn right. This approach is referred to as ‘response’ criterion they have found the reward in the same arm for
learning and memory and is mediated predominately by the at least 40 trials. This led to the question, why do high E
dorsal striatum, also known as the caudate/putamen (Packard female rats persist in using a hippocampus-based place/
and McGaugh, 1996). Thirdly, rats may also rely on the spatial memory system? It is well established that E
memory of a single, specific extramaze cue such as go toward improves hippocampus-based behaviors, but this does not
the door in the room, or an intramaze cue such as go down explain why these rats do not eventually switch to using
the arm that has a scratch on it. As would be expected, rats response memory like the males do.
use a combination of these memory systems under normal E interacts with DA to influence memory bias in female
circumstances when they are allowed to use any memory rats. Systemically administered DA D1 and D2 receptor antag-
system to learn where to go in a maze to find a reward (Chang onists are able to differentially shift the memory system used
and Gold, 2003). by female rats administered low but not high E (Quinlan
It was Tolman and colleagues in the 1940s (Tolman et al., et al., 2008). However, there is a limitation to the interpreta-
1946) who first proposed that rats can use either place or tion of those findings because DA antagonists were adminis-
response memory to solve a maze. They showed that initially, tered systemically. At that point, it could not be concluded
male rats more readily use place memory over response that E is acting via DA specifically in the striatum to shift
memory but are capable of using either one. We now know memory system bias. Another experiment correlated striatal
that male rats use a combination of both memory systems dopamine levels, measured via in vivo microdialysis, with
when both can be used to solve the maze (Chang and Gold, the amount of bias in using either place or response memory
2003) switching from an initial place memory system to a later (Hussain et al., 2016a). In that study it was found that while E
response memory system upon repetition of testing in the same levels were a significant predictor of whether a female rat
maze. It was later shown by Norman White and colleagues would use place or response memory, dorsal striatal dopa-
(e.g., McDonald and White, 1993, 1994) that selective inactiva- mine levels themselves were not a significant predictor of
tion of the hippocampus impairs the use of a place memory, memory system bias. Could estrogen be acting on dopamine
while inactivation of the dorsal striatum results in deficient elsewhere in the brain to affect which memory system is used
response memory (Packard and McGaugh, 1996). Spatial in females?
WM is impaired in rats with hippocampal damage but not Another study infused E directly into the PFC and tested
dorsal striatal damage (Kesner, 1990). Response memory of memory bias 15 min later. A dramatic shift in memory system
a directional turn, on the other hand, is attenuated in rats bias was observed. When E was infused into the PFC of low E
with dorsal striatal lesions but not hippocampal lesions rats, they all switched to using place memory but did not switch
(Cook and Kesner, 1988). During response tasks in which when vehicle was infused (Almey et al., 2014). The fact that E
dorsal striatal lesions significantly impair performance, rats has a rapid effect on memory system bias in the PFC indicated
with damaged hippocampi, but intact dorsal striatal function, that estrogen must be acting on membrane-associated E recep-
exhibit an enhanced performance compared to normal rats tors in the PFC and not intracellular receptors which would
(Matthews et al., 1995). This suggests a dissociation of the take hours.
neural bases for particular memory systems in which the effec- There has only been one study on women and multiple
tiveness of a particular strategy in solving a task is enhanced if memory system bias published to date. Women also show
relevant information from the alternative structure is made a bias on which memory system they use that is dependent
unavailable. upon phase of the menstrual cycle. Women tested in the
These studies, notably all using male rats, strongly suggest mid/late luteal phase, when progesterone is high, predomi-
the existence of structurally and functionally distinct nantly use spatial/place memory, whereas the opposite
multiple learning and memory systems in the brain. It is pattern is observed in women in the early follicular and ovula-
also important to note that it is not the mere loss of function tory phases (Hussain et al., 2016b). Surprisingly, it appears
of one structure that allows the other to take over, but that that progesterone is most important for memory system
when one structure is impaired, the learning and memory bias in women. The actions of progesterone in the PFC in
system subserved by the other brain structure appears to be women are unknown. We are not aware of any study investi-
enhanced. That is, lesion of the hippocampus, for example, gating multiple memory system bias in NHPs.
Ovarian Hormones and Prefrontal Cortex-Related Cognition 447

1.16.4 Effects of Ovarian Hormones on the PFC GluN2B-dependent is also influenced by endogenous
fluctuations of E2 (Galvin and Ninan, 2014). While E2
1.16.4.1 Receptor Expression
rapidly affects signaling in the PFC, suggesting nongenomic
All three estrogen receptors (ERa, ERb, GPER1) are expressed in mechanisms, it is currently unknown whether the effect on
the PFC of rats and monkeys (Almey et al., 2014; Crimins et al., PFC-dependent cognition is also nongenomic in nature.
2016; Pau et al., 1998). Among female rats, these receptors are Studies are needed to determine the mechanisms through
localized almost exclusively at extranuclear sites, including which E2 influences cognition in this region.
axons and axon terminals (Almey et al., 2014), suggesting A limited number of studies have examined the effect of
that estrogens alter presynaptic transmission in the PFC. progesterone treatment on synaptic plasticity in the PFC, and
GPER1 in the monkey PFC is also localized predominantly among these studies none have examined the independent
on presynaptic membranes (Crimins et al., 2016). ERa and effects of this hormone. One study examined the effect of
ERb mRNA are found in human PFC, although their distribu- combined E2 replacement (p.o., 25–30 pg ml1, serum) with
tion varies, with ERb found in layers V–VI, whereas ERa is either P (via implant) or medroxyprogesterone acetate (MPA;
restricted to layer V (Osterlund et al., 2000a; Osterlund et al., via implant) on synapse number in middle-aged OVX rats.
2000b). MPA is a synthetic progestin that is most commonly prescribed
Both progesterone receptor isoforms (PR-A and PR-B) are to women (Chisholm and Juraska, 2012). The number of
expressed in the frontal cortex of rats (Guerra-Araiza et al., synapses was inferred from the number of boutons expressing
2003), although the PR-B isoform appears to be more synaptophysin, a synaptic protein, which was measured in both
abundant than PR-A in this area (Guerra-Araiza et al., 2000). the PL and IL of the mPFC. While E2 alone had no effect on
synapse number, E2 combined with MPA treatment increased
the number of synaptophysin-expressing boutons. E2
1.16.4.2 Local Synthesis
combined with P had no effect on synapse number. These
While E2 is synthesized in the ovaries of females, there is also results provide some support for the idea that certain proges-
evidence that it is produced locally in the brain of both sexes, terone treatments may regulate synaptic number, although it
including in the PFC. Aromatase, which is the enzyme that is not clear if the results reflect independent or combined effects
converts testosterone to E2, is expressed in astrocytes in this of E2 and MPA. While MPA when combined with E2 may
region (Luchetti et al., 2011). Aromatase expression in the increase synapse number, it does not appear to influence
PFC appears to be higher in female than male rats (Wei et al., dendritic spine density. Progesterone given either cyclically or
2014). While E2 concentrations are detectable in the PFC, continuously (cyclic: 100 mg, given for 10 days after acute
levels are comparable between gonadally intact and OVX injection of E2, then every 28 days; continuous: 2.79–
female rats (Barker and Galea, 2009), further supporting the 3.40 ng ml1, serum via daily capsules) combined with E2
idea that E2 is synthesized locally in the PFC. Given that E2 (cyclic: 100 mg, i.m, given once every 28 days; or continuous:
levels were not detectable in males (intact and GDX), it seems 91.62–449.28 pg ml1, serum via silastic capsules) had no
plausible that sex differences exist in the local synthesis of E2 in effect on spine density in the DLPFC in aged OVX macaques
this region. (Ohm et al., 2012; Young et al., 2013). However, since synap-
tophysin is a presynaptic marker, it is possible that the effects of
P are restricted to presynaptic neurons.
1.16.4.3 Effects on Synaptic Plasticity and Signaling
in the PFC
1.16.4.4 Correlations between Changes in Neural Substrates
While cyclic E2 replacement (100 mg estradiol cypionate, i.m.
and Cognitive Performance
given every 3 weeks) improves WM in aged OVX macaques,
no effect is observed in young monkeys (Hao et al., 2007; Wang et al. (2010) provide some evidence for a role of ERa in
Rapp et al., 2003). However, dendritic spine density in layer PFC-dependent cognition by correlating receptor expression
III pyramidal neurons of the DLPFC (area 46) is enhanced by with DR task performance. In young OVX monkeys, DR task
this E2 regimen in both age groups (Hao et al., 2007). OVX performance was shown to correlate with presynaptic
is associated with reduced dendritic spine density in the expression of ERa (Wang et al., 2010). In aged OVX given
mPFC of female rats (Wallace et al., 2006). In addition to spine cyclic E2 replacement (100 mg ml1 estradiol cypionate, i.m.
density, cyclic E2 replacement (100 mg every 3 weeks, i.m.) is every 3 weeks), DR task performance was correlated with
also associated with increased expression of spinophilin, a post- postsynaptic expression of ERa within the postsynaptic
synaptic protein, in young macaques (Tang et al., 2004). density. A decrease in the expression of GPER1-containing
Spinophilin expression is also enhanced by continuous E2 nonperforated axospinous synapse density was also observed
replacement (via time-release pellets) in OVX rats (Khan in aged OVX macaque, which was partially restored following
et al., 2013). Taken together, these data suggest that E2 cyclic E2 replacement. However, this marker was not
increases synaptic density in the PFC. correlated with DR task performance.
In addition to increasing synaptic density, E2 replacement Multisynaptic boutons (MSBs), which are presynaptic
also appears to increase excitatory glutamatergic synapses, but protrusions that form multiple synapses with other neurons,
has no effect on GABAergic synapses in the PFC (Khan et al., are sensitive to OVX in aged monkeys in area 46 of the DLPFC.
2013). In the same study E2 replacement was shown to rapidly While OVX reduces the frequency of these boutons, cyclic E2
enhance ERK (2–7 min) and Akt (10–30 min) signaling in this replacement (100 mg ml1 estradiol cypionate, i.m. every
structure (Khan et al., 2013). Synaptic potentiation that is 3 weeks) restores MSBs to levels comparable to young and
448 Ovarian Hormones and Prefrontal Cortex-Related Cognition

aged premenopausal monkeys. MSBs in this region are also cognitive function in males, despite evidence that androgens
positively correlated with DR task performance (Hara et al., alter synaptic plasticity in PFC in male rats (Hajszan et al.,
2016), suggesting a potential role in E2 mediated improve- 2007) and that some PFC function is modulated by
ments in WM ability in aged OVX monkeys. androgens in humans (Janowsky et al., 2000), and rodents
(Kritzer et al., 2007; Wallin and Wood, 2015). A better
understanding of the effects of gonadal hormones and
1.16.5 Conclusion neuroestrogens throughout the life span in males and
females will allow the design of new interventions for
This chapter was organized around a set of cognitive constructs successful cognitive aging in men and women.
that form executive function, the mental processes orchestrated
by the PFC that are necessary for the cognitive control of
behavior. These cognitive processes, which manipulate WM References
through updating, selective attention, response inhibition, task
switching, and decision making, all show some degree of sensi- Almey, A., Cannel, E., Bertram, K., Filardo, E., Milner, T.A., Brake, W.G., 2014. Medial
tivity to estrogens in females, but the direction and magnitude prefrontal cortical estradiol rapidly alters memory system bias in female rats:
of these effects are far from being uniform. Differential effects ultrastructural analysis reveals membrane-associated estrogen receptors as
potential mediators. Endocrinology 155, 4422–4432.
of estrogens on PFC-dependent cognition are observed
Almey, A., Hafez, N.M., Hantson, A., Brake, W.G., 2013. Deficits in latent inhibition
according to a variety of task-, drug-, and population-related induced by estradiol replacement are ameliorated by haloperidol treatment. Front.
factors such as specific task demands, dose, duration and type Behav. Neurosci. 7, 136.
of E2, duration of estrogen loss, and age of the subjects. There Altamirano, L.J., Fields, H.L., D’Esposito, M., Boettiger, C.A., 2011. Interaction
are also species differences that are not clearly understood. For between family history of alcoholism and locus of control in the opioid regulation of
impulsive responding under the influence of alcohol. Alcohol. Clin. Exp. Res. 35,
example, most rodent studies report significant effects of 1905–1914.
estrogens on PFC-dependent function in young as well as Armstrong, K.M., Chang, M.H., Moore, T., 2009. Selection and maintenance of spatial
older animals. In contrast, NHP studies rarely find significant information by frontal eye field neurons. J. Neurosci. 29 (50), 15621–15629.
effects in younger subjects. As most human studies have been Bachevalier, J., Mishkin, M., 1986. Visual recognition impairment follows ventromedial
but not dorsolateral prefrontal lesions in monkeys. Behav. Brain Res. 20,
conducted in older women, it is not clear how age affects the
249–261.
E2 modulation of PFC function in women. Another Baddeley, A.D., Hitch, G.J., 1974. Working memory. In: Bower, G.A. (Ed.), The
important factor to consider is how E2 interacts with Psychology of Learning and Motivation: Advances in Research and Theory.
neurotransmitters such as dopamine, acetylcholine, and Academic Press, New York, pp. 47–89.
serotonin to influence PFC-dependent cognition. For example, Barker, J.M., Galea, L.A.M., 2009. Sex and regional differences in estradiol content in
the prefrontal cortex, amygdala and hippocampus of adult male and female rats.
it has been demonstrated that E2 can have enhancing or Gen. Comp. Endocrinol. 164 (1), 77–84.
impairing effects on WM depending on dopamine levels Baxter, M.G., Voytko, M.L., 1996. Spatial orienting of attention in adult and aged
(Jacobs and D’Esposito, 2011), and dopaminergic drugs rhesus monkeys. Behav. Neurosci. 110, 898–904.
can influence the effect of E2 on LI (Almey et al., 2013). Beason-Held, L.L., Rosene, D.L., Killiany, R.J., Moss, M.B., 1999. Hippocampal
formation lesions produce memory impairment in the rhesus monkey. Hippo-
While some studies examine how E2 interacts with
campus 9 (5), 562–574.
neurotransmitters to modulate cognition, more attention is Bechara, A., Damasio, A.R., Damasio, H., Anderson, S.W., 1994. Insensitivity to
needed to understand the discrepant results reported in the future consequences following damage to human prefrontal cortex. Cognition 50,
literature. 7–15.
Another fruitful avenue of research is to further our under- Bechara, A., Damasio, H., Tranel, D., Anderson, S.W., 1998. Dissociation of working
memory from decision making within the human prefrontal cortex. J. Neurosci. 18,
standing of the impact of neuroestrogens on cognition. Indeed, 428–437.
as suggested by others in the context of hippocampal function Berman, K.F., Schmidt, P.J., Rubinow, D.R., et al., 1997. Modulation of cognition-
(Prange-Kiel et al., 2013), estrogens synthesized in the brain specific cortical activity by gonadal steroids: a positron-emission tomography
may be more important for cognitive function than peripheral study in women. Proc. Natl. Acad. Sci. U.S.A. 94 (16), 8836–8841.
Bimonte-Nelson, H.A., Singleton, R.S., Williams, B.J., Granholm, A.C.E., 2004. Ovarian
estrogens. Interestingly, we still do not know whether E2
hormones and cognition in the aged female rat: II. Progesterone supplementation
synthesized in the brain declines with age in any species. reverses the cognitive enhancing effects of ovariectomy. Behav. Neurosci. 118 (4),
Furthering our knowledge in this area would likely provide 707–714.
crucial cues to understanding the complex effects of age and Bimonte-Nelson, H.A., Singleton, R.S., Hunter, C.L., Price, K.L., Moore, A.B.,
ovarian function loss on cognitive function. Recent data in Granholm, A.C.E., 2003. Ovarian hormones and cognition in the aged female rat: I.
Long-term, but not short-term, ovariectomy enhances spatial performance. Behav.
the macaque HPC (Higaki et al., 2012) revealed that aromatase Neurosci. 117, 1395–1406.
expression was upregulated in premenopausal OVX monkeys Bohacek, J., Daniel, J.M., 2010. The beneficial effects of estradiol on attentional
without changes in ERb expression, while ERb expression was processes are dependent on timing of treatment initiation following ovariectomy in
markedly upregulated in menopausal monkeys, with little middle-aged rats. Psychoneuroendocrinology 35 (5), 694–705.
Buschman, T.J., Miller, E.K., 2007. Top-down versus bottom-up control of attention in
change in aromatase expression. These data suggest that neuro-
the prefrontal and posterior parietal cortices. Science 315 (5820), 1860–1862.
estrogens are likely to play a crucial part in the modulation of Butts, K.A., Floresco, S.B., Phillips, A.G., 2013. Acute stress impairs set-shifting but
cognition throughout the life span. Another important issue is not reversal learning. Behav. Brain Res. 252, 222–229.
to further our knowledge of the effects of progestins on PFC- Cassaday, H.J., Nelson, A.J.D., Pezze, M.A., 2014. From attention to memory along
dependent function. With a few exceptions, the role of the dorsal-ventral axis of the medial prefrontal cortex: some methodological
considerations. Front. Syst. Neurosci. 8, 160.
progesterone in PFC-dependent processes has been largely Chang, Q., Gold, P.E., 2003. Switching memory systems during learning: changes in
overlooked. Finally, there is a paucity of data across species patterns of brain acetylcholine release in the hippocampus and striatum in rats.
on the effects of androgens (or estrogens) on PFC-mediated J. Neurosci. 23, 3001–3005.
Ovarian Hormones and Prefrontal Cortex-Related Cognition 449

Chisholm, N.C., Juraska, J.M., 2012. Effects of long-term treatment with estrogen and Gregoriou, G.G., Gotts, S.J., Zhou, H., Desimone, R., 2009. High-frequency long-range
medroxyprogesterone acetate on synapse number in the medial prefrontal cortex of coupling between prefrontal and visual cortex during attention. Science 324
aged female rats. Menopause 19 (7), 804–811. (5931), 1207–1210.
Christakou, A., Brammer, M., Giampietro, V., Rubia, K., 2009. Right ventro- Guerra-Araiza, C., Cerbón, M.A., Morimoto, S., Camacho-Arroyo, I., 2000. Proges-
medial and dorsolateral prefrontal cortices mediate adaptive decisions under terone receptor isoforms expression pattern in the rat brain during the estrotts
ambiguity by integrating choice utility and outcome evaluation. J. Neurosci. cycle. Life Sci. 66 (18), 1743–1752.
29, 11020–11028. Guerra-Araiza, C., Villamar-Cruz, O., González-Arenas, A., Chavira, R., Camacho-
Chudasama, Y., Muir, J.L., 2001. Visual attention in the rat: a role for the prelimbic Arroyo, I., 2003. Changes in progesterone receptor isoforms content in the rat
cortex and thalamic nuclei? Behav. Neurosci. 115, 417–428. brain during the oestrous cycle and after oestradiol and progesterone treatments.
Chudasama, Y., Robbins, T.W., 2006. Functions of frontostriatal systems in cognition: J. Neuroendocrinol. 15 (10), 984–990.
comparative neuropsychopharmacological studies in rats, monkeys and humans. Hajszan, T., Maclusky, N.J., Johansen, J.A., Jordan, C.L., Leranth, C., 2007. Effects of
Biol. Psychol. 73 (1), 19–38. androgens and estradiol on spine synapse formation in the prefrontal cortex of
Cohen, J.D., Perlstein, W.M., Braver, T.S., et al., 1997. Temporal dynamics of brain normal and testicular feminization mutant male rats. Endocrinology 148,
activation during a working memory task. Nature 386, 604–608. 1963–1967.
Collins, P., Roberts, A.C., Dias, R., Everitt, B.J., Robbins, T.W., 1998. Perseveration Hampson, E., Morley, E.E., 2013. Estradiol concentrations and working memory
and strategy in a novel spatial self-ordered sequencing task for nonhuman performance in women of reproductive age. Psychoneuroendocrinology 38 (12),
primates: effects of excitotoxic lesions and dopamine depletions of the prefrontal 2897–2904.
cortex. J. Cogn. Neurosci. 10, 332–354. Hao, J., Rapp, P.R., Janssen, W.G.M., Lou, W., Lasley, B.L., Hof, P.R., Morrison, J.H.,
Colzato, L.S., Hertsig, G., van den Wildenberg, W.P.M., Hommel, B., 2010. Estrogen 2007. Interactive effects of age and estrogen on cognition and pyramidal neurons in
modulates inhibitory control in healthy human females: evidence from the stop- monkey prefrontal cortex. Proc. Natl. Acad. Sci. U.S.A. 104 (27), 11465–11470.
signal paradigm. Neuroscience 167, 709–715. Hara, Y., Yuk, F., Puri, R., Janssen, W.G.M., Rapp, P.R., Morrison, J.H., 2016.
Cook, D., Kesner, R.P., 1988. Caudate nucleus and memory for egocentric localization. Cellular/molecular estrogen restores multisynaptic boutons in the dorsolateral
Behav. Neural Biol. 49, 332–343. prefrontal cortex while promoting working memory in aged rhesus monkeys.
Courtney, S.M., Petit, L., Maisog, J.M., Ungerleider, L.G., Haxby, J.V., 1998. An area J. Neurosci. 36 (3), 901–910.
specialized for spatial working memory in human frontal cortex. Science 279 Heaton, R.K., 1981. The Wisconsin Card Sorting Test Manual. Psychological
(5355), 1347–1351. Assessment Resources, Odessa.
Crimins, J.L., Wang, A.C.J., Yuk, F., et al., 2016. Diverse synaptic distributions of G Heekeren, H.R., Marrett, S., Ruff, D.A., Bandettini, P.A., Ungerleider, L.G., 2006.
protein-coupled estrogen receptor 1 in monkey prefrontal cortex with aging and Involvement of human dorsolateral prefrontal cortex in perceptual decision making
menopause. Cereb. Cortex 1–12. is independent of response modality. Proc. Natl. Acad. Sci. U.S.A. 103 (26),
Dalley, J.W., Cardinal, R.N., Robbins, T.W., 2004. Prefrontal executive and cognitive 10023–10028.
functions in rodents: neural and neurochemical substrates. Neurosci. Biobehav. Heidbreder, C.A., Groenewegen, H.J., 2003. The medial prefrontal cortex in the rat:
Rev. 28 (7), 771–784. evidence for a dorso-ventral distinction based upon functional and anatomical
Daniel, J.M., Hulst, J.L., Berbling, J.L., 2006. Estradiol replacement enhances working characteristics. Neurosci. Biobehav. Rev. 27 (6), 555–579.
memory in middle-aged rats when initiated immediately after ovariectomy but not Higaki, S., Takumi, K., Itoh, M., et al., 2012. Response of ERb and aromatase
after a long-term period of ovarian hormone deprivation. Endocrinology 147 (1), expression in the monkey hippocampal formation to ovariectomy and menopause.
607–614. Neurosci. Res. 72, 148–154.
Dias, R., Robbins, T.W., Roberts, A.C., 1996a. Dissociation in prefrontal cortex of Holmes, M.M., Wide, J.K., Galea, L.A.M., 2002. Low levels of estradiol facilitate,
affective and attentional shifts. Nature 380, 69–72. whereas high levels of estradiol impair, working memory performance on the radial
Dias, R., Robbins, T.W., Roberts, A.C., 1996b. Primate analogue of the Wisconsin arm maze. Behav. Neurosci. 116 (5), 928–934.
Card Sort Test: effects of excitotoxic lesions of the prefrontal cortex in the Hussain, D., Cossette, M.P., Brake, W.G., 2016a. High estradiol replacement reverses
marmoset. Behav. Neurosci. 110, 872–886. response memory bias in ovariectomized female rats regardless of dopamine levels
Donders, F.C., 1969. On the speed of mental processes. Acta Psychol. 30, 412–431. in the dorsal striatum. J. Neuroendocrinol. 28.
Dudchenko, P.A., 2004. An overview of the tasks used to test working memory in Hussain, D., Hanafi, S., Konishi, K., Brake, W.G., Bohbot, V.D., 2016b. Modulation of
rodents. Neurosci. Biobehav. Rev. 28 (7), 699–709. spatial and response strategies by phase of the menstrual cycle in women tested in
Duff, S.J., Hampson, E., 2000. A beneficial effect of estrogen on working memory in a virtual navigation task. Psychoneuroendocrinology 70, 108–117.
postmenopausal women taking hormone replacement therapy. Horm. Behav. 38 Hussain, D., Shams, W., Brake, W., 2014. Estrogen and memory system bias in
(4), 262–276. females across the lifespan. Transl. Neurosci. 5, 35–50.
Elliott, R., Dolan, R.J., Frith, C.D., 2000. Dissociable functions in the medial and lateral Jacobs, E., D’Esposito, M., 2011. Estrogen shapes dopamine-dependent cognitive
orbitofrontal cortex: evidence from human neuroimaging studies. Cereb. Cortex 10 processes: implications for women’s health. J. Neurosci. 31 (14), 5286–5293.
(3), 308–317. Jacobsen, C.F., 1936. The functions of the frontal association areas in monkeys.
Escobar, M., Oberling, P., Miller, R.R., 2002. Associative deficit accounts of disrupted Comp. Psychol. Monogr. 13, 1–60.
latent inhibition and blocking in schizophrenia. Neurosci. Biobehav. Rev. 26 (2), Janowsky, J.S., Chavez, B., Orwoll, E., 2000. Sex steroids modify working memory.
203–216. J. Cogn. Neurosci. 12 (3), 407–414.
Fellows, L.K., Farah, M.J., 2003. Ventromedial frontal cortex mediates affective Jones, B., Mishkin, M., 1972. Limbic lesions and the problem of stimulus-
shifting in humans: evidence from a reversal learning paradigm. Brain 126, reinforcement associations. Exp. Neurol. 36 (2), 362–377.
1830–1837. Kahn, J.B., Ward, R.D., Kahn, L.W., et al., 2012. Medial prefrontal lesions in mice
Floresco, S.B., Zhang, Y., Enomoto, T., 2009. Neural circuits subserving behavioral impair sustained attention but spare maintenance of information in working
flexibility and their relevance to schizophrenia. Behav. Brain Res. 204 (2), memory. Learn. Mem. 19 (11), 513–517.
396–409. Keenan, P.A., Ezzat, W.H., Ginsburg, K., Moore, G.J., 2001. Prefrontal cortex as the
Fuster, J.M., Alexander, G.E., 1971. Neuron activity related to short-term memory. site of estrogen’s effect on cognition. Psychoneuroendocrinology 26 (6), 577–590.
Science 173, 652–654. Kehagia, A.A., Murray, G.K., Robbins, T.W., 2010. Learning and cognitive flexibility:
Galvin, C., Ninan, I., 2014. Regulation of the mouse medial prefrontal cortical frontostriatal function and monoaminergic modulation. Curr. Opin. Neurobiol. 20
synapses by endogenous estradiol. Neuropsychopharmacology 39 (9), (2), 169–192.
2086–2094. Kesner, R.P., 1990. Memory for frequency in rats: role of the hippocampus and medial
Gibbs, R.B., 2000. Long-term treatment with estrogen and progesterone enhances prefrontal cortex. Behav. Neural Biol. 53, 402–410.
acquisition of a spatial memory task by ovariectomized aged rats. Neurobiol. Aging Khan, M.M., Dhandapani, K.M., Zhang, Q.G., Brann, D.W., 2013. Estrogen regulation
21 (1), 107–116. of spine density and excitatory synapses in rat prefrontal and somatosensory
Gibbs, R.B., Chipman, A.M., Hammond, R., Nelson, D., 2011. Galanthamine plus cerebral cortex. Steroids 78 (6), 614–623.
estradiol treatment enhances cognitive performance in aged ovariectomized rats. Knight, R.T., Grabowecky, M.F., Scabini, D., 1995. Role of human prefrontal cortex in
Horm. Behav. 60 (5), 607–616. attention control. Adv. Neurol. 66, 34–36.
Goldman-Rakic, P.S., 1996. The prefrontal landscape: implications of functional Korol, D.L., Malin, E.L., Borden, K.A., Busby, R.A., Couper-Leo, J., 2004. Shifts in
architecture for understanding human mentation and the central executive. Philos. preferred learning strategy across the estrous cycle in female rats. Horm. Behav.
Trans. R. Soc. Lond. Ser. B 351, 1445–1453. 45 (5), 330–338.
450 Ovarian Hormones and Prefrontal Cortex-Related Cognition

Kritzer, M.F., Brewer, A., Montalmant, F., Davenport, M., Robinson, J.K., 2007. Effects Moss, M.B., Killiany, R.J., Lai, Z.C., Rosene, D.L., Herndon, J.G., 1997. Recognition
of gonadectomy on performance in operant tasks measuring prefrontal cortical memory span in rhesus monkeys of advanced age. Neurobiol. Aging 18 (1),
function in adult male rats. Horm. Behav. 51, 183. 13–19.
Kromrey, S.A., Czoty, P.W., Nader, M.A., 2015. Relationship between estradiol and Muir, J.L., Everitt, B.J., Robbins, T.W., 1996. The cerebral cortex of the rat and visual
progesterone concentrations and cognitive performance in normally cycling female attentional function: dissociable effects of mediofrontal, cingulate, anterior dorso-
cynomolgus monkeys. Horm. Behav. 72, 12–19. lateral, and parietal cortex lesions on a five-choice serial reaction time task. Cereb.
Krug, R., Born, J., Rasch, B., 2006. A 3-day estrogen treatment improves prefrontal Cortex 6, 470–481.
cortex-dependent cognitive function in postmenopausal women. Psychoneur- Nakahara, K., Hayashi, T., Konishi, S., Miyashita, Y., 2002. Functional MRI of macaque
oendocrinology 31 (8), 965–975. monkeys performing a cognitive set-shifting task. Science 295 (5559),
Lacreuse, A., Chang, J., Metevier, C.M., LaClair, M., Meyer, J.S., Ferris, C.M., 2014. 1532–1536.
Oestradiol modulation of cognition in adult female marmosets (Callithrix jacchus). Ohm, D.T., Bloss, E.B., Janssen, W.G., et al., 2012. Clinically relevant hormone
J. Neuroendocrinol. 26 (5), 296–309. treatments fail to induce spinogenesis in prefrontal cortex of aged female rhesus
Lacreuse, A., Gullstrand, J., Fagot, J., 2016. Sex differences in inhibitory control in monkeys. J. Neurosci. 32 (34), 11700–11705.
socially-housed baboons (Papio papio). Behav. Brain Res. 312. Olton, D.S., Samuelson, R.J., 1976. Remembrance of places passed : spatial memory
Lacreuse, A., Verreault, M., Herndon, J.G., 2001. Fluctuations in spatial recognition in rats. J. Exp. Psychol. Anim. Behav. Process. 2 (2), 97–116.
memory across the menstrual cycle in female rhesus monkeys. Psychoneur- Osterlund, M.K., Grandien, K., Keller, E., Hurd, Y.L., 2000a. The human brain has
oendocrinology 26, 623–639. distinct regional expression patterns of estrogen receptor a mRNA isoforms derived
Lacreuse, A., Chhabra, R.K., Hall, M.J., Herndon, J.G., 2004. Executive function is less from alternative promoters. J. Neurochem. 75 (4), 1390–1397.
sensitive to estradiol than spatial memory: performance on an analog of the card Osterlund, M.K., Gustafsson, J.A., Keller, E., Hurd, Y.L., 2000b. Estrogen receptor beta
sorting test in ovariectomized aged rhesus monkeys. Behav. Processes 67 (2), (ERbeta) messenger ribonucleic acid (mRNA) expression within human forebrain:
313–319. distinct distribution pattern to ERalpha mRNA. J. Clin. Endocrinol. Metab. 85 (10),
Lacreuse, A., Herndon, J.G., 2003. Estradiol selectively affects processing of 3840–3846.
conspecifics’ faces in female rhesus monkeys. Psychoneuroendocrinology 28 (7), Owen, A.M., McMillan, K.M., Laird, A.R., Bullmore, E., 2005. N-back working memory
885–905. paradigm: a meta-analysis of normative functional neuroimaging studies. Hum.
Lacreuse, A., Herndon, J.G., Moss, M.B., 2000. Cognitive function in aged ovariec- Brain Mapp. 25 (1), 46–59.
tomized female rhesus monkeys. Behav. Neurosci. 114 (3), 506–513. Packard, M.G., McGaugh, J.L., 1996. Inactivation of hippocampus or caudate nucleus
Lacreuse, A., Wilson, M.E., Herndon, J.G., 2002. Estradiol, but not raloxifene, with lidocaine differentially affects expression of place and response learning.
improves aspects of spatial working memory in aged ovariectomized rhesus Neurobiol. Learn Mem. 65, 65–72.
monkeys. Neurobiol. Aging 23 (4), 589–600. Passetti, F., Chudasama, Y., Robbins, T.W., 2002. The frontal cortex of the rat and
Lacreuse, A., Wilson, M.E., Herndon, J.G., 2009. No effect of different estrogen visual attentional performance: dissociable functions of distinct medial prefrontal
receptor ligands on cognition in adult female monkeys. Physiol. Behav. 96 (3), subregions. Cereb. Cortex 12 (12), 1254–1268.
448–456. Pau, C.Y., Pau, K.Y.F., Spies, H.G., 1998. Putative estrogen receptor b and a mRNA
Lamar, M., Driscoll, I.R.A., 2007. Vulnerability of the orbitofrontal cortex to age- expression in male and female rhesus macaques. Mol. Cell. Endocrinol. 146,
associated structural and functional brain changes. Ann. N.Y. Acad. Sci. 59–68.
1121, 14. Petrides, M., 2000. Dissociable roles of mid-dorsolateral and anterior inferotemporal
Larsen, J.K., Divac, I., 1978. Selective ablations within the prefrontal cortex of the rat cortex in visual working memory. J. Neurosci. 20, 7496–7503.
and performance of delayed alternation. Physiol. Psychol. 6 (1), 15–17. Petrides, M., 2005. Lateral prefrontal cortex: architectonic and functional organization.
Lawrence, N.S., Jollant, F., O’Daly, O., Zelaya, F., Phillips, M.L., 2009. Distinct roles of Philos. Trans. R. Soc. B Biol. Sci. 360 (1456), 781–795.
prefrontal cortical subregions in the Iowa Gambling Task. Cereb. Cortex 19, Petrides, M., Alivisatos, B., Meyer, E., Evans, A.C., 1993. Functional activation of the
1134–1143. human frontal cortex during the performance of verbal working memory tasks.
Lebedev, M.A., Messinger, A., Kralik, J.D., Wise, S.P., 2004. Representation of Proc. Natl. Acad. Sci. U.S.A. 90, 878–882.
attended versus remembered locations in prefrontal cortex. PLoS Biol. 2 Petrides, M., Milner, B., 1982. Deficits on subject-ordered tasks after frontal- and
(11), e365. temporal-lobe lesions in man. Neuropsychologia 20, 249–262.
Lipatova, O., Wiener, N., Andrews, K., Kirshenbaum, A.P., Green, J.T., Petrides, M., Pandya, D.N., 1999. Dorsolateral prefrontal cortex: comparative
Toufexis, D.J., 2016. 17B-estradiol replacement in ovariectomized female rats cytoarchitectonic analysis in the human and the macaque brain and corticocortical
slows set 1 dorsolateral striatial-dependent learning and enhances learning of connection patterns. Eur. J. Neurosci. 11 (3), 1011–1036.
set 2 in an extradimensional set-shifting paradigm. Behav. Neurosci. 130 (1), Petrides, M., Pandya, D.N., 2002. Comparative cytoarchitectonic analysis in the
44–49. human and the macaque ventrolateral prefrontal cortex and corticocortical
Logan, G.D., Cowan, W.B., Davis, K.A., 1984. On the ability to inhibit simple and connection patterns in the monkey. Eur. J. Neurosci. 16, 291–310.
choice reaction time responses: a model and a method. J. Exp. Psychol. Hum. Petrides, M., Tomaiuolo, F., Yeterian, E.H., Pandya, D.N., 2012. The prefrontal cortex:
Percept. Perform. 10, 276–291. comparative architectonic organization in the human and the macaque monkey
Luchetti, S., Bossers, K., Van de Bilt, S., et al., 2011. Neurosteroid biosynthetic brains. Cortex 48 (1), 46–57.
pathways changes in prefrontal cortex in Alzheimer’s disease. Neurobiol. Aging 32 Pompili, A., Tomaz, C., Arnone, B., Tavares, M.C., Gasbarri, A., 2010. Working and
(11), 1964–1976. reference memory across the estrous cycle of rat: a long-term study in gonadally
Markowska, A.L., Savonenko, A.V., 2002. Effectiveness of estrogen replacement in intact females. Behav. Brain Res. 213 (1), 10–18.
restoration of cognitive function after long-term estrogen withdrawal in aging rats. Prange-Kiel, J., Schmutterer, T., Fester, L., et al., 2013. Endocrine regulation of
J. Neurosci. 22 (24), 10985–10995. estrogen synthesis in the hippocampus? Prog. Histochem. Cytochem. 48, 49–64.
Matthews, D.B., Simson, P.E., Best, P.J., 1995. Acute ethanol impairs spatial memory Quinlan, M.G., Duncan, A., Loiselle, C., Graffe, N., Brake, W.G., 2010. Latent inhibition
but not stimulus/response memory in the rat. Alcohol. Clin. Exp. Res. 19, is affected by phase of estrous cycle in female rats. Brain Cogn. 74 (3), 244–248.
902–909. Quinlan, M.G., Hussain, D., Brake, W.G., 2008. Use of cognitive strategies in rats: the
McDonald, R.J., White, N.M., 1993. A triple dissociation of memory systems: role of estradiol and its interaction with dopamine. Horm. Behav. 53, 185.
hippocampus, amygdala, and dorsal striatum. Behav. Neurosci. 107, 3–22. Ragozzino, M.E., Detrick, S., Kesner, R.P., 1999. Involvement of the prelimbic-
McDonald, R.J., White, N.M., 1994. Parallel information processing in the water maze: infralimbic areas of the rodent prefrontal cortex in behavioral flexibility for place and
evidence for independent memory systems involving dorsal striatum and hippo- response learning. J. Neurosci. 19, 4585–4594.
campus. Behav. Neural Biol. 61, 260–270. Rapp, P.R., Morrison, J.H., Roberts, J.A., 2003. Cyclic estrogen replacement improves
Meunier, M., Bachevalier, J., Mishkin, M., 1997. Effects of orbital frontal and anterior cognitive function in aged ovariectomized rhesus monkeys. J. Neurosci. 23 (13),
cingulate lesions on object and spatial memory in rhesus monkeys. Neuro- 5708–5714.
psychologia 35 (7), 999–1015. Reavis, R., Overman, W.H., 2001. Adult sex differences on a decision-making task
Mishkin, M., Delacour, J., 1975. An analysis of short-term visual memory in the previously shown to depend on the orbital frontal cortex. Behav. Neurosci. 115 (1),
monkey. J. Exp. Psychol. Anim. Behav. Process. 1 (4), 326–334. 196–206.
Moore, T.L., Schettler, S.P., Killiany, R.J., Rosene, D.L., Moss, M.B., 2012. Impair- Reimers, L., Buchel, C., Diekhof, E.K., 2014. How to be patient. The ability to wait for
ment in delayed nonmatching to sample following lesions of dorsal prefrontal a reward depends on menstrual cycle phase and feedback-related activity. Front.
cortex. Behav. Neurosci. 126 (6), 772–780. Neurosci. 8, 401.
Ovarian Hormones and Prefrontal Cortex-Related Cognition 451

Robbins, T.W., 2002. The 5-choice serial reaction time task: behavioural pharma- Voytko, M.L., Higgs, C.J., Murray, R., 2008. Differential effects on visual and spatial
cology and functional neurochemistry. Psychopharmacology 163, 362–380. recognition memory of a novel hormone therapy regimen of estrogen alone or
Roberts, J.A., Gilardi, K.V., Lasley, B., Rapp, P.R., 1997. Reproductive senescence combined with progesterone in older surgically menopausal monkeys. Neurosci-
predicts cognitive decline in aged female monkeys. Neuroreport 8 (8), ence 154 (4), 1205–1217.
2047–2057. Voytko, M.L., Murray, R., Higgs, C.J., 2009. Executive function and attention are
Rolls, E.T., 2004. The functions of the orbitofrontal cortex. Brain Cogn. 55, 11–29. preserved in older surgically menopausal monkeys receiving estrogen or estrogen
Rosvold, H.E., Mirsky, A.F., Sarasib, I., Bransome, E.D.B., 1956. A continuous plus progesterone. J. Neurosci. 29 (33), 10362–10370.
performance test of brain damage. J. Counsult. Psychol. 20 (5), 343–350. Wallace, M., Luine, V., Arellanos, A., Frankfurt, M., 2006. Ovariectomized rats show
Rueckert, L., Grafman, J., 1996. Sustained attention deficits in patients with right decreased recognition memory and spine density in the hippocampus and
frontal lesions. Neuropsychologia 34 (10), 953–963. prefrontal cortex. Brain Res. 1126 (1), 176–182.
Schiff, R., Bulpitt, C.J., Wesnes, K.A., Rajkumar, C., 2005. Short-term transdermal Wallin, K.G., Wood, R.I., 2015. Anabolic-androgenic steroids impair set-shifting and
estradiol therapy, cognition and depressive symptoms in healthy older women. A reversal learning in male rats. Eur. Neuropsychopharmacol. 25 (4), 583–590.
randomised placebo controlled pilot cross-over study. Psychoneuroendocrinology Wang, A.C.J., Hara, Y., Janssen, W.G.M., Rapp, P.R., Morrison, J.H., 2010.
30 (4), 309–315. Synaptic estrogen receptor-a levels in prefrontal cortex in female rhesus
Schmidt, R., Fazekas, F., Reinhart, B., et al., 1996. Estrogen replacement therapy in monkeys and their correlation with cognitive performance. J. Neurosci. 30 (38),
older women: a neuropsychological and brain MRI study. J. Am. Geriatr. Soc. 44, 12770–12776.
1307–1313. Wang, V.C., Neese, S.L., Korol, D.L., Schantz, S.L., 2009. Chronic estradiol
Seamans, J.K., Floresco, S.B., Phillips, A.G., 1995. Functional differences between the replacement impairs performance on an operant delayed spatial alternation task in
prelimbic and anterior cingulate regions of the rat prefrontal cortex. Behav. Neu- young, middle-aged, and old rats. Horm. Behav. 56 (4), 382–390.
rosci. 109 (6), 1063–1073. Wang, V.C., Neese, S.L., Korol, D.L., Schantz, S.L., 2011. Estradiol impairs response
Seo, H., Barraclough, D.J., Lee, D., 2007. Dynamic signals related to choice and inhibition in young and middle-aged, but not old rats. Neurotoxicol. Teratol. 33 (3),
outcomes in the dorsolateral prefrontal cortex. Cereb. Cortex 17 (Suppl. 1), 405–414.
110–117. Wang, V.C., Sable, H.J.K., Ju, Y.H., et al., 2008. Effects of chronic estradiol treatment
Sinopoli, K.J., Floresco, S.B., Galea, L.A.M., 2006. Systemic and local administration on delayed spatial alternation and differential reinforcement of low rates of
of estradiol into the prefrontal cortex or hippocampus differentially alters working responding. Behav. Neurosci. 122 (4), 794–804.
memory. Neurobiol. Learn. Mem. 86 (3), 293–304. Warburton, E.C., Brown, M.W., 2010. Findings from animals concerning when
Smith, C.T., Boettiger, C.A., 2012. Age modulates the effect of COMT genotype on interactions between perirhinal cortex, hippocampus and medial prefrontal cortex
delay discounting behavior. Psychopharmacology 222 (4), 609–617. are necessary for recognition memory. Neuropsychologia 48 (8), 2262–2272.
Smith, C.T., Sierra, Y., Oppler, S.H., Boettiger, C.A., 2014. Ovarian cycle effects on Wardak, C., Ibos, G., Duhamel, J.-R., Olivier, E., 2006. Contribution of the monkey
immediate reward selection bias in humans: a role for estradiol. J. Neurosci. 34 frontal eye field to covert visual attention. J. Neurosci. 26 (16), 4228–4235.
(16), 5468–5476. Wechsler, D., 2008. Wechsler Adult Intelligence Scale, Fourth Edition. Pearson, San
Smith, Y.R., Giordani, B., Lajiness-O’Neill, R., Zubieta, J.K., 2001. Long-term estrogen Antonio, TX.
replacement is associated with improved nonverbal memory and attentional Weed, M.R., Taffe, M.A., Polis, I., et al., 1999. Performance norms for a rhesus
measures in postmenopausal women. Fertil. Steril. 76 (6), 1101–1107. monkey neuropsychological testing battery: acquisition and long-term performance.
Squire, L.M., Noudoost, B., Schafer, R.J., Moore, T., 2013. Prefrontal contributions to Cogn. Brain Res. 8 (3), 185–201.
visual selective attention. Ann. Rev. Neurosci. 36, 451–466. Wei, J., Yuen, E.Y., Liu, W., Li, et al., 2014. Estrogen protects against the detrimental
Stroop, J.R., 1935. Studies of interference in serial verbal reactions. J. Exp. Psychol. effects of repeated stress on glutamatergic transmission and cognition. Mol.
18, 643–661. Psychiatry 19 (5), 588–598.
Tang, Y., Janssen, W.G., Hao, J., et al., 2004. Estrogen replacement increases Wide, J.K., Hanratty, K., Ting, J., Galea, L.A.M., 2004. High level estradiol impairs and
spinophilin-immunoreactive spine number in the prefrontal cortex of female rhesus low level estradiol facilitates non-spatial working memory. Behav. Brain Res. 155
monkeys. Cereb. Cortex 14 (2), 215–223. (1), 45–53.
Thomas, G.J., Brito, G.N.O., 1980. Recovery of delayed alternation in rats after lesions Wilson, C.R.E., Gaffan, D., Browning, P.G.F., Baxter, M.G., 2010. Functional locali-
in medial frontal cortex and septum. J. Comp. Physiol. Psychol. 94, 808–818. zation within the prefrontal cortex: missing the forest for the trees? Trends Neu-
Tolman, E.C., 1925. Purpose and cognition: the determiners of animal learning. rosci. 33 (12), 533–540.
Psychol. Rev. 32, 285–297. Winters, B.D., Saksida, L.M., Bussey, T.J., 2008. Object recognition memory:
Tolman, E.C., Ritchie, B.F., Kalish, D., 1946. Studies in spatial learning; place learning neurobiological mechanisms of encoding, consolidation and retrieval. Neurosci.
versus response learning. J. Exp. Psychol. 36, 221–229. Biobehav. Rev. 32 (5), 1055–1070.
Uban, K.A., Rummel, J., Floresco, S.B., Galea, L.A.M., 2012. Estradiol modulates Wise, S.P., 2009. Forward frontal fields: phylogeny and fundamental function. Trends
effort-based decision making in female rats. Neuropsychopharmacology 37 (2), Neurosci. 31 (12), 599–608.
390–401. Wolf, O.T., Heinrich, A.B., Hanstein, B., Kirschbaum, C., 2005. Estradiol or estradiol/
Uylings, H.B.M., Groenewegen, H.J., Kolb, B., 2003. Do rats have a prefrontal cortex? progesterone treatment in older women: no strong effects on cognition. Neurobiol.
Behav. Brain Res. 146 (1–2), 3–17. Aging 26 (7), 1029–1033.
Velázquez-Zamora, D.A., Garcia-Segura, L.M., González-Burgos, I., 2012. Effects of Wolf, C., Waksman, D., Finger, S., Almli, C.R., 1987. Large and small medial frontal
selective estrogen receptor modulators on allocentric working memory performance cortex lesions and spatial performance of the rat. Brain Res. Bull. 18 (1), 1–5.
and on dendritic spines in medial prefrontal cortex pyramidal neurons of ovari- Yoon, T., Okada, J., Jung, M., Kim, J., 2008. Prefrontal cortex and hippocampus
ectomized rats. Horm. Behav. 61 (4), 512–517. subserve different components of working memory in rats. Learn. Mem. 15,
Voytko, M.L., 2002. Estrogen and the cholinergic system modulate visuospatial 97–105.
attention in monkeys (Macaca fascicularis). Behav. Neurosci. 116 (2), 187–197. Young, M.E., Ohm, D.T., Janssen, W.G.M., Gee, N.A., Lasley, B.L., Morrison, J.H.,
Voytko, M.L., 2000. The effects of long-term ovariectomy and estrogen replacement 2013. Continuously delivered ovarian steroids do not alter dendritic spine density or
therapy on learning and memory in monkeys (Macaca fascicularis). Behav. Neu- morphology in macaque dorsolateral prefrontal cortical neurons. Neuroscience
rosci. 114 (6), 1078–1087. 255, 219–225.
This page intentionally left blank
1.17 Behavioral and Neuroendocrine Indicators of Well-being in Farm
and Laboratory Mammals
Gabriela González-Mariscal, Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala,
Tlaxcala, Mexico
Anne Sisto Burt, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
Raymond Nowak, INRA, UMR85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247
Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université François Rabelais de Tours, Tours, France; and
Institut Français du cheval et de l’Equitation, Nouzilly, France
Ó 2017 Elsevier Inc. All rights reserved.

1.17.1 Introduction 454


1.17.2 On Rodents 454
1.17.2.1 Regarding Lodging (Cages, Floors, Enrichment) 454
1.17.2.2 Regarding Environment and Procedures (Room Temperature, Noise, Cage Change, Blood Sampling) 455
1.17.2.3 Regarding Reactivity to Stress, as Modulated by Environment (Cages, Enrichment, Handling, Music) 455
1.17.2.4 Regarding Euthanasia Methods 456
1.17.3 On Rabbits 456
1.17.3.1 Regarding Lodging (Cages, Floors, Enrichment) 456
1.17.3.2 Regarding Environment (Temperature, Noise, Vibration) 458
1.17.3.3 Regarding Reduction in Reactivity to Stress by Neonatal Manipulations or Supplements in Adulthood 459
1.17.4 On Sheep and Goats 460
1.17.4.1 Regarding Behavioral Needs 460
1.17.4.1.1 Sexual Behavior 460
1.17.4.1.2 Welfare Issues Specific to Sexual Behavior 461
1.17.4.1.3 Maternity 462
1.17.4.1.4 Welfare Issues Specific to Maternal Behavior 463
1.17.4.1.5 Infancy 464
1.17.4.1.6 ‘Followers’ versus ‘Hiders’ 465
1.17.4.1.7 Welfare Issues Specific to the Young 465
1.17.4.2 Regarding Lodging (Cages, Floors, Nest Boxes, ‘Enrichment’) 467
1.17.4.2.1 Outdoor Facilities 467
1.17.4.2.2 Floor 467
1.17.4.2.3 Social Enrichment 467
1.17.4.2.4 Physical Enrichment 468
1.17.4.2.5 Human Contact 468
1.17.4.3 Regarding Environment (Light, Sound, Vibration, Temperature, Density of Animals) 469
1.17.4.3.1 Flocking 469
1.17.4.3.2 Dominance and Agonistic Behaviors 469
1.17.4.3.3 Social Isolation 469
1.17.4.3.4 Density of Animals 470
1.17.4.3.5 Heat and Cold Stress 471
1.17.4.3.6 Lighting and Noise 471
1.17.4.4 Regarding Modifications of Reactivity to Stress by Neonatal Manipulations 471
1.17.4.4.1 Prenatal Stress in Small Ruminants 471
1.17.4.4.2 Neonatal Stress in Small Ruminants 472
1.17.4.5 Regarding Methods to Quantify Parameters Presumably Indicative of Stress 472
1.17.4.6 Regarding Sacrifice Methods 473
1.17.5 On Dairy Cattle 473
1.17.5.1 Lodging and Handling during the Peripartum Period: Relevance for the Establishment of Maternal
Behavior and Nursing 473
1.17.5.1.1 Calf Lodging 474
1.17.5.2 Lodging of Dairy Cows 475
1.17.5.3 Regarding Environment 477
1.17.5.4 Behavioral Indicators of Well-being in Bovines 477
1.17.5.4.1 Handling of Adult Bovines 478
1.17.6 Conclusions and Discussion 479
Acknowledgment 480
References 480

Hormones, Brain, and Behavior, 3rd edition, Volume 1 http://dx.doi.org/10.1016/B978-0-12-803592-4.00017-1 453


454 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

1.17.1 Introduction From the above considerations in this chapter, we will


present studies in which specific biological indicators have
Animal husbandry and experimentation in the laboratory been measured in farm and laboratory mammals (i.e., bovines,
have created artificial environments where a variety of sheep, goats, rabbits, and rodents) under a variety of housing
mammalian species spend their entire lives. The question of conditions and husbandry practices. We hope that putting
whether such living conditions (and the procedures associ- together this information will reveal the consistency of findings
ated with them) are adequate for the well-being of such within and across species and will also allow a solid interpreta-
animals is, therefore, a valid one and constitutes a research tion of results within a broad scientific framework.
topic in itself. What do we mean by well-being? What biolog-
ical indicators can we use to measure it? An approach used to
investigate these questions has been to ask whether farm and/ 1.17.2 On Rodents
or laboratory environments generate stress in the animals
therein confined (e.g., see Moberg, 2000; Yehuda and Unlike the rest of mammals dealt with in this chapter, rodents
McEwen, 2004). The advantage of such an approximation is have been largely bred and used in laboratory settings. None-
that the stress response has been formally investigated since theless, similar concerns as those expressed for rabbits (raised
Hans Selye (1956) coined the term ‘General Adaptation and used both in the lab and on the farm; see the next section)
Syndrome.’ Consequently, a vast scientific literature exists have been addressed in the studies described below. Specifi-
on the behavioral, hormonal, immunological, and autonomic cally, the impact of housing conditions (in terms of animal
responses observed following the exposure of animals to density, floor type, temperature, environmental enrichment)
a variety of presumably ‘stressful’ stimuli. Although abundant and ‘standard’ manipulations of the animals (e.g., cage change,
information has, indeed, shown that some housing condi- injections) on corticosteroid secretion, physiological responses,
tions, handling procedures, noise, lighting schedules, etc. do and particular behaviors (presumably indicative of stress) have
change specific biological indicators in particular species, been the most explored topics. Two growing areas of study
the key question of whether such changes alter the well- relate to (1) the effect of manipulating young rodents on their
being of those animals requires a theoretical framework responsiveness to stressful situations as adults and (2) the
within which such results can be interpreted. possibility that stress is ‘communicable,’ i.e., that observing
Most researchers distinguish between ‘acute’ (i.e., short (presumably) stressful procedures performed on other rodents
lasting) and ‘chronic’ (i.e., lasting for several days) stress generates stress in the witnesses.
because the consequences of each one are different and the bio-
logical ‘meaning’ of these two types of stress responses varies
1.17.2.1 Regarding Lodging (Cages, Floors, Enrichment)
widely. Acute stress is generally considered to be adaptive;
that is, it is a generalized response that allows animals to react An enriched environment (provided with, e.g., ladders, balls,
‘as a whole’ to sudden changes in the environment. In healthy hiding places) plus a partner increases sleep, exploratory
animals once the perceived threat is over, hormonal and auto- behavior, body weight, and weight of thymus and spleen,
nomic responses return to baseline. Chronic stress, by contrast, relative to (male) rats housed singly in barren cages
can provoke distress, a state of (semi) permanent alteration in (Abou-Ismail and Mahboub, 2011). In pair-housed (female)
multiple systems that, in turn, leads to disease vulnerability, mice a high clustering of the elements added for enrichment
infertility, depression, and even death. This apparently simple increased aggression, stereotypic behavior, and displacement
dichotomy, however, is complicated by the observation that of ‘the other’ mouse from resources, but it had no effect on
repeated exposures to the same acute stressor or successive fecal corticosterone (CORT) metabolites (Akre et al., 2011).
contact with several different acute stressors can ‘add up’ and Are the effects of environmental enrichment due to novelty
lead to distress. or ‘complexity of the environment’? Do they exert long-
As we will illustrate below there is great variability among lasting consequences? Housing young mice in a variety of
farm and laboratory mammals regarding what they perceive enriched cages and transferring them back to standard cages
as ‘stressful.’ This should not be surprising in view of the fact after several weeks did not modify the incidence of stereoty-
that a given stimulus per se is not stressful; it becomes such pies or the performance in the elevated-plus maze (EPM) or
depending on its interpretation by the animal. This, of course, the open-field tests, relative to animals always housed in stan-
is a function of the central nervous system whose responsive- dard cages provided only with cotton wool (Gross et al.,
ness is modified (even within a species) by factors such as 2011). The effects provoked by an enriched environment are
age, sex, reproductive condition, early rearing, familiarity critically dependent on the strain used and the responses
with the environment, etc. Moreover, a number of studies measured. Adding different types of items to standard cages
have shown that using a single biological indicator to deter- effectively reduced mean arterial pressure in male rats from
mine if a given situation or procedure is ‘stressful’ may not the F344 but not the BN strain. Moreover, enrichment did
be appropriate as, for instance, the concentration of cortisol not affect fecal CORT, heart rate, or immunoglobulin A excre-
in blood may rise as a consequence of restraint, exercise, tion (Kemppinen et al., 2010).
mating, etc. Finally, we must emphasize that, in both the The extent to which stereotypies reflect stress was assessed
farm and the laboratory, we are dealing with domesticated by comparing the coincidence of such behaviors with CORT
mammals selected across many years for specific traits, a process secretion (following a stress reactivity test) in three lines of
that may (or not) have also influenced their responsiveness to mice: a commercial outbred stock (CD-1) and two lines
specific environmental conditions and handling by humans. selected for high (HR) or low (LR) stress reactivity. Although
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 455

acute CORT secretion followed the expected outcome (i.e., transferred in midpregnancy (highest CORT levels) or late
HR > commercial stock > LR), only CD-1 mice displayed gestation (poor nest building; Malmkvist and Palme, 2015).
stereotypies (Engel et al., 2011). These findings challenge In addition to cage change individually ventilated cage
the assumption that the display of such behaviors necessarily systems are increasingly being used in rodent colonies. The
indicates stress. Moreover, in mice raised in either standard preference of mice for different levels of air speeds and air
or enriched cages there was no evidence of a causal relation- changes inside the cage has been explored. Animals do react to
ship between the incidence of stereotypies (in the home drafts but they do not seem to be affected by a high number
cage) and perseverance (quantified in an extinction task; of air changes delivered without draft (Krohn and Hansen,
Gross et al., 2012). Yet, both rats and striped mice raised 2010).
in an enriched environment are less likely to develop stereo- Blood sampling is one of the most common experimental
typies when transferred to standard cages (Gross et al., 2012; procedures performed in rodents. Several methods have been
Jones et al., 2011). These findings would suggest a ‘protective’ devised to minimize stress, especially when the procedure is
effect of early enrichment on the emergence of stereotyped performed repeatedly on the same animal. Automated blood
behaviors. sampling (through a catheter implanted in the carotid artery)
Male rats housed in cages with wire-bottom floors showed provoked the smallest effects on all parameters measured while
a higher heart rate, more body temperature fluctuations, and mice subjected to cheek blood sampling lost more body
a reduction in locomotion and body weight than their coun- weight, had higher levels of CORT, and expressed more anxious
terparts housed in cages with bedding floors (Giral et al., behavior. Mice sampled from repeated puncturing in the tail
2011). The impact of type of bedding (aspen shaving vs chips) vein showed intermediate values (Teilmann et al., 2014).
on breeding, nest-building, and anxiety-related behaviors
was compared in four strains of mice. Aspen shaving favored
1.17.2.3 Regarding Reactivity to Stress, as Modulated by
nest building (thereby increasing the number of kits weaned/
Environment (Cages, Enrichment, Handling, Music)
litter) but had no effects on other behaviors (Jackson et al.,
2015). If given the choice mice, show a preference for specific There is an ample literature showing that, in rats, the early rear-
kinds of bedding by spending more time on softwood ing environment permanently modifies their stress reactivity in
shavings > coarse-grained chips > medium chips ¼ fine chips adulthood (for reviews see Champagne et al., 2003; Fleming
(Kirchner et al., 2012). Similarly, rats prefer wood shavings et al., 1999; Lomanowska and Melo, 2016). However, the
and paper bedding over sawdust and wire mesh (Van de effects vary depending on the method used to assess stress reac-
Weerd et al., 1996). tivity and the type of manipulation young animals were sub-
The stress generated by transport (as determined by fecal jected to. Thus, four different types of handling treatments
CORT) can be reduced in guinea pigs by providing huts (to provided to male rats across PP days 57–74 were compared:
hide in), but only if animals are pair-housed (Walters et al., (1) minimal (i.e., just cage cleaning); (2) passive hand
2012). (2 min); (3) tickling (2 min); or (4) restrained on back
Single-level versus two-level cages for rat housing improved (2 min). Responses of rats were assessed across days 77–87
their ‘affective state’ but only if animals were transferred from using the following tests: EPM and reactivity to human
the former to the latter. No differences were seen in several approach and to cat odor. Ease of weighing and injection was
behaviors, immune function, or metabolism in animals that also determined. Tickled rats showed less fear in the human
remained in either type of cage (Wheeler et al., 2015). approach test, but even the passive hand and restraint treat-
ments provoked this effect. No treatment facilitated handling
during weighing and no effects were seen in the EPM test or
1.17.2.2 Regarding Environment and Procedures (Room
Cat Odor Test (CAT) (Cloutier et al., 2012). The effect of tick-
Temperature, Noise, Cage Change, Blood Sampling)
ling young male rats (across days 32–41) was further explored
Cage change is perhaps the most common ‘laboratory proce- by quantifying the emission of ultrasonic vocalizations (USV)
dure’ to which rodents are routinely subjected to. This practice, (50 kHz, positive effect; 22 kHz, negative effect) and audible
although essential to maintain hygiene for both animals and squeals. Tickling rapidly induced a positive effect (i.e., it
humans, is not trivial as it entails a major change in the increased 50 kHz USV) when applied before and even immedi-
animals’ olfactory environment. Thus, the effect of the type of ately after injections (Cloutier et al., 2015).
cage rats were transferred into was assessed in Sprague-Dawley In an attempt to reduce stress reactivity during an experi-
and Wistar adult males. Heart rate and arterial blood pressure mental procedure adult ovariectomized rats were exposed to
were increased for up to 5 h following cage change, but the silence, ‘white’ noise (with all the frequencies), or a piano
largest effects were seen when rats were transferred into sonata. In the EPM and the light–dark box transition tests noise
a new, clean barren cage with a new lid. Interestingly, adding was found to be anxiogenic and music anxiolytic. Interestingly,
an enrichment object had practically no effect but keeping the anxiolytic effect of music was enhanced by injecting proges-
the old lid reduced ‘stress reactivity’ in both strains (Meller terone or allopregnanolone (Escribano et al., 2014).
et al., 2011). Pregnant rodents can be subject to an additional Experimental procedures are commonly done in rooms
cage change, i.e., maternity cages where they will give birth and where ‘other’ rodents are present. Consequently, those animals
nurse the litter. The gestational stage when this is performed are ‘unwilling witnesses’ to the fate of their peers. To determine
can have a major impact on maternal behavior, pup survival, if observing, hearing, or smelling the reactions of experimental
and stress reactivity. Minks transferred in early pregnancy built animals (across 4 weeks) has an impact on the witnesses, the
better nests and weaned more pups than did mothers thymus, spleen, adrenal gland, and body weight was
456 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

determined in adult rats, housed four per cage. Sleeping, or perirenal fat. However, evidence of dominance hierarchies
grooming, and agonistic behavior were also quantified. Rats emerged under both conditions, as evidenced by scarring (Bell
which witnessed managerial procedures during the light phase and Bray, 1984). Young bucks housed contiguously in two-
showed more indicators of stress than those exposed during the floor cages (that allowed looking at each other closely) ‘social-
dark phase (Abou-Ismail et al., 2015). Do these results indicate ized’ more by looking at the animal in the immediate vicinity
that the (presumed) stress of other animals is communicable (Negretti et al., 2004). A different impact of bicellular cages
(Makowska and Weary, 2013)? versus wired pens was seen in Leprino versus New Zealand
White (NZW) young bucks: mortality was highest in bicellular
cages for Leprinos but in pens for the NZW breed (Mugnai
1.17.2.4 Regarding Euthanasia Methods
et al., 2014).
Concerns about animal welfare in the laboratory have to The effect of animal density within a cage was explored by
include the ways by which animals are sacrificed at the end housing unfamiliar young bucks and does in cages of different
of an experiment. A recently introduced method involves the sizes (eight animals per cage). Hardly any differences were seen
use of CO2 inhalation. A combination of CO2 concentration in the animals’ behavior: only sternal lying increased with
and flow rate that minimizes dyspnea before the onset of insen- decreasing space. Placing a platform decreased social interac-
sitivity and, ultimately, death was explored in mice. Higher tions, and enriching the environment with a wooden stick
CO2 flow rates reduced the duration of dyspnea but, even decreased cage manipulation (Buijs et al., 2001). Food intake
under such conditions, mice do experience it, for about 30 s and weight gain were compared between fattening rabbits
before they become insensitive (Moody et al., 2014). housed in either of two cages: ‘low-density and enriched’
(with platform, hiding box, and gnawing material) or ‘high-
density and barren.’ Food intake was higher in enriched cages
1.17.3 On Rabbits only across the first 2 weeks, and aggression did not occur
under any condition. Average weight gain was not significantly
A growing concern for the well-being of rabbits kept in inten- different between the two cage types (Maertens et al., 2004).
sive production units or in small farms has promoted the study The combined effects of animal density and introduction of
of how specific variables impact the health, productivity, a gnawing stick were tested on productive parameters (live
growth, and behavior of animals of different ages and repro- weight, daily weight gain) and ‘spontaneous’ behaviors.
ductive conditions. Other works have investigated how specific Although productivity was not affected, more varied behaviors
manipulations or nutritional supplements ameliorate were observed in animals living two per cage. Addition of
unwanted consequences of environmental conditions or facil- gnawing stick reduced incidence of stereotypies, alertness,
itate animal handling. In the following subsections we will and aggression (Verga et al., 2004; Figure 1). Single-housed
review such studies, highlighting their common findings as versus group-housed NZW adult females were monitored for
well as the heterogeneity of the models explored. 12 weeks regarding immune response (humoral and delayed-
type hypersensitivity response), lymphocyte counts, circulating
corticosterone, food intake, growth rate, adrenal gland weight,
1.17.3.1 Regarding Lodging (Cages, Floors, Enrichment)
cross-sectional area of adrenal cortex, and social interactions.
The type of cage used to lodge rabbits (e.g., size, animal density The only differences between both groups were that group-
therein, floor material) has been, by far, the most investigated housed animals engaged in more behaviors (social,
variable. ‘Alternative’ cages (usually larger and provided or not
with a platform) have been compared to the conventional ones
in which rabbits are kept individually, either for fattening after 9
weaning or where lactating does nurse their kits (for review see
8 Enriched
Szendrö et al., 2016). Both pregnant and lactating does spend
about a quarter of their time on the provided platform, but 7 Control
this device creates hygienic problems inside the cage 6
Incidence (%)

(Alfonso-Carrillo et al., 2014). Young broilers, housed in


5
collective cages with a platform for several weeks, stand on it
at night and under it during the daytime, but the device does 4
not impact daily weight gain, mortality, morbidity, or body 3
lesions (Lang and Hoy, 2011). 2
In an attempt to mimic the ‘natural’ conditions where
1
rabbits live, i.e., underground colonies usually consisting of
males and females cohabiting according to a hierarchical order 0
Hops Sniffs Grooming Aggression Alert Cage bi ng
(Mykytowycyz, 1958a,b), various types of social arrangements
have also been explored. Rather than individual housing,
Figure 1 Effect of housing rabbits, from 35 to 75 days of age, in
groups of rabbits varying in age or reproductive condition are either ‘control’ or ‘enriched’ cages on the incidence of specific behav-
kept together in large pens. The development of young females iors. Modified from Verga, M., Zingarelli, I., Ferrante, V., et al., 2004.
reared in either same-sex or mixed-sex groups was not different Effect of housing and environmental enrichment on performance and
regarding weight gain, food intake, mortality, gonadotropin behaviour in fattening rabbits. In: Proceedings of the 8th World Rabbit
concentrations, or weight of ovaries, uterus, adrenals, kidneys, Congress, pp. 1283–1288.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 457

exploratory, play) that were not possible in the single-housed the nest boxes before kindling. Does kept in collective cages
rabbits. The former preferred small social groups (Whary showed a more varied behavioral repertoire relative to those
et al., 1993). The impact of individual cages versus collective housed individually. However, the interactions among the
pens on productivity was assessed in nulliparous NZW does former were not always ‘friendly’: a high incidence of aggres-
that were inseminated either artificially (AI) or by natural siveness, attack, and dominance was observed, especially in
mount. In the latter condition, the type of housing did not does ‘unfamiliar’ with the nest boxes. Reproductive perfor-
influence litter size, kit mortality, or kit weight at 14 days of mance and productivity were also lowest in such group as
age. However, when AI was used, a lower kindling rate and a low sexual receptivity was found in nearly 50% of animals
lower kit weight at weaning were found in animals housed in and fertility rates were 41%. Moreover, due to severe
collective pens. This was due largely to a high incidence of injury 8.3% of does in that group had to be replaced (Mugnai
pseudopregnancies (23% vs 0% in females housed individu- et al., 2009).
ally). Additionally, evidence of aggression (i.e., skin injuries) Other authors compared the productivity and behavior of
was found in 16–20% of does housed collectively (Rommers multiparous pregnant rabbits housed at a density of four
et al., 2006). does per cage, in standard versus in large cages. Reproductive
The reactions of lactating does kept in large communal pens performance (i.e., number of live/dead pups born, milk
with their litters differ from the ones observed in young or production, milk intake/kit, number of weaned kits, kit weight
adult animals housed together. In nature, in the laboratory, at weaning) was not affected by the type of cage (Dal Bosco
and on the farm lactating does nurse their kits inside a nest et al., 2004; Table 1). Rabbits housed in large cages showed
they construct with straw and their own body hair (for reviews more episodes of specific behaviors, e.g., moving, licking/
see González-Mariscal et al., 2007, 2016). Nursing usually scratching, lying down, sitting up, standing on hind legs.
occurs only once a day, with circadian periodicity, and lasts Females housed in standard cages showed more sniffing,
for 3–5 min (González-Mariscal et al., 2013a,b). In nature crouching, sitting hunched, standing alert, and biting bars
the site selected for building the maternal nest (and nursing (Dal Bosco et al., 2004; Table 2). In contrast, recently weaned
the kits) is determined by each individual doe, and it is located kits huddle spontaneously; that is, they prefer to be housed at
away from the communal burrow (Mykytowycz, 1958a,b). a high density (Matics et al., 2004). This condition resembles
Despite this natural pattern of ‘isolated nursing,’ attempts the one they experience in the maternal nest across lactation
have been made to house lactating does (and their litters) in and which is critical for conserving body heat and allowing
large communal pens. For instance, pregnant does were trans- a normal development (Bautista et al., 2003, 2008).
ferred from the communal pen they had shared with other
pregnant does to individual cages on prepartum day 1 and
kept there until lactation day 12. They were then placed back Table 1 Effect of housing rabbits in individual or collective
in communal pens together with the same (S) group of cages on specific reproductive parameters
(familiar) females or a group in which two or three of the
Type of cage
females were alien and mixed (M group) among the ‘familiar’
ones. Fecal corticosterone metabolites (FCM), lesions, and Parameter measured Individual Collective
agonistic behaviors were quantified before and after regroup-
ing. Lesions across days increased more in the M groups than Receptivity 81% 80%
Fertility 74% 70%
in the S groups. FCM raised after regrouping only in the M
Number of live-born kits 7.5  2.4 6.9  2.4
groups. These results indicate that familiarity with conspecifics
(mean  se)
provokes a more attenuated stress response after regrouping Milk intake per kit 19.3  3.5 20.5  3.5
than mixing with a few unfamiliar animals (Andrist et al., (g day1; mean  se)
2012). In another attempt to minimize agonistic interactions
among lactating does, they were transferred from the Modified from Dal Bosco, A., Mugnai, C., Castellini, C., et al., 2004.
A prototype of colony cage for improving the welfare of rabbit does:
communal pens they occupied throughout pregnancy to indi- preliminary results. In: Proceedings of the 8th World Rabbit Congress,
vidual cages across lactation days 1–12 and back to communal pp. 1229–1234.
pens. Does were individually sprayed (30 min before moving
them back to the communal pens) with water, 4.5% vinegar,
or fruit-scented alcohol. The presence of lesions, agonistic inter- Table 2 Effect of housing pregnant or lactating rabbits in individual
or collective cages on the display of specific behaviors
actions, glucose levels, and body temperature were measured.
On regrouping day 2, 43% of does had lesions; within days Type of cage
1–5 new lesions appeared in 60% of does; 32% were serious
lesions. Blood glucose concentrations and body temperature Percent time displaying Individual Collective
increased relative to baseline (i.e., before regrouping). Only General movement 22 26
a minor effect of spraying was observed: on regrouping day Eating–drinking 4.9 3.5
1, body temperature increased less in vinegar-scented does, Biting cage 8.2 1.4
compared to the other two groups. These results indicate that Sniffing 15.4 9.1
‘masking’ with alternative odors does not counteract agonistic Rearing 1.9 0.6
interactions among lactating does housed together (Andrist
Modified from Dal Bosco, A., Mugnai, C., Castellini, C., et al., 2004. A prototype of
et al., 2014). Another strategy used to reduce aggression was colony cage for improving the welfare of rabbit does: preliminary results. In:
to familiarize pregnant does, housed in collective pens, with Proceedings of the 8th World Rabbit Congress, pp. 1229–1234.
458 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

Enriching the animals’ housing environment has been using the following tests: open field, novel object, and social
attempted as a means to reduce ‘boredom’ and counteract runway. Semigroup housing consisted of rearing the kits in
anxiety-related behaviors, presumably provoked by barren cages. a litter with their siblings and mother in an individual cage
Three different types of gnawing blocks were added to the cages across lactation days 1–18, when they were transferred to collec-
of pregnant rabbits, housed in conventional wire cages, as an tive cages (four lactating does þ their own litters/pen) until
enrichment and dietary supplement. Litter weight or individual weaning. Kits reared under semigroup housing traveled less
kit weights were not significantly modified by the type of gnaw- distance in the open-field test and had a longer latency to leave
ing block provided, although a high variability in consumption the ‘start’ corner than control kits. In the novel-object test they
of the blocks occurred among females. Locomotion and intake reared less than control kits, but their latency to approach the
behavior in late pregnancy and at two time points in lactation object was not different. No differences were found in the social
were significantly increased when a gnawing block was available, runway test either (Buijs and Tuyttens, 2015). The impact of
but this did not modify the total number of behavioral transi- varying rabbit density in the cage and introducing a wooden
tions (Maertens et al., 2013). In multiparous lactating hybrid stick on the tonic immobility and emergence tests was studied
rabbits the effect of introducing a pinewood stick, straw in in young, hybrid, growing rabbits. Neither the stick nor the
a plastic bin, a compressed plywood block, or straw plus pine- animal density/cage had any impact on those tests or on weight
wood stick was compared. Across lactation more does were gain. Such factors, however, did increase locomotor activity and
engaged for more time in the group provided with straw þ ply- social interactions (Zucca et al., 2012a).
wood than with the pinewood stick (Rommers et al., 2014).
An important consideration has been given to the type of
cage floor, to avoid footpad injuries. Young rabbits (kept at three 1.17.3.2 Regarding Environment (Temperature, Noise,
different animal densities) spent more time over a wire net floor Vibration)
than over deep litter (Orova et al., 2008). Similarly, at low, Rabbits are raised worldwide in farms of different sizes and in
medium, or high ambient temperatures the lowest preference different latitudes/altitudes; they are transported from the
was for deep litter. Plastic floor was preferred at low temperature farms to the abattoirs; they are subjected to cage changes and
and wire floor at high. No differences among groups were seen to a variety of husbandry procedures. Thus, the impact of
on food intake, feed conversion ratio, or mortality (Gerencsér factors such as temperature, noise, vibration, and handling
et al., 2014). Broilers (8–12 weeks old) kept on bedding spent on productivity and some indicators of stress have been
less time resting and grooming and more time eating than did explored. For instance, Spanish Giant rabbits of both sexes
animals kept on wire floors (Ribikauskas et al., 2010). Providing (55–65 days old) were exposed for 4.5 h before slaughter to
plastic floor mats to does housed individually in wire cages heat (42  C), cold (5  C), noise (96 dB), or mixing with other
across five reproductive cycles significantly reduced the incidence rabbits. At slaughter, blood samples were taken to determine
of injured footpads (Rommers and de Jong, 2011). the concentration of cortisol, creatinine kinase, and lactate
In addition to determining the effects of housing conditions dehydrogenase; packed cell volume; and osmolarity. Cortisol
on productivity a few studies have explored the impact of such was significantly elevated (over values in the control group)
parameters on the behavior of rabbits in tests presumably in animals exposed to heat or noise while cold or mixing had
measuring anxiety or fear. Specifically, the effects of group no effect on this parameter (de la Fuente et al., 2007; Figure 2).
density (12 or 16 rabbits/m2) and floor type (wire or slat) Young bucks (12 weeks old) placed in plastic transport cages
were compared in the tonic immobility test (no differences
related to group density or floor type), the efficiency of feed
conversion (higher under high density), and the open-field 1.8 Control
test (rabbits under high density or wire net floor spent more 1.6 Heat
time exploring). The same work also found that growth param- Cold
eters in young rabbits were not affected by animal density 1.4
Noise
Cor sol, log 10 μg/dl

(Table 3; Trocino et al., 2004). The fearfulness of young female 1.2 Social mix
offspring raised under a semigroup housing system was assessed
1

0.8
Table 3 Effect of varying animal density in collective cages
on growth parameters in young rabbits 0.6
2
Animal density (rabbits/m ) 0.4

Parameter measureda 12 16 0.2


Initial weight (g) 616  39 620  39 0
Final weight (g) 2646  102 2664  102 Type of stressor
Weight increase (g day1) 48.3  2.0 48.7  2.0
Food intake (g day1) 149  6.4 148  6.4 Figure 2 Effect of five different types of potential stressors, applied
for 4.5 h prior to slaughter, to young adult male and female rabbits, on
a
mean  residual standard error. the concentration of cortisol in blood (mean  standard error). Modi-
Modified from Trocino, A., Xiccato, G., Queaque, P.I., et al., 2004.
fied from de la Fuente, J., Díaz, M.T., Ibáñez, M., et al., 2007. Physio-
Group housing of growing rabbits: effect of stocking density and cage
floor on performance, welfare, and meat quality. In: Proceedings of the logical response of rabbits to heat, cold, noise and mixing in the
8th World Rabbit Congress, pp. 1277–1282. context of transport. Anim. Welf. 16, 41–47.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 459

traveled by road for 2 h at 80 km h1 without air-conditioning In an attempt to optimize the use of artificial daylight
(ambient temperature ¼ 20  C; humidity 69%; Group A). without compromising fertility the effect of photoperiod dura-
Control groups were placed in transport cages (Group B) or tion on rabbit does’ productivity was determined. Starting at
in standard battery-style cages (Group C). Rectal temperature, 11 weeks of age does were housed under a long (16 h light:8 h
glucose, lactate, packed cell volume, and total proteins were dark; control group) or a short (8 h light:16 h dark) photope-
determined before (baseline), immediately, and at 6 and riod. The latter was ‘supplemented,’ 8 days prior to insemina-
24 h after transport. All parameters showed significant tion, by an extra hour of light applied in the middle of the
increases in Group A, relative to baseline and to both control dark phase (experimental group). The annual performance
groups. Baseline values of Group A were restored at 24 h (Fazio per doe, across five consecutive reproductive cycles, was supe-
et al., 2015). A temperature–humidity index (THI) of 30 or rior in the control group for number of kits born alive, and
more severely altered male and female rabbit physiology, number and weight of kits weaned (Gerencsér et al., 2011).
including reproduction parameters and the resistance to stress.
In females, conception rate, embryonic development, litter size,
1.17.3.3 Regarding Reduction in Reactivity to Stress by
litter weight, and milk production declined while age at
Neonatal Manipulations or Supplements in Adulthood
puberty onset and periweaning mortality increased as a conse-
quence of heat stress. In males, the factors affected are sperm Two main strategies have been used to minimize the conse-
count and motility, abnormal and dead sperm, testosterone quences of stress provoked by environmental factors or
concentration, fertility, sexual libido, and ejaculate volume. husbandry practices (see previous discussion): neonatal
In both sexes, feed conversion, feed intake, water metabolism, manipulations of kits and administration of specific nutritional
mineral balance, and energy balance were altered by a high THI supplements to young or adult rabbits of both sexes. Early
(Marai et al., 2002). Similarly, Rex rabbits placed under heat studies handled kits twice per day, at fixed times between 09:00
stress (or control temperature) for 9 weeks, followed by a return and 16:00 h, from days 1–10 or 11–20 postpartum. At
to control conditions, showed reductions in libido, sperm 98 days of age handled and nonhandled animals were tested
count, and semen volume. Heat stress also elevated apoptosis in the open-field test. No significant differences were found
in germ cells in seminiferous tubules. All parameters were between the groups, a finding the authors attributed to a large
restored to normal values upon return to control temperature interlitter variability (Kersten et al., 1989). Later studies,
(Pei et al., 2012). however, have shown that the kits’ brain is especially suscep-
To compare the effects provoked by high temperature versus tible to permanent changes in reactivity to stress when partic-
intense sound, young male hybrid rabbits aged ca. 42 days ular manipulations are given around the time of nursing. This is
were exposed to continuous heat (30  C) or intermittently to due to the fact that a state of arousal is generated spontaneously
noise (90 dB; from 8:30 to 12:30 h and again from 15:30 to in the kits shortly before the mother’s arrival for the single daily
19:30 h) for 1 week. Sound-exposed rabbits showed significant nursing bout (Hudson and Distel, 1982). Indeed, a light
increases in corticosterone across the whole trial while in manipulation of the kits, performed 15 min before or up to
animals exposed to heat such an increase was evident across 30 min after nursing (across the first postnatal week), reduced
the first 3 days, after which CORT values declined (Verde and the latency to approach humans and increased the frequency of
Piquer, 1986). contacts in tests performed in adulthood. Manipulating the kits
To determine the consequences of transport on pregnant at other times relative to nursing was ineffective at modifying
does, they were placed daily for 2 h, across gestation days their reactivity to stress later on (Bilkó and Altbäcker, 2000;
5–29, in or near a machine that generated vibration stress Pongrácz and Altbäcker, 1999; Figure 3). The effectiveness of
and sound, thus simulating transport. This procedure did not this strategy was further substantiated by the finding that just
affect maintenance of pregnancy, gestation length, litter size, introducing a human hand into the nest box for 5 min
perinatal mortality, or condition of the kits at birth (relative (without touching the kits), across postpartum days 1–7 within
to unexposed controls). Respiration rate increased from 65 30 min after nursing reduced, in 28-day-old kits, their latency to
to 120 min1 by moving animals from their cages and to approach humans and increased the number of approaches to
160/min1 by placing them in the machine. Noise alone the same extent as did the actual handling of the litter (Dúcs
provoked intermediate responses. Values returned to baseline et al., 2009). Moreover, such procedure also reduced the
after 20 min to 4 h (Stephens and Adams, 1982). latency to emerge from a dark box and the duration in the
The consequences of confinement within a small space were supine position (‘tonic immobility test’; Zucca et al., 2012b).
explored by placing rabbits for 15 min inside a restriction box A different type of manipulation that modifies the kits’ reac-
and measuring blood corticosterone concentration and temper- tivity to stress in adulthood is a 48 h mother–young separation
ature (skin and rectal). Corticosterone increased from 17.7 in early lactation. This so-called ‘biostimulation’ method is
(baseline) to 85.2 mg ml1 immediately after the restriction. used worldwide to restore estrus in lactating does (Alvariño
Skin temperature decreased from 35.3 to 34.8  C while rectal et al., 1998; Castellini et al., 1998; Theau-Clément and Mercier,
temperature rose from 39.2 to 39.7  C (Luzi et al., 2007). The 1999). It consists of increasing the interval between two succes-
effects provoked by exposing animals to a new environment sive nursing bouts from the usual 24 to 48 h. This increased
were explored in adult NZW rabbits of both sexes. The concen- time period without suckling provokes, in lactating does, an
trations of corticosterone, epinephrine, and norepinephrine, increased expression of estrous behaviors (scent-marking and
determined 10 min later, increased (relative to baseline) by lordosis; García-Dalmán and González-Mariscal, 2012), the
two- to threefold and two- to fourfold, respectively, while release of estradiol (Ubilla et al., 2000), and an increased
norepinephrine was hardly modified (Fenske et al., 1982). responsiveness of specific forebrain regions to mating stimuli
460 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

240 had a lower mortality rate and were significantly heavier than
220 the control group (Jezierski and Konecka, 1996). In a similar
200 paradigm NZW kits were handled six times per week, from
Latency to approach human (s)

birth until weaning (at 4 weeks of age), for 5 min in the after-
180
noon, i.e., not around nursing time. After weaning bucks
160
(housed in communal cages) were handled every other day
140 for 5 min. Across the next 12 weeks several tests were per-
120 formed to determine the effect of handling on the animals’
100 reactivity: human-approach latency, novel object, tonic immo-
80 bility, and open field. In general, results showed a marked effect
60 of handling in reducing emotional reactivity; that is, bucks were
more social, less sensitive to tonic immobility, made more and
40
faster contact with novel objects and humans, and moved more
20
in the open field (Verwer et al., 2009). Taken together, the
0
Control 30 min pre 15 min pre 30 min post 1 h post above results indicate that ‘windows of opportunity’ exist in
Manipula on me rela ve to nursing kits and in young adult rabbits at which the performance of
specific manipulations can permanently reduce the animal’s
Figure 3 Neonatal manipulation of kits at specific times before (pre-) reactivity to potentially stressful stimuli.
or after (post-) nursing, across postnatal days 1–7, reduces their In addition to such ‘prevention strategies’ some studies have
latency to approach humans, when tested at 28 days of age (mean - explored if providing particular nutritional supplements to
 standard error). Modified from Pongrácz, P., Altbäcker, V., 1999. The
adult animals can reduce the consequences of specific environ-
effect of early handling is dependent upon the state of the rabbit (Oryc-
tolagus cuniculus) pups around nursing. Dev. Psychobiol. 35, 241–251.
mental stressors. For instance, adult males housed under heat
conditions were supplemented with royal jelly (RJ) twice per
week for 20 weeks. RJ significantly increased: sperm count, total
(González-Mariscal et al., 2015). As a consequence of the doe’s sperm output, sperm motility, and the percentage of live and
‘biostimulation,’ the kits that missed a nursing bout (usually normal sperm, relative to control (i.e., nonsupplemented)
on lactation day 10) show a reduced release of corticosterone bucks. RJ also increased testosterone concentration, total
in response to a saline injection in adulthood (Brecchia et al., protein, albumin, globulin, glucose, and high-density lipopro-
2006; Figure 4). teins in plasma. RJ reduced total plasma lipids, triglycerides,
More prolonged manipulations (e.g., petting the kits daily cholesterol, and low-density lipoproteins (El-Hanoun et al.,
for 10 min from the age of 10 days up to 3 weeks) can provoke 2014). Similarly, the capacity of vitamin E or propolis to coun-
similar reductions in reactivity to stress in adulthood even if teract the impact of heat was explored in adult NZW bucks.
they are provided outside the perinursing period. In the ‘tonic Both supplements improved (relative to the control, nonsup-
immobility test’ the proportion of ‘bold’ (i.e., less time in plemented group) libido, sperm count, sperm viability, and
‘freezing’ posture) animals was 69% in the handled group testosterone concentration. They also increased number of
and 37% in control animals. Additionally, handled animals red blood cells, hematocrit value, and hemoglobin concentra-
tion. High-density lipoprotein and blood plasma glucose
were higher while cholesterol and triglycerides were lower in
40 both supplemented groups (Hashem et al., 2013). Another
supplement tried to ameliorate some of the adverse effects of
35 Control heat consequences is betaine. This agent, which has methyl
30 Separated donor and osmoprotective properties, decreased rectal temper-
Cor costerone (ug/dl)

ature and respiration rate; it also increased the humoral and


25 cell-mediated immune response and the serum concentrations
of T3, T4, total protein, globulin, and total lipids and enhanced
20
growth performance (Hassan et al., 2011). Polysaccharides
15 extracted from alfalfa and added to the regular diet of heat-
exposed NZW young males (40 days old) had a positive effect
10 on average daily weight gain, food intake, and food conversion
rate. Supplemented bucks also had lower cortisol concentra-
5
tions (Wei Liu et al., 2010).
0
0 30 60 90 120
Minutes postsaline, i.m.
1.17.4 On Sheep and Goats
Figure 4 Rabbit kits that skipped a single nursing episode in early 1.17.4.1 Regarding Behavioral Needs
lactation show a reduced release of corticosterone following an injec-
tion of saline (IM) in adulthood (mean  standard error). Modified from 1.17.4.1.1 Sexual Behavior
Brecchia, G., Bonanno, A., Dall’Aglio, C., et al., 2009. Neuroendocrine In wild and domesticated species of sheep and goats, reproduc-
responses in neonatal mother-deprived rabbits. Brain Res. 1304, tion is characterized by short breeding periods and long
105–112. periods of sexual segregation and inactivity, when females are
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 461

in anestrus. Both species are short-light breeders which means Even after 3 years of life in a flock made of members of only
that they usually reunite and breed when the light phase of the their own species, cross-fostered males maintain their social
photoperiod decreases (Chemineau et al., 1992). As the season and mating preferences for females of their mother’s species.
advances, males will begin to fight with one another. The fight Females shift their preference to the genetic species within
consists of head clashing and nudging behavior similar to that 1–2 years even though the social interest for female members
seen in courtship. Periodic reunion has two major conse- of their foster species does not vanish. Although this phenom-
quences: initiating breeding activity, by triggering the transition enon resembles ‘sexual imprinting,’ described in birds, it is
from anestrus to estrus, and synchronizing the sexual cycle in more likely to be a social learning determined by the parental
the females (Ungerfeld, 2007; Walkden-Brown and Martin, species. While the effect in males is very striking and looks irre-
1999). As a secondary consequence, this will also enable versible, the development of a strong social and mating prefer-
synchronous parturition, a defense mechanism that under ence for individuals of the foster species is never exclusive.
natural conditions buffers the impact of predation on Social learning is also important for males to develop hetero-
offspring. The induction of sexual activity in the females is sexual experience in order to prefer females over males as
called the ‘male effect.’ In responsive ewes and does the primary sexual partners. While isolated rams show some aberration in
reaction to the male is an immediate increase of LH pulse behavior when first exposed to estrous females, most are even-
frequency, which leads to ovulation within 24 h (Fabre-Nys tually able to mate. When reared in monosexual groups, some
and Gelez, 2007). Similarly, in both rams and bucks, exposure rams and bucks are sexually inhibited or show homosexual
to estrous females may stimulate an immediate secretion of LH, preferences and do so for long periods or permanently after
a phenomenon that has been termed the ‘female effect.’ maturity (Zenchak et al., 1981; Figure 5(a)). Homosexual
Although its functional significance remains unclear, it may behavior may be overcome by short exposure to ewes before
lead to behavioral changes that improve the success of mating puberty (Tilbrook and Cameron, 1990). Nonetheless, rams
(Walkden-Brown and Martin, 1999). Female sexual behavior is who do eventually mate with estrous ewes continue to exhibit
characterized by three components: (1) attractiveness: physical subnormal sexual activity. Bucks reared in groups of males
changes to attract the attention of the male; (2) proceptivity: develop homosexual behavior as well and display fewer
active search for and attraction toward the male; and (3) recep- agonistic interactions with other bucks at puberty (Price and
tivity: acceptance of mating attempts (Fabre-Nys and Gelez, Smith, 1984; Figure 5(b)). The dominant male initiates homo-
2007). Although attractiveness is not very explicit, rams find sexual behavior over the subordinate ones (Price et al., 1994).
ewes more attractive around the time when they are receptive Homosexual behavior does not interfere with the ability to
and when they are not shorn. During estrus, does appear rest- ejaculate in an artificial vagina (Orgeur et al., 1990) showing
less, show increased motor and vocal activity, and display that breeding failure relates more to the formation of
male-type behavior and reciprocal mounting. Ewes are less individual-specific sexual attachment with other males rather
demonstrative, but they do search actively the proximity of than with failure to develop normal mating skills. Conse-
males. The expression of such behavior is important when quently, early social experience must be viewed as critical for
males are not highly motivated, or when a large number of the behavioral development of the individual, and rearing juve-
ewes are in estrus in which case they compete for access to nile males in same-sex groups must be avoided.
the rams. Around the time of ovulation, ewes and does present Under laboratory conditions, the duration and time of the
a well-defined response to the courtship of the males mating season is controlled by humans and will not neces-
(Fabre-Nys and Gelez, 2007). There is no ritualized sequence sarily follow the natural breeding season of the animals. The
of events leading to mating, but the repertoire of behaviors impossibility to fulfill the sexual drive may be seen in some
expressed by both rams and bucks is very standardized and species as a potential source of frustration and consequently
similar. The male approaches the proceptive female which as a decrease in welfare (Webster, 1995). While in most
remains motionless, kicking her occasionally with its front species, the sexual drive of males is considered stronger than
leg. Alternatively, the two partners walk together in circles, that of females, in sheep and goats, it could be argued that
the female following the male and the male trying to place the females’ sexual drive is fairly strong as well because they
himself behind the female. There is considerable evidence search actively the proximity of males. Therefore, the frustra-
that olfactory cues from the female and the male are important tion of sexual restriction during the breeding season is a real
for sexual communication; however, both in sheep and in goats welfare issue. Because males and females are aroused in
the sexual partner is a multisensory stimulus, olfactory cues response to the sight and the smell of females in estrus, the
being only part of it (Walkden-Brown and Martin, 1999). segregation of sexes during the breeding season should not
be a source of excessive frustration. On the other hand, the
1.17.4.1.2 Welfare Issues Specific to Sexual Behavior confinement of males in a single sex group during the
1.17.4.1.2.1 Rearing Conditions in Early Age breeding season is likely to increase the amount of aggressive
Contact with the mother in early life can dramatically influence behavior and, therefore, injuries. Care should be taken to
sexual preferences. Sheep and goats cross-fostered at birth, but minimize fighting by rearing males (especially bucks) in indi-
raised in mixed-species groups, display play and grooming vidual pens while also allowing social contact with conspe-
behavior resembling that of their foster rather than their genetic cifics, an important issue due to the gregarious nature of
species (Kendrick et al., 2001). In adulthood, cross-fostered these ungulates. During the nonbreeding season the separa-
juveniles prefer to socialize and mate with partners of their tion of sexes should not be a problem because sheep and
foster mother’s species, even if raised with a conspecific of their goats, like most ungulates, naturally segregate outside of the
own species. This effect is weaker in females than in males. mating period.
462 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

(a) Rams
First approach Inves gate Preference
15 15 15
Number of responses
10 10 10

5 5 5

0 0
0
Normal Low response Normal Low response Normal Low response
Ewe Ram Ewe only Ram and ewe Ram only Ewe Ram

(b) Bucks
Subjects that mounted a male Subjects that did not mount a
Mean sexual responses

Mean sexual responses


12
male
10 6
8
6 4
4
2
2
0 0
Nudges Mounts Nudges Mounts
Estrous goat Diestrous goat Buck Estrous goat Diestrous goat Buck

Figure 5 Sexual behavior displayed by rams and bucks reared in unisexual groups. (a) First approach, olfactory investigation and preference
expressed by rams when given the choice between a ram and a ewe (Normal: rams displaying normal levels of sexual performance toward ewes; low
response: rams showing little interest in the ewes). (b) Responses of sexually active bucks when given the choice between an estrous goat, a dies-
trous goat, and a buck. Subjects are divided into two groups: whether they had been seen mounting a male or not in their home enclosure. Adapted
from Price, E.O., Smith, V.M., 1984. The relationship of male-male mounting to mate choice and sexual performance in male dairy goats. Appl. Anim.
Behav. Sci. 13, 71–82; Zenchak, J.J., Anderson, G.C., Schein, M.W., 1981. Sexual partner preference of adult rams (Ovis aries) as affected by social
experiences during rearing. Appl. Anim. Ethol. 7, 157–167.

1.17.4.1.3 Maternity conditions, females show restlessness, frequent vocalization,


1.17.4.1.3.1 Maternal Responsiveness and increased intolerance of conspecifics, and ewes isolate
Despite their strong social needs, as parturition approaches themselves if cubicles are provided in the group (Gonyou
ewes and goats will easily withdraw from their flock (Nowak and Stookey, 1985). As parturition approaches, females
et al., 2000; Poindron et al., 1993, 2007b). Isolation-seeking vocalize, paw the ground, circle, and try to lick their back.
behavior persists in most breeds of sheep even if domestication Parturition is most likely to occur during the day in goats, while
is associated with reduced time spent on the birth site (Dwyer ewes do not show any diurnal pattern. The amniotic fluids
and Lawrence, 2005; Figure 6). Even under farm or laboratory dripping from the vagina become suddenly attractive and
trigger intense licking of the contaminated bedding. Attraction
8 to these fluids incites females to stay on the birth site, and once
7 the newborn is expelled, it becomes the core of the mother’s
Days a er parturi on

6
attention. Removal of such fluids greatly disrupts the onset of
maternal care especially in primiparous animals. In sheep
5
and goats, like in many other mammals, it is the rapid variation
4 in progesterone and estrogens that takes place in the days or
3 hours preceding parturition that are responsible for the peripar-
2 turient behavioral changes. In addition, the mechanical stimu-
1 lation caused by the expulsion of the fetus, which leads to the
0 release of oxytocin, reinforces the expression of maternal
responsiveness (Dwyer, 2014; Poindron et al., 1993). Does
defend the area where they gave birth and their kid against
other females. This is rarely the case in sheep, and the strong
Figure 6 Maximum time spent on the birth site by various species maternal responsiveness of nearby periparturient ewes may
and genotypes of sheep. Adapted from Dwyer, C.M., Lawrence, A.B., result in lamb stealing, especially under indoor conditions.
2005. A review of the behavioural and physiological adaptations of hill Licking of the neonate usually starts from the head down to
and lowland breeds of sheep that favour lamb survival. Appl. Anim. the anal region and has important consequences. Firstly, it
Behav. Sci. 92, 235–260. removes much of the fluid present on the coat and helps reduce
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 463

heat loss, as young lambs and kids are both susceptible to chill- (Searby and Jouventin, 2003) and cues from the lambs’ face
ing (Dwyer et al., 2016). Secondly, it has a dual impact on the seem important for visual recognition (Alexander and Shillito,
newborn’s activity: licking of the head is soothing and triggers 1977). As lambs and kids grow older, recognition improves
head movement oral activity (teat-seeking) while grooming the and it seems very likely that mothers learn a composite image
back and anogenital area is associated with leg movement and of their young, including appearance, behavior, and voice.
may stimulate standing (Poindron et al., 1993). Finally, licking Once initiated, maternal behavior lasts for at least 3 months
of the neonate contributes to the establishment of the mother– after which lambs and kids are usually weaned by humans.
offspring bond, since this intimate relationship will make ewes However, if ewes and goats are left undisturbed, natural wean-
and does able to learn their offspring’s identity by smell. ing occurs when their young are between 4 and 6 months of age
Usually, the fetal membranes are licked off and consumed (Poindron et al., 1993; Miranda-de la Lama and Mattiello,
but the placenta is exceptionally eaten. Mothers emit simulta- 2010). Maternal memory and motivation seem to be rather
neously a very-low-pitched bleat or rumble, a call that is labile if mothers do not maintain close proximity with their
specific to the periparturient period or when they are in close young. Seven days after parturition, if lambs are temporarily
contact with their young. If mother and young are left undis- removed, maternal selectivity remains following 36 h of sepa-
turbed, maternal behavior develops normally and lasts for ration, but after 3 days, most ewes lose their maternal drive
several months. However, the maintenance of maternal and reject their young (Keller et al., 2005). Yet, vocal recogni-
behavior depends very strongly on the events occurring imme- tion of kids by their mothers persists for up to 13 months after
diately after parturition. Should mother and young be sepa- weaning, at a time when goats are already nursing a second
rated at birth, maternal behavior vanishes within hours. The generation of young (Briefer et al., 2012). The fact that mothers
ability of ewes and goats to remain responsive to a neonate is remembered the vocalizations of their weaned kids recorded
therefore limited to a sensitive period starting around parturi- 1.1 years earlier is quite remarkable and demonstrates the exis-
tion and ending a few hours later and is under the direct control tence of long-term memory.
of the neuroendocrinological changes that take place at partu-
rition (Dwyer, 2014; Poindron et al., 1993, 2007b). 1.17.4.1.4 Welfare Issues Specific to Maternal Behavior
1.17.4.1.4.1 Management of Pregnant and Parturient Ewes
1.17.4.1.3.2 Maternal Selectivity and Goats
Maternal responsiveness is soon followed by selective maternal The neonatal period is always of special concern for lambs and
care. It is characterized by the acceptance of suckling attempts goats as it is a period of high vulnerability (Dwyer, 2008a,
by their own young along with the rejection of alien ones. 2014; Dwyer et al., 2016; Nowak et al., 2000). Several aspects
Many ewes and does show selective nursing after only have to be taken into account to ensure maximum chance of
30 min of contact with their young but most are selective after survival and optimum development of the young. First of all,
2–4 h postpartum (Nowak et al., 2011; Poindron et al., proper nutrition during gestation is a key element since under-
2007b). There is some evidence that selectivity in goats may nutrition leads to reduced udder weight, delayed onset of lacta-
result from olfactory labeling of the kid though maternal tion, reduced colostrum yield, and impaired maternal behavior
licking and nursing (Gubernick, 1981), while in sheep, recogni- (Dwyer, 2014; Dwyer et al., 2016; Terrazas et al., 2009).
tion of the lamb relies on its distinctive individual odor. None- Improper nourishment may also lead to metabolic imbalance
theless, selective nursing in goats reflects learning of some (e.g., pregnancy toxemia) and death. Nutrient requirements
individual olfactory characteristics: even twins have different for sheep and goats are provided in detail in the document
odors (Poindron et al., 2007b). Selective nursing depends Nutrient Requirements of Small Ruminants: Sheep, Goats, Cervids
primarily on the main olfactory system since when ewes and and New World Camelids (NRC, 2007). Secondly, both ewes
goats are made anosmic, affecting only the main olfactory and goats tend to separate themselves from their flocks,
system, they subsequently nurse their own as well as alien a process that facilitates the development of the mother–young
young. If separated from their young, mothers express strong bond. Even under indoor conditions sheep display such
distress reactions with intense vocal (high-pitched bleats) and a behavior if cubicles are provided (Nowak et al., 2008). Any
motor activity (walking, circling). They stop vocalizing and disturbance during that period, especially with highly reactive
moving when reunited with their own lamb while they keep females, may jeopardize the onset of maternal behavior, and
being agitated in the presence of an alien young. Mothers primiparous mothers are more prone to react. Prolonged labor,
also identify their neonate from a distance by using auditory associated with pain and stress, increases the risk of hypoxia
and visual cues (Nowak et al., 2011; Poindron et al., 2007b). and brain trauma; it impairs maternal and infantile behavior
Nonolfactory recognition develops slightly later than olfactory and it also diminishes the chance of survival of the newborn.
recognition, but it is fully effective by 6 h postpartum. The A swift parturition is, therefore, important and should it be pro-
neural basis involved in these two types of recognition longed, it is important to intervene and pull the fetus out.
processes seems to be independent since anosmic ewes can Under crowded situations, care should also be taken to facili-
discriminate between their own and an alien young from tate the development of a bond and to avoid mother–young
a distance even though they are willing to nurse any lamb separation due to excessive human intervention or to interfer-
(Ferreira et al., 2000). Olfaction alone as a means of recogni- ence by other females. In some cases, when several ewes give
tion declines in importance with time, and after several days birth simultaneously, lamb stealing by preparturient ewes
it is probably used only for close contact discrimination. The occurs (Dwyer, 2014; Nowak et al., 2000). Unfortunately, these
respective roles of hearing and vision have not been clarified lambs may be rejected once the ‘alien’ ewe has given birth
yet, although ewes do recognize the voice of their young to her own lamb and they are not taken back by their own
464 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

mother: a fatal rejection. It is often advised in highly stocked (Nowak et al., 2011; Poindron et al., 2007a). While at
areas to isolate postparturient females with their neonates in 12–24 h identification of the mother is mostly based on
individual pens to favor early mother–young interactions, cues that lambs can perceive at close quarters, lambs can
especially in case of multiple births. However, restricting the clearly locate their mother from a distance of several meters
movements of postparturient females increases the risk that when they are 3 days old. Olfactory cues are not necessary
they lie on their offspring and suffocate them to death. There- for newborn lambs to discriminate between two ewes at close
fore, the pen should be wide enough to allow the animals to quarters even though they do recognize maternal odors at
move freely (1.8–2.0 m2). It is also recommended to isolate birth (Vince, 1993). Instead, both auditory and visual cues
females only if parturition is well advanced or terminated are used (Nowak et al., 2011). This reflects the fact that initial
and just for the time needed to establish the mother–young orientation toward the mother is based on general maternal
bond (24–48 h). Then, mothers and newborn can be released signals since they are the differences between acceptance
and subsequent separations are no longer risky. behaviors (low-pitched bleats signal acceptance) and rejec-
tion behaviors (high-pitched bleats indicate aggression) that
1.17.4.1.4.2 Fostering make lambs and kids choose a specific adult (Poindron
There may be a number of occasions when neonates cannot be et al., 2007a; Terrazas et al., 2002). Step by step neonates learn
reared by their own mother (no expression of maternal various individual features from their mother which results in
behavior, rejection, insufficient milk yield, death of mother). an improved ability to recognize her from a distance. The role
While under unsupervised conditions such young are likely of hearing seems to be as important in lambs as in kids once
to die from starvation, close supervision at lambing may often they are several days old (Briefer and McElligott, 2011; Searby
save them. A way to save them is to force another maternal and Jouventin, 2003), but there is no doubt that infants form
female to accept these orphans by triggering the development a multisensory image of their mother. Bit by bit this specific
of another bond. Fostering has been more or less successful responsiveness to individual maternal cues leads to a specific
in sheep using three methods: (1) bonding an alien lamb to attachment process that consolidates into an enduring
a ewe during the period of maternal responsiveness, i.e., before emotional bond, characterized by a tendency to seek and
maternal selectivity takes place; (2) transferring the mother’s maintain closeness, especially during times of stress (Nowak
own lamb characteristics to the one to be fostered; and et al., 2011; Gaudin et al., 2015). This bond with the mother
(3) allowing a second bond to form progressively with the alien is based on the infant’s need for safety, security, and
lamb by cohabitation (Nowak et al., 2008). In all cases the protection.
procedure consists of penning a periparturient ewe (or goat)
with an additional young, helping it have access to the udder, 1.17.4.1.5.2 Relation with Humans
and preventing excessive expression of selective maternal Farm animals develop an important social network with inter-
behavior until it is fully accepted. This may take a few days. specific partners, such as humans. Unlike other forms of social
Should this method not be successful, lambs can be artificially attachment, the relationship sheep and goats can develop with
fed (see the later discussion) humans is not well understood. It seems that both species
develop intra- and interspecific relationships but not in
1.17.4.1.5 Infancy a concomitant manner since the presence of the mother
1.17.4.1.5.1 Attachment with the Mother reduces strongly the lamb’s motivation to interact with
Lambs and kids are born in a well-advanced stage of physical a human. However, when newborns are separated from their
and behavioral development which allows them to interact mother and reared under artificial feeding conditions, the
with their mother on a multisensory basis. The activity of human becomes a salient figure and this may facilitate the
the newborn, which has to be able to stand steadily before ori- management of lambs and kids under laboratory conditions
enting toward the body of the mother, needs to be coordi- as well as on the farm (Nowak and Boivin, 2015; Miranda-de
nated with that of the mother. Most neonates stand within la Lama and Mattiello, 2010). Usually, human–animal rela-
30 min after birth. As their coordination improves, they tionships are facilitated by positive social contacts (stroking,
nose the side of their mother’s body until a teat is found, bottle-feeding) provided by a specific caregiver in the first few
usually within the first hour. They are guided by various weeks after birth. In both species, stroking alone reinforces
cues emanating from the mother’s body, and in sheep many the motivation to interact with the stockperson (Boivin and
ewes may also help the young by lowering their back and Braastad, 1996; Tallet et al., 2005). Stroking is appeasing, it
bending a hind leg to facilitate access to the teat (Vince, induces relaxing postures in lambs, and it decreases heart rate
1993). Both kids and lambs learn the position and shape of due to an activation of the parasympathetic system. Behavioral
the udder very quickly, a learning process that is probably sus- tests comparing responses directed toward a familiar caregiver
tained by the dynamics of mother–young interactions and the and unfamiliar or familiar humans show that lambs do search
reinforcing properties of suckling. This spontaneous attrac- specifically the proximity of their human caregiver. Differential
tiveness to the mother’s udder, reinforced by the fact that emotional responses in the presence (calmness) or the absence
suckling is displayed at hourly intervals, brings the newborn of the partner (agitation) are also expressed (Figure 7). The
in contact with maternal cues and behaviors required for the clear and durable signs of appeasement shown by lambs in
establishment of individual recognition. Most lambs and the presence of their caregiver suggest that the human can be
kids develop a clear preference for their dam by 12–24 h of perceived as an affective substitute in artificially reared animals
age, and their ability to discriminate their mother from an (Nowak and Boivin, 2015). The same phenomenon may apply
alien ewe improves markedly over the first few days of life to young goats (Miranda-de la Lama and Mattiello, 2010).
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 465

Number or zones crossed Number of zones crossed


30 40
*
25 35
30
20 25
15 20
10 15
10
5 5
0 0
Isola on Reunion Separa on Isola n Reunion Separa

Number of vocaliza ons Number of vocaliza ons


40 40
35 * * 35
30 30 *
25 25
20 20
15 15
10 10
5 5
0 0
Isola on Reunion Separa on Isola n Reunion Separa

Familiar caregiver Unfamiliar person Familiar person

Figure 7 Affinity of lambs to their caregiver. Lambs were tested alone (isolation), then with a person (reunion), and alone again once the person
left the pen (separation). The subjects’ reactions were observed in two comparative situations: familiar caregiver versus unfamiliar person (left histo-
gram) or familiar caregiver versus familiar person (right histogram). The number of zones crossed and the number of vocalizations emitted indicate
that lambs are soothed during reunion and agitated after separation. Appeasement during reunion and distress after separation is more important
when the test is performed with the familiar caregiver.

1.17.4.1.6 ‘Followers’ versus ‘Hiders’ combination of humidity, wind, and inadequate colostrum
One of the main differences between sheep and goats is that intake that weakens them (Obst and Day, 1968; Figure 8(a)).
lambs follow their mother within hours after birth and, there- Newborn lambs and kids seek shelter more readily than their
fore, are classified as ‘followers,’ while goat kids remain tempo- mothers, which are insulated by their full fleece or hair.
rarily hidden from their mother while she forages. Timing and Providing sheltered areas may, therefore, protect the young
duration of the hiding phase varies from a few days to several from the deleterious effects of wind and rain and contribute
weeks, but it is a general trend that remains even when animals to their thermal comfort. Under indoor conditions, weak hypo-
are reared indoors (Miranda-de la Lama and Mattiello, 2010; thermic lambs should be put temporarily under a heating lamp
Poindron et al., 2007b). The mother leaves her kid for periods or a warm air blower that will help them maintain an adequate
of up to 8 h before returning to the precise location where the body temperature, after which they can be put back with their
kid is lying. During that stage, mother–young separation does mother. While cold stress is a common source of thermal
not trigger any distress response. In contrast to goats, lambs discomfort, it should be borne in mind that young ruminants
stay close to their mother for the first few days after birth and are also vulnerable to heat stress. Lambs respond to high
display distress behavior should they be separated. Both types temperature by increased respiratory rate, irregular breathing,
of mother–young relationships are believed to be antipredator and periods of apnea (Riesenfeld et al., 1996). Indeed, severe
strategies. In one case hiding allows neonates to remain out of heat waves can lead to death after several days. Therefore,
sight; in the other, the young imitate the flight reaction of their should lambs and kids be kept in buildings, effective ventila-
mother (or other adults) as the flock runs away from a potential tion is essential to allow free air circulation and decrease
source of danger. Over time, kids and lambs tend to associate ambient temperature.
and form bonds with age mates and to engage in play behavior.
1.17.4.1.7.2 Colostrum Needs
1.17.4.1.7 Welfare Issues Specific to the Young Ruminants are born markedly hypogammaglobulinemic, the
1.17.4.1.7.1 Thermal Discomfort mother providing passive immunological protection through
At birth, lambs and kids lose heat because their coat is covered the immunoglobulins contained in the colostrum. Passive
with amniotic fluid. A direct consequence may be hypothermia immunity must be acquired during a very narrow time window
as the ambient temperature can fall from 39  C in the uterus to since in newborn lambs, gut closure to immunoglobulin
10  C or less outdoors, if parturition takes place in winter absorption occurs between 24 and 36 h of birth (Dwyer
(Dwyer, 2008a; Dwyer et al., 2016). Once dried and properly et al., 2016; Nowak and Poindron, 2006). Consequently,
fed, the neonates are resistant to cold weather and can maintain lambs and kids must find the teat and suckle as soon after birth
homeothermia even below freezing temperature. It is the as possible in order to receive an adequate amount of
466 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

(a) (b)
90 18–26 °C
80 0–10 °C

% mortality 0–48 h a er birth


70
80 60
60 50

Hours
40
40 30
10–16 −1 ) 20
20 s
1–8
d (km 10
0 0
ee 0
> 1.5 0.5–0 0 sp
ind 0 150 210 290
W
Rainfall (mm day−1) Colostrum intake (ml)

Figure 8 (a) Lamb mortality during the first 48 h of life according to rainfall and wind speed. (b) Average periods for which carbohydrates (lactose
from colostrum, glycogen from the liver and the skeletal muscle) available during the first day after birth can sustain heat production in lambs at
different ambient temperatures and colostrum intakes. Adapted from Obst, J.M., Day, H.R., 1968. The effect of inclement weather on mortality of
Merino and Corriedale lambs on Kangaroo Island. Proc. Aust. Soc. Anim. Prod. 8, 239–242 and Mellor, D.J., Cockburn, F., 1986. A comparison of
energy metabolism in the new-born infant, piglet and lamb. Q. J. Exp. Physiol. 71, 361–379.

colostrum. Lambs born outdoors need 280 ml kg1 body experience a marked emotional stress represented by the loss
weight during the first day, while 210 ml kg1 should be of their attachment figure, the mother, and a nutritional stress
adequate for lambs born indoors (Mellor, 1990). Insufficient provoked by the abrupt transition from maternal milk to
colostrum intake may lead to exhaustion of energy reserves a commercial milk substitute. Artificial rearing of lambs and
and hypothermia, which in severe cases is fatal (Mellor and kids requires close attention and a high standard of supervi-
Cockburn, 1986; Figure 8(b)). Care should be taken that sion. First of all, it is essential that all animals receive an
mothers have sufficient colostrum yield and that neonates adequate supply of colostrum. Their body temperature should
display proper suckling activity. If this is not the case, lambs be monitored to avoid hypothermia and, if needed, a heating
and kids should be supplemented with an external source of lamp should be provided. Where automatic feeding equipment
colostrum. Usually, cow, sheep, and goat colostrum are equally is used, lambs and kids must be trained to suck from the artifi-
efficient and have interspecific positive actions; however, cial rubber teats several times per day until fully independent.
commercial colostrum is not as satisfying to ensure proper Thereafter, an adequate amount of milk substitute must be
immunological protection (Argüelloa et al., 2004). provided, and from the second week onward, palatable and
nutritious solid food should be given. Despite close attention
1.17.4.1.7.3 Behavioral Needs and human care, the welfare of artificially fed lambs is now
Sheep and goats are social animals. Immediately after birth, the questioned. Compared to young reared by their mother, those
core of their attention is the mother. Care should be taken that lambs display abnormal behaviors (cross-sucking activity) and
the mother–young bond is not disrupted, especially with twins have higher basal cortisol levels. When tested in an open field,
and triplets which are more likely to lose contact with their artificially fed lambs emit less distress vocalization and their
mother. Concerning newborn kids, because of their natural cortisol response increases more sharply than that in lambs
tendency to hide even under housing management (Miranda- reared with their mother. They also have decreased immuno-
de la Lama and Mattiello, 2010; Poindron et al., 2007b), logical responses and a high incidence of diarrhea (Napolitano
providing shelter will satisfy their behavioral need. If animals et al., 2008). Attempts to reduce the detrimental effects of early
have to be penned at parturition, it is important not to isolate separation focus on the emotional and nutritional aspects of
the mother–infant dyad beyond the first few days after parturi- artificial rearing. It is not recommended to leave a lamb alone,
tion. With time, lambs and kids interact socially with peers and except for specific experimental reasons and only for short
adults, a process that plays an important role in shaping the periods of time. However, even in a group, peers do not
behavioral development of juveniles: the mother facilitates compensate for the loss of the attachment figure. In this case,
the transmission of acquired social behaviors, environmental a human can provide a ‘maternal substitute’ (Boivin and
preferences, and consumption of solid food while sexual play Braastad, 1996; Boivin et al., 1997). The necessary condition
with peers has a strong impact on later sexual preferences is for the human to perform gentle stroking and feeding, two
(Dwyer, 2008a; Nowak et al., 2008). forms of physical attention that trigger the affinity for the care-
giver. Regarding nutrition it is known that mixing ewe milk and
1.17.4.1.7.4 Mother–Young Separation a milk substitute during the first week and then moving gradu-
Separation of mother and young occurs fairly frequently in ally to only milk substitute reduces some problems. Ewe–lamb
small ruminant husbandry either temporarily during manage- separation at weaning on the farm occurs generally at 3 months
ment procedures or permanently at birth, to rear lambs and of age, which is considered early weaning compared to natural
kids artificially, and at weaning when it is time to raise adults conditions (Poindron et al., 1993). It results in a temporary
and juveniles in separate flocks. Artificially reared lambs increase in cortisol levels, distressed behavior, altered social
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 467

interaction, and disrupted circadian rhythmicity (Dwyer, in order to limit heat loss, (2) designed to allow neonates to
2008a). Care should be taken not to wean the lambs too early; take full advantage of the sun, and (3) sufficiently familiar to
otherwise, growth rate is retarded and lambs have an increased encourage ewes to isolate from other sheep at lambing
susceptibility to disease. Nonetheless, maternal deprivation while minimizing the risk of mother–young separation
appears to be worse when performed at 2 days (artificial rear- (Pollard, 2006).
ing) than at 3 months of age (early weaning).
1.17.4.2.2 Floor
Sheep and goats are reared under various floor surfaces including
1.17.4.2 Regarding Lodging (Cages, Floors, Nest Boxes, compacted soil, straw bedding, concrete, concrete- or wood-
‘Enrichment’) slatted floors, not naming all the types of surfaces they can be
1.17.4.2.1 Outdoor Facilities in contact with when reared outdoors. All of them are considered
Among large domestic animals, sheep and goats present the to be suitable as long as they are kept dry and clean and they are
widest diversity of breeds, and they live in the largest variety nonskid surfaces. However, some studies show that while
of habitats. They are found from tropical to temperate regions, unsheared ewes show no clear flooring preferences at low
and they can face very hot or very cold climatic constraints. ambient temperatures (1.4–8.6  C), sheared ewes choose softer
They are reared in sheds, in fenced meadows, in open moun- floors with low thermal conductivity (Figure 9). Wooden floors
tain pastures, in arid steps driven by nomadic people, and are preferred over expanded metal ones, straw to wooden floor,
even in remote rangelands with hardly any human supervi- and straw to expanded metal floor (Færevik et al., 2005). Goats
sion (for sheep see Dwyer, 2008b; Kilgour et al., 2008). kept under moderate temperatures (10–12  C) prefer expanded
From such diversity recommendations for ‘standard’ manage- metal to solid wood, while solid wood and mattress are equiva-
ment practices or housing cannot be derived. The general lent, and straw the least preferred flooring (Figure 10). During
guideline is to take into consideration local conditions and cold periods (8–12  C), mattress and solid wood are the most
to ensure that animals are properly cared for. Sheep and goats preferred types of flooring, expanded metal intermediate, and
well adapted to local conditions usually take advantage of surprisingly, straw is not perceived as attractive by goats (Bøe
natural shelter, and the use of habitat is strongly influenced et al., 2007). Thus both species do express a need for flooring
by season or weather conditions (Dwyer, 2008b). Nonethe- with lower thermal conductivity under low temperature
less, under outdoor conditions local climate and extreme constraints. While the welfare of sheared ewes is improved if
temperatures should be incentive for providing shelter. Too straw is provided during the first weeks after shearing, softness
often animals are fenced off in barren paddocks with nothing does not have any effect on the choices of goats. The fact that
to protect them from adverse weather conditions. Various in the wild goats are adapted to rest on rocky hills may explain
types of shelter may be used, from artificial settings to more their lack of interest in soft surfaces.
natural windbreakers like shrubs, gramineous plants (e.g.,
Phalaris) which are not ingested by sheep, or tall deciduous 1.17.4.2.3 Social Enrichment
trees. Such shelter reduces wind velocity, protects adult Sheep and goats are highly gregarious species; they live in
animals from rain and snowstorms, and provides valuable flocks of various sizes, and therefore, they should never be
shade during hot sunny days (Gregory, 1995). With shade, reared in social isolation. There is surprisingly no evidence
sheep can support ambient temperatures of up to 50  C of what could be an optimal group size that satisfies the social
(Dwyer, 2008b). Because sheep give birth normally in winter, needs of these mammals. Under natural conditions, the
wind shelter can reduce mortality rates by up to 3% in single preferred group size in sheep is influenced by the type of breed
lambs and 37% in twins. It is recommended that, to be effec- (Dwyer, 2008b). In stable flocks, social positive relationships
tive against lamb mortality, shelter should be (1) very dense, improve group affinity, and when reared in paddocks, sheep

Figure 9 Preference for lying areas expressed by ewes before and after shearing. Adapted from Færevik, G., Andersen, I.L., Bøe, K.E., 2005. Prefer-
ences of sheep for different types of pen flooring. Appl. Anim. Behav. Sci. 90, 265–276.
468 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

Figure 10 Preference for lying areas expressed by dairy goats under moderate (12–12  C) and cold temperature periods (8–12  C). Adapted from
Bøe, K.E., Andersen, I.L., Buisson, L., 2007. Flooring preferences in dairy goats at moderate and low ambient temperature. Appl. Anim. Behav. Sci.
108, 45–57.

do form subgroups. While sheep display little affiliative newborn needs to be taken into account. Thus, small hiding
behavior with each other, goats groom, sniff, and lick each boxes have been provided (50  15  25 cm) and newborn
other (Miranda-de la Lama and Mattiello, 2010). It has been kids use them for 4–7 days (Lickliter, 1984). Moreover, because
empirically suggested that the minimal group size is four or goats are known to climb on a variety of objects, providing such
five individuals for adult sheep, but the familiarity with the devices may contribute to their well-being. Environmental
environment may influence this number. One study reports enrichment consisting of old tires and wooden railway sleepers
that when 1-month-old lambs are put in a novel environ- to climb on, and PVC piping to mouth and butt, increased
ment, the minimal group size for suppressing nearly all the weight gain in a feedlot of castrated goats by reducing aggres-
distress vocalizations is four individuals (Rault et al., 2011). sive behavior at the feed trough (Flint and Murray, 2001). It
A second study reveals that while separation from mates in is recommended that ample climbing space is provided to
the familiar pen increases respiration rate, cortisol concentra- avoid dominant animals from displacing subordinates.
tion, number of steps, vocalizations, and urinations, these
reactions decline to basal levels once a single companion 1.17.4.2.5 Human Contact
ewe is present. There is no additional advantage to having The development of a positive human–animal relationship in
two companion ewes (Carbajal and Orihuela, 2001). No small ruminants has obvious beneficial effects by lowering
equivalent data are available for goats, but if adults need to their level of fear or increasing their level of confidence in
be housed in groups, care must be taken to minimize harmful people. Gentle stroking by a familiar caregiver is a strategy
social behavior such as aggression. that helps sheep and goats calm down in potentially stressful
situations (e.g., isolation: Boivin and Braastad, 1996; Boivin
1.17.4.2.4 Physical Enrichment et al., 1997). Additional benefits of gentle stroking have been
Because ewes show a strong tendency to isolate from the flock observed on milk production in goats and on lambs’ immune
when they are about to give birth outdoors, providing such system and cortisol response to handling (Caroprese et al.,
a possibility in an enclosed building is beneficial for the early 2009b; Miranda-de la Lama and Mattiello, 2010). However,
mother–young interactions and bonding. Also ewes that had a relationship with a human takes place more easily when
been previously exposed to 1.2  1.8 m cubicles use them lambs are reared without their mother. The bond with the care-
more readily than naïve animals (Figure 11(a)). After giving giver can be equivalent to the filial attachment developed by
birth, the ewes occupying 1.2  1.8 m cubicles remain longer mother-reared lambs, and the underlying mechanisms medi-
on the birth site and closer to their lambs, are rarely disturbed ating these processes may be similar (Nowak and Boivin,
by other ewes, and have fewer lambs separated or stolen from 2015). Although the existence of a sensitive period is still
them (Gonyou and Stookey, 1985; Figure 11(b)). In goats debated, affinity with the caregiver develops more rapidly if
living within a confined group, the strong hiding drive of the positive interactions take place immediately after birth.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 469

Figure 11 (a) Importance of previous experience on the percentage of parturient ewes using a 1.2  1.8 m cubicle to isolate themselves. Ewes had
never experienced such cubicles before, or had been exposed and did or did not use them. (b) Percentage of parturient ewes that were interfered
with by other ewes whether they had no hiding place (control), had access to cubicles and lambed outside, or had access to cubicles and lambed in
them. Adapted from Gonyou H., Stookey, J.M., 1985. Behavior of parturient ewes in group-lambing pens with and without cubicles. Appl. Anim.
Behav. Sci. 14, 163–171.

1.17.4.3 Regarding Environment (Light, Sound, Vibration, 1.17.4.3.2 Dominance and Agonistic Behaviors
Temperature, Density of Animals) Within an established flock of sheep, many expressions of
dominance are not necessarily associated with overtly aggres-
1.17.4.3.1 Flocking
sive behavior. Sheep maintain hierarchy through subtle activi-
Both goats and sheep show a strong desire to remain with
ties, like head movements and eye contact, rarely head-butting.
their group mates, and they become very vocal when sepa-
Subordinate sheep usually move away and do not retaliate after
rated from the flock. They may separate themselves into
receiving aggressions (Nowak et al., 2008). Dominance is more
subgroups consisting of family units or of animals from
obvious in a flock of goats, which are a much more aggressive
different origins when mixed together to form bigger flocks
species, and the presence of horns is an important determinant
(Miranda-de la Lama and Mattiello, 2010; Nowak et al.,
of dominance. Accordingly, most agonistic interactions are
2008). Unlike the mother–infant dyad, little is still known
brief feints or rushes in which the dominant animal lowers
about recognition between adult individuals. Sheep, and in
its head and points its horns at the subordinate. However,
all likelihood goats as well, are predominantly visual
long fights may occur in which horns and head are clashed
animals. Cohesion of the group is maintained by each indi-
together repeatedly when two animals are of equal or undeter-
vidual adjusting its behavior or position relative to the
mined rank. If the conflict escalates, aggressive behavior
postures or activities of other members. This leads to
involves one goat standing up on its hind legs, lowering its
synchronized feeding activity, resting, spreading of alertness
head, and striking it against its opponent’s head. When space
states, and tendency to follow a leader. But above all, sheep
is limited and butting cannot be performed, biting is a more
are known to recognize individuals on the basis of visual
frequent means to maintain individual distances (Miranda-de
cues: up to 50 different sheep faces can be discriminated in
la Lama and Mattiello, 2010). Usually, the level of aggression
an operant conditioning task at one time, and such recogni-
is higher among animals kept indoors than among individuals
tion may last for up to 2 years (Kendrick, 2008). Sheep can
living outdoors because of the difference in the amount of
also discriminate conspecifics, based on a similar test para-
space available and the impossibility for subordinates to avoid
digm, using olfactory cues contained in urine, wool, feces,
dominants. There are a number of consequences for subordi-
saliva, and secretions from the interdigital, inguinal, and
nate individuals. When access to food is limited, subordinate
infraorbital pouches. How these senses are used in group
sheep are likely to have access to poorer-quality forage or
cohesion and interindividual communication remains
they may be displaced from the feeding-trough if access is
unknown. The tendency to aggregate varies between breeds,
limited. Subordinates may also be displaced from shaded
and mountain breeds usually tolerate greater individual
areas, males may mate the less preferred females, and females
distances than sheep from lowland breeds. Leadership is an
may have their lambs stolen by dominant subjects.
additional component of sheep and goat social behavior,
and it is expressed by animals that initiate a movement in
the group. Leadership is never associated with dominance. 1.17.4.3.3 Social Isolation
Rather, it is negatively correlated with the tendency to join Sheep and goats are highly gregarious, and separation from the
the flock; thus, it is generally the most independent indi- flock is perceived as very stressful. Immediately after being
vidual that initiates movement. isolated, they show increased behavioral and vocal activity
470 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

(Price and Thos, 1980). Various physiological responses are individual in dairy ewes is associated with a decreased propor-
also observed after sheep are isolated in a pen, specifically, tion of animals walking and an enhanced incidence of aggres-
increases in heart rate, number of neutrophils, concentration sions (Caroprese et al., 2009a). In addition, immune response
of catecholamines, cortisol and prolactin, plus a decreased and milk production are increased in dairy ewes when space
number of lymphocytes (Cockram et al., 1994; Minton et al., allowance increases from 1 to 2 m2 per ewe illustrating an
1992; Parrott et al., 1994). The physiological responses vanish improvement in health condition and well-being (Sevi et al.,
with repetitive testing, but the behavioral changes are still 1999). Compared to sheep, goats are reported to have a signif-
visible when sheep are isolated for the seventh time. Goats icantly higher rate of aggressive interactions; they display biting
do not habituate to social isolation either (Sieberta et al., to maintain individual distances if space allowance does not
2011; Figure 12(a)). While paradigms used to investigate the allow butting (Miranda-de la Lama and Mattiello, 2010).
effect of social isolation usually confound the direct effects of Increased interindividual competition, which results in denied
isolation with other factors (e.g., being moved to a novel of access to the feeding-trough, leads to longer time spent queuing
environment, e.g., Cockram et al., 1994), removing group and shorter time spent feeding, and an increased number of
mates from a room also triggers behavioral distress and eleva- displacements and aggressive interactions (Jørgensen et al.,
tion of cortisol in the ewe left alone (Guesdon et al., 2012; 2007; Loretz et al., 2004). Reduced available space impairs
Figure 12(b)). If sheep have to be put in individual pens, social the growing performance in young lambs as well (Gonyou
contact must be maintained by the presence of peers in adja- and Stookey, 1985; Horton et al., 1991). In animals reared in
cent pens. It has also been suggested that mirrors may serve pens, space allowance should allow them to lie down, stand
as a social surrogate under total social isolation; however, up, circle, and feed without restriction. For instance,
they do not completely abolish the physiological stress a maximum of three adult ewes per square meter of feeding-
response (Parrott et al., 1988). trough is usually recommended to ensure proper access to
food; however, this should be adjusted according to the size
1.17.4.3.4 Density of Animals of the breed and whether ewes are pregnant or not. In Europe
Crowding, which implies an excessive density of animals in ewes are allowed 1–1.4 m2 per head according to their size and
a restricted space, is typically found indoors, but its potential physiological status, rams between 0.5 and 2 m2, and lambs
deleterious effects are difficult to evaluate. Nonetheless, it is 0.25–0.90 m2 according to their size. Similar housing condi-
a fact that sheep confinement within enclosures activates the tions may apply to goats (Sevi et al., 2009). However, it is
hypothalamic–pituitary–adrenal (HPA) axis with an increase uncertain if these figures are chosen on objective grounds and
of cortisol and the display of stereotypic behaviors (Caroprese, truly satisfy the needs of sheep. In Europe recommendations
2008). Reduction of the lying area (1.0, 0.75, and 0.5 m2 per for animals used for scientific purposes are provided by the
ewe) alters the resting patterns of pregnant ewes (Bøe et al., Directive, 2010/63/EU (Directive, 2010), and they differ from
2006). Reduction in space allowance from 3 to 1.5 m2 per those concerning animals reared on farms and given by the

(a) Goats (b) Ewes


Vocaliza on
Vocaliza on 70
Number of high bleats
Number of high bleats

120 60
100 50
80 40
60 30
40 20
20 10
0 0
1 2 3 4 5 6 Session 1 Session 2 Session 3
Isola on sessions
Rearing on back legs Pawing with foreleg
Number of occurrences

20
Number of occurrences

8
15
6
10
4
2 5

0 0
1 2 3 4 5 6 Session 1 Session 2 Session 3
Isola on sessions Before A er

Figure 12 (a) Distress behavior in goats submitted to social isolation on six consecutive days. (b) Distress behavior before and after isolation in
ewes submitted to three consecutive testing sessions. Vocal and locomotor activities show that subjects do not habituate. Adapted from Sieberta, K.,
Langbein, J., Schön, P.C., et al., 2011. Degree of social isolation affects behavioural and vocal response patterns in dwarf goats (Capra hircus). Appl.
Anim. Behav. Sci. 131, 53–62 and Guesdon, V., Ligout, S., Delagrange, P., et al., 2012. Multiple exposures to familiar conspecific withdrawal is
a novel robust stress paradigm in ewes. Physiol. Behav. 105, 203–208.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 471

Department for Environment, Food and Rural Affairs in the UK considered to be optimal in the range of 6–19  C for adults
(DEFRA, 2000) and by the Guide des Bonnes Pratiques Ovines and 14–20  C for lambs. Natural physiological adaptation
in France (GBPO, 2011). Apparently these space allowance occurs between 9 and 31  C in adults and between 2 and
ranges do not raise major practical problems for farmers in 25  C in lambs. Beyond these limits animals are considered
terms of production nor any ethical concerns in terms of to be under thermal stress.
research. Nonetheless, as sheep and goats cannot escape from
their pen to seek shelter or avoid unpleasant environmental 1.17.4.3.6 Lighting and Noise
conditions, care must be taken not to expose them to excessive Sheep and goats are seasonal breeders, and as such they both
heat, cold, noise, or dampness. are very sensitive to changes in the duration of light. It is the
circadian rhythmicity that controls the secretion of sexual
1.17.4.3.5 Heat and Cold Stress hormones via the release of melatonin. As a consequence, arti-
Sheep and goats are rather resistant to climatic extremes, and ficial control of light–dark ratio results in the control of repro-
they are found in tropical regions as well as in mountain duction in lightproof buildings (Chemineau et al., 1992). In
pastures. However, under very hot conditions they reduce a more general sense, light intensity and color affect sheep
their locomotion and food consumption, seek shade, and behavior, but data in that domain are scarce. Hypoactivity
feed preferentially at nighttime. In bare areas they congregate with a decrease in the time spent feeding is observed in lambs
together side by side (Silanikove, 2000). Such a peculiar exposed to low light intensity (10 lx); conversely, they are
behavior occurs when the heat being taken by the body hyperactive at high light intensity (1000 lx). Green light
exceeds the ability to dissipate it. Thus, by huddling together, appears to increase the time spent standing and feeding
individuals reduce the intake of direct solar energy per body compared to white, red, and blue lights, and body weight
surface and thermoregulate in a better way than when they gain is improved (Caroprese, 2008). We also know very little
stand alone. There is little information considering specifi- about the effect of noise on sheep and goats. Cortisol may
cally the effects of heat stress on recognized welfare indicators increase moderately in lambs under some conditions (2 kHz
compared to the impact on productivity (Caroprese, 2008; and 75, 85, 95 dB) but not under others (100–6300 Hz and
Silanikove, 2000; Marai et al., 2007; Sevi and Caroprese, 85 dB) even though walking activities increase in the latter
2012). Goats seem to be less affected by heat than sheep, condition. In pregnant ewes, exposure to loud noise (2 kHz
which reflects their adaptation to harsh environments: 88% and 95 dB) results in shorter active behavior including eating,
of goats are found in Asia and Africa, mostly in tropical and but without change in cortisol levels or immune responses.
subtropical areas. In addition to reducing feed intake in sheep, Despite the scarcity of information, available data suggest
heat stress exposure (above 30  C) disturbs water metabo- that lambs and ewes are not particularly affected by intense
lism, energy and mineral balances, enzymatic reactions, illumination nor by loud noises (Caroprese, 2008). In any
hormonal secretion (cortisol increase and decrease in thyroid case, it is recommended that loud noise be avoided and that
hormone activity), and blood metabolites and increases pulse sheep housed indoors must be exposed to natural daylight
rate, respiration rate, skin and rectal temperature. However, cycle using either artificial or natural light, except for breeding
breeds native to tropical regions are better adapted to heat animals under a controlled light regime. Should supplemented
than temperate breeds (Dwyer, 2008b). In pregnant ewes light be added, illumination intensity should be set to provide
a reduction in total embryo cell number, placentome size, 75–100 lx (Caroprese, 2008).
and lamb birth weight is observed under extreme heat.
Lactating ewes exposed to direct solar radiation (maximum
1.17.4.4 Regarding Modifications of Reactivity to Stress
range: 33–37  C) exhibit high concentrations of nonesterified
by Neonatal Manipulations
fatty acids in blood (due to enhanced mobilization of body
reserves), depressed lymphocyte proliferation, and reduced 1.17.4.4.1 Prenatal Stress in Small Ruminants
mammary defense capacity. Sheep are extremely cold-hardy Aversive events taking place in early life can alter the ability to
and rarely experience environmental conditions below their cope with subsequent stressful circumstances. Compared to the
lower critical temperature (the minimum body temperature vast amount of research on prenatal and neonatal stress in
before bodily functions are altered). By contrast, because of rodents, only a limited number of experiments have been per-
their short and wet fleece, and their small size, lambs and formed on small ruminants. Work on sheep and goats has
kids are particularly vulnerable to hypothermia (Dwyer shown that aversive prenatal experiences may influence post-
et al., 2016). Soon after birth, their body temperature falls natal physiological and behavioral responses. Kids exposed to
by 1–2  C before stabilizing and then rising progressively to repeated prenatal stress develop heavier adrenal medullas
reach an hour later the initial value (39  C). Their physiolog- and higher HPA axis activity at 1 month, and they are more
ical adaptive mechanisms to increase heat production are reactive in open-field tests; females seem to be more sensitive
limited and include nonshivering thermogenesis involving than males (Duvaux-Ponter et al., 2003; Roussel et al., 2005).
brown adipose tissue metabolism and shivering. The second Similarly, lambs born to mothers stressed during pregnancy
mechanism usually occurs in older animals under cold and show increased exploratory behavior and locomotion at
wet conditions and metabolizes muscle glycogen. In case of 8 months of age compared to control lambs (Roussel et al.,
unfavorable environmental conditions, shelter should be 2004). While no behavioral differences are observed at an
provided when animals are kept outside, and heating lamps earlier age, basal cortisol concentrations are nonetheless higher
are provided indoors for small hypothermic lambs. Under in 25-day-old prenatally stressed lambs compared to controls.
dry and no wind conditions thermal well-being in sheep is Some authors report a longer-term effect on cognitive and
472 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

emotional aspects since at the age of 1.5–2.5 months lambs circulating cortisol. The autonomic SAM is also activated at
subjected to prenatal stress are more fearful, do not perform detection of some threat and causes the release of catechol-
as well in a T-maze, and show pessimistic-like judgment in amines from the adrenal medulla. However, the release of
a cognitive bias test (Coulon et al., 2015). The effects are not adrenaline and noradrenaline is far more difficult to detect
necessarily comparable between studies and species since the as the dynamics of secretion is more rapid than for cortisol
types of stressors vary (e.g., pregnant mothers exposed to (Parrott et al., 1994, 1998). Because catecholamines influence
social isolation, transport, or dog) and postnatal evaluation peripheral systems, they can be monitored by measurement of
involves a diversity of testing situations (e.g., isolation tests, heat rate, respiration rate, body temperature, or blood pres-
surprise tests, dark–light test, novel environment). For sure. Temporary rise in cardiac rate and cortisol indicates
instance, social instability – which triggers aggression among a normal response to a threat, and if the stressor is short
pregnant goats – has minor effect in terms of the behavioral lasting, the physiological responses eventually decline to base-
development of the kids. Compared to young born into line. However, standard stress responses are difficult to charac-
stable social groups, they seem to be less fearful at 5 weeks of terize in sheep as they may be influenced by breed, age,
age and have lower basal cortisol levels (Andersen et al., experience, physiological state, gender, and considerable inter-
2008). Whether these effects persist beyond infancy is individual variation. Prolonged or repeated exposure to
unknown. a stressor causes a specific form of adaptation with reduced
activation of the HPA axis (e.g., Cockram et al., 1994), which
1.17.4.4.2 Neonatal Stress in Small Ruminants is believed to help the individual cope with the actual situation
In dairy goats, kids are taken away from their mothers the since excessive amounts of cortisol can be detrimental. As
moment they are born and reared artificially. In sheep, many a consequence, lower levels of cortisol may be indicative of
lambs follow the same fate for various reasons: nonmaternal chronic stress. However, cortisol by itself cannot be always
ewe, insufficient milk yield, large litter size. Artificial rearing interpreted as indicative of a state of stress since it can increase
of lambs is not without consequences (Napolitano et al., in times of excitement or positive emotional states (e.g.,
2008). It is associated with immediate behavioral disturbances, mating). Consequently, its concentration in blood must be
such as increased vocalization and redirected sucking on pen associated with other physiological or behavioral evidence to
mates, as well as increased basal neonatal cortisol and oxytocin support an indication of stress. By and large, small ruminants
levels indicative of stress. Such lambs also have a lower anti- avoid the stressor and display agitation and increased vocaliza-
body titer to keyhole limpet hemocyanin in the following tion. Behavior should, nonetheless, be analyzed cautiously as
weeks after injection, illustrating a marked depression of it varies according to the context and does not always correlate
immunological responses and suggesting reduced level of cyto- with physiological responses. For instance, when tested in an
kines stimulating B cell activity. Moreover, when tested in isola- open-field arena, behavioral and physiological responses
tion at 3 weeks of age they show reduced locomotion and expressed by lambs reared with or without their mother may
vocalization and an increased cortisol response (Napolitano differ or not according to the context (Napolitano et al.,
et al., 2008). Yet, an increased cortisol response is not system- 2008). Another well-described response of sheep and goats
atically observed if lambs are tested at an older age (Moberg exposed to stressors is the alteration of the immune function
and Wood, 1982). The behavioral consequences of early social (Cockram et al., 1994; Minton et al., 1992; Sejian and
experience are more consistent and can still be observed at Srivastava, 2010). The main characteristic is an alteration in
42 days; they are even more accentuated if lambs had been leukocyte population, usually an increase in the number of
reared in total isolation. In an open-field test, artificially reared neutrophils and a decrease in the number of lymphocytes
lambs kept in isolation express a nonfunctional stereotyped suggestive of immunosuppression. Under chronic stress, alter-
behavior called flank touch and described as ‘lamb reaching ation in immune system has an effect on disease resistance. For
back and touching its flank with its muzzle’ (Moberg and instance, stress associated with weaning or transport and
Wood, 1982). This self-directed behavior may be displayed in indoor housing reduces resistance to parasite infestation
substitution of proximity searching and suckling that is nor- (Sotiraki et al., 1999). Small ruminants are less likely to
mally directed toward the mother. The long-term effect of perform stereotyped behavior, but for animals kept indoors,
maternal depravation has never been investigated in small a number of abnormal behaviors were reported such as
ruminants. ground searching, rail-standing, bar-chewing, or pen-butting
(Marsden and Wood-Gush, 1986a,b). Sheep may also show
other forms of abnormal behavior such as cross-sucking and
1.17.4.5 Regarding Methods to Quantify Parameters
wool-pulling. Cross-sucking occurs in artificially reared lambs
Presumably Indicative of Stress
where some individuals suck the navels or scrotum of conge-
The physiological responses to stress relate to the functioning ners. This activity is common in the initial days, but it may
of the HPA system, the sympathetic-adrenal medullary system last until weaning and it is not without consequences. The
(SAM), and some assessment of the immune system. The HPA lamb being sucked may be disturbed and develop navel infec-
axis is a neuroendocrine response triggered by external or tions, while the animal performing sucking, poorly adapted to
internal factors that act on the hypothalamus. Specific neurons artificial feeding, may reduce the ingested amounts of reconsti-
of the paraventricular nucleus are involved, and in sheep, tuted milk (Napolitano et al., 2008). Wool-pulling is exclu-
isolation, restraint, or exposure to a dog activates neurons sively observed in indoor-housed sheep. It is frequent at
producing CRH, AVP, and enkephalin (Cook, 2004; Minton, high stocking density and reduced by increased provision of
1994; Rivalland et al., 2007). This provokes an increase in roughage (Vasseur et al., 2006).
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 473

1.17.4.6 Regarding Sacrifice Methods and perform as much licking as unassisted cows; this indicates
Euthanasia may be necessary for experimental reasons or no delayed onset or impaired expression of maternal behavior
because of natural causes (traumatic injury, incurable disease). in dams given assistance at delivery. However, although maternal
There are no specific regulations concerning this issue but there behavior was unaffected by a difficult delivery, dairy calves born
are guidelines which are accepted worldwide, e.g., the American following difficult calvings have lower vigor in the first 3 h after
Veterinary Medical Association in the USA (Leary et al., 2013), the birth than unassisted calves. Moreover, Barrier et al. (2013)
Euthanasia of Experimental Animals published by the European found that calves that survive a dystocia experience have lower
Commission (Close et al., 1997). Only two methods are passive immunity transfer, higher mortality, and higher indica-
considered to be suitable for sheep and goats: chemical tors of physiological stress (salivary cortisol). This might have
methods with the use of injectable agents such as barbiturates longer-term effects on the health and survival of the calves.
and physical methods such as captive bolt or electric stunning Reproductive disorders such as lack of estrus, embryonic
followed by exsanguination. Other methods may be used loss, or early abortion can reflect prolonged or short-term
under specific conditions. Barbiturates (pentobarbitone, poor welfare (Algers et al., 2009). Stress during parturition or
sodium pentobarbitone) act by depressing the central nervous in early lactation can lead directly to poor well-being, particu-
system in three steps: from consciousness to unconsciousness, larly dystocia and genital infections, associated with pain or
then deep anesthesia, and finally death. The jugular vein is the inflammatory reactions. The weeks immediately after parturi-
normal intravenous route of injection; shearing the neck prior tion are important since cows are at a high risk for both meta-
the injection facilitates the procedure. It is important to ensure bolic and infectious diseases (Ingvartsen et al., 2003). Both are
that the animal is dead before disposing of the body. The use of related to inadequate nutrition and are one of the most serious
captive bolt is an acceptable physical method for small rumi- welfare issues affecting dairy cows (von Keyserlingk et al.,
nants. Projection of the shot should be on a line running 2009). Several behavioral changes are observed in sick animals,
between the bases of the ears and aiming toward the throat. such as reduced activity, feeding, grooming, and rumination,
Electric stunning is an acceptable form of euthanasia as long which are used as behavioral indicators of a lack of well-
as the research facility has a slaughter room. In both cases being (Borderas et al., 2008; von Keyserlingk et al., 2009).
adjunctive methods such as exsanguination should be applied Cows are ‘hiders’ and they maintain their calves separated
immediately following the loss of consciousness of the animal. from the rest of the group for the first days after parturition
Operation training, experience, and confidence are required, (Kilgour and Dalton, 1984). Calves reared under natural condi-
but shooting may only be suitable if the operator has an appro- tions develop a bond with their mothers through grooming,
priate firearms license. Potassium chloride and magnesium lactation, and vocalizations. Most dairy cattle productions
sulfate are effective methods to ensure death in sheep and goats discourage maternal behavior, with the exception of milk
previously rendered unconscious by mechanical methods or production. When domestic cattle are permitted to raise their
administration of anesthetics. Because lambs and kids are calves, their behavior is similar to that of wild ungulates. These
born in an advanced stage of development, the same methods behaviors permit the cow to bond with her calf, protect it,
of euthanasia should be applied on them. and nourish until the breakup of this bond at weaning
(von Keyserlingk and Weary, 2007). Colostrum is essential
for the calf, and several studies indicate that many calves do
1.17.5 On Dairy Cattle not ingest sufficient quantities. Management practices that
maximize the calf’s opportunities to ingest colostrum and
Dairy cows have had the opportunity to live in a friendlier envi- minimize the contact with pathogens will have beneficial
ronment than some other animals in intensive productions, effects on their welfare. Albright (1983) demonstrated that
having spaces to lie down, eat, drink, and walk and having colostrum (even when bottle fed) has a better chance to be
the freedom to decide when to do it. Unfortunately, this does absorbed when the calf is next to its mother and that grooming
not mean that they are free from welfare concerns. Calves are helps the calf to drink with ease (Figure 13).
separated from their mothers at birth or a few days later, Unfortunately, the calf’s separation from the cow in the first
housed individually, lacking the opportunity for social contact, days after parturition has behavioral, health, and production
and artificially fed. Cows also lack the opportunity to graze; effects, on both the mother and the calf. There is evidence
they can suffer from metabolic problems, lameness, and other that 5-min contact with a calf immediately postpartum is suffi-
infectious diseases. cient for the formation of a strong, specific maternal bond with
that calf. Hudson and Mullord (1977) found that this bond
lasts, even if the calf is removed for 12 h and then returned.
1.17.5.1 Lodging and Handling during the Peripartum Period:
After 24 h of separation, the cow still shows signs of distress,
Relevance for the Establishment of Maternal Behavior and
but can no longer recognize her own calf. If no contact is
Nursing
allowed between mother and young for periods of up to 5 h
Parturition is a difficult process. The calf’s development and postpartum, this maternal bond is not formed in half of the
survival is dependent upon being strong and vigorous at birth animals. These results suggest that there is a sensitive period
and receiving appropriate maternal care. However, difficulty at for the development of maternal responsiveness in dairy cattle.
delivery can result in less vigorous offspring and maternal care Hopstee et al. (1995) found only mild effects immediately after
can be altered, probably as a consequence of exhaustion, pain, the calves were removed. After separation cows vocalized
and human intervention. Barrier et al. (2012) have found that initially and moved to the feeding rack and started feeding
assisted cows at parturition do not take longer to lick the calf shortly after that. Heart rate effects were restricted to the first
474 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

30 70
1 day
Primiparous 60 2 weeks
25
Mul parous

20 50

# vocaliza ons
Minutes

40
15

30
10
20
5
10
0
1 2 3 4 5 6 0
Days 1h 18 h 24 h

Figure 13 Time licking their calves in primiparous and multiparous Figure 14 Calf vocalization after 1, 18, or 24 h of separation, when
cows. Redrawn from von Keyserlingk, M.A.G., Weary, D.M., 2007. calves were separated at 1 day or 2 weeks of age. Data obtained from
Maternal behavior in cattle. Horm. Behav. 52, 106–113. Flower, F.C., Weary, D.M., 2003. The effects of early separation on the
dairy cow and calf. Anim. Welf. 12, 339–348.

minutes after separation and cortisol effects were not found. living in elevated crates in a closed building. In the last 20 years
They concluded that multiparous dairy cows respond only hutches have become more popular.
mildly immediately after their calves had been removed. Friend et al. (1985a,b) worked on different tests to measure
Lidfors (1996) found that separation after 4 days has an the well-being of calves under different housing systems
effect on activity, vocalization, and rumination in the cows (Table 4). Chronic deprivation of movement that does not
and on the activity and oral behavior of the calves. Stehulova allow natural behaviors such as locomotion, rest, grooming,
et al. (2008) concluded, from the study of specific behavioral exploration, and social interaction can be accompanied by
reactions, that separating dairy cows from their calves is more behavioral and physiological changes indicative of stress
stressful for both parties if it is performed on days 4 and 7 (Friend et al., 1985a,b; Sisto and Friend, 2001). Calves housed
than on day 1. The behavioral response to separation was in crates and hutches tend to stumble more frequently than
more intense and lasted longer in both of the animals when those housed in groups, with more space and allowed to exer-
they had visual and auditory contact after separation. The heart cise (Friend et al., 1985a,b; Figure 15).
rate response of cows was not influenced by the age of the Individually confined calves do not show normal social
calves at separation or postseparation contact, but the heart interaction patterns and are, therefore, unable to compete for
rate increase in the separated calves lasted longer the more food with calves reared in groups (Broom and Leaver, 1978).
they had been with their mothers. Indeed, calves that stayed This is reflected in a lower weight gain and higher cortisol levels
with their mothers had less incidence of diarrhea, better weight at transport (Trunkfield, 1990). Calves reared in groups show
gain, and better maternal ability than calves separated a few fewer fear reactions when confronted with new stimuli at
days after parturition (Flower and Weary, 2003), findings sug- 3 months of age, suggesting that individual management prac-
gesting that production and welfare benefits are greater when tices compromise calf welfare by inhibiting their normal
calves stay longer with the cows. Even though late separation behavior patterns such as grouping in crèches (Donovan,
would be a challenge for the milk industry, this study explains 1992). The European Union has legislated that calves older
how a natural imprinting between the mother and the calf can than 8 weeks have to be housed in groups, and in organic farms
be used to improve production and welfare (Figure 14). they must be housed in groups since the first week of age
(Jensen, 2003). Socialization in calves is considered a priority.
1.17.5.1.1 Calf Lodging
After the second week, calves, under natural conditions, are left Table 4 Physiological changes in calves under different housing
in ‘crèches’ or ‘kindergartens,’ where one or two cows take care systems
of all the calves. This facilitates social behavior (Phillips, 2008). Treatment
Play can be used as a welfare indicator, since calves play longer
when they are well fed and healthy (Houpt, 2005). Calves Elevated crate Hutch Group pen
reared in intensive farms live under very different conditions
Basal cortisol 5.4 4.8 3.4
to those encountered in nature, and this can be an important ACTH challenge 103 81 75
source of welfare problems. Calves under these conditions Lymphocytes (%) 61 69 71
can be housed in groups or individually, with different space Neutrophil/lymphocyte 0.69 0.45 0.36
allowance and levels of climatic control. Milk production
systems have undergone several changes in the last 50 years. Based on data from Friend, T.H., Dellmeier, G.R., Gbur, E.E., 1985a. Comparison of
four methods of calf confinement. I. Physiology. J. Anim. Sci. 60, 1095–1101 and
In the 1960s, when farms grew and became more specialized, Friend, T.H., Dellmeier, G.R., Gbur, E.E., 1985b. Comparison of four methods of calf
calves went from living in small groups near their mothers to confinement. II. Behavior. J. Anim. Sci. 60, 1102–1109.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 475

12 concentration is elevated, suggesting that milk flavor is what


Galloping
motivates suckling (de Pasillé et al., 1997; Figure 16). When
10 lactose levels are elevated, nonnutritive suckling is also
elevated, and when lactose levels are reduced, you get the oppo-
8 site effect (de Pasillé and Rushen, 1998), suggesting that the
most important factor is the sugar concentration in the milk
Incidence

6 and not lactose per se. Rushen and de Pasillé (1995) examined
if suckling reduced the motivation to suckle using dry teats after
4 the calf drank its milk and found that there was an inhibitory
effect on nonnutritive suckling. They also found that the moti-
2 vation to suckle diminished after 10 min. de Pasillé and Caza
(1997) found that calves which got dry teats after feeding
0 preferred them over their group mates, reducing cross-suckling
Elevated crate Hutch Group pen
by 75%. Jung and Lidfors (1999) reported similar results.
Many have tried to extinguish the suckling instinct by trying
Figure 15 Behavioral changes during an open-field test in different
housing systems. Based on data from Friend, T.H., Dellmeier, G.R., to teach the calves to drink from a bucket in the first days of life,
Gbur, E.E., 1985a. Comparison of four methods of calf confinement. but this increases, rather than decreasing, the problems. The
I. Physiology. J. Anim. Sci. 60, 1095–1101 and Friend, T.H., Dellmeier, same happens when bottles with big nipple holes are used.
G.R., Gbur, E.E., 1985b. Comparison of four methods of calf confine- Cross-suckling is less frequent when a teat or nipple is used
ment. II. Behavior. J. Anim. Sci. 60, 1102–1109. (bottles or buckets with nipples) compared to only using
buckets. When calves take longer to drink the milk through
the nipple, they engage in less nonnutritive suckling (Haley
1.17.5.1.1.1 Calf Feeding et al., 1998). Offering water through a nipple or straw after
Feeding behavior of artificially reared calves is very different feeding reduces, but does not eliminate, nonnutritive suckling
from that of naturally reared calves. Besides the lack of the (Haley et al., 1998; de Pasillé, 2001).
natural stimulus associated with the cow’s udder and maternal
behaviors, calves are usually fed only twice a day and they
consume, at each bout, considerably more milk than what 1.17.5.2 Lodging of Dairy Cows
they would in any of the 4–10 feedings seen under natural Housing systems differ all over the world depending on climate,
conditions (bouts lasting 8–12 min; Wilt, 1985; Jensen, economy, and personal preferences of the farmers. In many
2003). The ways by which milk is offered to artificially reared countries dairy cows are kept on pastures year-round or during
calves promote the development of crossed suckling (i.e., suck- good weather. In the United States, dairy cows are usually kept
ling of ears, tail, prepuce, and other parts of the body). Some in large lots with cement or dirt floors, some with free stalls
researchers have compared the behavior of calves fed using (cubicles) or tie stalls. Algers et al. (2009), following a request
different methods, while others have investigated the motiva- from the European Commission, analyzed the most important
tional basis of nonnutritive suckling by manipulating different hazards regarding housing conditions and found that poor
aspects of milk feeding and the opportunity to suckle. Milk cubicle design and lack of space in cubicle houses and tie stalls
producers hesitate at the possibility of maintaining calves in had the highest estimated risk values. Inadequate ventilation,
groups due to the problem of crossed suckling as they believe
it could lead to milk stealing in adult cows (Spinka and
160
Illmann, 1992; Keil et al., 2000). Rings and metal plates are
placed on their noses when these animals are identified in 140
order to control the behavior, but this practice raises concerns
regarding the animal’s welfare (Keil et al., 2000). To be able 120
to improve group management and control these unwanted
% observa ons

100
behaviors, we need to understand what makes calves want to
suckle. 80
de Pasillé and Rushen (1995) have extensively studied
calves’ motivation to suckle. Contrary to what most people 60
think nonnutritive suckling is greater after feeding and not
40
before, and therefore it is not motivated by hunger as long as
the calves are well fed. Even when their milk rations were cut 20
in half nonnutritive suckling did not increase. This suggests
that neither the quantity of milk in the stomach nor the oral 0
Milk Water Milk×2
sensation of ingesting milk inhibits nonnutritive suckling.
When calves do not drink milk or water there is some nonnu- Figure 16 Percentage of nonnutritive suckling after a milk meal,
tritive suckling, but much less than what is observed after water, or increased concentration of milk replacer. Adapted from de
drinking milk (de Pasillé et al., 1992). Injecting small quanti- Pasillé, A.M., Rushen, J., Jensen, M., 1997. Some aspects of milk
ties of milk into the mouth of a calf produces a great amount that elicit non-nutritive sucking in the calf. Appl. Anim. Behav. Sci. 53,
of nonnutritive suckling, as well as when milk replacement 167–173.
476 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

temperature, and humidity were the highest-ranked hazards in designed cubicles (Haley et al., 2000; Manteca, 2009). Errors
straw yards. However, the risk estimate values in straw yards in stall design and maintenance have long been recognized as
were much lower than in cubicles and tie stalls. significant risk factors for mastitis, hock abrasions, teat trauma,
Studying the normal behavior of cows helps to improve and and entrapment injuries of dairy cows. Moreover, if the stalls
promote the well-being of the animals. To do so it is important are uncomfortable, the cow may choose to lie down in
not only to see the size of the cows, but also to observe their a slurry-covered alleyway and put herself at risk for environ-
movement sequences when lying down. The problem with mental mastitis infection (Nordlund and Cook, 2003).
cubicles is that some are poorly designed; this, together with Floors also play an important role in foot health. Concrete
the recent increase in the size of the animals, makes them floors can be slippery for cows, and when they are rough, they
poorly suited to fit the behavioral needs of the cows. They damage the hoof and animals have to alter their gait (Phillips
need to lie down in a comfortable, dry, clean environment as and Morris, 2000; Telezhenko and Bergsten, 2005). There is
deprivation of rest can affect their behavior (Cooper et al., increasing evidence for an association between standing time
2007). Cows lie down for 7–12 h (or more) during a 24-h on concrete and the induction of laminitis (Nordlund and
period, and if they cannot find a comfortable lying space, Cook, 2003; Espejo et al., 2006). Behavior also seems to play
they stand or lie in dirty passageways which will lead to poor a role in foot problems. For instance, half-standing in the
health issues, specially foot and udder problems (Wierenga cubicle (perching) is performed more frequently by low-
and Hopster, 1990; Arney and Aland, 2012). Lying is an impor- ranking cows, as they use cubicles to hide from dominant indi-
tant behavior for cows, as it enables them to rest and to avoid viduals (Metz and Wierenga, 1987; Potter and Broom, 1987).
confrontations. Moreover, a reduction in lying time affects their Galindo et al. (2000) found that low-ranking cows performed
welfare since the cow rests and ruminates when it is lying this behavior for a longer time and had a greater reduction in
down; the cow’s hoofs rest and dry off and blood circulation heel depth of the hind digits. This predisposed them to infec-
to the udder increases by up to 30% (Bewley, 2011). Uncom- tion in the interdigital space and heel, causing clinical lame-
fortable stalls result in less frequent or shorter-duration resting ness. Loberg et al. (2004) found that when given access to an
periods, and consequently, there is an increased standing time outdoor paddock, adult cows used the time to walk, trot, and
on concrete surfaces (Nordlund and Cook, 2003). explore the environment, and these behaviors had a positive
As the cow lies down, she puts about two-thirds of her effect on the claw conformation.
body weight on her front knees. These drop freely to the floor Regula et al. (2004) found that cows housed in free stalls
from a height of 20–30 cm, so it is very important to have with access to a pen, yard, or pasture where they could regularly
adequate bedding to avoid injury. Wet knees mean wet exercise had lower odds of presenting lameness, hock joint
bedding, and cows do not like to lie down in dirty stalls alterations, and teat injuries, therefore requiring fewer overall
(Manteca, 2009). The time a cow takes to lie down is medical treatments (Figure 17). Many cows have subclinical-
a good indicator of welfare or lack thereof. For example, if it level claw disorders, and welfare is an important issue on
takes her 5 min or less (on average), the welfare is good; lameness in dairy cows. Together with mastitis, lameness is
moderate, from 5 to 6.3 min, and bad if it is longer than recognized the most important health problem having a nega-
6.3 min (Manteca, 2009; Bewley, 2011). tive effect on the welfare of dairy cows (Somers et al., 2003).
It is important to consider lunge space to allow the cow to ‘Housing conditions affect animals throughout their lives’
lie down and stand up without difficulty. Cows that are not (Rushen et al., 2008), and it is extremely important to be aware
using all of the stalls or enter them in reverse indicate poorly of this and to take measures to correct any problem.

180

160 Reduced feed (Kg)

140 Lost milk (Kg)

120

100

80

60

40

20

0
Hock Hoof Teat injury Systemic Local Difficult Very
injury lesions mas s mas s calving difficult
calving

Figure 17 Effect of different diseases on cumulative reduction of feed intake and cumulative loss of milk production. Based on data presented in
Table 2.1 of Rushen, J., de Pasillé, A.M., von Keyserlingk, M.A.G., et al., 2008. The Welfare of Cattle. Springer, The Netherlands.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 477

1.17.5.3 Regarding Environment This leaves animals with an unfulfilled motivation to perform
these natural foraging activities. Object-licking, which is
Alleviating heat stress is critical to milk production. Heat
a repetitive licking or biting of fences, walls, or pen fixtures,
stress from high environmental temperatures can reduce
has also been reported in dairy cattle (Seo et al., 1998; Bashaw
food intake and milk production by more than 25% and it
et al., 2001).
is estimated that for every 1-lb decrease in dry matter intake
When measuring welfare we usually focus on the absence of
2 lbs of milk production is lost (Garcia, 2006). When cows
stress rather than on positive behaviors. Adequate environ-
have heat stress, they lie down less, and the risk for mastitis
mental stimulation favors good welfare and positive indicators
is higher (Manteca, 2009).
such as play (Napolitano et al., 2009). Animals are highly moti-
Water, often overlooked as an important nutrient, is
vated to play only if their primary needs are satisfied, while
extremely important to the modern, high-yield dairy cow, and
illness and injury are associated with the absence of play
insufficient or poor-quality water is an obvious welfare concern
(Friend et al., 1985a,b; de Pasillé and Rushen, 1995; Boissy
(Meyer et al., 2004). Water intake is closely related to feeding
et al., 2007; Napolitano et al., 2009). In calves, play is mainly
intake and thus animal productivity. Cattle are sensitive to the
expressed as locomotive play, that includes galloping, bucking,
palatability of water and prefer to drink clean water without
and kicking, and social play, which includes play fighting and
contamination (Garcia, 2006; Manteca, 2009). Water trough
nonreproductive mounting (Emmering, 2004; Napolitano
design can affect both preferences and consumption by lactating
et al., 2009). Although also described in adult cattle, play is
and nonlactating dairy cattle. Dairy cows prefer and drink more
more frequent in calves.
from larger troughs (Pinheiro Machado Filho et al., 2004).
Cattle, under natural or extensive conditions, are highly
synchronized as a result of allomimetic motivation (Bouissou
et al., 2001). Therefore, a high degree of synchronization
1.17.5.4 Behavioral Indicators of Well-being in Bovines
within the group may indicate a positive welfare state (Galindo
The time spent performing normal behaviors, such as eating and Broom, 1993). When cows have enough space and their
and resting, versus abnormal behaviors, such as stereotypies, primary needs are satisfied, all animals can feed and rest
can tell much about the welfare of the animals (Broom, without competition, they usually synchronize, and it could
1991; Sherman and Serpell, 2008). Automated monitoring of be said that their welfare is good. Their synchrony should
normal behaviors such as feeding, drinking, or walking can exceed 80% to consider a positive evaluation of the welfare
help identify sick animals before clinical signs appear or cows state of the herd (Napolitano et al., 2009; Figure 18).
that are in heat to correctly time artificial insemination. Tech- Self-licking and scratching have been also proposed as indi-
nology is advancing, but we must not rely on the automatic cators of adequate living conditions. A reduced expression of
measurement of behavioral indicators just because it is easy these behaviors has been related to slippery floors, thus reflect-
(Rushen et al., 2012). ing poor housing conditions (Winckler et al., 2002). Allog-
Stereotypies are important indicators of poor welfare, and rooming plays a key role with regard to the so-called
in dairy cattle we mainly find oral stereotypies such as sociopositive interactions. Allogrooming seems to serve both
tongue-rolling (Sambraus, 1985; Bergeron et al., 2006) due hygienic (body care) and social functions as it is thought to
to diets that take too little time to find, chew, or ruminate. play a major role in reinforcing social bonds and in reducing

Behavior indoors
100 Behavior outdoors
90
80
70
60
% of cows

50
40
30
20
10
0
1 3 5 7 9 11 13 15 17 19 21 23 25 27 29 31
Time of day

Figure 18 Feeding synchronization in the same group of cows when kept indoors (free stall) or outdoors (at pasture). Redrawn from data of
Galindo, F.A., Broom, D.M., 2000. The relationships between social behaviour of dairy cows and the occurrence of lameness in three herds. Res. Vet.
Sci. 69, 75–79.
478 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

tension in groups of animals. Soothing effects regarding Table 5 Examples of physiological indicators of well-being used in
a reduction in heart rate have been demonstrated in cattle dairy cattle
(Sato et al., 1993; Boissy et al., 2007; Napolitano et al.,
Heart rate Broom (1991), Mohr et al. (2002), Hagen
2009). Sato et al. (1993) found that all animals were groomed
et al. (2005), and Stenhulova et al. (2008)
but that only 75% performed grooming. Plasma cortisol Friend et al. (1985a), Hopster et al. (1999),
and Sisto and Friend (2001)
1.17.5.4.1 Handling of Adult Bovines Hair cortisol Comin et al. (2012) and González de la Vara
The way animals are handled by people has a major effect on et al. (2011)
their welfare, and unfortunately, until recently this issue had Fecal cortisol Palme et al. (2000), Möstl and Palme (2002),
been neglected (Rushen et al., 2008; Waiblinger et al., 2006). and Morrow et al. (2002)
Salivary cortisol Barrier et al. (2013)
Dairy cattle that are roughly handled often become frightened
ACTH challenge Friend et al. (1985a,b) and González de la
of humans, and this may result in a substantial drop in milk
Vara et al. (2011)
production. Rushen et al. (1999) found that the mere presence Temperature Sisto (2002)
of an aversive handler during milking may be sufficient to cause Infrared thermography Stewart et al. (2007)
the cows to ‘hold back’ milk, through a stress-induced suppres-
sion of oxytocin secretion. Many people believe that cattle are
not sensitive and are difficult to move, and this leads to rough
handling. Through classical conditioning the cows learn to
associate rough handling with the handler and soon show Table 6 Examples of behavioral indicators of well-being used in
fear (Rushen et al., 1999). Munksgaard et al. (1999) studied dairy cattle
the role of the color of overalls as a cue used by cows to discrim-
inate between people. The cows kept a longer distance when Maternal (licking of calf) Flower and Weary (2003) and Barrier et al.
the handler wore the color worn during the aversive treatment (2012)
than when the handler wore the color worn during the gentle Vocalizations Lidford (1996) and Flower and weary (2003)
Play Friend et al. (1985a,b), de Pasille and Rushen
treatment. The cows also responded according to the color of
(1995), Sisto and Friend (2001), Emmering
the clothes when worn by an unfamiliar person, although (2004), Houpt (2005), Boissy et al. (2007),
they kept a shorter distance from the unfamiliar person than and Napolitano et al. (2009)
from the handler. Rushen et al. (1998) mention that one way Synchronization Galindo et al. (2000), Boissou et al. (2001),
to reduce the magnitude of the fear that cows develop toward and Napolitano et al. (2009)
their handlers as a result of aversive handling may be to apply Grooming and Sato et al. (1993), Winckler et al. (2003),
those procedures outside the cows’ home stalls. Flight distance allogrooming Boissy et al. (2007), and Napolitano et al.
is, therefore, a good indicator of animal welfare (Figure 19). (2009)
Good management consists of much more than handling the Fear (flight distance) Rushen et al. (1999, 2008), Rushen (1996),
animals properly. Everyday routine animal care tasks, such as Rushen et al. (1999, 2008), Munksgaard
et al. (1999), and Waiblinger et al. (2006)
cleaning, are very important in ensuring good animal welfare
Social interactions Broom and Leaver (1978), Friend and
(Rushen et al., 2008). Dellmeier (1988), Galindo et al. (2000),
Tables 5–8 summarize the main behavioral, neuroendo- and Phillips (2008)
crine, immunological, and physiological indicators that have Feeding Witt (1985), Sisto (2002), Jensen (2003),
been used in bovines to determine their well-being. Garcia (2006), Borderas (2008), and von
Keyserlingk (2009)
Drinking Sisto (2002), Meyer et al. (2004), Pinheiro
6 et al. (2004), Garcia (2006), and Manteca
(2009)
Gentle handler Feeding and drinking Rushen (2012)
5 Aversive handler automated measures
Cross-suckling Spinka (1992), Haley et al. (1998),
4 Rushen and de Pasillé (1992, 1995),
de Pasillé et al. (1997), de Pasillé and
Caza (1997), de Pasillé and Rushen
Meters

3
(1998), Jung and Lidfords (1999), and
de Pasillé (2001)
2 Lying, resting Haley et al. (2000), Sisto (2002), Manteca
(2009), Cooper et al. (2007), and Bewley
1 (2011)
Standing Wierenga and Hopster (1990), Sisto (2002),
Nordlund et al. (2003), and Arney and Alan
0 (2012)
Perching Metz and Wierenga (1987), Potter and Broom
Figure 19 Flight distance in the presence of a gentle or an aversive (1987), and Galindo et al. (2000)
handler after 19 times of handling. Adapted from Rushen, J., de Pasillé, Stereotypies Sambraus (1985), Seo et al. (1998), Bashaw
A.M., von Keyserlingk, M.A.G., et al., 2008. The Welfare of Cattle. et al. (2001), and Bergeron et al. (2006)
Springer, The Netherlands.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 479

Table 7 Examples of health indicators used in dairy cattle welfare always consider each species’ needs, for example, isolation, nest
building. To what an extent is this a welfare concern? Recom-
Passive immunity Barrier et al. (2013)
mendations for ‘optimal’ lodging of mammals of different
Lymphocytes Friend et al. (1985a,b) and Sisto and Friend (2001)
reproductive states need to balance the impact of, e.g., type
Lymphocyte/ Friend et al. (1985a,b)
neutrophil of cage, animal density, ‘enriched’ environment versus its
Diarrhea Flower and Weary (2003) consequences on other important issues, such as hygiene,
Lameness Galindo et al. (2000), Phillips and Morris (2000), fertility, ‘peaceful cohabitation.’
Somers et al. (2003), Nordlund (2003), Bergsten An important finding that has consistently emerged across
(2003), Loberg et al. (2004), Regula et al. (2004), species is the existence of the so-called ‘critical periods’ or
Telezhenko and Bergsten (2005), and Espejo ‘windows of opportunity,’ i.e., particular times in the early
et al. (2006) postnatal period during which specific manipulations of
Mastitis Somers et al. (2003) the young can permanently alter their reactivity to poten-
Injuries Nordlund et al. (2003)
tially stressful stimuli in adulthood. In rodents this is a partic-
ularly intense area of research, and important advances have
been made on the subcellular mechanisms underlying this
Table 8 Examples of other indicators of well-being used in dairy phenomenon (Champagne et al., 2003; Fleming et al.,
cattle 1999; Lomanowska and Melo, 2016). By contrast, few
studies have been done on this issue in nonrodent mammals
Weight gain Flower and Weary (2003) (Jensen, 2014; Rodenburg, 2014). Much work is needed to
Precollected registered data, such as Algiers (2009)
define the duration of ‘critical periods’ in particular species
percentage of cows with repeated
inseminations, stillbirths, or mortality
and the type of early-life manipulations that effectively
reduce the stress generated by handling, transportation,
lodging, etc. On the other hand, practices like cross-
fostering in bovines, sheep, and goats provide evidence of
1.17.6 Conclusions and Discussion plasticity regarding social preferences in later life. From this
information management strategies can be implemented to
Throughout this chapter we have reviewed the ways by which reduce stress responses to, e.g., moving flocks, mixing
lodging, environmental conditions, and handling can impact animals in pens, transportation, etc. Moreover, bonding
the well-being and stress reactivity of rodents, rabbits, goats, with humans is well known to occur in several domesticated
sheep, and dairy cattle kept in the laboratory or on the farm. mammals, and this phenomenon can also be used to facili-
Although the studies performed are numerous and varied, tate management in the lab and on the farm. Furthermore,
when viewed as a whole the field seems to be lacking in unifor- animal–human bonding is a fascinating topic in itself and
mity of the work performed; allowing a multiplicity of interpre- the neurobiological mechanisms underlying it should be
tations in some findings; concentrating on a particular age, sex, investigated in their own right.
or reproductive condition; rarely including both behavioral An important consideration should be given to the housing
and neuroendocrine indicators in the same work. For instance, and management of recently weaned and ‘adolescent’ animals.
what do the results of the open-field test mean.reduced fear- Specific issues such as individual versus group housing, type of
fulness? Larger motivation to explore? How are these possible feed, size of cages (or pens), mixing with adults, etc. have been
interpretations related to welfare? How about the display of scarcely addressed in ‘classic’ Behavioral Neuroendocrinology.
stereotypies? Are they indicative of stress or boredom? They Yet, we know that (peri) adolescent mammals are a category
are not always displayed in association with increased CORT. in their own right and that experiences during such period
Moreover, is boredom a psychological stressor (Panksepp, can modify a variety of behaviors and neuroendocrine
2011)? Similar reflections can be made regarding studies in responses in adulthood (Schulz et al., 2009).
which animals are ‘asked’ to choose between two conditions. A puzzling, yet consistent, phenomenon emerging from
For instance, if an animal ‘prefers’ a certain type of bedding the need to help animals deal with environmental stress is
or cage, does that indicate that ‘the other’ choices are not in the effect of the so-called ‘nutritional supplements.’ A variety
accord with the animal’s well-being? of them have been tried, several of them do reduce
The social organization of a mammalian species has immunological and endocrine parameters indicative of heat
a profound impact on the type of lodging that is ‘optimal’ for stress, but little is known regarding their mechanism of
it in the lab and on the farm. Most species housed therein are action. Clearly, this can be a fruitful area of research in
social, and one should rethink if ‘larger is better’; that is, space Psychoneuroimmunoendocrinology.
allowance should consider the species’ habits which, in turn, It is essential to ‘bridge’ from lab to farm and back. The
are modulated by factors such as sex and reproductive state. experimental models, environmental conditions, and findings
Female rabbits, cows, and goats are very social, but when it are complementary to each other and mutually enriching.
comes to ‘acting maternal,’ they retreat and they do not Sadly, the ‘compartmentalization’ of science (i.e., ‘basic vs
welcome encounters with other males or females. Indeed, applied’ research; ‘mainstream’ vs ‘local’ journals; evermore
maternal behavior is always ‘peculiar,’ in the sense that the specialized scientific meetings) does not favor such interaction.
activities performed by mothers – and aimed at their offspring Many issues related to the well-being of farm and laboratory
– are unique to the lactation period (and late pregnancy). Yet, animals are rich topics for investigation in Behavioral Neuroen-
the setups provided for lactating mothers and offspring do not docrinology. The authors hope that, from reading our chapter,
480 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

students and researchers will become interested in exploring Bell, D.J., Bray, G.C., 1984. Effects of single- and mixed-sex caging on postweaning
new aspects of the animals they already work with or venture development in the rabbit. Lab. Anim. 18, 267–270.
Benson, G.J., Rollin, B.E., 2004. The Well-being of Farm Animals. Blackwell, London.
into establishing connections with investigators that study
Bergeron, R., Badnell-Waters, A.J., Lambton, S., et al., 2006. Stereotypic oral
the same species, but from a different perspective. behaviour in captive ungulates: foraging, diet and gastrointestinal function.
Stereotypic Anim. Behav. Fundam. Appl. Welf. 2, 19–41.
Bergsten, C., 2003. Causes, risk factors, and prevention of laminitis and related claw
Acknowledgment lesions. Acta Veterinaria Scand. 98 (Suppl. 3), 157–166.
Bewley, J., 2011. DeLaval. Milkproduction.com.
Bewley, J., Palmer, R.W., Jackson-Smith, D.B., 2001. An overview of experience of
The authors thank Francisco Torres Quintanar for his generous help in Wisconsin dairy farmers who modernized their operations. J. Dairy Sci. 84,
the preparation of figures. 717–729.
Bilkó, A., Altbäcker, V., 2000. Regular handling early in nursing period eliminates fear
response toward human beings in wild and domestic rabbits. Dev. Psychobiol. 36,
78–87.
References Bøe, K.E., Andersen, I.L., Buisson, L., 2007. Flooring preferences in dairy goats at
moderate and low ambient temperature. Appl. Anim. Behav. Sci. 108, 45–57.
Abou-Ismail, U.A., Mahboub, H.D., 2011. The effects of enriching laboratory cages Bøe, K.E., Berg, S., Andersen, I.L., 2006. Resting behaviour and displacement in
using various physical structures on multiple measures of welfare in singly-housed ewes-effects of reduced lying space and pen shape. Appl. Anim. Behav. Sci. 98,
rats. Lab. Anim. 45, 145–153. 249–259.
Abou-Ismail, U.A., Mohamed, R.A., El-Kholya, S.Z., 2015. The effects of witnessing Boissy, A., Manteuffel, G., Bak Jensen, M., et al., 2007. Assessment of positive
managemental procedures during the light versus the dark phase of the light cycle emotions in animals to improve their welfare. Physiol. Behav. 92, 375–397.
on behaviour, performance and welfare of laboratory rats. Appl. Anim. Behav. Sci. Boivin, X., Braastad, B.O., 1996. Effects of handling during temporary isolation after
162, 47–57. early weaning on goat kids’ later response to humans. Appl. Anim. Behav. Sci.
Akre, A.K., Bakken, M., Hovland, A.L., Palme, R., Mason, G., 2011. Clustered envi- 48, 61–71.
ronmental enrichments induce more aggression and stereotypic behaviour than do Boivin, X., Nowak, R., Després, G., et al., 1997. Discrimination between shepherds by
dispersed enrichments in female mice. Appl. Anim. Behav. Sci. 131, 145–152. lambs reared under artificial conditions. J. Anim. Sci. 75, 2892–2898.
Albright, J., 1983. Feeding behavior of dairy cattle. J. Dairy Sci. 76, 485–498. Borderas, T.F., Rushen, J., de Passillé, A.M., 2008. Behavioral effects of endotoxin-
Alexander, G., Shillito, E.E., 1977. The importance of odour, appearance and voice in induced fever in dairy calves. J. Anim. Sci. 86, 2920–2927.
maternal recognition of the young in Merino sheep (Ovis aries). Appl. Anim. Ethol. Bouissou, M.F., Boissy, A., Le Neindre, P., et al., 2001. The social behaviour of cattle.
3, 127–135. In: Keeling, L., Gonyou, H. (Eds.), Social Behaviour in Farm Animals. CABI
Alfonso-Carrillo, C., Martín, E., De Blas, C., et al., 2014. Effect of cage type on the Publishing, Wallingford, UK, pp. 113–145.
behaviour pattern of rabbit does at different physiological stages. World Rabbit Sci. Brecchia, G., Bonanno, A., Galeati, G., et al., 2006. Hormonal and metabolic adap-
22, 59–69. tation to fasting: effects on the hypothalamic- pituitary-ovarian axis and repro-
Algers, B., Blokhuis, H.J., Botner, A., et al., 2009. Scientific opinion on welfare of dairy ductive performance of rabbit does. Domest. Anim. Endocrinol. 31, 105–122.
cows in relation to metabolic and reproductive problems based on a risk Brecchia, G., Bonanno, A., Dall’Aglio, C., et al., 2009. Neuroendocrine responses in
assessment with special reference to the impact of housing, feeding, management neonatal mother-deprived rabbits. Brain Res. 1304, 105–112.
and genetic selection. Scientific Opinion of the Panel on Animal Health and Animal Briefer, E.F., McElligott, A.G., 2011. Mutual mother–offspring vocal recognition in an
Welfare. EFSA J. 1140, 1–75. ungulate hider species (Capra hircus). Anim. Cogn. 14, 585–598.
Alvariño, J.M.R., Del Arco, J.A., Bueno, A., 1998. Effect of mother– litter separation on Briefer, E.F., Padilla de la, T., McElligot, A.G., 2012. Mother goats do not forget their
reproductive performance of lactating rabbit females inseminated on day 4 or 11 kids’ calls. Proc. R. Soc. B Biol. Sci. 279, 3749–3755.
post-partum. World Rabbit Sci. 6, 191–194. Broom, D.M., 1991. Animal welfare: concepts and measurement. J. Anim. Sci. 69,
Andersen, I.L., Roussel, S., Ropstad, E., et al., 2008. Social instability increases 4167–4175.
aggression in groups of dairy goats, but with minor consequences for the goats’ Broom, D.M., Leaver, J.D., 1978. The effects of group housing or partial isolation on
growth, kid production and development. Appl. Anim. Behav. Sci. 114, later social behaviour in calves. Anim. Behav. 26, 1255–1263.
132–148. Buijs, S., Keeling, L.J., Tuyttens, F.A.M., 2001. Behaviour and use of space in
Anderson, C.O., Denenberg, V.H., Zarrow, M.X., 1973. Effects of handling and social fattening rabbits as influenced by cage size and enrichment. Appl. Anim. Behav.
isolation upon the rabbit’s behavior. Behaviour 43, 165–175. Sci. 134, 229–238.
Andrist, C.A., Bigler, L., Würbel, H., et al., 2012. Effects of group stability on Buijs, S., Tuyttens, F.A.M., 2015. Evaluating the effect of semi-group housing of rabbit
aggression, stress and injuries in breeding rabbits. Appl. Anim. Behav. Sci. 142, does on their offspring’s fearfulness: can we use the open-field test? Appl. Anim.
182–188. Behav. Sci. 162, 58–66.
Andrist, C.A., Bigler, L., Würbel, H., et al., 2014. Masking odour when regrouping Carbajal, S., Orihuela, A., 2001. Minimal number of conspecifics needed to minimize
rabbit does: effect on aggression, stress and lesions. Livest. Sci. 170, 150–157. the stress response of isolated mature ewes. J. Appl. Anim. Welf. Sci. 4,
Argüelloa, A., Castroa, N., Zamoranoa, M.J., et al., 2004. Passive transfer of immunity 249–255.
in kid goats fed refrigerated and frozen goat colostrum and commercial sheep Caroprese, M., 2008. Sheep housing and welfare. Small Ruminant Res. 76, 21–25.
colostrum. Small Ruminant Res. 54, 237–241. Caroprese, M., Annicchiarico, G., Schena, L., et al., 2009a. Influence of space
Arney, A., Aland, A., 2012. Contemporary issues in farm animal housing and allowance and housing conditions on the welfare, immune response and production
management cattle housing and welfare. In: Jakobsson, C. (Ed.), Sustainable performance of dairy ewes. J. Dairy Res. 76, 66–73.
Agriculture, Ecosystem Health and Sustainable Agriculture Book, vol. 1. The Baltic Caroprese, M., Casamassima, D., Rassu, S.P.G., et al., 2009b. Monitoring the on-farm
University Programme, Uppsala, pp. 324–329. welfare of sheep and goats. Ital. J. Anim. Sci. 8 (Suppl. 1), 343–354.
Barrier, A.C., Haskell, M.J., Birch, S., et al., 2013. The impact of dystocia on dairy calf Castellini, C., Canali, C., Boiti, C., 1998. Effect of mother–litter separation for 24 h, by
health, welfare, performance and survival. Vet. J. 195, 86–90. closing the nestbox or change of cage, on rabbit doe reproduction performance.
Barrier, A.C., Ruelle, E., Haskell, M.J., et al., 2012. Effect of a difficult calving on the World Rabbit Sci. 6, 199–203.
vigour of the calf, the onset of maternal behaviour, and some behavioural indicators Champagne, F.A., Francis, D.D., Mar, A., Meaney, M.J., 2003. Variations in maternal
of pain in the dam. Prev. Vet. Med. 103, 248–256. care in the rat as a mediating influence for the effects of environment on devel-
Bashaw, J.M., Tarou, L.R., Maki, T.S., et al., 2001. A survey assessment of variables opment. Physiol. Behav. 79, 359–371.
related to stereotypy in captive giraffe and okapi. Appl. Anim. Behav. Sci. 73, Chemineau, P., Malpaux, B., Delgadillo, J.A., et al., 1992. Control of sheep and goat
235–247. reproduction: use of light and melatonin. Anim. Reprod. Sci. 30, 157–184.
Bautista, A., Drummond, H., Martínez-Gómez, M., et al., 2003. Thermal benefit of Close, B., Banister, K., Baumans, et al., 1997. Euthanasia of Experimental Animals.
sibling presence in the newborn rabbit. Dev. Psychobiol. 43, 208–215. European Commission. Office for Official Publications of the European Communi-
Bautista, A., García-Torres, E., Martínez-Gómez, M., et al., 2008. Do newborn ties, Luxembourg.
domestic rabbits Oryctolagus cuniculus compete for thermally advantageous Cloutier, S., Panksepp, J., Newberry, R.C., 2012. Playful handling by caretakers reduces
positions in the litter huddle? Behav. Ecol. Sociobiol. 62, 331–339. fear of humans in the laboratory rat. Appl. Anim. Behav. Sci. 140, 161–171.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 481

Cloutier, S., Wahl, K.L., Panksepp, J., et al., 2015. Playful handling of laboratory rats is Flint, M., Murray, P.J., 2001. Lot-fed goats - the advantages of using an enriched
more beneficial when applied before than after routine injections. Appl. Anim. environment. Aust. J. Exp. Agric. 41, 473–476.
Behav. Sci. 164, 81–90. Flower, F.C., Weary, D.M., 2001. Effects of early separation on the dairy cow and calf:
Cockram, M.S., Ranson, M., Goddard, P.J., et al., 1994. The behavioural, endocrine 2. Separation at 1 day and 2 weeks after birth. Appl. Anim. Behav. Sci. 70,
and immune responses of sheep to isolation. Anim. Sci. 58, 389–399. 275–284.
Comin, A., Veronesi, M.C., Montillo, M., et al., 2012. Hair cortisol level as a retro- Flower, F.C., Weary, D.M., 2003. The effects of early separation on the dairy cow and
spective marker of hypothalamus-pituitary-adrenal axis activity in horse foals. calf. Anim. Welf. 12, 339–348.
Veterinary Rec. 194, 131–132. Friend, T., Dellmeier, G., 1988. Common practices and problems related to artificially
Cook, C.J., 2004. Stress induces CRF release in the paraventricular nucleus, and both rearing calves: an ethological analysis. Appl. Anim. Behav. Sci. 20, 47–62.
CRF and GABA release in the amygdala. Physiol. Behav. 82, 751–762. Friend, T.H., Dellmeier, G.R., Gbur, E.E., 1985a. Comparison of four methods of calf
Cooper, M.D., Arney, D.R., Phillips, C.J.C., 2007. Two- or four- hour lying deprivation confinement. I. Physiology. J. Anim. Sci. 60, 1095–1101.
on the behaviour of lactating dairy cows. J. Dairy Sci. 90, 1149–1158. Friend, T.H., Dellmeier, G.R., Gbur, E.E., 1985b. Comparison of four methods of calf
Coulon, M., Nowak, R., Andanson, S., Petit, B., Levy, F., Boissy, A., 2015. Effects of confinement. II. Behavior. J. Anim. Sci. 60, 1102–1109.
prenatal stress and emotional reactivity of the mother on emotional and cognitive de la Fuente, J., Díaz, M.T., Ibáñez, M., et al., 2007. Physiological response of
abilities in lambs. Dev. Psychobiol. 57, 626–636. rabbits to heat, cold, noise and mixing in the context of transport. Anim. Welf.
Dal Bosco, A., Mugnai, C., Castellini, C., et al., 2004. A prototype of colony cage for 16, 41–47.
improving the welfare of rabbit does: preliminary results. In: Proceedings of the 8th Galindo, F.A., Broom, D.M., 1993. The relationships between social behaviour of dairy
World Rabbit Congress, pp. 1229–1234. cows and the occurrence of lameness. Cattle Pract. 1, 360–364.
de Pasillé, A.M., Rushen, J., Jensen, M., 1997. Some aspects of milk that elicit non- Galindo, F.A., Broom, D.M., 2000. The relationships between social behaviour of dairy
nutritive sucking in the calf. Appl. Anim. Behav. Sci. 53, 167–173. cows and the occurrence of lameness in three herds. Res. Vet. Sci. 69, 75–79.
DEFRA, 2000. Code of Recommendations for the Welfare of Livestock. Sheep. Galindo, F., Broom, D.M., Jackson, P.G.G., 2000. A note on possible link between
Department for Environment Food and Rural Affairs, London. behavior and the occurrence of lameness in dairy cows. Appl. Anim. Behav. Sci.
Directive, 2010. Directive 2010/63/EU of the European Parliament and of the Council 67, 335–341.
of 22 September 2010 on the protection of animals used for scientific purposes. Garcia, A., 2006. Dealing with heat stress in dairy cows. Ext. Extra (ExEx) 4024, 1–3.
Off. J. Eur. Union L276/33–L276/78. García-Dalmán, C., González-Mariscal, G., 2012. Major role of suckling stimulation for
Dixon, L.M., Hardiman, J.R., Cooper, J.J., 2010. The effects of spatial restriction on inhibition of estrous behaviors in lactating rabbits: acute and chronic effects. Horm.
the behavior of rabbits (Oryctolagus cuniculus). J. Vet. Behav. 5, 302–308. Behav. 61, 108–113.
Donovan, G.A., 1992. Management of cow and newborn calf at calving. In: Van Gaudin, S., Chaillou, E., Cornilleau, F., et al., 2015. Daughters are more strongly
Horn, H.H., Wilcox, C.J. (Eds.), Large Dairy Herd Management. American Dairy attached to their mother than sons: a possible mechanism for early social
Science Association, Champaign, IL. segregation. Anim. Behav. 102, 33–43.
Dúcs, A., Bilkó, Á., Altbácker, V., 2009. Physical contact while handling is not GBPO (Guide des Bonnes Pratiques Ovines), 2011. Un guide de conseils et de
necessary to reduce fearfulness in the rabbit. Appl. Anim. Behav. Sci. 121, 51–54. recommandations pour une démarche de progrès. Chambre d’Agriculture de la
Duvaux-Ponter, C., Roussel, S., Tessier, J., et al., 2003. Physiological effects of Nievre, Nevers. http://www.nievre.chambagri.fr/uploads/media/GUIDE_BONNES_
repeated transport in pregnant goats and their offspring. Anim. Res. 52, 553–566. PRATIQUES_01.pdf.
Dwyer, C.M., 2008a. The welfare of the neonatal lamb. Small Ruminant Res. 76, 31–41. Gerencsér, Z., Matics, Z., Nagy, I., et al., 2011. Effect of lighting schedule on
Dwyer, C.M., 2008b. Environment and the sheep. Breed adaptations and welfare production of rabbit does. World Rabbit Sci. 19, 209–216.
implication. In: Dwyer, C. (Ed.), The Welfare of Sheep. Springer Science & Business Gerencsér, Z., Szendrö, K., Szendrö, Z., et al., 2014. Effect of floor type on behavior
Media, University of Queensland, Australia, pp. 41–79. and productive performance of growing rabbits. Livest. Sci. 165, 114–119.
Dwyer, C.M., 2014. Maternal behavior and lamb survival: from neuroendocrinology to Giral, M., García-Olmo, D.C., Kramer, K., 2011. Effects of wire-bottom caging on heart
practical application. Animal 8, 102–112. rate, activity and body temperature in telemetry-implanted rats. Lab. Anim. 45,
Dwyer, C.M., Conington, J., Corbiere, F., et al., 2016. Improving neonatal survival in 247–253.
small ruminants: science into practice. Animal 10, 449–459. Gonyou, H., Stookey, J.M., 1985. Behavior of parturient ewes in group-lambing pens
Dwyer, C.M., Lawrence, A.B., 2005. A review of the behavioural and physiological with and without cubicles. Appl. Anim. Behav. Sci. 14, 163–171.
adaptations of hill and lowland breeds of sheep that favour lamb survival. Appl. González de la Vara, M.R., Valdez, R.A., Lemus, V., et al., 2011. Effects of adreno-
Anim. Behav. Sci. 92, 235–260. corticotropic hormone challenge and age on hair cortisol concentrations in dairy
El-Hanoun, A.M., Elkomy, A.E., Fares, W.A., et al., 2014. Impact of royal jelly to cattle. Can. J. Vet. Res. 75, 216–221.
improve reproductive performance of male rabbits under hot summer conditions. González-Mariscal, G., Caba, M., Martínez-Gómez, M., et al., 2016. Mothers and
World Rabbit Sci. 22, 241–248. offspring: the rabbit as a model system in the study of mammalian maternal
Emmering, H., 2004. Behavioural Indicators of Good Welfare in Dairy Cows: An behavior and sibling interactions. Horm. Behav. 77, 30–41.
Exploratory Approach (Ph.D. thesis). University of Kassel, Germany. González-Mariscal, G., García-Dalmán, C., Jiménez, A., 2015. Biostimulation and
Engel, A.K.J., Gross, A.N., Richter, S.H., et al., 2011. Variation in stress reactivity nursing modify mating-induced c-FOS immunoreactivity in the female rabbit
affects cage-induced stereotypies in female CD-1 (ICR) mice. Appl. Anim. Behav. forebrain. Brain Res. 1608, 66–74.
Sci. 133, 101–108. González-Mariscal, G., Lemus, A.C., Vega-González, A., et al., 2013a. Litter size
Escribano, B., Quero, I., Feijóo, M., et al., 2014. Role of noise and music as anxiety determines circadian periodicity of nursing in rabbits. Chronobiol. Int. 30,
modulators: relationship with ovarian hormones in the rat. Appl. Anim. Behav. Sci. 711–718.
152, 73–82. González-Mariscal, G., McNitt, J.I., Lukefahr, S.D., 2007. Maternal care of rabbits in
Espejo, L.A., Endres, M.I., Salfer, J.A., 2006. Prevalence of lameness in high- the lab and on the farm: endocrine regulation of behavior and productivity. Horm.
producing Holstein cows housed in freestall barns in Minnesota. J. Dairy Sci. Behav. 52, 86–91.
89, 3052–3058. González-Mariscal, G., Toribio, A., Gallegos-Huicochea, J.A., et al., 2013b. The
Fabre-Nys, C., Gelez, H., 2007. Sexual behavior in ewes and other domestic rumi- characteristics of suckling stimulation determine the daily duration of mother-young
nants. Horm. Behav. 52, 18–25. contact and milk output in rabbits. Dev. Psychobiol. 55, 809–817.
Færevik, G., Andersen, I.L., Bøe, K.E., 2005. Preferences of sheep for different types Grandin, T. (Ed.), 1998. Genetics and the Behavior of Domestic Animals. Academic
of pen flooring. Appl. Anim. Behav. Sci. 90, 265–276. Press, San Diego.
Fazio, F., Casella, S., Giudice, E., et al., 2015. Evaluation of secondary stress Gregory, N.G., 1995. The role of shelterbelts in protecting livestock: a review. N.Z. J.
biomarkers during road transport in rabbit. Livest. Sci. 173, 106–110. Agric. Res. 38, 423–450.
Fenske, M., Fuchs, E., Probst, B., 1982. Corticosteroid and catecholamine plasma Gross, A.N., Engel, A.K.J., Würbel, H., 2011. Simply a nest? Effects of different
levels in rabbits stressed repeatedly by exposure to a novel environment or by enrichments on stereotypic and anxiety-related behaviour in mice. Appl. Anim.
injection of (1-24) ACTH or insulin. Acta Endocrinol. 99 (Suppl. 246), 110. Behav. Sci. 134, 239–245.
Ferreira, G., Terrazas, A., Poindron, P., et al., 2000. Learning of olfactory cues is not Gross, A.N., Richter, S.H., Engel, A.K.J., et al., 2012. Cage-induced stereotypies,
necessary for early lamb recognition by the mother. Physiol. Behav. 69, 405–412. perseveration and the effects of environmental enrichment in laboratory mice.
Fleming, A.S., O’Day, D.H., Kraemer, G.W., 1999. Neurobiology of mother–infant Behav. Brain Res. 234, 61–68.
interactions: experience and central nervous system plasticity across development Gubernick, D.J., 1981. Maternal “imprinting” or maternal “labelling” in goats. Anim.
and generations. Neurosci. Biobehav. Rev. 23, 673–685. Behav. 28, 124–129.
482 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

Guerrero-Casado, J., Ruiz-Aizpurua, L., Carpio, A.J., et al., 2013. Factors affecting Kendrick, K.M., 2008. Sheep senses, social cognition and capacity for consciousness.
wild rabbit production in extensive breeding enclosures: how can we optimise In: Dwyer, C. (Ed.), The Welfare of Sheep. Springer Science & Business Media,
efforts? World Rabbit Sci. 21, 193–199. University of Queensland, Australia, pp. 135–157.
Guesdon, V., Ligout, S., Delagrange, P., et al., 2012. Multiple exposures to familiar Kendrick, K.M., Haupt, M.A., Hinton, M.R., et al., 2001. Sex differences in the
conspecific withdrawal is a novel robust stress paradigm in ewes. Physiol. Behav. influence of mothers on the sociosexual preferences of their offspring. Horm.
105, 203–208. Behav. 40, 322–338.
Hagen, K., Langbein, J., Schmied, C., et al., 2005. Heart rate variability in dairy cows – Kersten, A.M.P., Meijsser, F.M., Metz, J.H.M., 1989. Effect of early handling on later
influences of breed and milking system. Physiol. Behav. 85, 195–204. open-field behaviour of rabbits. Appl. Anim. Behav. Sci. 24, 157–167.
Haley, D., Rushen, J., Duncan, I., et al., 1998. Effects of resistance to milk flow and Kilgour, R.J., Waterhouse, T., Dwyer, C.M., et al., 2008. Farming systems for sheep
provision of hay on non-nutritive sucking by dairy calves. J. Dairy Sci. 81, production and their effect on welfare. In: Dwyer, C. (Ed.), The Welfare of Sheep.
2165–2172. Springer Science & Business Media, University of Queensland, Australia,
Haley, D.B., Rushen, J., de Passillé, A.M., 2000. Behavioural indicators of cow pp. 213–265.
comfort: activity and resting behaviour of dairy cows in two types of housing. Can. Kilgour, R., Dalton, C., 1984. Livestock Behaviour: A Practical Guide. HarperCollins
J. Anim. Sci. 80, 257–263. Publishers Ltd, Granada: London.
Harkness, J.E., 1987. Rabbit behavior as related to environmental stress. J. Appl. Kirchner, J., Hackbarth, H., Stelzer, H.D., et al., 2012. Preferences of group-housed
Rabbit Res. 10, 227–236. female mice regarding structure of softwood bedding. Lab. Anim. 46, 95–100.
Hashem, N.M., Abd El-Hady, A., Hassan, O., 2013. Effect of vitamin E or propolis Kraft, V., Deeny, A.A., Blanchet, H.M., et al., 1994. Recommendations for the health
supplementation on semen quality, oxidative status and hemato-biochemical monitoring of mouse, rat, hamster, guinea pig and rabbit breeding colonies. Lab.
changes of rabbit bucks during hot season. Livest. Sci. 157, 520–526. Anim. 28, 1–12.
Hassan, R.A., Ebeid, T.A., Abd El-Lateif, A.I., et al., 2011. Effect of dietary betaine Krohn, T.C., Hansen, A.K., 2010. Mice prefer draught-free housing. Lab. Anim. 44,
supplementation on growth, carcass and immunity of New Zealand White rabbits 370–372.
under high ambient temperature. Livest. Sci. 135, 103–109. Lang, C., Hoy, S., 2011. Investigations on the use of an elevated platform in group
Hawkins, P., 2002. Recognizing and assessing pain, suffering and distress in labo- cages by growing rabbits. World Rabbit Sci. 19, 95–101.
ratory animals. A survey of current practice in the UK with recommendations. Lab. Leary, S., Underwood, W., Anthony, R., et al., 2013. AVMA Guidelines for Euthanasia
Anim. 36, 378–395. of Animals. American Veterinary Medical Association, Schaumburg, Illinois.
Hawkins, P., 2011. A guide to defining and implementing protocols for the welfare Lickliter, R.E., 1984. Hiding behavior in domestic goat kids. Appl. Anim. Behav. Sci.
assessment of laboratory animals: eleventh report of the BVAAWF/FRAME/RSPCA/ 12, 245–251.
UFAW joint working group on refinement. Lab. Anim. 45, 1–13. Lidfors, L.M., 1996. Behavioural effects of separating the dairy calf immediately or
Hopstee, H., O’Connell, J.M., Harry, J., 1995. Acute effects of cow-calf separation on 4 days post-partum. Appl. Anim. Behav. Sci. 49, 269–283.
heart rate, plasma cortisol and behaviour in multiparous dairy cows. Appl. Anim. Loberg, J., Telezhenko, E., Bergsten, C., et al., 2004. Behaviour and claw health in
Behav. Sci. 44, l–8. tied dairy cows with varying access to exercise in an outdoor paddock. Appl. Anim.
Hopster, H., van der Werf, J., Erkens, J., Blokhuis, H., 1999. Effects of repeated Behav. Sci. 89, 1–16.
jugular puncture on plasma cortisol concentrations in loose-housed dairy cows. J. Lomanowska, A.M., Melo, A.I., 2016. Deconstructing the function of maternal stim-
Anim. Sci. 77, 708–714. ulation in offspring development: insights from the artificial rearing model in rats.
Horton, G.M.J., Malinowski, K., Burgher, C.C., et al., 1991. The effect of space Horm. Behav. 77, 224–236.
allowance and sex on blood catecholamines and cortisol, feed consumption and Loretz, C., Wechsler, B., Hauser, R., et al., 2004. A comparison of space requirements
average daily gain in growing lambs. Appl. Anim. Behav. Sci. 32, 197–204. of horned and hornless goats at the feed barrier and in the laying area. Appl. Anim.
Houpt, K., 2005. Domestic Animal Behavior. Blackwell Publishing, Iowa. Behav. Sci. 87, 275–283.
Hudson, R., Distel, H., 1982. The pattern of behaviour of rabbit pups in the nest. Luzi, F., Ludwig, N., Gargano, M., et al., 2007. Evaluation of skin temperature change
Behaviour 79, 255–271. as stress indicator in rabbit through infrared thermography. Ital. J. Anim. Sci. 6
Hudson, S., Mullord, M., 1977. Investigations of maternal bonding in dairy cattle. Appl. (Suppl. 1), 769.
Anim. Ethol. 3, 271–276. Maertens, L., Buijs, S., Davoust, C., 2013. Gnawing blocks as cage enrichment and
Ingvartsen, K.L., Dewhurst, R.J., Friggens, N.C., 2003. On the relationship between dietary supplement for does and fatteners: intake, performance and behaviour.
lactational performance and health: is it yield or metabolic imbalance that cause World Rabbit Sci. 21, 185–192.
production diseases in dairy cattle? A position paper. Livest. Prod. Sci. 83, Maertens, L., Tyuttens, F., Van Poucke, E., 2004. Group housing of broiler rabbits:
277–308. performances in enriched vs barren pens. In: Proceedings of the 8th World Rabbit
Jackson, E., Demarest, K., Eckert, W.J., et al., 2015. Aspen shaving versus chip Congress, pp. 1247–1250.
bedding: effects on breeding and behavior. Lab. Anim. 49, 46–56. Makowska, I.J., Weary, D.M., 2013. Assessing the emotions of laboratory rats. Appl.
Jensen, P., 2014. Behaviour epigenetics – The connection between environment, Anim. Behav. Sci. 148, 1–12.
stress and welfare. Appl. Anim. Behav. Sci. 157, 1–7. Malmkvist, J., Palme, R., 2015. Early transfer of mated females into the maternity unit
Jensen, M.B., 2003. The effects of feeding method, milk allowance and social factors reduces stress and increases maternal care in farm mink. Appl. Anim. Behav. Sci.
on milk feeding behaviour and cross-sucking in grouped housed dairy calves. Appl. 167, 56–64.
Anim. Behav. Sci. 80, 191–206. Manteca, X., 2009. Etología Veterinaria. Multimédicas Ediciones Veterinarias, España.
Jezierski, T.A., Konecka, A.M., 1996. Handling and rearing results in young rabbits. Marai, I.F.L., El-Darawanya, A.A., Fadielb, A., et al., 2007. Physiological traits as
Appl. Anim. Behav. Sci. 46, 243–250. affected by heat stress in sheepdA review. Small Ruminant Res. 71, 1–12.
Jones, M.A., Mason, G., Pillay, N., 2011. Early environmental enrichment protects Marai, I.F.M., Habeeb, A.A.M., Gad, A.E., 2002. Rabbits’ productive, reproductive and
captive-born striped mice against later development of stereotypic behaviour. Appl. physiological performance traits as affected by heat stress: a review. Livest. Prod.
Anim. Behav. Sci. 135, 138–145. Sci. 78, 71–90.
Jørgensen, G.H.M., Andersen, I.L., Bøe, K.E., 2007. Feed intake and social interac- Marsden, D., Wood-Gush, D.G.M., 1986a. A note on the behaviour of individually-
tions in dairy goatsdThe effects of feeding space and type of roughage. Appl. penned sheep regarding their use for research purposes. Anim. Prod. 42,
Anim. Behav. Sci. 107, 239–251. 157–159.
Jung, J., Lidfors, L., 1999. Effects of milk amount, milk flow rate and access to Marsden, D., Wood-Gush, D.G.M., 1986b. Abnormal behaviour versus production
a rubber teat on the behaviour of dairy calves. In: Proceedings of the performance criteria in the assessment of welfare in housed lambs. Appl. Anim.
International Symposium on Suckling, Hamra Farm, Alfa Laval Agri, Tumba, Behav. Sci. 15, 90–91.
Sweden, p. 51. Matics, Z., Szendro, Z., Bessei, W., et al., 2004. The free choice of rabbits among
Keil, N.M., Audigé, L., Langhans, W., 2000. Factors associated with intersuckling in identically and differently sized cages. In: Proceedings of the 8th World Rabbit
Swiss dairy heifers. Prev. Veterinary Med. 45, 305–323. Congress, pp. 1251–1256.
Keller, M., Meurisse, M., Lévy, F., 2005. Mapping of brain networks involved in Mc Ewen, B.S., 2004. Protection and damage from acute and chronic stress. In:
consolidation of lamb recognition memory. Neuroscience 133, 359–369. Yehuda, R., Mc Ewen, B. (Eds.), Biobehavioral Stress Response. The New York
Kemppinen, N., Hau, J., Meller, A., et al., 2010. Impact of aspen furniture and Academy of Sciences, New York, pp. 1–7.
restricted feeding on activity, blood pressure, heart rate and faecal corticosterone Meller, A., Kasanen, I., Ruksenas, et al., 2011. Refining cage change routines:
and immunoglobulin A excretion in rats (Rattus norvegicus) housed in individually comparison of cardiovascular responses to three different ways of cage change in
ventilated cages. Lab. Anim. 44, 104–112. rats. Lab. Anim. 45, 167–173.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 483

Mellor, D., 1990. Meeting colostrum needs of newborn lambs. Pract. 12, 242–244. Orova, Z., Szendro, Zs., Matics, Z., et al., 2008. Free choice of growing rabbits
Mellor, D.J., Cockburn, F., 1986. A comparison of energy metabolism in the new-born between deep litter and wire net floor in pens. In: Proceedings of the 8th World
infant, piglet and lamb. Q. J. Exp. Physiol. 71, 361–379. Rabbit Congress, pp. 1263–1265.
Metz, J.H.M., Wierenga, H.K., 1987. Behavioural criteria for the design of housing Palme, R., Robia, C., Baumgartner, W., et al., 2000. Transport stress in cattle as
systems for cattle. In: Cattle Housing Systems, Lameness and Behaviour. Martinus reflected by an increase in faecal cortisol metabolite concentrations. Veterinary
Nijhoff, Dordrecht, pp. 14–25. Rec. 146, 108–109.
Meyer, U., Everinghoff, M., Gädeken, D., et al., 2004. Investigations on the water Panksepp, J., 2011. The basic emotional circuits of mammalian brains: do animals
intake of lactating dairy cows. Livest. Prod. Sci. 90, 117–121. have affective lives? Neurosci. Biobehav. Rev. 35, 1791–1804.
Minton, J.E., 1994. Function of the hypothalamic-pituitary-adrenal axis and the Parrott, R.F., Houpt, K.A., Misson, B.H., 1988. Modification of the responses of sheep to
sympathetic nervous system in models of acute stress in domestic farm animals. isolation stress by the use of mirror panels. Appl. Anim. Behav. Sci. 19, 331–338.
J. Anim. Sci. 72, 1891–1898. Parrott, R.F., Misson, B.H., De La Riva, C.F., 1994. Differential stressor effects on the
Minton, J.E., Coppinger, T.R., Reddy, P.G., et al., 1992. Repeated restraint and concentrations of cortisol, prolactin and catecholamines in the blood of sheep. Res.
isolation stress alters adrenal and lymphocyte functions and some leukocyte Veterinary Sci. 56, 234–239.
differentiation antigens in lambs. J. Anim. Sci. 70, 1126–1132. Parrott, R.F., Hall, S.J.G., Lloyd, D.M., 1998. Heart rate and stress hormone responses
Miranda-de la Lama, G.C., Mattiello, S., 2010. The importance of social behavior for of sheep to road transport following two different loading procedures. Anim. Welf.
goat welfare in livestock. Small Ruminant Res. 90, 1–10. 7, 257–267.
Moberg, G.P., 2000. Biological response to stress: implications for animal welfare. In: de Pasillé, A.M., 2001. Sucking motivation and related problems in calves. Appl. Anim.
Moberg, G.P., Mench, J.A. (Eds.), The Biology of Animal Stress. CAB International, Behav. Sci. 72, 175–187.
New York, pp. 1–21. de Pasillé, A.M., Rushen, J., 1998. Identifying milk components that elicit non-nutritive
Moberg, G.P., Wood, V.A., 1982. Effect of differential rearing on the behavioral and sucking by calves. J. Dairy Sci. 81 (Suppl. 1), 104.
adrenocortical response of lambs to a novel environment. Appl. Anim. Ethol. 8, de Pasillé, A.M., Caza, N., 1997. Cross-sucking by calves occurs alter meals and is
269–279. reduced when calves suck a dry teat. J. Dairy Sci. 80 (Suppl. 1), 229.
Mohr, E., Langbein, J., Nürnberg, G., 2002. Heart rate variability: a non invasive de Pasillé, A.M., Metz, J.H.M., Mekking, P., et al., 1992. Nonnutritive sucking by the
approach to measure stress in calves and cows. Physiol. Behav. 75, 251–259. calf and postprandial secretion of insulin, CCK and gastrin. Physiol. Behav. 54,
Moody, C.M., Chua, B., Weary, D.M., 2014. The effect of carbon dioxide flow rate on 1069–1073.
the euthanasia of laboratory mice. Lab. Anim. 48, 298–304. de Pasillé, A.M., Rushen, J., 1995. Effects of spatial restriction and behavioural
Morrow, C.J., Kolver, E.S., Verkerk, G.A., Matthews, L.R., 2002. Fecal glucocorticoid deprivation on open- field responses, growth and adrenocortical reactivity of calves
metabolites as a measure of adrenal activity in dairy cattle. General Comp. (abstr.). In: Rutter, S.M., Rushen, J., Randle, H.D., et al. (Eds.), Proceedings of the
Endocrinol. 126, 229–241. 29th International Congress of ISAE. UFAW, Potters Bar, UK, p. 207.
Möstl, E., Palme, R., 2002. Hormones as indicators of stress. Domest. Anim. Endo- Pei, Y., Wu, Y., Cao, J., et al., 2012. Effects of chronic heat stress on the reproductive
crinol. 23, 67–74. capacity of male Rex rabbits. Livest. Sci. 146, 13–21.
Mugnai, C., Dal Bosco, A., Cardinali, A., et al., 2014. Effect of pasture availability and Phillips, C., 2008. Cattle Behaviour and Welfare. John Wiley & Sons, pp. 30–37.
genotype on welfare, immune function, performance and meat characteristics of Phillips, C.J.C., Morris, I.D., 2000. The locomotion of dairy cows on concrete floors
growing rabbits. World Rabbit Sci. 22, 29–39. that are dry, wet or covered with a slurry of excreta. J. Dairy Sci. 83, 1767–1772.
Mugnai, C., Dal Bosco, A., Castellini, C., 2009. Effect of different rearing systems and Pinheiro Machado Filho, L.C., Teixeira, D.L., Weary, D.M., et al., 2004. Designing
pre-kindling handling on behaviour and performance of rabbit does. Appl. Anim. better water troughs: dairy cows prefer and drink more from larger troughs. Appl.
Behav. Sci. 118, 91–100. Anim. Behav. Sci. 89, 185–193.
Munksgaard, L., de Passillé, A.M., Rushen, J., et al., 1999. Dairy cows’ use of colour Poindron, P., Gilling, G., Hernandez, H., et al., 2007a. Preference of 12-h-old kids for
cues to discriminate between people. Appl. Anim. Behav. Sci. 65, 1–11. their mother goat is impaired by pre-partum-induced anosmia in the mother.
Mykytowycz, R., 1958a. Social behaviour of an experimental colony of wild rabbits Animal 1, 1328–1334.
Oryctolagus cuniculus (L.). I. Establishment of the colony. CSIRO Wildl. Res. 3, Poindron, P., Lévy, F., Keller, M., 2007b. Maternal responsiveness and maternal
7–25. selectivity in domestic sheep and goats: the two facets of maternal attachment.
Mykytowycz, R., 1958b. Social behaviour of an experimental colony of wild rabbits Dev. Psychobiol. 49, 54–70.
Oryctolagus cuniculus (L.). II. First breeding season. CSIRO Wildl. Res. 4, 1–13. Poindron, P., Nowak, R., Lévy, F., et al., 1993. Development of exclusive mother-
Napolitano, F., De Rosab, G., Sevi, A., 2008. Welfare implications of artificial rearing young bonding in sheep and goats. Oxf. Rev. Reprod. Biol. 15, 311–364.
and early weaning in sheep. Appl. Anim. Behav. Sci. 110, 58–72. Poindron, P., Terrazas, A., de La Luz Navarro Montes de Oca, M., et al., 2007c.
Napolitano, F., Knierim, U., Grasso, F., et al., 2009. Positive indicators of cattle welfare Sensory and physiological determinants of maternal behavior in the goat (Capra
and their applicability to on-farm protocols. Ital. J. Anim. Sci. 8 (Suppl. 1), 355–365. hircus). Horm. Behav. 52, 99–105.
Negretti, P., Alabani, A., Finzi, A., 2004. Location and social behaviour of young rabbit Pollard, J.C., 2006. Shelter for lambing sheep in New Zealand: a review. N.Z. J. Agric.
bucks. In: Proceedings of the 8th World Rabbit Congress, pp. 1257–1262. Res. 49, 395–404.
Nordlund, K., Cook, N.B., 2003. A flowchart for evaluating dairy cow freestalls. Bov. Pongrácz, P., Altbäcker, V., 1999. The effect of early handling is dependent upon the
Pract. 37, 89–93. state of the rabbit (Oryctolagus cuniculus) pups around nursing. Dev. Psychobiol.
Nowak, R., Boivin, X., 2015. Filial attachment in sheep: similarities and differences 35, 241–251.
between ewe-lamb and human-lamb relationships. Appl. Anim. Behav. Sci. 16, Potter, M.J., Broom, D.M., 1987. The behaviour and welfare of cows in relation to
12–28. cubicle house design. In: Wierenga, H.K., Petersen, D.J. (Eds.), Cattle Housing
Nowak, R., Poindron, P., 2006. From birth to colostrum: early steps leading to lamb Systems, Lameness and Behaviour, Current Topics in Veterinary Medicine and
survival. Reprod. Nutr. Dev. 46, 431–446. Animal Science, vol. 40. Martinus Nijhoff, Dordrecht, pp. 129–147.
Nowak, R., Keller, M., Lévy, F., 2011. Mother-young relationships in sheep: a model Price, E.O., Smith, V.M., 1984. The relationship of male-male mounting to mate choice
for a multidisciplinary approach of the study of attachment in mammals. J. Neu- and sexual performance in male dairy goats. Appl. Anim. Behav. Sci. 13, 71–82.
roendocrinol. 23, 1042–1053. Price, E.O., Thos, J., 1980. Behavioral responses to short-term social isolation in
Nowak, R., Porter, R.H., Blache, D., Dwyer, C.M., 2008. Behaviour and the welfare of sheep and goats. Appl. Anim. Ethol. 6, 331–339.
the sheep. In: Dwyer, C. (Ed.), The Welfare of Sheep. Springer Science & Business Price, E.O., Borgwardt, R., Blackshaw, J.K., et al., 1994. Effect of early experience on
Media, University of Queensland, Australia, pp. 135–157. the sexual performance of yearling rams. Appl. Anim. Behav. Sci. 42, 41–48.
Nowak, R., Porter, R.H., Lévy, F., et al., 2000. Role of mother-young interactions in the Rault, J.L., Boissy, A., Boivin, X., 2011. Separation distress in artificially-reared lambs
survival of offspring in domestic mammals. Rev. Reprod. 5, 153–163. depends on human presence and the number of conspecifics. Appl. Anim. Behav.
NRC, 2007. Nutrient Requirements of Small Ruminants: Sheep, Goats, Cervids and Sci. 132, 42–50.
New World Camelids. National Academy Press, Washington, DC. Regula, G., Danuser, J., Spycher, B., et al., 2004. Health and welfare of dairy cows in
Obst, J.M., Day, H.R., 1968. The effect of inclement weather on mortality of Merino different husbandry systems in Switzerland. Prev. Vet. Med. 66, 247–264.
and Corriedale lambs on Kangaroo Island. Proc. Aust. Soc. Anim. Prod. 8, Ribikauskas, V., Ribikauskaskiene_ , D., Skurdeniene_ , I., 2010. Effect of housing system
239–242. (wire cage versus group-housing) and in-house air quality parameters on the
Orgeur, P., Mimouni, P., Signoret, J.P., 1990. The influence of rearing conditions on behaviour of fattening rabbits. World Rabbit Sci. 18, 243–250.
the social relationships of young male goats (Capra hircus). Appl. Anim. Behav. Sci. Riesenfeld, T., Hammarlund, K., Norsted, T., et al., 1996. Irregular breathing in young
27, 105–113. lambs and newborn infants during heat stress. Acta Paediatr. 85, 467–470.
484 Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals

Rivalland, E.T.A., Clarke, I.J., Turner, A.I., et al., 2007. Isolation and restraint stress Spinka, M., Illmann, G., 1992. Suckling behaviour of young dairy calves with their own
results in differential activation of corticotrophin-releasing hormone and arginine and alien mothers. Appl. Anim. Behav. Sci. 33, 165–173.
vasopressin neurons in sheep. Neuroscience 145, 1048–1058. Stehulova, I., Lidfors, L., Spinka, M., 2008. Response of dairy cows and calves to early
Rodenburg, T.B., 2014. The role of genes, epigenetics and ontogeny in behavioural separation: effect of calf age and visual and auditory contact after separation. Appl.
development. Appl. Anim. Behav. Sci. 157, 8–13. Anim. Behav. Sci. 110, 144–165.
Rommers, J.M., de Jong, I.C., 2011. Technical note: plastic mats prevent footpad Stephens, D.B., Adams, C.E., 1982. Observations on the effects of vibration stress and
injuries in rabbit does. World Rabbit Sci. 19, 233–237. sound on pregnancy, parturition and respiration in the rabbit. Lab. Anim. 16,
Rommers, J.M., Boiti, C., De Jong, I., et al., 2006. Performance and behaviour of 341–347.
rabbit does in a group-housing system with natural mating or artificial insemination. Stewart, M., Webster, J.R., Verkerk, G.A., et al., 2007. Non-invasive measurement
Reprod. Nutr. Dev. 46, 677–687. of stress in dairy cows using infrared thermography. Physiol. Behav. 92,
Rommers, J.M., Bracke, M.B.M., Reuvekamp, B., Gunnink, H., de Jong, I.C., 2014. 520–525.
Cage enrichment: rabbit does prefer straw or a compressed wooden block. World Szendrö, Z., McNitt, J.I., Matics, Z., Mikó, A., Gerencsér, Z., 2016. Alternative and
Rabbit Sci. 22, 301–309. enriched housing systems for breeding does: a review. World Rabbit Sci. 24, 1–14.
Roussel, S., Boissy, A., Montigny, D., et al., 2005. Gender-specific effects of prenatal Tallet, C., Veissier, I., Boivin, X., 2005. Human contact and feeding as rewards for the
stress on emotional reactivity and stress physiology of goat kids. Horm. Behav. 47, lamb’s affinity to their stockperson. Appl. Anim. Behav. Sci. 94, 59–73.
256–266. Teilmann, A.C., Kalliokoski, O., Sorensen, D.B., et al., 2014. Manual versus automated
Roussel, S., Hemsworth, P.H., Boissy, A., et al., 2004. Effects of repeated stress during blood sampling: impact of repeated blood sampling on stress parameters and
pregnancy in ewes on the behavioural and physiological responses to stressful events behavior in male NMRI mice. Lab. Anim. 48, 278–291.
and birth weight of their offspring. Appl. Anim. Behav. Sci. 85, 259–276. Telezhenko, E., Bergsten, C., 2005. Influence of floor type on the locomotion of dairy
Rushen, J., 1996. Using aversion learning techniques to assess the mental state, cows. Appl. Anim. Behav. Sci. 93, 183–197.
suffering, and welfare of farm animals. J. Anim. Sci. 74, 1990–1995. Telezhenko, E., Lidfors, L., Bergsten, C., 2007. Dairy cow preferences for soft or hard
Rushen, J., Chapinal, N., de Pasillé, A.M., 2012. Automated monitoring of behavioural- flooring when standing or walking. J. Dairy Sci. 90, 3716–3724.
based animal welfare indicators. Anim. Welf. 21, 339–350. Terrazas, A., Nowak, R., Serafín, N., et al., 2002. Twenty-four-hour-old lambs rely
Rushen, J., de Pasillé, A.M., 1992. The scientific assessment of the impact of housing more on maternal behavior than on the learning of individual characteristics to
on animal welfare: a critical review. Can. J. Anim. Sci. 72, 721–743. discriminate between their own and an alien mother. Dev. Psychobiol. 40,
Rushen, J., de Pasillé, A.M., 1995. The motivation of non –nutritive sucking in calves, 408–418.
Bos taurus. Anim. Behav. 49, 1503–1510. Terrazas, A., Robleto, V., Serafín, N., et al., 2009. Differential effects of undernutrition
Rushen, J., de Passillé, A.M., Munksgaard, L., 1999. Fear of people by cows and during pregnancy on the behaviour of does and their kids at parturition and on the
effects on milk yield, behavior and heart rate at milking. J. Dairy Sci. 82, 720–727. establishment of mutual recognition. Animal 3, 294–306.
Rushen, J., de Pasillé, A.M., von Keyserlingk, M.A.G., et al., 2008. The Welfare of Theau-Clément, M., Mercier, P., 1999. Effect of a doe–litter separation on rabbit
Cattle. Springer, The Netherlands. doe reproductive performance and growth of the young. World Rabbit Sci. 7,
Rushen, J., Munksgaard, L., de Passillé, A.M., et al., 1998. Location of handling and 177–179.
dairy cows’ responses to people. Appl. Anim. Behav. Sci. 55, 259–267. Tilbrook, A.J., Cameron, A.W.N., 1990. The contribution of the sexual behaviour of
Sambraus, H.H., 1985. Mouth-based anomalous syndromes. In: Ethology of Farm rams to successful mating of ewes under field conditions. In: Oldham, C.M.,
Animals. Elsevier, Amsterdam, pp. 391–422. Martin, G.B., Purvis, I.W. (Eds.), Reproductive Physiology of Merino Sheep. The
Sato, S., Hatae, K., Tarumizu, K., 1993. The influence of social factors on allogrooming University of Western Australia, School of Agriculture, Perth, pp. 143–160.
in cows. Appl. Anim. Behav. Sci. 38, 235–244. Trocino, A., Xiccato, G., Queaque, P.I., et al., 2004. Group housing of growing rabbits:
Schulz, K.M., Molenda-Figueira, H.A., Sisk, C.L., 2009. Back to the future: the effect of stocking density and cage floor on performance, welfare, and meat
organizational-activational hypothesis adapted to puberty and adolescence. Horm. quality. In: Proceedings of the 8th World Rabbit Congress, pp. 1277–1282.
Behav. 55, 597–604. Trunkfield, H.R., 1990. The Effects of Previous Housing Experience on Calf Responses
Searby, A., Jouventin, P., 2003. Mother-lamb acoustic recognition in sheep: to Housing and Transport (Ph.D. thesis). University of Cambridge.
a frequency coding. Proc. R. Soc. Ser. B Biol. Sci. 270, 1765–1771. Ubilla, E., Rebollar, P.G., Pazo, D., Esquifino, A., Alvariño, J.M.R., 2000. Effects of
Sejian, V., Srivastava, R.S., 2010. Pineal–adrenal–immune system relationship under doe-litter separation on endocrinological and productivity variables in lactating
thermal stress: effect on physiological, endocrine, and non-specific immune rabbits. Livest. Prod. Sci. 67, 67–74.
response in goats. J. Physiol. Biochem. 66, 339–349. Ungerfeld, R., 2007. Socio-sexual signaling and gonadal function: opportunities for
Selye, H., 1956. The Stress of Life. McGraw Hill, New York. reproductive management in domestic ruminants. J. Fertil. Steril. 64 (Suppl.),
Seo, T., Sato, S., Kosaka, K., et al., 1998. Tongue-playing and heart rate in calves. 207–221.
Appl. Anim. Behav. Sci. 58, 179–182. Van de Weerd, H.A., Van den Broek, F.A.R., Baumans, V., 1996. Preference for
Sevi, A., Caroprese, M., 2012. Impact of heat stress on milk production, immunity and different types of flooring in two rat strains. Appl. Anim. Behav. Sci. 46, 251–261.
udder health in sheep: a critical review. Small Ruminant Res. 107, 1–7. Vasseur, S., Paul, D.R., Atkinson, A.S.J., et al., 2006. Effects of dietary fibre and
Sevi, A., Casamassima, D., Pulina, G., et al., 2009. Factors of welfare reduction in feeding frequency on wool biting and aggressive behaviours in housed Merino
dairy sheep and goats. Ital. J. Anim. Sci. 8 (Suppl. 1), 81–101. sheep. Aust. J. Exp. Agric. 46, 777–782.
Sevi, A., Massa, S., Annicchiarico, G., Dell’Aquila, S., Muscio, A., 1999. Effect of Veissier, I., Boissy, A., 2007. Stress and welfare: two complementary concepts that
stocking density on ewes’ milk yield, udder health and microenvironment. J. Dairy are intrinsically related to the animal’s point of view. Physiol. Behav. 92, 429–433.
Res. 489–499. Verde, M.T., Piquer, J.G., 1986. Effect of stress on the corticosterone and ascorbic
Sherman, B.L., Serpell, J.A., 2008. Training veterinary students in animal behavior to acid (Vitamin C) content of the blood plasma of rabbits. J. Appl. Rabbit Res. 9,
preserve the human–animal bond. Purdue Conference on the Human–Animal 181–185.
J. Vet. Med. Educ. 35, 496–502. Verga, M., Zingarelli, I., Ferrante, V., et al., 2004. Effect of housing and environmental
Sieberta, K., Langbein, J., Schön, P.C., et al., 2011. Degree of social isolation affects enrichment on performance and behaviour in fattening rabbits. In: Proceedings of
behavioural and vocal response patterns in dwarf goats (Capra hircus). Appl. Anim. the 8th World Rabbit Congress, pp. 1283–1288.
Behav. Sci. 131, 53–62. Verwer, C.M., van Merongen, G., van den Bos, R., et al., 2009. Handling effects on
Silanikove, N., 2000. Effects of heat stress on the welfare of extensively managed body weight and behaviour of group housed male rabbits in a laboratory setting.
domestic ruminants. Livest. Prod. Sci. 67, 1–18. Appl. Anim. Behav. Sci. 117, 93–102.
Sisto, A.M., 2002. Effect of Seasons on Coping Strategies of Low and High Producing Vince, M.A., 1993. Newborn lambs and their dams: the interaction that leads to
Dairy Cows (Ph.D. thesis). Texas A&M University, USA. sucking. Adv. Study Behav. 22, 239–268.
Sisto, A.M., Friend, T.H., 2001. The effect of confinement on motivation to exercise in von Keyserlingk, M.A.G., Weary, D.M., 2007. Maternal behavior in cattle. Horm.
young dairy calves. Appl. Anim. Behav. Sci. 73, 83–91. Behav. 52, 106–113.
Somers, J.G.C.J., Frankena, K., Noordhuizen-Stassen, E.N., et al., 2003. Prevalence von Keyserlingk, M.A.G., Rushen, J., de Passillé, A.M., et al., 2009. Invited review: the
of claw disorders in Dutch dairy cows exposed to several floor systems. J. Dairy welfare of dairy cattledkey concepts and the role of science. J. Dairy Sci. 92,
Sci. 86, 2082–2093. 4101–4111.
Sotiraki, S.T., Leontides, L.S., Himonas, C.A., 1999. The effect of transportation and Waiblinger, S., Boivin, X., Pedersen, V., et al., 2006. Assessing the human-animal
confinement stress on egg production by Dicrocoelium dendriticum in sheep. relationship in farmed species: a critical review. Appl. Anim. Behav. Sci. 101,
J. Helminthol. 73, 337–339. 185–242.
Behavioral and Neuroendocrine Indicators of Well-being in Farm and Laboratory Mammals 485

Walkden-Brown, S.W., Martin, G.B., 1999. Role of male-female interaction in Wilt, J.G., 1985. Behaviour and Welfare of Veal Calves in Relation to Husbandry
regulating reproduction in sheep and goats. J. Reprod. Fertil. 52 (Suppl.), Systems (PhD thesis). Institute of Agriculture Engineering, The Netherlands.
243–257. Winckler, C., Buehnemann, A., Seidel, K., 2002. Social behaviour of commercial dairy
Walters, S.L., Torres-Urbano, C.J., Chichester, L., et al., 2012. The impact of huts on herds as a parameter for on-farm welfare assessment. In: Proceedings of the 36th
physiological stress: a refinement in post-transport housing of male guinea pigs International Congress. ISAE, The Netherlands.
(Cavia porcellus). Lab. Anim. 46, 220–224. Winckler, C., Capdeville, J., Gebresenbet, G., et al., 2003. Selection of parameters
Webster, J., 1995. Animal Welfare. A Cool Eye towards Eden. Blackwell Scientific for on-farm welfare-assessment protocols in cattle and buffalo. Anim. Welf. 12,
Publications, Oxford. 619–624.
Wei Liu, H., Fang Dong, X., Ming Tong, J., Zhang, Q., 2010. Alfalfa polysaccharides Yehuda, R., McEwen, B.S. (Eds.), 2004. Biobehavioral Stress Response. The New York
improve the growth performance and antioxidant status of heat-stressed rabbits. Academy of Sciences, New York, pp. 1–7.
Livest. Sci. 131, 88–93. Zenchak, J.J., Anderson, G.C., Schein, M.W., 1981. Sexual partner preference of adult
Whary, M., Peper, R., Borkowski, G., et al., 1993. The effects of group housing on the rams (Ovis aries) as affected by social experiences during rearing. Appl. Anim.
research use of the laboratory rabbit. Lab. Anim. 27, 330–341. Ethol. 7, 157–167.
Wheeler, R.R., Swan, M.P., Hickman, D.L., 2015. Effect of multilevel laboratory rat Zucca, D., Marelli, S.P., Redaelli, V., et al., 2012a. Effect of environmental enrichment
caging system on the well-being of the singly-housed Sprague-Dawley rat. Lab. and group size on behaviour and live weight in growing rabbits. World Rabbit Sci.
Anim. 49, 10–19. 20, 89–95.
Wierenga, H.K., Hopster, H., 1990. The significance of cubicles for the behaviour of Zucca, D., Redaelli, V., Marelli, S.P., et al., 2012b. Effect of handling in pre-weaning
dairy cows. Appl. Anim. Behav. Sci. 26, 309–337. rabbits. World Rabbit Sci. 20, 97–101.
This page intentionally left blank
HORMONES, BRAIN,
AND BEHAVIOR

THIRD EDITION
This page intentionally left blank
HORMONES, BRAIN,
AND BEHAVIOR

THIRD EDITION

EDITORS-IN-CHIEF
Donald W. Pfaff
The Rockefeller University, New York, NY, USA

Marian Joëls
University Medical Center Utrecht, Utrecht, The Netherlands;
and University Medical Center Groningen, Groningen, The Netherlands

VOLUME 2
NON-MAMMALIAN HORMONE-BEHAVIOR SYSTEMS

VOLUME EDITOR
Jacques Balthazart
University of Liège, Liège, Belgium

AMSTERDAM • BOSTON • HEIDELBERG • LONDON • NEW YORK • OXFORD


PARIS • SAN DIEGO • SAN FRANCISCO • SINGAPORE • SYDNEY • TOKYO
Academic Press is an imprint of Elsevier
Academic Press is an imprint of Elsevier
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
125, London Wall, EC2Y, 5AS
525 B Street, Suite 1800, San Diego, CA 92101-4495, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States

Copyright Ó 2017 Elsevier Inc. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means, electronic or mechanical, including
photocopying, recording, or any information storage and retrieval system, without permission in writing from the publisher. Details on how
to seek permission, further information about the Publisher’s permissions policies and our arrangements with organizations such as the
Copyright Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the Publisher (other than as may be
noted herein).

Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our understanding, changes in
research methods, professional practices, or medical treatment may become necessary.

Practitioners and researchers may always rely on their own experience and knowledge in evaluating and using any information, methods,
compounds, or experiments described herein. In using such information or methods they should be mindful of their own safety and the
safety of others, including parties for whom they have a professional responsibility.

To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any liability for any injury and/or
damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods,
products, instructions, or ideas contained in the material herein.

Library of Congress Cataloging-in-Publication Data


A catalog record for this book is available from the Library of Congress

British Library Cataloguing-in-Publication Data


A catalogue record for this book is available from the British Library

ISBN: 978-0-12-803592-4

For information on all publications visit our website


at http://store.elsevier.com

Printed and bound in Canada

Publisher: Oliver Walter


Acquisition Editor: Susan Dennis
Content Project Manager: Michael Nicholls
Associate Content Project Manager: Marise Willis
Designer: Greg Harris
CONTENTS OF VOLUME 2

Editorial Board vii


Contributors to Volume 2 ix
Preface of the First Edition xi
Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease.
An Introduction to the Second Edition xiii
Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease.
An Introduction to the Third Edition xv

Non-mammalian Vertebrates: Fishes


2.01 Neuroendocrinology of Social Behavior in Teleost Fish 3
David Gonçalves, Ana S Félix, and Rui F Oliveira
2.02 Social Regulation of Sex: How the Brain Controls Reproductive Circuits 19
Russell D Fernald
2.03 Socially Controlled Sex Change in Fishes 31
John Godwin and Melissa Lamm
2.04 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish
as Model Systems 47
Ni Y Feng and Andrew H Bass
2.05 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 69
Kent D Dunlap, Ana C Silva, G Troy Smith, and Harold H Zakon

Non-mammalian Vertebrates: Amphibians


2.06 Neuroendocrine Control of Social Behavior in Frogs 101
Walter Wilczynski
2.07 The Vertebrate Brain Stem as the Primary Site of Behavior Command:
Insights from an Amphibian 117
Emma Coddington
2.08 Hormones and Vocal Systems: Insights from Xenopus 131
Erik Zornik and Darcy B Kelley
2.09 Endocrinology of Complex Life Cycles: Amphibians 145
Robert J Denver

v
vi Contents of Volume 2

Non-mammalian Vertebrates: Reptiles


2.10 Hormones, Brain, and Behavior in Reptiles 171
David Kabelik and David Crews

Non-mammalian Vertebrates: Birds


2.11 Neuroendocrine Regulation of Reproductive Behavior in Birds 217
Gregory F Ball and Jacques Balthazart
2.12 Neural and Hormonal Control of Birdsong 255
Barney A Schlinger and Eliot A Brenowitz
2.13 Rapid Effects of Estrogens on Avian Brain and Social Behavior 291
L Remage-Healey, S A Heimovics, K K Soma, and C A Cornil
2.14 The Function of Behavior as Assessed by Phenotypic Engineering with Testosterone 305
Nicole M Gerlach and Ellen D Ketterson
2.15 Ecophysiological Studies of HormoneeBehavior Relations in Birds: Future Challenges
in a Changing World 321
John C Wingfield

Non-mammalian Invertebrates
2.16 Actions of Developmental Hormones in Adult Social Insects 349
Susan E Fahrbach, Ashton M Trawinski, and Rodrigo A Velarde
2.17 Diverse Functions of Insect Biogenic Amines as Neurotransmitters, Neuromodulators, and
Neurohormones 367
Wendi S Neckameyer and Sandra M Leal
2.18 Regulation of Honeybee Worker (Apis mellifera) Life Histories by Vitellogenin 403
Gyan P Harwood, Kate E Ihle, Heli Salmela (nee Havukainen), and Gro V Amdam
2.19 Endocrine Influences on Insect Societies 421
A R Hamilton, H Shpigler, G Bloch, D E Wheeler, and G E Robinson
2.20 Hormonal Regulation of Behavioral and Phenotypic Plasticity in Bumblebees 453
Abraham Hefetz and Christina M Grozinger
2.21 Stressed Out Insects II. Physiology, Behavior, and Neuroendocrine Circuits Mediating
Stress Responses 465
Erik C Johnson
EDITORIAL BOARD

EDITORS-IN-CHIEF
Donald W. Pfaff
The Rockefeller University, New York, NY, USA

Marian Joëls
University Medical Center Utrecht, Utrecht, The Netherlands;
and University Medical Center Groningen, Groningen, The Netherlands

VOLUME EDITORS
Gabriela González-Mariscal (Volume 1: Mammalian Hormone-Behavior Systems)
CINVESTAV-Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico

Jacques Balthazart (Volume 2: Non-Mammalian Hormone-Behavior Systems)


University of Liège, Liège, Belgium

E. Ronald de Kloet (Volume 3: Molecular and Cellular Mechanisms)


Leiden University Medical Center, Leiden, The Netherlands

Stafford Lightman (Volume 4: Clinically Important Hormone Effects on Brain and Behavior)
University of Bristol, Bristol, UK

Anthony P. Auger (Volume 5: Development of Hormone-Behavior Relationships)


University of Wisconsin, Madison, WI, USA

Catherine J. Auger (Volume 5: Development of Hormone-Behavior Relationships)


University of Wisconsin, Madison, WI, USA

vii
This page intentionally left blank
CONTRIBUTORS TO VOLUME 2

Gro V Amdam John Godwin


School of Life Sciences, Arizona State University, Tempe, North Carolina State University, Raleigh, NC, USA
AZ, USA; and Norwegian University of Life Sciences,
Aas, Norway David Gonçalves
University of St. Joseph, Macao, China
Gregory F Ball
University of Maryland, College Park, MD, USA Christina M Grozinger
Center for Pollinator Research, Center for Chemical
Jacques Balthazart Ecology, Huck Institutes of the Life Sciences, The
University of Liège, Liège, Belgium Pennsylvania State University, University Park, PA,
Andrew H Bass USA
Cornell University, Ithaca, NY, USA
A R Hamilton
G Bloch University of Illinois, Urbana, IL, USA
The Hebrew University of Jerusalem, Jerusalem, Israel
Gyan P Harwood
Eliot A Brenowitz School of Life Sciences, Arizona State University, Tempe,
University of Washington, Seattle, WA, USA AZ, USA
Emma Coddington Abraham Hefetz
Willamette University, Salem, OR, USA George S. Wise Faculty of Life Sciences, Tel Aviv
C A Cornil University, Tel Aviv, Israel
GIGA Neurosciences, University of Liege, Liege, Belgium
S A Heimovics
David Crews University of St. Thomas, St. Paul, MN, USA
University of Texas at Austin, Austin, TX, USA
Kate E Ihle
Robert J Denver University of Zurich, Zurich, Switzerland
The University of Michigan, Ann Arbor, MI, USA
Erik C Johnson
Kent D Dunlap Wake Forest University, Winston-Salem, NC, USA
Trinity College, Hartford, CT, USA
David Kabelik
Susan E Fahrbach
Rhodes College, Memphis, TN, USA
Wake Forest University, WinstoneSalem, NC, USA
Ana S Félix Darcy B Kelley
Gulbenkian Institute of Science, Oeiras, Portugal; and Columbia University, New York, NY, USA
ISPA e University Institute, Lisbon, Portugal Ellen D Ketterson
Ni Y Feng Indiana University, Bloomington, IN, USA
Cornell University, Ithaca, NY, USA
Melissa Lamm
Russell D Fernald North Carolina State University, Raleigh, NC, USA
Stanford University, Stanford, CA, USA
Sandra M Leal
Nicole M Gerlach Harris-Stowe State University, St. Louis, MO, USA
University of Florida, Gainesville, FL, USA

ix
x Contributors to Volume 2

Wendi S Neckameyer G Troy Smith


Saint Louis University School of Medicine, St. Louis, Indiana University, Bloomington, IN, USA
MO, USA
K K Soma
Rui F Oliveira University of British Columbia, Vancouver, BC, Canada
Gulbenkian Institute of Science, Oeiras, Portugal;
ISPA e University Institute, Lisbon, Portugal; and Ashton M Trawinski
Champalimaud Neuroscience Program, Lisbon, Portugal Wake Forest University, WinstoneSalem, NC, USA

L Remage-Healey Rodrigo A Velarde


Center for Neuroendocrine Studies, University of University of Buenos Aires, Buenos Aires, Argentina
Massachusetts, Amherst, MA, USA D E Wheeler
G E Robinson University of Arizona, Tucson, AZ, USA
University of Illinois, Urbana, IL, USA
Walter Wilczynski
Heli Salmela (nee Havukainen) Neuroscience Institute and Center for Behavioral
Centre of Excellence in Biological Interactions, Neuroscience, Georgia State University, Atlanta, GA,
University of Helsinki, Helsinki, Finland USA
Barney A Schlinger John C Wingfield
Brain Research Institute, University of California, Los University of California, Davis, CA, USA
Angeles, CA, USA
Harold H Zakon
H Shpigler University of Texas, Austin, TX, USA
University of Illinois, Urbana, IL, USA
Erik Zornik
Ana C Silva Reed College, Portland, OR, USA
Unidad Bases Neurales de la Conducta, Instituto de
Investigaciones Biologicas Clemente Estable,
Montevideo, Uruguay; and Universidad de la Republica,
Montevideo, Uruguay
PREFACE OF THE FIRST EDITION

Because of the large number of neuroactive substances discovered by chemical endocrinologists and the
prominent roles of nuclear hormone receptors as ligand-activated transcription factors, scientists studying
hormoneebrainebehavior relations have made discoveries and achieved explanations of behavior that place
their field foremost in neurobiology. The purpose of these volumes is to review the current state of this
knowledge inclusively. That means covering true molecular genetic approaches as well as neuroanatomical,
electrophysiological, zoological, neurochemical, developmental, and behavioral studies. The medical impor-
tance of this work is clear from the last section of this treatise. The editors intend these reviews to be
comprehensive; if there are any gaps in the coverage, a second edition will correct them.

Acknowledgments

Lucy Frank and Carol Oliver, at The Rockefeller University, organized all the volumes. Noelle Gracy and Mica
Haley handled things efficiently at Academic Press. Jasna Markovac at Academic Press presided over all stages of
this project. The editors thank all of these folks for their intelligent and gracious efforts.

Donald W. Pfaff
The Rockefeller University, New York, NY, USA

xi
This page intentionally left blank
HORMONAL EFFECTS ON SPECIFIC BEHAVIORS, ON GLOBAL
CNS STATES, AND ON HUMAN DISEASE. AN INTRODUCTION
TO THE SECOND EDITION

In 2002, there appeared the first edition of Hormones, Brain, and Behavior, dedicated to three generations of
productive scientists in our field, as it had developed at that point. With five volumes, totaling about 4100
pages, it represented a massive effort by over 200 scientists. The volumes were well received.
At least four developments have taken place since 2002 and justify a second edition. Two of them are
technical: the increasingly sophisticated application of molecular biological techniques and biophysical tech-
niques to the understanding of mechanisms for hormone/behavior relations. Third, we have entered an era of
translational research. Laboratory scientists are under increasing pressure to show how their work might lessen
the burden of disease upon the citizenry. The fourth development is the most subtle one and can be explained as
follows.
Some contemporary approaches to the hormonal controls of various forms of behavior can be distin-
guished sharply from the major theme in neuroscientific research in the past century. I characterize that
classical theme as a hunt for specificity. In the spinal cord, for example, the work of neurophysiologists such as
Sir Charles Sherrington (1906) and Sir John Eccles (1957) sought to explain why a particular reflex response
occurred specifically as a result of one particular stimulus and not another. At the level of the cerebral cortex,
work typified by that of David Hubel and Wiesel (1977) in the visual system and Vernon Mountcastle (1957)
in the somatosensory system described cells whose responses required stimuli of a specific type in a well-
defined receptive field. My own work, explaining mechanisms for lordosis behavior (Pfaff, 1999), fell into that
tradition.
In contrast, new approaches to changes of global states of the central nervous system involve the
regulation of large classes of behavioral responses to large sets of adequate stimuli. Thus, hormonal
influences on mood, exploratory tendencies, arousal, alertness, and attention would not be specific in the
manner displayed by neurons in the visual cortex, but they would be incredibly important for main-
taining mental and physical health. Our field of hormones and behavior increasingly embraces these
concepts.
In order to add new material, discovered since 2002, with special emphasis on clinical research, we had
to adopt the following strategy. We asked authors who had contributed to the first edition to reduce
markedly the length of their treatments of findings already covered in 2002. Therefore, the reader of this
current (second) edition will want to refer to the first edition as well, for the kind of comprehensive review
we intend.
Taking the first and the second editions of Hormones, Brain, and Behavior together will afford the reader
a tremendous number of facts, ranging from molecular genomics of hormone action, through biophysics,
neuroanatomy, and endocrinology to animal behavior and human disease. But this bulky coverage of primary
research findings is not, across the scope of several volumes, intended to be logically systematic. A parallel effort
in the text Principles of Hormone Behavior Relations takes a step in that direction.

References
Eccles, J., 1957. The Physiology of Nerve Cells. Johns Hopkins University Press, Baltimore, MD.
Hubel, D., Wiesel, T., 1977. Functional architecture of macaque monkey visual cortex. Proc. R. Soc. Lond. 198, 1e59.

xiii
xiv Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease

Mountcastle, V., 1957. Modality and topographic properties of single neurons of cat’s somatic sensory cortex. J. Neurophysiol. 20, 408e434.
Pfaff, D., 1999. Drive: Neural and Molecular Mechanisms of a Motivated Behavior. MIT Press, Cambridge, MA.
Sherrington, C., 1906. The Integrative Action of the Nervous System. Yale University Press, New Haven, CT (Reprinted 1947).

Further Reading
Pfaff, D., Phillips, M.I., Rubin, R.T., 2004. Principles of Hormone/Behavior Relations. Academic Press/Elsevier, San Diego.

Donald W. Pfaff
The Rockefeller University, New York, NY, USA
HORMONAL EFFECTS ON SPECIFIC BEHAVIORS, ON GLOBAL CNS
STATES, AND ON HUMAN DISEASE. AN INTRODUCTION TO THE
THIRD EDITION

In 2002, the first edition of Hormones, Brain, and Behavior appeared, dedicated to three generations of productive
scientists in our field, as it had developed at that point. With five volumes, totaling about 4100 pages, it rep-
resented a massive effort by over 200 scientists. In 2009 the volumes were updated, in the second edition. Both
sets of five volumes were well received.
At least four developments have taken place since 2009 which justify a third edition. Two are technical: the
application of increasingly sophisticated molecular biological techniques and biophysical techniques to the
understanding of mechanisms for hormone/behavior relations. Third, we have entered an era of translational
research. Laboratory scientists are under increasing pressure to show how their work might lessen the burden of
disease upon the citizenry. The fourth development is the most subtle one and can be explained as follows.
Some contemporary approaches to the hormonal controls of various forms of behavior can be distinguished
sharply from the major theme in neuroscientific research in the past century. We characterize that classical theme
as a ‘hunt for specificity.’ In the spinal cord, for example, the work of neurophysiologists such as Sir Charles
Sherrington (1906) and Sir John Eccles (1957) sought to explain why a particular reflex response occurred as
a result of one particular stimulus and not another. At the level of the cerebral cortex, work typified by that of
David Hubel and TorstenWiesel (1977) in the visual system and Vernon Mountcastle (1957) in the somato-
sensory system described cells whose responses required stimuli of a specific type in a well-defined receptive field.
Pfaff’s own work, explaining mechanisms for lordosis behavior (Pfaff, 1999), fell into that tradition.
In contrast, new approaches to changes of global states of the central nervous system involve the regulation of
large classes of behavioral responses to large sets of adequate stimuli. Experimental work on steroid hormone
regulation of responses to stress provide an example of this type of brain research (reviewed, Joels et al., 2013). Thus,
hormonal influences on mood, exploratory tendencies, arousal, alertness, and attention would not be specific in the
manner displayed by neurons in the visual cortex, but they would be incredibly important for maintaining mental
and physical health. Our field of hormones and behavior increasingly embraces these concepts.
In order to add new material, discovered since 2009, with special emphasis on clinical research, we had to
adopt the following strategy. We asked authors who had contributed to the first and second editions to reduce
markedly the length of their treatments of findings already covered in 2009; therefore, the reader of this current
(third) edition may want to refer to the earlier editions as well, for the kind of comprehensive review we intend.
In lieu of these earlier sections, the authors expanded information on more recent concepts and experiments.
Taking all editions of Hormones, Brain, and Behavior together will afford the reader a tremendous number of
facts, ranging from molecular genomics of hormone action, through biophysics, neuroanatomy, and endo-
crinology to animal behavior and human disease. But this bulky coverage of primary research findings is not,
across the scope of several volumes, intended to be logically systematic.
A parallel effort in the text Principles of Hormone Behavior Relations (the second edition of which will appear
soon) takes a step in that direction.

References
Eccles, J., 1957. The Physiology of Nerve Cells. Johns Hopkins University Press, Baltimore, MD.
Hubel, D., Wiesel, T., 1977. Functional architecture of macaque monkey visual cortex. Proc. R. Soc. Lond. 198, 1e59.

xv
xvi Hormonal Effects on Specific Behaviors, on Global CNS States, and on Human Disease

Joels, M., Pasricha, N., Karst, H., 2013. The interplay between rapid and slow corticosteroid actions in brain. Eur. J. Pharmacol. 719, 44e52.
Mountcastle, V., 1957. Modality and topographic properties of single neurons of cat’s somatic sensory cortex. J. Neurophysiol. 20, 408e434.
Pfaff, D., 1999. Drive: Neural and Molecular Mechanisms of a Motivated Behavior. MIT Press, Cambridge, MA.
Sherrington, C., 1906. The Integrative Action of the Nervous System. Yale University Press, New Haven, CT (Reprinted 1947).

Further Reading
Pfaff, D., Phillips, M.I., Rubin, R.T., 2004. Principles of Hormone/Behavior Relations. Academic Press/Elsevier, San Diego.

Donald W. Pfaff
The Rockefeller University, New York, NY, USA
Marian Joëls
University Medical Center Utrecht, Utrecht, The Netherlands
PERMISSION ACKNOWLEDGMENT

The following material is reproduced with kind permission of Oxford University Press
Figure 1 of Regulation of Honeybee Worker (Apis mellifera) Life Histories by Vitellogenin
www.oup.com

i
NON-MAMMALIAN
VERTEBRATES: FISHES
This page intentionally left blank
2.01 Neuroendocrinology of Social Behavior in Teleost Fish
David Gonçalves, University of St. Joseph, Macao, China
Ana S Félix, Gulbenkian Institute of Science, Oeiras, Portugal; and ISPA – University Institute, Lisbon, Portugal
Rui F Oliveira, Gulbenkian Institute of Science, Oeiras, Portugal; ISPA – University Institute, Lisbon, Portugal; and Champalimaud
Neuroscience Program, Lisbon, Portugal
Ó 2017 Elsevier Inc. All rights reserved.

2.01.1 Introduction 3
2.01.2 The Social Decision-Making Network 3
2.01.2.1 Short History of the Concept 3
2.01.2.2 Homologies between Teleost Fish and Mammalian Brain Areas in the SDMN 4
2.01.2.3 Functional Analysis of the SDMN in Teleost Fish 5
2.01.3 Endocrine Regulation of Social Behavior in Teleost Fish 6
2.01.3.1 Aggressive Behavior 6
2.01.3.2 Mating Behavior 9
2.01.3.3 Parental Care 11
2.01.3.4 Prosocial Behavior 11
2.01.4 Hormones as Social Semiochemicals 12
2.01.5 Conclusions 14
Acknowledgments 14
References 14

2.01.1 Introduction 2.01.2 The Social Decision-Making Network


2.01.2.1 Short History of the Concept
Animals need to interact with other members from the same
species in order to survive and reproduce successfully, and In 1999, Newman challenged the neuroscientific community
the set of behaviors used in these social interactions are by proposing the existence of a core set of brain areas that
labeled social behaviors. From a functional perspective, these collectively regulate social behavior in mammals. Each one
social behaviors can be classified into major groups accord- of these areas is reciprocally connected with the others,
ing to the type of interaction in which they are expressed, contains sex steroid hormone receptors, and it is involved
namely, aggressive, mating, parental, and prosocial. Interest- in the activation or regulation of several social behaviors. It
ingly, despite their different functions, all these types of was designated as social behavior network (SBN), and it is
social behaviors seem to share a common underlying mech- composed of six limbic areas: the lateral septum (LS), the
anism, composed of a neural network, recently named the medial extended amygdala (medial amygdala, meAMY, and
social decision-making network (SDMN) (O’Connell and bed nucleus of the stria terminalis, BNST), the medial pre-
Hofmann, 2012a, 2011), whose overall state parallels the optic area (POA), the anterior hypothalamus (AH), the
expression of the specific type of social behavior expressed ventromedial and ventrolateral hypothalamus (VMH), all
in a given moment in time, such that the state of the network localized in the forebrain, and the midbrain periaqueductal
is a better predictor of social behavior than the activity of gray and tegmentum (PAG/CG), lying in the midbrain. Her
a specific brain region (Goodson and Kabelik, 2009; Teles model was based on a considerable amount of behavioral,
et al., 2015). Moreover, the fact that all nodes in this network neuroanatomical, and neuroendocrine evidences from
express receptors for steroid hormones and for neuropeptides diverse studies in rodents and other mammals, which used
opens the possibility for its neuromodulation and concomi- electrical stimulation, neuropharmacological manipulations,
tantly for the regulation of its behavioral output by these specific brain lesions, and detection of immediate early gene
agents. Thus, by acting on the SDMN, hormones can regulate (IEG) expression. Together, these data show that common
the expression of social behavior and integrate it with the areas jointly influence sexual, parental, or even aggressive
organismal state of the individual. Hormones can also regu- behavior, counteracting the idea of one area (or even a sepa-
late the social behavior of animals other than the one in rate minicircuit) is determining a specific behavior. Instead,
which they are produced when they act as hormonal phero- all these areas represent the nodes of a neuroanatomical
mones. In such cases, hormones are released into the envi- network, whose dynamic activation patterns are responsible
ronment and act on receptors located in sensory tissues of for multiple behaviors. For instance, male sexual behavior
other individuals triggering changes on their behavior. In would be the result of successive behavioral responses such
this chapter we will use the conceptual framework sketched as sniffing, mounting, ejaculation, or grooming, which alto-
above to illustrate how hormones regulate social behaviors gether are activated by this integrated circuit and modulated
in teleost fish. by environmental stimuli and sex steroids. Newman also

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00020-1 3


4 Neuroendocrinology of Social Behavior in Teleost Fish

highlighted that species and sex differences in social behav- region (vTn), anterior tuberal nucleus (aTn), all localized in
iors are a consequence of brain organization and connec- the forebrain, in addition to the PAG, lying in the midbrain
tivity divergences, influenced by variations in hormone (O’Connell and Hofmann, 2011). On the other hand, the mes-
sensitivity along development, on this central network. olimbic reward system is presumably constituted by the dorsal
Later, Goodson (2005) expanded the same framework to (Vd) and central (Vc) part of the ventral telencephalon, the
other nonmammalian vertebrates describing important medial part of the dorsal telencephalon (Dm), the lateral part
evidences for birds and teleost fish, and providing foundations of the dorsal telencephalon (Dl), the posterior tuberculum
for the evolutionarily conservation of the SBN in vertebrates. (TPp) on the midbrain, and also Vv/Vl and Vs, that are also
He also contributed to a better insight into this network by add- nodes of the SBN (O’Connell and Hofmann, 2011; Figure 1).
ing the role of peptidergic neuromodulation such as arginine Both Vv and Vl seem to be homologous of mammalian LS
vasotocin (AVT, homologue of mammalian arginine vaso- region. Cholinergic neurons were detected only in this telence-
pressin) or isotocin (IT, homologue of oxytocin) on social phalic area. It is reciprocally connected to several important
behavior and specifically as an integrating component of SBN. nuclei, and it expresses sex steroid receptors (see Wullimann
More recently, O’Connell and Hofmann (2012a, 2011) and Mueller, 2004; O’Connell and Hofmann, 2011 for more
proposed that social behavior would be regulated by an even details). It is also involved in reproductive behavior since elec-
wider network. The SDMN would include SBN and the meso- trical stimulation of Vv in Sockeye salmon, Oncorhynchus nerka,
limbic reward system. The latter system is the brain circuit elicits females’ digging and spawning while Vv and Vs ablation
responsible for the evaluation of the salience of a stimulus in the goldfish Carassius auratus impairs male ejaculation (Kyle
(via dopaminergic signaling) prior to the behavioral response, and Peter, 1982; Satou et al., 1984). Vs is putatively homolo-
which in turn is elicited by the former network. Thus, the gous of meAMY/BNST based on developmental evidences,
reinforcing/rewarding component of social behavior as as well as neurochemical and connectivity similarities (see
a substantial feature of an individuals’ adaptive response to Wullimann and Mueller, 2004; O’Connell and Hofmann,
the environment is the main argument of the authors. The 2011 for further details), even though some consider that
mammalian mesolimbic reward system is constituted by the ventral telencephalon (Vp) should also be included within
striatum (Str), the nucleus accumbens (NAcc), the ventral pal- this node (Goodson and Kingsbury, 2013). The teleost POA
lidum (VP), the basolateral amygdala (blAMY), the hippo- homology is well established. This node that is imperative for
campus (Hyp), the ventral tegmental area (VTA), and the LS the regulation of sexual, parental, and aggressive behaviors
and the meAMY/BNST, overlapping with nodes of the SBN. (Demski and Knigge, 1971; Macey et al., 1974; Satou et al.,
The authors performed a comparative analysis of the two neural 1984; Wong, 2000) is localized in the hypothalamus along
circuits in five vertebrate lineages: mammals, birds, reptiles, the third ventricle, just like in mammals. It is divided into three
amphibians, and teleost fish. Putative brain homologies were subregions according to cell size: parvocellular, magnocellular,
described based on neuronal connections, the presence of and gigantocellular, and it is reciprocally connected with the
steroid hormone receptors, gene expression, neurochemistry, telencephalon and other hypothalamic regions (reviewed in
developmental and behavioral studies. Concurrently, they O’Connell and Hofmann, 2011). vTn is the putative homolo-
provided a very useful resource to study the neural substrates gous of AH because it is localized between the POA and the
responsible for social behavior in vertebrates and a relevant ventral hypothalamus, it receives and sends projections to
framework to make species comparisons. several hypothalamic regions, and it expresses sex steroid recep-
Nevertheless, attention must be drawn to the fact that tors (O’Connell and Hofmann, 2011 reviewed this informa-
some proposed homologies are not complete, instead they tion). However, functional studies are yet not available to
are only partial. In that sense, in a recent review, Goodson confirm this homology. aTn was proposed to be the teleost
and Kingsbury (2013) proposed the inclusion of the paraven- equivalent of VMH, although only a subset of aTn cells are
tricular nucleus (PVN) of the hypothalamus within the actually homologous (Goodson and Kingsbury, 2013). Lying
mammalian POA node (i.e., POA/PVN), in order to compre- in the ventrocaudal region of the hypothalamus, it is recipro-
hend vasopressin–oxytocin nonapeptide neurons crucial for cally connected with several parts of the telencephalon and
the regulation of social behavior. With this incorporation the contains sex steroid hormone receptors (reviewed in O’Connell
POA/PVN mammalian node would be similar to the anam- and Hofmann, 2011) but functional studies are as well
niotes POA node. limited. PAG is also present in teleosts, located near the torus
On the other hand, for some nonmammalian species, semicircularis and receiving and sending projections to several
homologies of the SDMN nodes are not clear, and functional other nuclei (see O’Connell and Hofmann, 2011 for more
studies are still missing, especially for the mesolimbic reward information). Functionally, it is associated with social commu-
system (Goodson and Kingsbury, 2013). So, despite being nication, specifically, in sound production of the plainfin
strongly supported in mammals, for other taxa, the SDMN midshipman Porichthys notatus (Kittelberger et al., 2006). Vc is
must be cautiously evaluated and tested. comparable to Str in mammals, while Vd seems homologous
to NAcc, but this is only supported by neurochemical studies
and some hodological evidences (consider Wullimann and
2.01.2.2 Homologies between Teleost Fish and Mammalian
Mueller, 2004; O’Connell and Hofmann, 2011 for more
Brain Areas in the SDMN
details). Unfortunately, a homologous for the mammalian
In teleosts, the SBN is assumed to be composed by the ventral VP node has not yet been identified. Dm is the putative homol-
(Vv) and lateral (Vl) part of ventral telencephalon, supracom- ogous of blAMY based on developmental, tract tracing, and
misural part of the ventral pallium (Vs), POA, ventral tuberal lesions studies, demonstrating its implication in emotional
Neuroendocrinology of Social Behavior in Teleost Fish 5

Brain
SDMN
Dl Dm
POA

Sensory interface

Motor output
Input Vd Vs PAG Behavior

Vc Vv/Vl aTn

vTn
TPp VP?

IT AVT

Peripheral hormones

Sex steroids Glucocorcoids


Body

Figure 1 Representation of the interaction between hormones and the social decision-making network (SDMN) within teleosts’ social behavior:
putative nodes of the mesolimbic reward system in white – dorsal (Vd) and central (Vc) part of the ventral telencephalon, medial part of the dorsal
telencephalon (Dm), lateral part of the dorsal telencephalon (Dl), posterior tuberculum (TPp) – and the social behavior network in black – medial pre-
optic area (POA), ventral tuberal region (vTn), anterior tuberal nucleus (aTn), and midbrain periaqueductal gray (PAG). Ventral and lateral (Vv/Vl) part
of ventral telencephalon and supracommisural part of the ventral pallium (Vs), overlapping nodes of the social behavior network and the mesolimbic
reward system, are in gray. A homologous for the mammalian ventral pallidum (VP) node has not yet been identified. AVT, arginine vasotocin; IT,
isotocin.

learning, as shown in mammals (reviewed in O’Connell and selected nodes of the SDMN while animals were in different
Hofmann, 2011; Portavella et al., 2002). The homologous of behavioral states (Teles et al., 2015). The aim of this work
Hyp is considered to be the Dl mainly because of its involve- was to test for functional brain specialization or alternatively
ment in spatial learning (Dl lesions of C. auratus impair for functional connectivity and, for example, determine
maplike memory representations, Rodríguez et al., 2002), whether expression of social behavior is explained by the acti-
besides some of its hodological features (reviewed in vation of a single node or by the overall combination of the
O’Connell and Hofmann, 2011). Finally, even though amphib- activity in the various nodes. They quantified IEG expression
ians and teleosts lack a midbrain dopaminergic population, in the brains of winners and losers of agonistic fights, as well
TPp, located in the ventral diencephalon, has been suggested as in individuals who only experienced mirror fights and
to present homologies to VTA, the A10 dopaminergic cell compared them with a reference noninteracting group. IEG-
group, because of its dopaminergic ascending projections to transcription patterns of c-fos and egr-1 as measured by qPCR
Vd and conserved neurochemical patterns (see O’Connell and were used as markers of neuronal activity in the Vv, Vs, POA,
Hofmann, 2011 for details). However, this seems to be refutable Dm, and Dl. The similarity of IEG activation between groups
as a recent study on the zebrafish, Danio rerio, ‘projectome’ and areas showed that there were no patterns of localized
underlines that posterior tuberculum cells (DC2 and DC4–6 activity in a specific nucleus. Instead socially driven behavioral
cell groups) seem homologous to A11 mammalian dopamine states demonstrated patterns of functional connectivity across
neurons based on transcription factor conservation and projec- the nodes. The notion of an SDMN is therefore supported in
tion patterns (Tay et al., 2011). Actually ascending projections a teleost fish.
to telencephalon are scarce (and only from DC2 and DC4 On the other hand, a considerable number of studies
neuronal cells), while the most important dopaminergic centered on the behavioral responses of teleost fishes have
connections between the subpallium and the ventral dienceph- documented the activation of specific SDMN nodes, hence
alon are descending (Tay et al., 2011). Consequently, the exis- establishing their involvement in the regulation of social
tence of a mesolimbic reward system in fish is questionable behavior. For instance, Desjardins et al. (2010) focused on
since the connection between the VTA and forebrain regions is how mate information impacts female neural activity. In this
considered the core of the dopaminergic reward system work, they measured IEG (c-fos and egr-1) expression levels
(Bromberg-Martin et al., 2010; Spanagel and Weiss, 1999). by qPCR in gravid females of the Burton’s mouthbrooder
cichlid, Astatotilapia burtoni, that saw their preferred males
winning or losing a fight. They looked specifically to the Vv
2.01.2.3 Functional Analysis of the SDMN in Teleost Fish
(LS), POA, vTn (AH), aTn (VMH), PAG, Dm, and Dl. Results
A recent study in zebrafish functionally tested the SDMN demonstrate that the POA and VMH, two nuclei known to be
concept. In this study, IEG expression was determined along involved in the control of reproduction, are highly activated
6 Neuroendocrinology of Social Behavior in Teleost Fish

when females see their preferred male winning, whereas IEG there is now substantive information on the role of these
response in the mammalian LS homologue region (a nucleus hormones as modulators of social behavior. However, in
associated with anxiety) is elicited when females see the male some cases, the expression of social behavior seems to be inde-
losing. pendent from hormones produced in the periphery, and
O’Connell et al. (2013) directed their interest to how indi- the role of brain-synthesized hormones, in particular neuroste-
viduals integrate social information. These researchers pre- roids and neuropeptides, has been receiving increasing atten-
sented A. burtoni males with different social stimuli and tion (for a review see Gonçalves and Oliveira, 2010; Oliveira
discovered that visual information (seeing a female or and Gonçalves, 2008).
a male) is sufficient to elicit c-fos transcription in dopaminergic Regardless of the source, still little is known on how molec-
neurons of Vc, and this transcription is significantly correlated ular and cellular mechanisms of hormonal action in the brain
to aggressive behavior in the case of exposure to an intruder modify social behavior in fish. The recent concept of the
male. These data suggest that Vc seems to be involved in assess- SDMN is useful as it allows studies on the neuroendocrine
ing stimulus visual valence. Another interesting survey was modulation of social behavior to be focused in relevant brain
also carried out in this species. Since A. burtoni males can revers- areas. As described above, recent work has addressed the inter-
ibly switch between dominant and subordinate status and play between social environment and activation of the nodes
rapidly present distinct phenotypes, investigators examined of the SDMN using IEG as proxies of neuronal activity (e.g.,
IEG levels in several brain areas of males ascending or descend- Desjardins et al., 2010; Teles et al., 2015). However,
ing in social status, as compared with control individuals studies investigating the regulatory role of hormones in the func-
(Maruska et al., 2013a,b). In socially ascending males, both tionality of the SDMN are still scarce in spite of the fact that
c-fos and egr-1 levels were higher than in control males in all major modulatory effects of hormones in the SDMN are ex-
the SDMN nuclei (Vv, Vs, POA, vTn, aTn, Dm, and Dl) pected, as evidenced by the widespread distribution of hormone
(Maruska et al., 2013b). Descending males presented different receptors in its nodes. In particular, estrogen (P. notatus, Forlano
activation patterns for c-fos and egr-1 across the same areas. c-fos et al., 2005; Micropogonias undulatus, Hawkins et al., 2005;
expression levels were increased in the Vs, POA, and aTn D. rerio, Menuet et al., 2002; A. burtoni, Munchrath and
by comparison with controls while egr-1 mRNA levels were Hofmann, 2010; Dicentrarchus labrax, Muriach et al., 2008),
higher in the Vv, Vs, vTn, Dm, and Dl (Maruska et al., progesterone (A. burtoni, Munchrath and Hofmann, 2010),
2013a). Another relevant study used the monogamous cichlid androgen (P. notatus, Forlano et al., 2010; C. auratus, Gelinas
Amatitlania nigrofasciata as a model to study the influence of IT and Callard, 1997; A. burtoni, Harbott et al., 2007; Munchrath
in parental care (O’Connell et al., 2012). The authors and Hofmann, 2010), vasotocin (A. burtoni, Huffman et al.,
compared males housed with their mate (control males), single 2012; rock hind, Epinephelus adscensionis, Kline et al., 2011),
fathers who had the mate removed or lone males (mate and and IT (A. burtoni, Huffman et al., 2012) receptors are widely
offspring removed), and quantified c-fos expression in Vv, distributed along the areas of the SDMN (see also Diotel et al.,
POA, and the central part of the dorsal telencephalon (Dc). 2011).
At the behavioral level, only single fathers increased paternal Below we review the evidence for the endocrine modulation
care immediately after removal of their mate, and they also of different types of social behavior in fish, addressing their
presented significantly higher IEG activity levels in Vv possible mechanisms of action at the brain level.
compared to lone males, as well as increased c-fos expression
in the parvocellular POA IT neurons. Together these data
2.01.3.1 Aggressive Behavior
suggest that IT promotes paternal care after mate removal
and that Vv and POA are important brain areas in this process. Different categories of hormones, notoriously gonadal steroids,
Finally, a very interesting study with P. notatus, where reproduc- corticosteroids, and nonapeptide hormones, have been associ-
tive behavior is intimately associated with social acoustic ated with the regulation of aggressive behavior in fish. For
signals, measured c-fos activation in several brain nuclei gonadal steroids, early evidence demonstrated that male domi-
including vTn, aTn, and TPp (Petersen et al., 2013). The authors nance correlated with circulating androgen levels, in particular
report a significant increase of IEG expression in aTn and TPp with the nonaromatizable androgen 11-ketotestosterone
of males exposed to acoustic signals of other males compared (11KT; for a review, see Oliveira and Gonçalves, 2008; Oliveira
to control males, showing the importance of these nuclei in et al., 2002). Classical castration–androgen replacement experi-
social reproductive communication in this species. ments suggested an effect of gonadal androgens in aggression,
and a meta-analysis confirmed that exogenous administration
of androgens promotes aggression in fish (Hirschenhauser and
2.01.3 Endocrine Regulation of Social Behavior in Oliveira, 2006). In one of the best studied models in this
Teleost Fish respect, the cichlid A. burtoni, a change from a submissive to
a dominant status in males increases the expression of the
The pioneering work of Arnold Berthold on the endocrine GnRH1 gene and the concomitant production of its peptide in
regulation of sexual behavior in animals demonstrated the neurons of the POA, inducing gonadal development and a sur-
influence of a ‘blood-borne product’ released in peripheral ge in plasma 11KT levels within 30 min (Francis et al., 1993;
glands on behavior (Berthold, 1849). Likewise, early studies Maruska and Fernald, 2010; White et al., 2002). Interestingly,
on the endocrine regulation of behavior in fish focused on in this species a submissive experience seems to have more rapid
the role of hormones produced in the periphery, mainly and more profound physiological consequences than a domi-
gonadal steroids, prostaglandins, and corticosteroids, and nant one as aggressive behaviors were reduced more promptly
Neuroendocrinology of Social Behavior in Teleost Fish 7

in males undergoing a descent in social status than they emerged weakly electric fish Gymnotus omarorum (Jalabert et al., 2015).
in animals ascending in social status (White et al., 2002). Simi- This would suggest that aromatization of T into E2 is needed
larly, in zebrafish males, losing a fight induces a more to promote male aggressive displays in these species. Clearly,
pronounced change in future aggressive behavioral displays more data are needed to interpret the divergent results across
and in the neurogenomic state of the whole brain than winning species and understand what the general pattern in fish is.
(Oliveira et al., 2011, 2016). The above data suggest that a dominance experience (e.g.,
Aggression in females has been much less investigated winning fights) activates the hypothalamic–pituitary–gonadal
although, interestingly, the role of androgens in the modulation axis, increasing the secretion of gonadal androgens that in
of aggressive displays seems to be more consistent than turn act on different tissues to promote the expression of
for males. In the Mozambique tilapia, plasma testosterone a dominant phenotype, including increased aggression, via
(T) levels in females peak during a phase of the reproductive effects on the brain (and the reverse for submissive experi-
cycle that matches increased aggression (Oliveira and Canário, ences). This feedback between the environment and behavior,
2000). In the cichlid Neolamprologus pulcher, females and males translated via neuroendocrine modulation by androgens,
jointly defend a territory all year round and females were shown would allow animals to dynamically adjust their behavior
to be more aggressive than males in response to a territorial and physiology to a particular social context (Oliveira, 2004).
intrusion and to experience a higher increase in androgen levels However, the hypothesis that high levels of aggression are
(including 11KT, usually undetected in females, Desjardins et al., maintained through a positive effect of gonadal androgens in
2006). Similarly, in the blue acara, Andinoacara pulcher, T admin- the brain is contradicted by other studies. The majority of
istration increased aggressive behavior in females (Munro and studies where males were gonadectomized during the breeding
Pitcher, 1985) and daily injections of T to females of the Siamese season, thus reducing the circulating levels of androgens, failed
fighting fish Betta splendens for a period of 9 weeks increased to find a significant effect on male aggression (reviewed in Gon-
aggression directed toward males, although it decreased aggres- çalves and Oliveira, 2010). As an example, in the Mozambique
sion directed toward females (Badura and Friedman, 1988). tilapia gonadectomy impaired the expression of reproductive
Finally, in all-female groups of A. burtoni, it was observed behaviors in males, including nest building and courtship
that females start to exhibit typical male behaviors, including displays, but did not affect aggressive behavior toward a
territorial and aggressive displays (Renn et al., 2012). In this conspecific male (Almeida et al., 2014). Also, in the social
experiment, dominant females had higher T levels when sex-changing bluehead wrasse Thalassoma bifasciatum, gonadec-
compared with subordinates and T levels correlated with aggres- tomy did not prevent female-to-male behavioral change,
sive displays like chasing and threatening. including an increase in the expression of aggressive behavior,
In birds and mammals, the central effects of androgens on when females were given an opportunity to occupy a vacant
aggressive behavior seem to depend on the aromatization of territory (Godwin et al., 1996). Finally, variation in aggressive
T into estradiol (E2) (Trainor et al., 2006). In fish, however, behavior has been shown to occur in immature individuals
in spite of aromatase being abundant in nuclei of the SDMN and animals outside the breeding season, questioning the
(e.g., Forlano et al., 2001), there are conflicting results on the hypothesis of aggression being modulated by gonadal andro-
role of aromatization in aggression. Several lines of evidence gens in these contexts. For example, in the G. omarorum, high
suggest a direct action of androgens in the expression of aggres- levels of aggression were decoupled from 11KT levels outside
sive behavior in fish: (1) androgen receptors are widely distrib- the reproductive season (Jalabert et al., 2015) and in the
uted in nuclei of the telencephalon and diencephalon, damselfish Stegastes nigricans, also a year-round territorial
including in areas of the SDMN (e.g., Harbott et al., 2007); species, androgen levels did not increase when an aggressive
(2) differences in plasma androgen levels between aggressive challenge was presented to males (Ros et al., 2014).
and less aggressive phenotypes seem to be more evident for The contradictory results found for the effect of sex steroids
the nonaromatizable androgen 11KT than for T (Oliveira, on aggressive behavior have driven the search for alternative
2005) and the peripheral administration of 11KT has been modulators of aggression. A pathway that has also been shown
shown to promote aggression in teleosts (e.g., Rodgers et al., to relate with the neuroendocrine modulation of aggression in
2013); (3) in some species, peripheral administration of fishes is the hypothalamic–pituitary–interrenal axis. In the
estrogens has an inhibitory effect on male aggression (e.g., abovementioned study in S. nigricans, where androgens failed
three-spined stickleback, Gasterosteus aculeatus, Bell, 2001; to respond to an aggressive challenge, cortisol levels in the
B. splendens, Clotfelter and Rodriguez, 2006; D. rerio, Colman plasma increased after males were presented with intra- and
et al., 2009; Filby et al., 2012; peacock blenny, Salaria pavo, interspecific challenges, and its concentration was strongly
Gonçalves et al., 2007; A. pulcher, Munro and Pitcher, 1985; correlated with aggressive behaviors (Ros et al., 2014). Also,
sand goby, Pomatoschistus minutus, Saaristo et al., 2010); in the cichlid A. pulcher, cortisol administration increased
and (4) whole brain aromatase activity was shown to be aggression toward a model intruder (albeit not toward a mirror
inversely correlated with aggression in females of the sex- image, Munro and Pitcher, 1985). In juvenile rainbow trout,
changing blue-banded goby, Lythrypnus dalli (Black et al., Oncorhynchus mykiss, cortisol administration failed to promote
2005), suggesting that a higher availability of T (or a decrease aggression 1 h after being administered, but pharmacological
in E2 synthesis) promotes aggression. Contrarily, in the cichlid blockage of GR and MR receptors reduced aggression levels,
A. burtoni, E2 administration increased male aggression suggesting that basal levels of cortisol were contributing to
(O’Connell and Hofmann, 2012b) and pharmacologically aggressive behavior via activation of intracellular GR and MR
blocking aromatization with Fadrozole decreased these behav- receptors (Schjolden et al., 2009). Nevertheless, prolonged
iors in the same species (Huffman et al., 2013) and also in the (48 h) exposure to cortisol reduced aggression, suggesting
8 Neuroendocrinology of Social Behavior in Teleost Fish

a complex time-dependent effect of this hormone in aggressive higher levels of expression of AVT in the gigantocellular
behavior (Øverli et al., 2002). layer but lower levels in the parvocellular layer, as compared
AVT and IT have also been implicated in the regulation of with nonterritorial males. The authors suggested that
aggression, and more generally social behavior, in fishes gigantocellular neurons might be more related with the
(reviewed in Godwin and Thompson, 2012). AVT neurons modulation of dominance-related traits, including the
occur in the POA and project to the neurohypophysis, releasing expression of aggressive behavior, while parvocellular cells
AVT into circulation when activated, but also project to many may relate to the activation of the stress axis or submissive
other brain regions, including the ventral telencephalon, thal- behaviors. Following this model, Godwin and Thompson
amus, and mesencephalon (for details on the neuroanatomy (2012) suggest that AVT projections from the POA may
of the AVT system in fish, see Godwin and Thompson, 2012; regulate ‘sociosexual circuits,’ including those related with
Huffman et al., 2012; Thompson and Walton, 2013). Within aggression, by modulating neuronal action in central brain
the POA, three subpopulations of AVT neurons can be identi- regions, including nodes of the SDMN. On the other hand,
fied; parvocellular, magnocellular, and gigantocellular, and AVT would also be able to promote submissive and
they have been suggested to play different roles in osmoregula- escape behaviors by producing peripheral changes that
tion and modulation of behavior (Greenwood et al., 2008). feedback to the brain. These effects could be induced either
Receptors for both AVT and IT have been found throughout directly, via modulation of hindbrain autonomic nuclei that
nuclei of the SDMN, suggesting a direct neuromodulatory regulate peripheral states, or indirectly, as, for example,
action of these neuropeptides in nodes of this brain network through the demonstrated capacity of AVT to stimulate the
(Huffman et al., 2012; Lema, 2010). stress axis (Baker et al., 1996). As a consequence, peripheral,
Following studies in mammals that associated AVP with or even central, administration of AVT may have variable
increased expression of aggressive behavior, AVT has also been effects in behavior as it activates the targets of multiple
implicated in the regulation of aggression in fishes, although subpopulations of AVT neurons. In addition, and as
with inconsistent results between species. A positive effect of demonstrated in mammalian models, the effects of AVP/AVT
AVT on aggression has been described for some species, as, in behavior are greatly dependent on tissue sensitivity which
for example, nonterritorial phase males of T. bifasciatum (Semsar may vary across phenotypes, developmental stages, or seasons
et al., 2001), males of the damselfish Stegastes leucostictus due to variation in the abundance of its receptors (e.g., Walton
(Santangelo and Bass, 2006), and males of A. nigrofasciata et al., 2010). Future studies manipulating the AVT system
(Oldfield and Hofmann, 2011) while a suppressive effect of using techniques such as optogenetics, transgenics, or
AVT on aggression has been described for other species, as, for others may offer an opportunity to investigate the exact
example, territorial phase males of T. bifasciatum (Semsar et al., function of the different subpopulations of AVT neurons and
2001), males of the brown ghost knifefish, Apteronotus leptorhyn- subtypes of receptors in the modulation of aggression, and
chus (Bastian et al., 2001), males of Amargosa pupfish, Cyprino- other categories of social behavior, in fish.
don nevadensis amargosae (Lema and Nevitt, 2004; but see Lema The effect of IT on aggressive behavior in fish has been less
et al., 2015), and males and females of D. rerio (Filby et al., investigated. In the plainfin midshipman, IT administered to
2010). These contradictory results may have different, but not the POA-AH elicited fictive aggressive vocalizations in a neuro-
necessarily exclusive, explanations. In studies with butterfly fish physiological preparation of parasitic (sneaker) males but not
of the genus Chaetodon, it was shown that a territorial species of territorial males (Goodson and Bass, 2000). On the other
had larger AVT-immunoreactive somata within the POA area hand, IT administered to males of the beaugregory damselfish
and higher AVT fiber densities within a number of had no effect in aggressive displays (Santangelo and Bass,
telencephalic nuclei than a nonterritorial species and that 2006), and similar results were found in females and males
aggression correlated positively with the number of POA of the cichlid N. pulcher (Reddon et al., 2012). Further studies
gigantocellular AVT cells and negatively with the number are needed before a role for IT in the modulation of fish aggres-
and size of POA parvocellular AVT cells (Dewan and Tricas, sion can be established.
2011; Dewan et al., 2008). In the C. nevadensis amargosae, Finally, hormones involved in somatic growth, in particular
telencephalic proAVT mRNA levels were found to be elevated the growth hormone (GH) and somatostatin (SS), have also
in subordinate males and to correlate with aggression. These been proposed as modulators of aggressive behavior in fish.
males also had higher V1a1 receptor transcript levels in the These hormones are synthesized at the level of the hypothal-
telencephalon and hypothalamus, as compared with amus projecting to somatotropes in the pituitary. In
dominant males. On the other hand, the levels of proAVT A. burtoni, SS seems to inhibit the expression of aggressive
mRNA were fourfold higher in the hypothalamus of behavior in a dose-dependent fashion and independently of
dominant males, which also had higher levels of any potential effect in gonadal androgen secretion (Trainor
hypothalamic V1a2 receptor transcript abundance (Lema and Hofmann, 2006). In the rainbow trout, peripheral admin-
et al., 2015). This may be interpreted as hypothalamic AVT istration of GH was shown to increase male aggression, but this
playing a role in the expression of aggressive behavior in was interpreted as an indirect effect as GH also increased swim-
dominant males via the activation of the V1a2 receptor, ming activity that promoted agonistic encounters (Jönsson
while AVT action in forebrain targets would promote et al., 1998). Later, Jönsson et al. (2003) confirmed this
aggression only in subordinate animals. Greenwood et al. hypothesis by administering GH directly into the third ventricle
(2008) showed an opposite pattern of AVT mRNA of juvenile rainbow trout and observing also an increase in
expression in A. burtoni parvocellular and magnocellular swimming activity. Interestingly, in the A. burtoni study, only
subpopulations of AVT cells, with territorial males having chasing behavior, and not threatening behavior, was affected
Neuroendocrinology of Social Behavior in Teleost Fish 9

by SS. As SS is known to inhibit the release of GH, the inhibi- maintained (e.g., G. aculeatus, Páll et al., 2002; T. bifasciatum,
tory effects of SS in aggressive displays could be explained by Semsar and Godwin, 2003). Also, exogenous administration
a decrease in general locomotor activity induced by a reduction of androgens either to gonadectomized or to intact males has
in GH levels. These data are also contradictory to the findings variable effects on male mating displays (for a list of studies,
of Hofmann and Fernald (2000) showing that dominant see Oliveira and Gonçalves, 2008). For example, androgen
males have larger SS immunoreactive neurons in the POA as administration to gonadectomized males of M. americana
compared with subordinate animals, suggesting that SS restored sexual displays, with 11KT being more effective than
administration should increase aggression if it is directly T (Salek et al., 2001), while 11KT administration to intact
related with the endocrine regulation of these behaviors. males of the rock-pool blenny Parablennius parvicornis failed
to promote sexual behavior (Ros et al., 2004). Finally, pharma-
cologically blocking androgen receptors decreased male
2.01.3.2 Mating Behavior
nesting behavior in G. aculeatus (Sebire et al., 2008) and male
The brain regions associated with the expression of sexual courtship displays in the guppy Poecilia reticulata (Baatrup
behaviors have been extensively described in vertebrates, and Junge, 2001), in agreement with the hypothesis that
including fish (for a related review, see Forlano and Bass, male mating behaviors are directly facilitated by androgens.
2011). Notoriously, the POA and AH are known to be central The administration of androgens to juveniles, parasitic
brain regions for the control of reproduction as they contain ‘sneaker’ males or females, all phenotypes with lower plasma
the GnRH neurons that command the release of the gonadotro- androgen levels than males, likewise produced variable results.
pins LH and FSH from the pituitary, regulating gonadal develop- In the peacock blenny, T implants inhibited female-like
ment and secondarily gonadal steroid secretion. These regions displays in castrated parasitic males but failed to promote nest-
also synthesize neuropeptides relevant for reproduction and ing male behaviors (Gonçalves et al., 2007), while in C. auratus
are rich in sex steroid receptors. Early studies highlighted the T and 11KT implants given to intact females induced the full-
role of this area in reproduction by showing that electrical stim- suite of male sexual behaviors (Stacey and Kobayashi, 1996).
ulation of the POA induced reproductive behaviors in male The variable results obtained between studies of the effects of
bluegill sunfish Lepomis macrochirus (Demski and Knigge, androgens on male sexual behavior surely have multiple causes,
1971), results later confirmed in the hime salmon O. nerka including differences in hormone concentrations, type of
(Satou et al., 1984). As mentioned above, females of the cichlid androgens and antiandrogens used, hormone delivery mode,
A. burtoni, observing their preferred male winning fights acti- species-specific differences, season when experiments were per-
vated the POA (as measured by IEG expression) and also the formed or duration of exposure to the hormone, just to mention
Vm, another area implicated in female sexual displays in verte- a few. Nevertheless, the overall pattern suggests that androgens
brates (Desjardins and Fernald, 2010). In a study in female have a positive effect on male sexual displays also in fish.
Medaka Oryzias latipes, mating induced widespread c-fos expres- The central effects of androgens on male displays and in
sion in the POA, telencephalon, optic tectum, and cerebellum particular their potential action in the nodes of the SDMN
(Okuyama et al., 2011), suggesting the implication of a wide- are still poorly understood. In mammals, aromatization of
spread set of brain nuclei in female mating behavior. In addition T into E2 in the brain plays a crucial role in the regulation
to these central brain regions, hormones can also modulate of male sexual behavior (reviewed in Ball and Balthazart,
reproductive displays by acting in sensory or effector systems. 2004; Baum, 2003). However, although aromatase is abun-
As an example, in female midshipman, E2 acts in the inner dant in the areas of the SDMN and partly colocalizes with
ear’s sacculus to increase the degree of temporal encoding of androgen receptors (Forlano et al., 2010; e.g., Gelinas and
the frequency content of male vocalizations (Sisneros et al., Callard, 1997; Harbott et al., 2007), the evidence for aroma-
2004), thus synchronizing female phonotaxis and receptivity tization playing a role in the activation of male sexual
with maturation of the ovaries (for a review, see Sisneros, 2009). displays in fish via local conversion of T into E2 is less
Both female and male reproductive behaviors are expected obvious than in birds or mammals. In fact, in some studies
to be coordinated with gonadal function, and thus hormones the nonaromatizable 11KT seems to have a more effective
of gonadal origin, in particular sex steroids for males and sex role in the induction or recovery of male sexual displays
steroids and prostaglandins for females, have been seen as than the aromatizable T (e.g., Stacey and Kobayashi, 1996),
main candidates for endocrine regulation of reproductive and pharmacologically blocking aromatization with Fadro-
behaviors in fish. zole was shown to inhibit male displays in P. reticulata
The main androgens detected in fish plasma are T, 11KT, (Hallgren et al., 2006) but not in A. burtoni (Huffman
and 11b-hydroxytestosterone (Borg, 1994). Males have usually et al., 2013). Also, exposure to estrogens or xenoestrogens
higher plasma levels of 11KT than females while T levels often generally reduces male sexual displays (e.g., P. reticulata,
do not differ between sexes (Borg, 1994; Lokman et al., 2002). Bayley et al., 1999; C. auratus, Bjerselius et al., 2001; Colman
The impact of manipulating androgen levels in male reproduc- et al., 2009; D. rerio, Pradhan and Olsson, 2015). Further-
tive displays is highly variable (see Oliveira and Gonçalves, more, while androgens masculinize the electric organ
2008 for a review). While gonadectomy is effective in reducing discharge signal in ghost knifefishes, estrogens feminize it
plasma androgen levels (e.g., Almeida et al., 2014; Gonçalves (reviewed in Smith, 2013). In zebrafish, E2 seems to femi-
et al., 2007; Salek et al., 2001), in some cases a concomitant nize the male brain and 11KT to masculinize the female
reduction in reproductive behaviors occurs (e.g., Oreochromis brain, as assessed by gene transcriptomic profiling (Pradhan
mossambicus, Almeida et al., 2014; G. aculeatus, Hoar, 1962; and Olsson, 2015), further supporting a direct action of
Morone americana, Salek et al., 2001) while in others they are androgens on male reproductive behavior.
10 Neuroendocrinology of Social Behavior in Teleost Fish

Male sexual displays are often more elaborated than female including the possibility of direct neural communication
displays, and consequentially there are more published studies between the gonads and the brain via the vagal nerve, which
testing the effect of endocrine manipulations in sexual behavior would induce neural synthesis of PGF2a and the activation
in males than in females. The regulation of female sexual of female reproductive displays (Juntti et al., 2016), or the acti-
behavior was initially hypothesized to be controlled by gonadal vation of brain PGF2a synthesis by other ovarian hormones.
hormones and determined by the mode of reproduction For example, in mammals, E2 has been found to promote
(Stacey, 1981). In internal fertilizers, sexual behavior and fertil- the synthesis of prostaglandins both in the uterus (PGF, Ham
ization are temporally dissociated, and sex steroids were et al., 1975) and in the POA (PGE2, Amateau and McCarthy,
proposed to regulate female displays. By contrast, in external 2002). It seems possible that the above-described positive
fertilizers, female sexual behavior was considered to be mostly effects of both E2 and PGF2a in the activation of sexual
restricted to oviposition, which may be regarded as homologous displays in ovariectomized females of S. pavo could occur via
to parturition. Thus ovarian prostaglandins, which induce a direct effect of PGF2a in the brain and to a stimulatory
uterine contractions in mammals and oviposition in fishes effect of E2 in the neural synthesis of PGF2a. Studies investi-
(Jalabert and Szöllösi, 1975), were proposed to modulate gating in further detail the interconnection between sex
female spawning behaviors. This idea was originally proposed steroids and prostaglandins in fish, in particular the effects of
based mainly on data for P. reticulata, an internal fertilizer, gonadal steroids in brain PGF2a synthesis and receptor expres-
and C. auratus, an external fertilizer, but new evidence suggests sion, and the modulation by PGF2a of SDMN nodes look
that the sex steroid and prostaglandin pathways may actually like promising venues for future research.
be complementary. In a sex-role reversed population of the Nevertheless, evidence that the expression of sexual
peacock blenny, an external fertilizer, females court males with behavior in fish does not rely on hormones of gonadal origin
very elaborate displays (Gonçalves et al., 1996), providing an was obtained by Godwin et al. (1996) who showed that, in
opportunity to test the effects of endocrine manipulations in the sex-changing wrasse T. bifasciatum, females could rapidly
female sexual behavior. Ovariectomy was effective in quantita- occupy a territory left vacant by the removal of the dominant
tively reducing the expression of female courtship displays and male and express male courtship and spawning displays in
nuptial coloration 2 weeks after the removal of the ovaries but the absence of gonads. Hypothalamic abundance of proAVT
still a majority of ovariectomized females (9 out of 13) courte- mRNA in the brain of these females during sex change increases
d at least once a nesting male, showing that removal of the fourfold when compared with nonchanging females (Godwin
ovaries does not completely suppress the expression of sexual et al., 2000) and is higher in ovariectomized dominant
behavior (Gonçalves et al., 2014). Interestingly, both the steroid females than in subordinate females (Semsar and Godwin,
E2 and the prostaglandin PGF2a restored female sexual displays 2003), suggesting that AVT may be a key peptide regulating
(but not nuptial coloration), although PGF2a was more effec- the transition into male sexual displays in this species.
tive than E2 in this respect. In the same population, parasitic However, the effect of AVT seems to be context dependent as
males that mimic female displays occur and the removal of AVT administration failed to induce male sexual behaviors
the testes in these males has the paradoxical effect of promoting when a dominant male was present (Semsar and Godwin,
even further the expression of female-like behaviors (Gonçalves 2004). Interestingly, 11KT administration also promoted
et al., 2007). This is probably explained by the fact that andro- male displays in subordinate ovariectomized females but did
gens suppress female-like displays in sneaker males (Gonçalves not change AVT hypothalamic levels (Semsar and Godwin,
et al., 2007; Oliveira et al., 2001), and removing the testes 2004, 2003), suggesting that gonadal androgens modulate
reduces circulating androgen levels, releasing this inhibition male reproductive displays via a pathway independent from
(Gonçalves et al., 2007). Because E2 levels are very low in AVT. The positive effects of AVT on male sexual displays are
sneaker males and E2 administration does not promote confirmed by studies in other species (e.g., A. leptorhynchus,
female-like displays (Gonçalves et al., 2007), other neuroendo- Bastian et al., 2001; white perch, M. americana, Salek et al.,
crine mechanisms are proposed to regulate these behaviors (see 2001). For example, in the gymnotiform Brachyhypopomus gau-
below). derio, AVT stimulated the production of electric signals used for
Prostaglandins have been shown to promote female courtship by direct action on the hindbrain pacemaker cells
displays in other externally fertilizing species, including the (Perrone et al., 2010). However, in the cichlid A. burtoni, block-
paradise fish, Macropodus opercularis (Villars et al., 1985), the ing the V1a receptor inhibited aggression and stimulated court-
black acara Cichlasoma bimaculatum (Cole and Stacey, 1984), ship displays in subordinate males that ascended to become
the barb Puntius gonionotus (Liley and Tan, 1985), the cichlid dominant, suggesting that AVT impairs male reproductive
A. burtoni (Kidd et al., 2013) and, notoriously, the goldfish displays in this context, although the same manipulation did
C. auratus (reviewed in Kobayashi et al., 2002). Recently, Juntti not produce any effects in stable dominant or subordinate
et al. (2016) confirmed this role of PGF2a in A. burtoni by fish (Huffman et al., 2015).
showing that the expression of a putative PGF2a receptor in AVT has also been shown to promote female displays. In
areas such as vagal lobe and POA increase during spawning a sex-role reversed population of the peacock blenny S. pavo,
and that the activation of this receptor is needed for spawning AVT mRNA levels were higher in the courting morphs, i.e., in
behavior to occur. This evidence suggests that ovarian prosta- females and in female-mimicking parasitic males, than in
glandins act in external fertilizing teleost species, and probably noncourting nesting males (Grober et al., 2002). Accordingly,
also in amphibians, as a short-duration endogenous messenger AVT administration promoted sexual displays in females and
to synchronize sexual behavior with the presence of mature in parasitic males but not in nesting males (Carneiro et al.,
oocytes in the ovaries. However, alternative explanations exist 2003).
Neuroendocrinology of Social Behavior in Teleost Fish 11

Taken together, these results suggest that AVT promotes Schradin and Anzenberger, 1999), including fish (reviewed in
sexual behavior in fishes and, following the model proposed Whittington and Wilson, 2013). The first study on the effects
by Godwin and Thompson (2012), this is probably achieved of this hormone in fish paternal behavior was conducted in
via modulation of central brain areas, including nodes of the the ocellated wrasse Symphodus ocellatus where PRL administra-
SDMN, by AVT projections from the POA. tion was shown to promote egg fanning behavior in nesting
males (Fiedler, 1962). These results were corroborated by
studies in other species, both in males (e.g., blue discus, Sym-
2.01.3.3 Parental Care
physodon aequifasciata, Blüm and Fiedler, 1965; L. macrochirus,
The wide diversity of modes of reproduction in fishes extends Kindler et al., 1991; G. aculeatus, Páll et al., 2004) and females
to the patterns of care to eggs or juveniles, and examples of (e.g., S. aequifasciata, Blüm and Fiedler, 1965). However, there
no care, paternal, maternal, biparental, or even alloparental are also studies where the expected positive effect of PRL on
care can be found (Breder and Rosen, 1966). This offers an parental behavior was not observed. In the cooperatively
excellent opportunity to study the proximal mechanisms of breeding cichlid N. pulcher, PRL mRNA levels in the pituitary
parental care in vertebrates but not much is known on the brain were not elevated in breeding females as compared with
substrates of parental care in fish. In mammals, the POA, thal- nonbreeding females, and PRL administration to both males
amus, BNST, and the LS have been associated with the expres- and females did not promote parental behavior (Bender
sion of paternal care (for a review, see Dulac et al., 2014). In et al., 2008). In the Nile tilapia Oreochromis niloticus, pituitary
fish, the putative homologue for the LS is the Vv, but an equiv- and plasma levels of the two PRL isoforms described in fish
alent area to the BNST is still ambiguous. Also, Dc, the central also did not differ between female mouthbrooding eggs and
part of the dorsal telencephalon, has been suggested to relate nonincubating females (Tacon et al., 2000). Nevertheless,
with paternal care in bluegill sunfish (Demski and Knigge, PRL II showed high variation during the mouthbrooding
1971). In the only study so far investigating the neural period, and the authors did not exclude a possible role of
substrates of parental care in fish using IEG, it was shown in PRL in the regulation of maternal behavior. In a comparative
the cichlid A. nigrofasciata that parental males have a higher study between a monogamous and a polygynous species of
activation of the Vv, but not of the POA or Dc, as compared cichlids of the genus Herichthys, gene expression levels of PRL
with nonparental males (O’Connell et al., 2012). and of a PRL receptor in brain macroareas were not associated
Paternal care is more common than maternal care (Breder with paternal care (Oldfield et al., 2013).
and Rosen, 1966), and thus the endocrine regulation of Interestingly, sex steroids have been found to interact with
parental behavior has been mainly investigated in males. A PRL, raising the possibility of modulation of parental behavior
conceptual framework for the relationship between androgens by sex steroids occurring indirectly via effects in PRL. Estrogens
and parental care was proposed by Wingfield (1990), following have been found to promote the secretion of PRL from pitui-
the observation in birds that male androgen levels decrease tary glands (e.g., O. mossambicus Barry and Grau, 1986; rainbow
during the parenting phase as compared with the mating phase, trout O. mykiss, Williams and Wigham, 1994). Onuma et al.
even in animals that continue to reproduce after the initiation (2005) report variable effects of E2, T, and 11KT in PRL
of parental care (Wingfield, 1984). This hypothesis postulates mRNA expression levels in pituitary cell cultures of masu
that androgen levels should be lower during the paternal phase, salmon depending on gender and time in the reproductive
when social instability is usually reduced, as compared to the season. During the prespawning stage, E2, T, and 11KT
mating phase, when males need to compete for territories increased the expression of PRL mRNA while opposite effects
and sexual partners. In fish, androgen levels drop during the were detected during the spawning stage, suggesting that sex
parental phase thus supporting this prediction of the challenge steroids may regulate both positively and negatively PRL secre-
hypothesis (Oliveira et al., 2002). However, there are many tion. This study also highlights the importance of integrating
exceptions to this pattern (e.g., Rodgers et al., 2006; Ros variation in tissue sensitivity to modulatory agents to under-
et al., 2003), and exogenous administration of androgens to stand their mode of action.
parenting males failed to have the expected suppressive effect Similar to what has been described for oxytocin in
in parental behavior in some species (T propionate implants mammals, IT has also been found to regulate paternal care in
in L. macrochirus, Rodgers et al., 2012; e.g., 11KT implants in fish. In the monogamous cichlid, A. nigrofasciata, c-fos expres-
Parablennius sanguinolentus parvircornis, Ros et al., 2004). In sion was higher in POA parvocellular IT neurons in fathers
the context of the challenge hypothesis, the regulation of than in nonfathers, and the administration of an IT receptor
paternal behavior by androgens is interpreted as a trade-off antagonist blocked paternal care (O’Connell et al., 2012).
between parental behavior and androgen-induced aggression. Interestingly, IT fibers and IT receptors are present in the Vv,
The decrease in androgen levels postulated to occur during a brain area observed to be more activated in fathers than in
the parental phase would decrease the frequency of aggressive nonfathers, raising the possibility of stimulatory effects of IT
behaviors, releasing more energy and time to parental duties. on parental behavior occurring via modulation of neuronal
Androgens may thus be better seen as secondary modulators signals in this brain region (O’Connell et al., 2012).
of parental behaviors, more related with aggressive displays,
while other hormones are expected to have a more direct regu-
2.01.3.4 Prosocial Behavior
latory action on specific parental care displays.
Prolactin (PRL) has been suggested as a prime candidate for The idea that social bonding, or affiliative behavior, can be regu-
the endocrine modulation of both paternal and maternal lated by evolutionary conserved brain and neurochemical
behavior across vertebrates (e.g., Bachelot and Binart, 2007; systems across vertebrates is relatively new. In fact, since the
12 Neuroendocrinology of Social Behavior in Teleost Fish

ground-breaking research on the role of AVP and oxytocin in Information on the brain areas targeted by AVT or IT to
the regulation of social behavior in rodents (reviewed in Young regulate social bonding is still very scarce but Godwin and
et al., 2011), evidence has been accumulating showing that Thompson (2012) suggest forebrain nodes of the SDMN to
these nonapeptides, and their nonmammalian homologues, be likely candidates for AVT modulation; indeed motor output
play an important role in social bonding, affiliative behavior, pathways descending from these regions show dense AVT
and attention to social stimuli also in other taxa, including innervation and project into multiple central targets. New
fish (reviewed in Godwin and Thompson, 2012). In goldfish, studies selectively manipulating subpopulations of AVT cells
intracerebroventricular administration of AVT and IT has oppo- will be able to test this hypothesis.
site effects in social behavior, with AVT-reducing approach
behavior toward a conspecific and IT increasing the duration
of this behavior (Thompson and Walton, 2004). The inhibitory 2.01.4 Hormones as Social Semiochemicals
effects of AVT seemed to be mediated by AVT cell projections
from the POA to the hindbrain (Thompson and Walton, When hormones are released into the environment, either
2009). This was confirmed by showing that the behavioral actively or passively, they convey information about the sender
effects of AVT infusions into the fourth ventricle were more that becomes potentially available to other conspecifics, and
potent than the effects of infusions into the third ventricle thus can be seen as social semiochemicals (i.e., chemical cues
(Thompson et al., 2008). Accordingly, it was demonstrated of conspecific origin). If released hormones have evolved to
that seasonal changes in behavioral responsiveness to AVT are convey information about the sender and trigger a specific
associated with changes in hindbrain sensitivity to AVT, as and adaptive response in the receivers, then they can be viewed
measured by the expression of the V1a AVT receptor in this as pheromones (Sorensen, 2014; Wyatt, 2010). Thus, the
brain region (Walton et al., 2010). In the cichlid N. pulcher, evolution of hormonal pheromones has been proposed to
peripheral administration of IT seemed to increase responsive- follow a cue-signal continuum, where different evolutionary
ness to socially relevant information during aggressive contests states can be recognized (Sorensen and Scott, 1994; Sorensen
as IT-treated fish fought in accordance with the size of the and Stacey, 1999; Wisenden, 2014; Figure 2). In the ancestral
opponent while control animals fought according to their state, senders release hormones into the environment, but
intrinsic aggressive levels (Reddon et al., 2012). In the potential receivers are unable to detect them. In a second state,
monogamous cichlid A. nigrofasciata, the peripheral receivers evolved the capacity to detect and respond adaptively
administration of an AVP/IT receptor antagonist to males to the hormone; hence they are now spying on senders based
reduced affiliative behavior although it did not prevent pair on the hormone that acts as a chemical cue for the internal
bonding nor did it disrupt pair bonding after pairs had been state of senders. Finally, a third evolutionary state may evolve
established (Oldfield and Hofmann, 2011). In the cleaner if the selective pressure imposed by spying on senders leads
wrasse, Labroides dimidiatus, AVT inhibited interspecific to the evolution of specialized production and/or release of
cleaning activities while it did not affect other social the hormonal pheromones, which becomes a signal (i.e.,
conspecific behaviors (Soares et al., 2012). In the same study, true pheromone) according to animal communication termi-
IT administration failed to affect social behavior. In contrast nology (Wisenden, 2014).
with previous studies, in N. pulcher, IT administered An apparently easy way to discriminate between spying and
intraperitoneally reduced affiliative behavior and, in a second true signaling would be to look for the occurrence of special-
study, brain IT levels were found to be negatively correlated ized structures in the production and/or release of phero-
with these behaviors (Reddon et al., 2015). mones. Many such structures have been described among

(a) Ancestral state (b) Spying state (c) CommunicaƟon state


No communicaƟon Change in receiver Change in sender

Hormone released into water Hormone release unchanged Change in hormone and/or its
release

Chemical Cue Signal


Response

No mechanism
h for
f detecƟon/
d / response. EvoluƟon of detecƟon and response. Response off the
h receiver selects
l ffor
signal specializaƟon

Figure 2 Stages in the evolution of chemical signaling in fish: (a) ancestral state, in which fish release a hormone that is not detected by conspe-
cifics; (b) spying state, where conspecifics are able to detect and respond adaptively to the hormone; and (c) communication state, characterized by
a response of the receiver which selects for signal specialization.
Neuroendocrinology of Social Behavior in Teleost Fish 13

teleost fish, such as the seminal vesicles in catfish (Clarias garie- spying on reproductive state of females in order to increase
pinus) that release female attractants (Resink et al., 1989), the their reproductive success (Stacey and Sorensen, 2002;
hypertrophied mesorchial glands in the testes of gobies that Wisenden and Stacey, 2005). However, subsequently it has
secrete steroids that attract females (e.g., Colombo et al., been found that these hormones are mainly released through
1980; Murphy et al., 2001), or anal glands in blennies, which the urine and that female goldfish increase the frequency of
consist of a transformation of the first rays of the anal fin pulses of urine when in the presence of a male, in particular
into a sex pheromone-producing gland (Serrano et al., if in the presence of oviposition substrate (Appelt and
2008a,b). Interestingly, in the case of both gobies and blennies, Sorensen, 2007), indicating a specialization in the sender for
i.e., in species in which male alternative reproductive tactics the release of the signal, compatible with a true signaling
occur and both territorial and sneaker males are present, only scenario. This form of control of signal release is also present
territorial males develop the glands that produce female attrac- in other species where hormonal pheromones are released
tants (blennies: Gonçalves et al., 1996; gobies: Locatello et al., through the urine and males adjust the urination rate in
2002), suggesting a secondary loss of the pheromone- response to the presence of receptive/preovulatory females
producing tissue in sneakers. However, the absence of such (e.g., Mozambique tilapia, O. mossambicus, Almeida et al.,
specialized structures cannot by itself rule out the ability to 2005; Burton’s mouthbrooder, A. burtoni, Maruska and Fernald,
release/store pheromones, and thus apparent cases of chemical 2012; swordtail, Xiphophorus birchmanni, Rosenthal et al.,
spying in fish may just reflect our failure to detect these more 2011). Thus, the role of hormones as pheromones may be
subtle specializations. A good example of such scenario is the more common than initially thought.
goldfish (C. auratus), where, despite lacking any obvious Most of the examples of hormonal pheromones provided
specialization for production and release of pheromones, above are related to reproduction and to their effect on the
females release sequentially two hormonal pheromones: (1) behavior and physiology of the opposite sex. However, evidence
a preovulatory pheromone, consisting of progestins (17,20-b- has accumulated indicating a role for chemical cues in other
dihydroxy-4-pregen-3-one and its sulfated form) and androste- aspects of fish social behavior such as intrasexual aggression,
nedione, that has a primer effect on sperm production in parental care, and affiliative behaviors (Keller-Costa et al.,
males; and (2) a postovulatory pheromone, consisting of pros- 2015; e.g., Sorensen and Baker, 2014). Unfortunately, for most
taglandins (prostaglandin F2-a and 15-keto-PGF2-a), that trig- of these other cases of chemical communication, the chemical
gers male courtship and makes ovulated females attractive to identity of the cue/signal is not known, and thus hormones
the males (reviewed in Stacey and Sorensen, 2002). Given cannot be directly implicated. One particular function that has
that these female pheromones are the result of passive release been the focus of recent research is the role of chemical commu-
into the water of sex hormones and their metabolites involved nication in the regulation of male–male aggression in cichlids
in female ovulation (progestins and androstenedione) and in (Keller-Costa et al., 2015). In the Mozambique tilapia, males
the regulation of female reproductive behavior (prostaglan- also release urine during agonistic interactions in pulses of short
dins), and there are no specialization for the production and/ duration and those that become subordinate stop releasing
or their release in the scope of chemical communication, this urine (Figure 3; Barata et al., 2007). If urination is surgically
system has initially been considered a classic example of males prevented, agonistic interactions escalate and more overt

Figure 3 Examples (1–3) of behavior of two territorial tilapia males (a) and (b) interacting for 45 min (submissive: white; not aggressive: light gray;
aggressive displays: dark gray; highly aggressive: black) and their corresponding release of urine pulses (urination). In (1), male (a) started aggres-
sive behavior and urination (time point around 25 min), subsequently male (b) initiated aggressive displays and then agonistic interaction escalated
to high symmetrical aggression. In (2), both males increased their urination frequency and aggressivity almost at the same time (within 10–15) and
maintained agonistic behavior until the end of experiment, although urine pulses decreased significantly. In (3), after the release of several urine pul-
ses from both males and a short period of symmetrical high aggression, male (a) became submissive whereas male (b) continued with agonistic
displays; both males stopped urine release at this time point (around 18 min). Adapted from Barata, E.N., Hubbard, P.C., Almeida, O.G., Miranda, A.,
Canário, A.V.M., 2007. Male urine signals social rank in the Mozambique tilapia (Oreochromis mossambicus). BMC Biol. 5, 1–11.
14 Neuroendocrinology of Social Behavior in Teleost Fish

aggressive behaviors are expressed (Keller-Costa et al., 2012). Almeida, O.G., Miranda, A., Hubbard, P.C., Frade, P., Barata, E.N., Canário, A.V.M.,
Moreover, in stable dominance hierarchies the olfactory 2005. Urine as a social signal in the Mozambique tilapia (Oreochromis mos-
sambicus). Chem. Senses 30, i309–i310.
potency of the urine is correlated with the male’s social rank
Amateau, S.K., McCarthy, M.M., 2002. A novel mechanism of dendritic spine plasticity
(Barata et al., 2007), and urine of dominant and subordinate involving estradiol induction of prostaglandin-E2. J. Neurosci. 22, 8586–8596.
males triggers different patterns of gene expression in olfactory Appelt, C.W., Sorensen, P.W., 2007. Female goldfish signal spawning readiness by
brain regions of male receivers (Simões et al., 2015). Together altering when and where they release a urinary pheromone. Anim. Behav. 74,
these results strongly suggest the presence of a chemical signal 1329–1338.
Baatrup, E., Junge, M., 2001. Antiandrogenic pesticides disrupt sexual character-
in the urine used in social status assessment in this species. istics in the adult male guppy (Poecilia reticula). Environ. Health Perspect. 109,
Interestingly, dominant males also store large volumes of urine 1063–1069.
(Barata et al., 2007), having hypertrophied urinary bladders Bachelot, A., Binart, N., 2007. Reproductive role of prolactin. Reproduction 133,
(Keller-Costa et al., 2012), which indicates a specialization for 361–369.
Badura, L.L., Friedman, H., 1988. Sex reversal in female Betta splendens as a function
urine release supportive of the evolution of a true dominance
of testosterone manipulation and social influence. J. Comp. Psychol. 102,
pheromone rather than just a dominance cue. The chemical 262–268.
identity of this putative dominance pheromone has not Baker, B.I., Bird, D.J., Buckingham, J.C., 1996. In the trout, CRH and AVT synergize to
been established yet, but it is known to have multiple compo- stimulate ACTH release. Regul. Pept. 67, 207–210.
nents, present both in polar and nonpolar urine fractions Ball, G.F., Balthazart, J., 2004. Hormonal regulation of brain circuits mediating male
sexual behavior in birds. Physiol. Behav. 83, 329–346.
(Keller-Costa et al., 2016) and to be distinct from the sex Barata, E.N., Hubbard, P.C., Almeida, O.G., Miranda, A., Canário, A.V.M., 2007. Male
pheromone that has primer physiological effects on females urine signals social rank in the Mozambique tilapia (Oreochromis mossambicus).
(Keller-Costa et al., 2014). Similar evidence for the occurrence BMC Biol. 5, 1–11.
of putative dominance pheromones has also been collected for Barry, T.P., Grau, E.G., 1986. Estradiol-17 beta and thyrotropin-releasing hormone
stimulate prolactin release from the pituitary gland of a teleost fish in vitro. Gen.
two other cichlid species (Burton’s mouthbrooder, Crapon de
Comp. Endocrinol. 62, 306–314.
Caprona, 1980; Maruska and Fernald, 2012; Nile tilapia, Bastian, J., Schniederjan, S., Nguyenkim, J., 2001. Arginine vasotocin modulates
Giaquinto and Volpato, 1997; Gonçalves-de-Freitas et al., a sexually dimorphic communication behavior in the weakly electric fish Apter-
2008), suggesting that pheromones may be also a widespread onotus leptorhynchus. J. Exp. Biol. 204, 1909–1923.
phenomena in this teleost family. Baum, M.J., 2003. Activational and organizational effects of estradiol on male
behavioral neuroendocrine function. Scand. J. Psychol. 44, 213–220.
Bayley, M., Nielsen, J.R., Baatrup, E., 1999. Guppy sexual behavior as an effect
biomarker of estrogen mimics. Ecotoxicol. Environ. Saf. 43, 68–73.
2.01.5 Conclusions Bell, A.M., 2001. Effects of an endocrine disrupter on courtship and aggressive
behaviour of male three-spined stickleback, Gasterosteus aculeatus. Anim. Behav.
62, 775–780.
To conclude, it is now clear that hormones exert powerful Bender, N., Taborsky, M., Power, D.M., 2008. The role of prolactin in the regulation of
modulatory effects on social behavior by acting on a core of brood care in the cooperatively breeding fish Neolamprologus pulcher. J. Exp. Zool.
forebrain and midbrain areas that underlie the expression of A Ecol. Genet. Physiol. 309, 515–524.
Berthold, A.A., 1849. Transplantation in the hoden. Arch. Anat. Physiol. Wiss. Med.
these behaviors in fishes. However, the exact mechanism
42–46.
through which hormones change the functional connectivity Bjerselius, R., Lundstedt-Enkel, K., Olsén, H., Mayer, I., Dimberg, K., 2001. Male
of the SDMN to affect behavior remains poorly understood, goldfish reproductive behaviour and physiology are severely affected by exogenous
and this is a promising area for research. Studies manipulating exposure to 17b-estradiol. Aquat. Toxicol. 53, 139–152.
hormone levels and investigating the effect in the neuroge- Black, M.P., Balthazart, J., Baillien, M., Grober, M.S., 2005. Socially induced and
rapid increases in aggression are inversely related to brain aromatase activity in
nomic states and neuronal output of nodes of the SDMN a sex-changing fish, Lythrypnus dalli. Proc. Biol. Sci. 272, 2435–2440.
will help to elucidate how hormones modulate the expression Blüm, V., Fiedler, K., 1965. Hormonal control of reproductive behavior in some
of social behaviors. Also, different hormonal systems are cichlid fish. Gen. Comp. Endocrinol. 5, 186–196.
known to interact with each other, and studies addressing the Borg, B., 1994. Androgens in teleost fishes. Comp. Biochem. Physiol. Part C Comp.
109, 219–245.
cross-talk between endocrine systems are valuable. Finally,
Breder, C.M., Rosen, D.E., 1966. Modes of Reproduction in Fishes. T.F.H. Publica-
hormones may also act on social behavior by acting as phero- tions, New Jersey.
mones, and their role in the regulation of social interactions Bromberg-Martin, E.S., Matsumoto, M., Hikosaka, O., 2010. Dopamine in motiva-
has started to be unveiled. tional: rewarding, aversive, and alerting. Neuron 68, 815–834.
Carneiro, L.A., Oliveira, R.F., Canário, A.V.M., Grober, M.S., 2003. The effect of
arginine vasotocin on courtship behaviour in a blenniid fish with alternative
reproductive tactics. Fish Physiol. Biochem. 28, 241–243.
Acknowledgments Clotfelter, E.D., Rodriguez, A.C., 2006. Behavioral changes in fish exposed to phy-
toestrogens. Environ. Pollut. 144, 833–839.
Cole, K.S., Stacey, N.E., 1984. Prostaglandin induction of spawning behavior in
During the writing of this review, RFO and ASF were supported by
Cichlasoma bimaculatum (Pisces Cichlidae). Horm. Behav. 18, 235–248.
Fundação para a Ciência e a Tecnologia (grants EXCL/BIA-ANM/0549/
Colman, J.R., Baldwin, D., Johnson, L.L., Scholz, N.L., 2009. Effects of the synthetic
2012 and SFRH/BD/102892/2014, respectively) and DG by Fundo para estrogen, 17alpha-ethinylestradiol, on aggression and courtship behavior in male
o Desenvolvimento das Ciências e da Tecnologia de Macau (grant 012/ zebrafish (Danio rerio). Aquat. Toxicol. 91, 346–354.
2012/A1). Colombo, L., Marconato, A., Belvedere, P.C., Friso, C., 1980. Endocrinology of teleost
reproduction: a testicular steroid pheromone in the black goby, Gobius jozo L.
Bolletino Zool. 47, 355–364.
Crapon de Caprona, M., 1980. Olfactory communication in a cichlid fish, Hap-
References lochromis burtoni. Z. Tierpsychol. 52, 113–134.
Demski, L.S., Knigge, K.M., 1971. The telencephalon and hypothalamus of the bluegill
Almeida, O., Canário, A.V.M., Oliveira, R.F., 2014. Castration affects reproductive but (Lepomis macrochirus): evoked feeding, aggressive and reproductive behavior with
not aggressive behavior in a cichlid fish. Gen. Comp. Endocrinol. 207, 34–40. representative frontal sections. J. Comp. Neurol. 143, 1–16.
Neuroendocrinology of Social Behavior in Teleost Fish 15

Desjardins, J.K., Fernald, R.D., 2010. What do fish make of mirror images? Biol. Lett. Goodson, J.L., Kabelik, D., 2009. Dynamic limbic networks and social diversity in
6, 744–747. vertebrates: from neural context to neuromodulatory patterning. Front. Neuro-
Desjardins, J.K., Hazelden, M.R., Van Der Kraak, G.J., Balshine, S., 2006. Male and endocrinol. 30, 429–441.
female cooperatively breeding fish provide support for the “Challenge Hypothesis.” Goodson, J.L., Kingsbury, M.A., 2013. What’s in a name? Considerations of homol-
Behav. Ecol. 17, 149–154. ogies and nomenclature for vertebrate social behavior networks. Horm. Behav. 64,
Desjardins, J.K., Klausner, J.Q., Fernald, R.D., 2010. Female genomic response to 103–112.
mate information. Proc. Natl. Acad. Sci. U.S.A. 107, 21176–21180. Greenwood, A.K., Wark, A.R., Fernald, R.D., Hofmann, H.a, 2008. Expression of
Dewan, A.K., Maruska, K.P., Tricas, T.C., 2008. Arginine vasotocin neuronal arginine vasotocin in distinct preoptic regions is associated with dominant and
phenotypes among congeneric territorial and shoaling reef butterflyfishes: subordinate behaviour in an African cichlid fish. Proc. Biol. Sci. 275,
species, sex and reproductive season comparisons. J. Neuroendocrinol. 20, 2393–2402.
1382–1394. Grober, M.S., George, A.A., Watkins, K.K., Carneiro, L.A., Oliveira, R.F., 2002.
Dewan, A.K., Tricas, T.C., 2011. Arginine vasotocin neuronal phenotypes and their Forebrain AVT and courtship in a fish with male alternative reproductive tactics.
relationship to aggressive behavior in the territorial monogamous multiband but- Brain Res. 57, 423–425.
terflyfish, Chaetodon multicinctus. Brain Res. 1401, 74–84. Hallgren, S.L.E., Linderoth, M., Olsén, K.H., 2006. Inhibition of cytochrome p450 brain
Diotel, N., Do Rego, J.L., Anglade, I., Vaillant, C., Pellegrini, E., Vaudry, H., Kah, O., aromatase reduces two male specific sexual behaviours in the male Endler guppy
2011. The brain of teleost fish, a source, and a target of sexual steroids. Front. (Poecilia reticulata). Gen. Comp. Endocrinol. 147, 323–328.
Neurosci. 19. Ham, E.a, Cirillo, V.J., Zanetti, M.E., Kuehl, F.a, 1975. Estrogen-directed synthesis of
Dulac, C., O’Connell, L.A., Wu, Z., 2014. Neural control of maternal and paternal specific prostaglandins in uterus. Proc. Natl. Acad. Sci. U.S.A. 72, 1420–1424.
behaviors. Science 345, 765–770. Harbott, L.K., Burmeister, S.S., White, R.B., Vagell, M., Fernald, R.D., 2007. Androgen
Fiedler, K., 1962. Die Wirkung von Prolactin auf das Verhalten des Lippfisches receptors in a cichlid fish, Astatotilapia burtoni: structure, localization, and
Crenilabrus ocellatus (Forksal). Zool. Jahrb. Abt. Anat. Zool. Physiol. Tiere 69, expression levels. J. Comp. Neurol. 504, 57–73.
609–620. Hawkins, M.B., Godwin, J., Crews, D., Thomas, P., 2005. The distributions of the
Filby, A.L., Paull, G.C., Hickmore, T.F., Tyler, C.R., 2010. Unravelling the neuro- duplicate oestrogen receptors ER-ba and ER-bb in the forebrain of the Atlantic
physiological basis of aggression in a fish model. BMC Genomics 11, 498. croaker (Micropogonias undulatus): evidence for subfunctionalization after gene
Filby, A.L., Paull, G.C., Searle, F., Ortiz-Zarragoitia, M., Tyler, C.R., 2012. Environmental duplication. Proc. R. Soc. B Biol. Sci. 272, 633–641.
estrogen-induced alterations of male aggression and dominance hierarchies in fish: Hirschenhauser, K., Oliveira, R.F., 2006. Social modulation of androgens in male
a mechanistic analysis. Environ. Sci. Technol. 46, 3472–3479. vertebrates: meta-analyses of the challenge hypothesis. Anim. Behav. 71,
Forlano, P.M., Bass, A.H., 2011. Neural and hormonal mechanisms of reproductive- 265–277.
related arousal in fishes. Horm. Behav. 59, 616–629. Hoar, W.S., 1962. Hormones and the reproductive behaviour of the male three-spined
Forlano, P.M., Deitcher, D.L., Bass, A.H., 2005. Distribution of estrogen receptor alpha stickleback (Gasterosteus aculeatus). Anim. Behav. 10, 247–266.
mRNA in the brain and inner ear of a vocal fish with comparisons to sites of Hofmann, H.A., Fernald, R.D., 2000. Social status controls somatostatin neuron size
aromatase expression. J. Comp. Neurol. 483, 91–113. and growth. J. Neurosci. 20, 4740–4744.
Forlano, P.M., Deitcher, D.L., Myers, D.A., Bass, A.H., 2001. Anatomical distribution Huffman, L.S., Hinz, F.I., Wojcik, S., Aubin-Horth, N., Hofmann, H.A., 2015. Arginine
and cellular basis for high levels of aromatase activity in the brain of teleost fish: vasotocin regulates social ascent in the African cichlid fish Astatotilapia burtoni.
aromatase enzyme and mRNA expression identify glia as source. J. Neurosci. 21, Gen. Comp. Endocrinol. 212, 106–113.
8943–8955. Huffman, L.S., O’Connell, L.a, Hofmann, H.a, 2013. Aromatase regulates aggression
Forlano, P.M., Marchaterre, M., Deitcher, D.L., Bass, A.H., 2010. Distribution of in the African cichlid fish Astatotilapia burtoni. Physiol. Behav. 112-113, 77–83.
androgen receptor mRNA expression in vocal, auditory, and neuroendocrine circuits Huffman, L.S., O’Connell, L.a., Kenkel, C.D., Kline, R.J., Khan, I.a., Hofmann, H.a,
in a teleost fish. J. Comp. Neurol. 518, 493–512. 2012. Distribution of nonapeptide systems in the forebrain of an African cichlid
Francis, R.C., Soma, K., Fernald, R.D., 1993. Social regulation of the brain-pituitary- fish, Astatotilapia burtoni. J. Chem. Neuroanat. 44, 86–97.
gonadal axis. Proc. Natl. Acad. Sci. U.S.A. 90, 7794–7798. Jalabert, B., Szöllösi, D., 1975. In vitro ovulation of trout oocytes: effect of prosta-
Gelinas, D., Callard, G.V., 1997. Immunolocalization of aromatase- and androgen glandins on smooth muscle-like cells of the theca. Prostaglandins 9, 765–778.
receptor-positive neurons in the goldfish brain. Gen. Comp. Endocrinol. 106, Jalabert, C., Quintana, L., Pessina, P., Silva, A., 2015. Extra-gonadal steroids
155–168. modulate non-breeding territorial aggression in weakly electric fish. Horm. Behav.
Giaquinto, P.C., Volpato, G.L., 1997. Chemical communication, aggression, and 72, 60–67.
conspecific recognition in the fish Nile tilapia. Physiol. Behav. 62, 1333–1338. Jönsson, E., Johansson, V., Björnsson, B., jorn, T., Winberg, S., 2003. Central nervous
Godwin, J., Crews, D., Warner, R.R., 1996. Behavioural sex change in the absence of system actions of growth hormone on brain monoamine levels and behavior of
gonads in a coral reef fish. Proc. Biol. Sci. 263, 1683–1688. juvenile rainbow trout. Horm. Behav. 43, 367–374.
Godwin, J., Sawby, R., Warner, R.R., Crews, D., Grober, M.S., 2000. Hypothalamic Jönsson, E., Johnsson, J.I., Björnsson, B.T., 1998. Growth hormone increases
arginine vasotocin mRNA abundance variation across sexes and with sex change in aggressive behavior in juvenile rainbow trout. Horm. Behav. 33, 9–15.
a coral reef fish. Brain Behav. Evol. 55, 77–84. Juntti, S.A., Hilliard, A.T., Kent, K.R., Kumar, A., Nguyen, A., Jimenez, M.A.,
Godwin, J., Thompson, R.R., 2012. Nonapeptides and social behavior in fishes. Horm. Loveland, J.L., Mourrain, P., Fernald, R.D., 2016. A neural basis for control of
Behav. 61, 230–238. cichlid female reproductive behavior by prostaglandin F2a. Curr. Biol. 26,
Gonçalves, D., Alpedrinha, J., Teles, M.C., Oliveira, R.F., 2007. Endocrine control of 943–949.
sexual behavior in sneaker males of the peacock blenny Salaria pavo: effects of Keller-Costa, T., Canário, A.V.M., Hubbard, P.C., 2015. Chemical communication in
castration, aromatase inhibition, testosterone and estradiol. Horm. Behav. 51, cichlids: a mini-review. Gen. Comp. Endocrinol. 221, 64–74.
534–541. Keller-Costa, T., Hubbard, P.C., Paetz, C., Nakamura, Y., da Silva, J.P., Rato, A.,
Gonçalves, D., Costa, S.S., Teles, M.C., Silva, H., Ingles, M., Oliveira, R.F., 2014. Barata, E.N., Schneider, B., Canario, A.V.M., 2014. Identity of a tilapia pheromone
Oestradiol and prostaglandin F2 regulate sexual displays in females of a sex-role released by dominant males that primes females for reproduction. Curr. Biol. 24,
reversed fish. Proc. R. Soc. B 281, 20133070. 2130–2135.
Gonçalves, D., Oliveira, R.F., 2010. Hormones and sexual behavior of teleost fishes. In: Keller-Costa, T., Lopes, O.S., Almeida, O., Hubbard, P.C., Iacovella, A., Lima, M.,
Norris, D.O. (Ed.), Hormones and Reproduction of Vertebrates – Volume 1-Fishes. Barata, E.N., Canário, A.V.M., 2012. Muscular hypertrophy of urinary bladders in
Elsevier, New York, pp. 119–147. dominant tilapia facilitates the control of aggression through urinary signals.
Gonçalves, E.J., Almada, V.C., Oliveira, R.F., Santos, A.J., 1996. Female mimicry as Behaviour 149, 953–975.
a mating tactic in males of the blenniid fish Salaria pavo. J. Mar. Biol. Assoc. U.K. Keller-Costa, T., Saraiva, J.L., Hubbard, P.C., Barata, E.N., Canário, A.V.M., 2016.
76, 529–538. A multi-component pheromone in the urine of dominant male Tilapia (Oreochromis
Gonçalves-de-Freitas, E., Teresa, F.B., Gomes, F.S., Giaquinto, P.C., 2008. Effect of mossambicus) reduces aggression in rivals. J. Chem. Ecol. 42, 173–182.
water renewal on dominance hierarchy of juvenile Nile tilapia. Appl. Anim. Behav. Kidd, M.R., Dijkstra, P.D., Alcott, C., Lavee, D., Ma, J., O’Connell, L.A., Hofmann, H.A.,
Sci. 112, 187–195. 2013. Prostaglandin F2alpha facilitates female mating behavior based on male
Goodson, J.L., 2005. The vertebrate social behavior network: evolutionary themes and performance. Behav. Ecol. Sociobiol. 67, 1307–1315.
variations. Horm. Behav. 48, 11–22. Kindler, P.M., Bahr, J.M., Gross, M.R., Philipp, D.P., 1991. Hormonal regulation of
Goodson, J.L., Bass, A.H., 2000. Forebrain peptides modulate sexually polymorphic parental care behavior in nesting male bluegills: do the effects of Bromocriptine
vocal circuitry. Nature 403, 769–772. suggest a role for prolactin? Physiol. Zool. 61, 310–322.
16 Neuroendocrinology of Social Behavior in Teleost Fish

Kittelberger, J.M., Land, B.R., Bass, A.H., 2006. Midbrain periaqueductal gray and Okuyama, T., Suehiro, Y., Imada, H., Shimada, A., Naruse, K., Takeda, H., Kubo, T.,
vocal patterning in a teleost fish. J. Neurophysiol. 96, 71–85. Takeuchi, H., 2011. Induction of c-fos transcription in the medaka brain (Oryzias
Kline, R.J., O’Connell, L.A., Hofmann, H.A., Holt, G.J., Khan, I.A., 2011. The distri- latipes) in response to mating stimuli. Biochem. Biophys. Res. Commun. 404,
bution of an avt v1a receptor in the brain of a sex changing fish, Epinephelus 453–457.
adscensionis. J. Chem. Neuroanat. 42, 72–88. Oldfield, R.G., Harris, R.M., Hendrickson, D.A., Hofmann, H.A., 2013. Arginine
Kobayashi, M., Sorensen, P.W., Stacey, N.E., 2002. Hormonal and pheromonal control vasotocin and androgen pathways are associated with mating system variation in
of spawning behavior in the goldfish. Fish Physiol. Biochem. 26, 71–84. North American cichlid fishes. Horm. Behav. 64, 44–52.
Kyle, A.L., Peter, R.E., 1982. Effects of forebrain lesions on spawning behaviour in the Oldfield, R.G., Hofmann, H.A., 2011. Neuropeptide regulation of social behavior in
male goldfish. Physiol. Behav. 28, 1103–1109. a monogamous cichlid fish. Physiol. Behav. 102, 296–303.
Lema, S.C., 2010. Identification of multiple vasotocin receptor cDNAs in teleost fish: Oliveira, R., Gonçalves, D., 2008. Hormones and social behaviour of teleost fish. In:
sequences, phylogenetic analysis, sites of expression, and regulation in the Fish Behaviour. Science Publishers, pp. 61–150.
hypothalamus and gill in response to hyperosmotic challenge. Mol. Cell. Endocrinol. Oliveira, R.F., 2004. Social modulation of androgens in vertebrates: mechanisms and
321, 215–230. function. In: Slater, P.J.B., Rosenblatt, J.S., Snowdown, C.T., Roper, T.J. (Eds.),
Lema, S.C., Nevitt, G.A., 2004. Exogenous vasotocin alters aggression during agonistic Advances in the Study of Behavior. Academic Press, New York, pp. 165–239.
exchanges in male Amargosa River pupfish (Cyprinodon nevadensis amargosae). Oliveira, R.F., 2005. Neuroendocrine mechanisms of alternative reproductive tactics in
Horm. Behav. 46, 628–637. fish. Fish. Physiol. 24, 297–357.
Lema, S.C., Sanders, K.E., Walti, K.A., 2015. Arginine vasotocin, isotocin and non- Oliveira, R.F., Canário, A., 2000. Hormones and social behavior of cichlid fishes:
apeptide receptor gene expression link to social status and aggression in sex- a case study in the Mozambique tilapia. J. Aquaric. Aquatic Sci. IX, 187–207.
dependent patterns. J. Neuroendocrinol. 27, 142–157. Oliveira, R.F., Carneiro, A., Gonçalves, D.M., 2001. 11-Ketotestosterone inhibits the
Liley, N.R., Tan, E.S.P., 1985. The induction of spawning behaviour in Puntius alternative mating tactic in sneaker males of the peacock blenny, Salaria pavo.
gonionotus (Bleeker) by treatment with prostaglandin PGF2a. J. Fish Biol. 26, Brain Behav. Evol. 2001, 28–37.
491–502. Oliveira, R.F., Hirschenhauser, K., Carneiro, L.A., Canario, A.V.M., 2002. Social
Locatello, L., Mazzoldi, C., Rasotto, M.B., 2002. Ejaculate of sneaker males is modulation of androgen levels in male teleost fish. Comp. Biochem. Physiol. B
pheromonally inconspicuous in the black goby, Gobius niger (Teleostei, Gobiidae). Biochem. Mol. Biol. 132, 203–215.
J. Exp. Zool. 293, 601–605. Oliveira, R.F., Silva, J.F., Simões, J.M., 2011. Fighting zebrafish: characterization of
Lokman, P.M., Harris, B., Kusakabe, M., Kime, D.E., Schulz, R.W., Adachi, S., aggressive behavior and winner-loser effects. Zebrafish 8, 73–81.
Young, G., 2002. 11-Oxygenated androgens in female teleosts: prevalence, Oliveira, R.F., Simões, J.M., Teles, M.C., Oliveira, C.R., Becker, J.D., Lopes, J.S.,
abundance, and life history implications. Gen. Comp. Endocrinol. 129, 1–12. 2016. Assessment of fight outcome is needed to activate socially driven tran-
Macey, M.J., Pickford, G.E., Peter, R.E., 1974. Forebrain localization of the spawning scriptional changes in the zebrafish brain. Proc. Natl. Acad. Sci. U.S.A. 113,
reflex response to exogenous neurohypophysial hormones in the killfish, Fundulus E654–E661.
heteroclitus. J. Exp. zool. 190, 269–280. Onuma, T., Ando, H., Koide, N., Okada, H., Urano, A., 2005. Effects of salmon GnRH
Maruska, K.P., Fernald, R.D., 2010. Behavioral and physiological plasticity: rapid and sex steroid hormones on expression of genes encoding growth hormone/
changes during social ascent in an African cichlid fish. Horm. Behav. 58, prolactin/somatolactin family hormones and a pituitary-specific transcription factor
230–240. in masu salmon pituitary cells in vitro. Gen. Comp. Endocrinol. 143, 129–141.
Maruska, K.P., Fernald, R.D., 2012. Contextual chemosensory urine signaling in an Øverli, Ø., Kotzian, S., Winberg, S., 2002. Effects of cortisol on aggression and
African cichlid fish. J. Exp. Biol. 215, 68–74. locomotor activity in rainbow trout. Horm. Behav. 42, 53–61.
Maruska, K.P., Becker, L., Neboori, A., Fernald, R.D., 2013a. Social descent with Páll, M.K., Liljander, M., Borg, B., 2004. Prolactin diminishes courtship behaviour and
territory loss causes rapid behavioral, endocrine and transcriptional changes in stimulates fanning in nesting male three-spined sticklebacks, Gasterosteus acu-
the brain. J. Exp. Biol. 216, 3656–3666. leatus. Behav. 141, 1511–1519.
Maruska, K.P., Zhang, A., Neboori, A., Fernald, R.D., 2013b. Social opportunity causes Páll, M.K., Mayer, I., Borg, B., 2002. Androgen and behavior in the male three-spined
rapid transcriptional changes in the social behaviour network of the brain in an stickleback, Gasterosteus aculeatus I.– changes in 11-ketotestosterone levels
African cichlid fish. J. Neuroendocrinol. 25, 145–157. during the nesting cycle. Horm. Behav. 41, 377–383.
Menuet, A., Pellegrini, E., Anglade, I., Blaise, O., Laudet, V., Kah, O., Pakdel, F., 2002. Perrone, R., Batista, G., Lorenzo, D., Macadar, O., Silva, A., 2010. Vasotocin actions
Molecular characterization of three estrogen receptor forms in zebrafish: binding on electric behavior: interspecific, seasonal, and social context-dependent differ-
characteristics, transactivation properties, and tissue distributions. Biol. Reprod. ences. Front. Behav. Neurosci. 4, 1–12.
66, 1881–1892. Petersen, C.L., Timothy, M., Kim, D.S., Bhandiwad, A.A., Mohr, R.A., Sisneros, J.A.,
Munchrath, L.A., Hofmann, H.A., 2010. Distribution of sex steroid hormone receptors Forlano, P.M., 2013. Exposure to advertisement calls of reproductive competitors
in the brain of an African cichlid fish, Astatotilapia burtoni. J. Comp. Neurol. 518, activates vocal-acoustic and catecholaminergic neurons in the plainfin midshipman
3302–3326. fish, Porichthys notatus. PLoS One 8.
Munro, A.D., Pitcher, T.J., 1985. Steroid hormones and agonistic behavior in a cichlid Portavella, M., Vargas, J.P., Torres, B., Salas, C., 2002. The effects of telencephalic
teleost, Aequidens pulcher. Horm. Behav. 19, 353–371. pallial lesions on spatial, temporal, and emotional learning in goldfish. Brain Res.
Muriach, B., Carrillo, M., Zanuy, S., Cerdá-Reverter, J.M., 2008. Distribution of Bull. 57, 397–399.
estrogen receptor 2 mRNAs (Esr2a and Esr2b) in the brain and pituitary of the sea Pradhan, A., Olsson, P.-E., 2015. Zebrafish sexual behavior: role of sex steroid
bass (Dicentrarchus labrax). Brain Res. 1210, 126–141. hormones and prostaglandins. Behav. Brain Funct. 11, 23.
Murphy, C.A., Stacey, N.E., Corkum, L.D., 2001. Putative steroidal pheromones in the Reddon, A.R., O’Connor, C.M., Marsh-Rollo, S.E., Balshine, S., 2012. Effects of
round goby, Neogobius melanostomus: olfactory and behavioral responses. isotocin on social responses in a cooperatively breeding fish. Anim. Behav. 84,
J. Chem. Ecol. 27, 443–470. 753–760.
Newman, S.W., 1999. The medial extended amygdala in male reproductive behavior. A Reddon, A.R., O’Connor, C.M., Marsh-Rollo, S.E., Balshine, S., Gozdowska, M.,
node in the mammalian social behavior network. Ann. N.Y. Acad. Sci. 877, Kulczykowska, E., 2015. Brain nonapeptide levels are related to social status and
242–257. affiliative behaviour in a cooperatively breeding cichlid fish. R. Soc. Open Sci. 2,
O’Connell, L.A., Hofmann, H.A., 2011. The vertebrate mesolimbic reward system and 140072.
social behavior network: a comparative synthesis. J. Comp. Neurol. 519, Renn, S.C.P., Fraser, E.J., Aubin-Horth, N., Trainor, B.C., Hofmann, H.A., 2012.
3599–3639. Females of an African cichlid fish display male-typical social dominance behavior
O’Connell, L.A., Hofmann, H.A., 2012a. Evolution of a vertebrate social decision- and elevated androgens in the absence of males. Horm. Behav. 61, 496–503.
making network. Science 336, 1154–1157. Resink, J.W., Voorthuis, P.K., Van Den Hurk, R., Peters, R.C., Van Oordt, P.G.W.J.,
O’Connell, L.A., Hofmann, H.A., 2012b. Social status predicts how sex steroid 1989. Steroid glucuronides of the seminal vesicle as olfactory stimuli in African
receptors regulate complex behavior across levels of biological organization. catfish, Clarias gariepinus. Aquaculture 83, 153–166.
Endocrinology 153, 1341–1351. Rodgers, C.M.C., Neff, B.D., Knapp, R., 2012. Effects of exogenous testosterone on
O’Connell, L.A., Matthews, B.J., Hofmann, H.A., 2012. Isotocin regulates paternal care parental care behaviours in male bluegill sunfish (Lepomis macrochirus). Ethology
in a monogamous cichlid fish. Horm. Behav. 61, 725–733. 118, 636–643.
O’Connell, L.A., Rigney, M.M., Dykstra, D.W., Hofmann, H.A., 2013. Neuroendocrine Rodgers, C.M.C., Neff, B.D., Knapp, R., 2013. Androgen-mediated nurturing and
mechanisms underlying sensory integration of social signals. J. Neuroendocrinol. aggressive behaviors during paternal care in bluegill sunfish (Lepomis macro-
25, 644–654. chirus). Horm. Behav. 63, 454–461.
Neuroendocrinology of Social Behavior in Teleost Fish 17

Rodgers, E.W., Earley, R.L., Grober, M.S., 2006. Elevated 11-ketotestosterone during Sorensen, P.W., Baker, C., 2014. Species-specific pheromones and their roles in
paternal behavior in the Bluebanded goby (Lythrypnus dalli ). Horm. Behav. 49, shoaling, migration, and reproduction. In: Sorensen, P.W., Wisenden, B.D. (Eds.),
610–614. Fish Pheromones and Related Cues. John Wiley & Sons, Inc., pp. 11–32.
Rodríguez, F., López, J.C., Vargas, J.P., Gómez, Y., Broglio, C., Salas, C., 2002. Sorensen, P.W., Scott, A.P., 1994. The evolution of hormonal sex pheromones in
Conservation of spatial memory function in the pallial forebrain of reptiles and ray- teleost fish: poor correlation between the pattern of steroid release by goldfish
finned fishes. J. Neurosci. Official J. Soc. Neurosci. 22, 2894–2903. and olfactory sensitivity suggests that these cues evolved as a result of
Ros, A.F.H., Bruintjes, R., Santos, R.S., Canario, A.V.M., Oliveira, R.F., 2004. The role chemical spying rather than signal specialization. Acta Physiol. Scand. 152,
of androgens in the trade-off between territorial and parental behavior in the 191–205.
Azorean rock-pool blenny, Parablennius parvicornis. Horm. Behav. 46, 491–497. Sorensen, P.W., Stacey, N.E., 1999. Evolution and specialization of fish hormonal
Ros, A.F.H., Canario, A.V.M., Couto, E., Zeilstra, I., Oliveira, R.F., 2003. Endocrine pheromones. In: Johnston, R.E., Müller-Schwarze, D., Sorensen, P.W. (Eds.),
correlates of intra-specific variation in the mating system of the St. Peter’s fish Advances in Chemical Signals in Vertebrates. Kluwer Academic/Plenum Publishers,
(Sarotherodon galilaeus). Horm. Behav. 44, 365–373. New York, Boston, MA, pp. 15–47.
Ros, A.F.H., Vullioud, P., Bruintjes, R., Vallat, A., Bshary, R., 2014. Intra- and inter- Spanagel, R., Weiss, F., 1999. The dopamine hypothesis of reward: past and current
specific challenges modulate cortisol but not androgen levels in a year-round status. Trends Neurosci. 22, 521–527.
territorial damselfish. J. Exp. Biol. 217, 1768–1774. Stacey, N., Kobayashi, M., 1996. Androgen induction of male sexual behaviors in
Rosenthal, G.G., Fitzsimmons, J.N., Woods, K.U., Gerlach, G., Fisher, H.S., 2011. female goldfish. Horm. Behav. 30, 434–445.
Tactical release of a sexually-selected pheromone in a Swordtail fish. PLoS One 6, Stacey, N.E., 1981. Hormonal regulation of female reproductive behavior in fish. Am.
e16994. Zool. 21, 305–316.
Saaristo, M., Craft, J.A., Lehtonen, K.K., Lindström, K., 2010. Exposure to 17alpha- Stacey, N.E., Sorensen, P.W., 2002. Hormonal pheromones in fish. In: Pfaff, D.,
ethinyl estradiol impairs courtship and aggressive behaviour of male sand gobies Arnold, D., Etgen, A., Fahrbach, S., Rubin, R. (Eds.), Hormones, Brain, and
(Pomatoschistus minutus). Chemosphere 79, 541–546. Behavior, vol. 2. Academic Press, New York, pp. 375–435.
Salek, S.J., Sullivan, C.V., Godwin, J., 2001. Courtship behavior of male white perch, Tacon, P., Baroiller, J.F., Le Bail, P.Y., Prunet, P., Jalabert, B., 2000. Effect of egg
Morone americana: evidence for control by androgens. Comp. Biochem. Physiol. A deprivation on sex steroids, gonadotropin, prolactin, and growth hormone profiles
Mol. Integr. Physiol. 130, 731–740. during the reproductive cycle of the mouthbrooding cichlid fish Oreochromis
Santangelo, N., Bass, A.H., 2006. New insights into neuropeptide modulation of niloticus. Gen. Comp. Endocrinol. 117, 54–65.
aggression: field studies of arginine vasotocin in a territorial tropical damselfish. Tay, T.L., Ronneberger, O., Ryu, S., Nitschke, R., Driever, W., 2011. Comprehensive
Proc. Biol. Sci. 273, 3085–3092. catecholaminergic projectome analysis reveals single-neuron integration of
Satou, M., Oka, Y., Kusunoki, M., Matsushima, T., Kato, M., Fujita, I., Ueda, K., 1984. zebrafish ascending and descending dopaminergic systems. Nat. Commun.
Telencephalic and preoptic areas integrate sexual behavior in hime salmon 2, 171.
(landlocked red salmon, Oncorhynchus nerka): results of electrical brain stimulation Teles, M.C., Almeida, O., Lopes, J.S., Oliveira, R.F., 2015. Social interactions elicit
experiments. Physiol. Behav. 33, 441–447. rapid shifts in functional connectivity in the social decision-making network of
Schjolden, J., Basic, D., Winberg, S., 2009. Aggression in rainbow trout is inhibited by zebrafish. Proc. R. Soc. B Biol. Sci. 282, 20151099.
both MR and GR antagonists. Physiol. Behav. 98, 625–630. Thompson, R.R., Walton, J.C., 2004. Peptide effects on social behavior: effects of
Schradin, C., Anzenberger, G., 1999. Prolactin, the hormone of paternity. News vasotocin and isotocin on social approach behavior in male goldfish (Carassius
Physiol. Sci. 14, 223–231. auratus). Behav. Neurosci. 118, 620–626.
Sebire, M., Allen, Y., Bersuder, P., Katsiadaki, I., 2008. The model anti-androgen Thompson, R.R., Walton, J.C., 2009. Vasotocin immunoreactivity in goldfish brains:
flutamide suppresses the expression of typical male stickleback reproductive characterizing primitive circuits associated with social regulation. Brain Behav. Evol.
behaviour. Aquat. Toxicol. 90, 37–47. 73, 153–164.
Semsar, K., Godwin, J., 2003. Social influences on the arginine vasotocin system are Thompson, R.R., Walton, J.C., 2013. Social regulatory functions of vasotocin and
independent of gonads in a sex-changing fish. J. Neurosci. Off. J. Soc. Neurosci. isotocin in fish. In: Choleris, E., Pfaff, D.W., Kavaliers, M. (Eds.), Oxytocin, Vaso-
23, 4386–4393. pressin and Related Peptides in the Regulation of Behavior. Cambridge University
Semsar, K., Godwin, J., 2004. Multiple mechanisms of phenotype development in the Press, Cambridge, pp. 75–96.
bluehead wrasse. Horm. Behav. 45, 345–353. Thompson, R.R., Walton, J.C., Bhalla, R., George, K.C., Beth, E.H., 2008.
Semsar, K., Kandel, F.L., Godwin, J., 2001. Manipulations of the AVT system shift A primitive social circuit: vasotocin-substance P interactions modulate social
social status and related courtship and aggressive behavior in the bluehead behavior through a peripheral feedback mechanism in goldfish. Eur. J. Neurosci.
wrasse. Horm. Behav. 40, 21–31. 27, 2285–2293.
Serrano, R.M., Barata, E.N., Birkett, M.A., Hubbard, P.C., Guerreiro, P.S., Trainor, B.C., Hofmann, H.A., 2006. Somatostatin regulates aggressive behavior in an
Canário, A.V.M., 2008a. Behavioral and olfactory responses of female Salaria pavo African cichlid fish. Endocrinology 147, 5119–5125.
(Pisces: Blenniidae) to a putative multi-component male pheromone. J. Chem. Trainor, B.C., Kyomen, H.H., Marler, C.A., 2006. Estrogenic encounters: how inter-
Ecol. 34, 647–658. actions between aromatase and the environment modulate aggression. Front.
Serrano, R.M., Lopes, O., Hubbard, P.C., Araújo, J., Canário, A.V.M., Barata, E.N., Neuroendocrinol. 27, 170–179.
2008b. 11-Ketotestosterone stimulates putative sex pheromone production in Villars, T.A., Hale, N., Chapnick, D., 1985. Prostaglandin-F2a stimulates reproductive
the male peacock blenny, Salaria pavo (Risso 1810). Biol. Reprod. 79, behavior of female paradise fish (Macropodus opercularis). Horm. Behav. 19,
861–868. 21–35.
Simões, J.M., Barata, E.N., Harris, R.M., O’Connell, L.A., Hofmann, H.A., Oliveira, R.F., Walton, J.C., Waxman, B., Hoffbuhr, K., Kennedy, M., Beth, E., Scangos, J.,
2015. Social odors conveying dominance and reproductive information induce Thompson, R.R., 2010. Behavioral effects of hindbrain vasotocin in goldfish are
rapid physiological and neuromolecular changes in a cichlid fish. BMC Genomics seasonally variable but not sexually dimorphic. Neuropharmacology 58, 126–134.
16, 1–13. White, S.a, Nguyen, T., Fernald, R.D., 2002. Social regulation of gonadotropin-
Sisneros, J.A., 2009. Adaptive hearing in the vocal plainfin midshipman fish: getting in releasing hormone. J. Exp. Biol. 205, 2567–2581.
tune for the breeding season and implications for acoustic communication. Integr. Whittington, C.M., Wilson, A.B., 2013. The role of prolactin in fish reproduction. Gen.
Zool. 4, 33–42. Comp. Endocrinol. 191, 123–136.
Sisneros, J.A., Forlano, P.M., Deitcher, D.L., Bass, A.H., 2004. Steroid-dependent Williams, A.J., Wigham, T., 1994. The regulation of prolactin cells in the rainbow trout
auditory plasticity leads to adaptive coupling of sender and receiver. Science (Oncorhynchus mykiss). 1. Possible roles for thyrotropin-releasing hormone (TRH)
305, 404–407. and oestradiol. Gen. Comp. Endocrinol. 93, 388–397.
Smith, G.T., 2013. Evolution and hormonal regulation of sex differences in the elec- Wingfield, J.C., 1984. Androgens and mating systems: testosterone-induced polygyny in
trocommunication behavior of ghost knifefishes (Apteronotidae). J. Exp. Biol. 216, normally monogamous birds. Auk 101, 665–671.
2421–2433. Wingfield, J.C., Hegner, R.E., Dufty, A.M.J., Ball, G.F., 1990. The challenge hypoth-
Soares, M.C., Bshary, R., Mendonça, R., Grutter, A.S., Oliveira, R.F., 2012. Arginine esis: theoretical implications for patterns of testosterone secretion, mating
vasotocin regulation of interspecific cooperative behaviour in a cleaner fish. PLoS systems, and breeding strategies. Am. Nat. 136, 829–846.
One 7, 1–10. Wisenden, B.D., 2014. The cue–signal continuum. In: Sorensen, P.W., Wisenden, B.D.
Sorensen, P.W., 2014. Introduction to pheromones and related chemical cues in (Eds.), Fish Pheromones and Related Cues. John Wiley & Sons, Inc., pp. 149–158
fishes. In: Sorensen, P.W., Wisenden, B.D. (Eds.), Fish Pheromones and Related Wisenden, B.D., Stacey, N.E., 2005. Fish Semiochemicals and the Evolution of
Cues. John Wiley & Sons, Inc., pp. 1–9. Communication Networks. Cambridge University Press.
18 Neuroendocrinology of Social Behavior in Teleost Fish

Wong, C.J., 2000. Electrical stimulation of the preoptic area in Eigenmannia: Wyatt, T.D., 2010. Pheromones and signature mixtures: defining species-wide signals
evoked interruptions in the electric organ discharge. J. Comp. Physiol. A 186, and variable cues for identity in both invertebrates and vertebrates. J. Comp.
81–93. Physiol. A 196, 685–700.
Wullimann, M.F., Mueller, T., 2004. Teleostean and mammalian forebrains contrasted: Young, K., Gobrogge, K., Liu, Y., Wang, Z., 2011. The neurobiology of pair-bonding:
evidence from genes to behavior. J. Comp. Neurol. 475, 143–162. insight from a socially monogamus rodent. Front. Neuroendocrinol. 32, 53–69.
2.02 Social Regulation of Sex: How the Brain Controls Reproductive Circuits
Russell D Fernald, Stanford University, Stanford, CA, USA
Ó 2017 Elsevier Inc. All rights reserved.

2.02.1 Social Dominance Influences Reproductive Physiology in Many Vertebrate Species 19


2.02.1.1 Social System of Astatotilapia burtoni: Reproductive Consequences of Status 20
2.02.2 How Does Social Status Regulate the HPG Axis? 21
2.02.3 How Is GnRH1 Released in a Pulsatile Fashion? 23
2.02.4 Can Males Be Deceptive about Their Reproductive State? 24
2.02.5 Genomic Consequences of Female Mate Choice 26
2.02.6 What Social Information Influences Changes in Status? 27
2.02.6.1 Attention Hierarchies in Male Astatotilapia burtoni 27
2.02.6.2 Transitive Inference in Astatotilapia burtoni 28
2.02.7 Conclusions 28
Acknowledgments 28
References 28

2.02.1 Social Dominance Influences Reproductive Bshary and Würth, 2001), cooperate (e.g., Pitcher et al.,
Physiology in Many Vertebrate Species 1986), learn in a social context (Laland et al., 2003), eavesdrop
on interactions among conspecifics to gain social insights
Animals experience of life on the planet varies according to (McGregor, 1993), show transitive inference (Grosenick et al.,
their sensory systems that are differentially responsive to 2007), and generally display a wide range of social activities
unique perceptual worlds. Correspondingly, their responses comparable in some instances with those of primates (e.g.,
depend on their abilities to act in that private world. Von Bshary et al., 2002). In addition to their social sophistication,
Uexküll (1921) described the perceptual world as the ‘umwelt’ fish species have proven important and quite useful for under-
of a species, and since then, scientists have discovered new standing mechanisms controlling social behavior. With several
dimensions of the umwelts for many species. While Von Uex- fish genomes now sequenced, it has become much easier to
küll conceptualized his idea of the umwelt idea in relation to trace genetic and neurobiological mechanisms responsible for
physical stimuli, Lorenz extended this concept, recognizing sophisticated social interactions in fishes.
that animals also have a social ‘umwelt’ (Lorenz, 1937) Why might fish social behavior provide general insights
through which signals from other individuals influence about hormone and brain mechanisms of social interactions?
behavior. Lorenz’ influential article, ‘Companions as factors Stable social systems in any animal species require the individ-
in the bird’s environment,’ (1937) showed that behavioral uals to behave predictably. However, what an individual does
scientists should include other individuals in considering the at any moment in time depends on its status relative to other
umwelt and, importantly, the overall social context of the individuals, its reproductive state, and its recent behavioral
behavior. Such interactions are particularly important for interactions. In addition, environmental factors (e.g., preda-
sexual behavior in which multimodal signals orchestrate repro- tors, prey, or resource competitors) also need to be incorpo-
ductive activities. Since behavior is the ultimate arbiter of rated into any plan for behavioral action. So, to be successful
animal survival, response of animals during their interactions in a social group, individuals must be aware of the immediate
with others and with their environment shapes the phenotype. behavior of others and use that information to regulate their
Behavior, in turn, depends on intricate physiological, cellular, own activity. But what exactly does an individual need to
and molecular adaptations forged during evolution and modi- know and how do they acquire the knowledge that lets them
fied during development. Ultimate questions about the evolu- act appropriately? It is possible that the apparently subtle social
tion and control of behavior and especially interactions that interactions can be explained and understood in terms of
sculpt behavior require understanding the causal mechanisms contingencies. That is, a set of ‘if-then’ rules with associated
responsible as animals interact with one another, preferably probabilities might suffice in many situations to explain the
in a natural setting. Yet, the vast majority of experiments are behavior of animals in social groups. Since it has been argued
performed on isolated individuals, often in domesticated that species in the fish taxon have demonstrated many but not
species. Clearly little can be learned about how evolution has all the social skills that arguably led to the evolution of
shaped social behavior by analyzing individual animals since complex brain structures in primates (Bshary et al., 2002),
social interactions cannot occur. how could we proceed to exploit fish social skills in under-
Fish species have emerged as important models for studies standing neural mechanisms? Here I will describe results
of how hormones influence social behavior and its neural from experiments using the relationship between social domi-
underpinnings because of the many examples of sophisticated nance, hormones, and reproduction to understand how social
behavioral interactions regulating sex. For example, it has been signals regulate the hypothalamic–pituitary–gonadal (HPG)
shown that fish might exploit Machiavellian strategies (e.g., axis in Astatotilapia burtoni. I also explore the range of social

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00021-3 19


20 Social Regulation of Sex: How the Brain Controls Reproductive Circuits

requirements for A. burtoni to be reproductively successful and rapidly turns on his bright body colors (Fernald and Hirata,
describe evidence about how social behavior sculpts the brain 1977b; Burmeister et al., 2005) and will quickly begin perform-
in ways that depend on the developmental stage, social circum- ing many of the 17 distinct behaviors characteristic of dominant
stance, and environmental context. males upon his social ascent. Over a few days, the reproductive
system of the ascending male is remodeled rendering the male
reproductively competent, producing changes that are evident
2.02.1.1 Social System of Astatotilapia burtoni: Reproductive
at several levels along the HPG axis (Maruska and Fernald,
Consequences of Status
2014). In A. burtoni, as in all vertebrates, reproduction is
In their natural habitat, the shallow shore pools and river estu- controlled by GnRH1-containing neurons in the hypothalamus
aries of Lake Tanganyika (Fernald and Hirata, 1977a; Coulter, that deliver the eponymously named GnRH decapeptide to the
1991), A. burtoni live in a leklike social system in which domi- pituitary. When a male ascends (ND/D), delivery of this mole-
nant males vigorously defend contiguous territories (Fernald cule sets in motion a cascade of actions ultimately resulting in
and Hirata, 1977b). Social communication among these fish reproductive competence and release of sex steroids from the
appears to depend primarily on visual signals (Fernald, 1984; gonads. The GnRH neurons increase in volume by eightfold
see below). Astatotilapia burtoni males live as one of two revers- (Davis and Fernald, 1990), extend their dendrites (Fernald,
ible, socially controlled phenotypes: reproductively competent 2012), and rapidly increase production of GnRH mRNA
dominant (D) males and reproductively incompetent (Burmeister et al., 2005) and GnRH peptide (White et al.,
nondominant (ND) males (see Figure 1). D males are brightly 2002). However, when a D male is moved into a social system
colored, aggressively defend territories, and actively court with larger D males (>5% longer), it abruptly loses its color
females (Fernald and Hirata, 1977b). In striking contrast, ND (<1 min) and joins other ND males and females in a school.
males have a dull coloration, mimic female behavior, and Its GnRH-containing neurons in the preoptic area (POA)
school with females and other ND males except when fleeing shrink to one-eighth their volume and produce less GnRH
from an attacking D male. mRNA and peptide, causing hypogonadism and loss of
These obvious external differences are reflected in major reproductive competence (2 weeks) (Davis and Fernald,
physiological and neural adaptations in response to differences 1990; Francis et al., 1993). Similarly, androgen, estrogen, and
in social status. As animals transition from one phenotype to GnRH receptor mRNA expression levels depend on social
the other, some changes including expression of the black bar status (Au et al., 2006; Burmeister et al., 2007; Harbott et al.,
through the eye, brightening of the body color, and switching 2007) as do electrical properties of the GnRH neurons
behavioral repertoires are expressed in minutes while concom- themselves (Greenwood and Fernald, 2004).
itant physiological and neural changes happen over days. Astatotilapia burtoni D males perform 17 distinct behavioral
ND males attend closely to the unfolding social scene, patterns during social interactions (Fernald, 1977). D males dig
assessing when they might be able to gain a territory by defeat- a pit in their territory, exchange threat displays with neigh-
ing a resident male. When this happens, there is typically boring territorial males, chase ND animals from their territo-
a dramatic fight during which males engage in mouth-to-mouth ries, and solicit and court females. D males display bright
biting, hitting each other with their bodies, and nipping at each coloration patterns, particularly during courtship. A D male
other’s fins. If the ND male successfully defeats the resident, he will lead a female toward his territory, typically using large

Figure 1 Sketch of an observation site along the edge of a shore pool on the north end of Lake Tanganyika, near Bujumbura, Burundi, Africa. Solid
dots represent grid stakes spaced at 50 cm intervals that label grids (1–5; A–D) for identification. Dark circles represent spawning pit locations of
dominant males. Lighter-colored outlines circumscribe the approximate territories of individuals. Male territories are arrayed over the food source of
detritus. Nondominant males and females school together near the territorial area. Based on Fernald, R.D., Hirata, N.R., 1977a. Field study of Hap-
lochromis burtoni: habitats and co habitants. Environ. Biol. Fish. 2, 299–308.
Social Regulation of Sex: How the Brain Controls Reproductive Circuits 21

movements of his tail and court by quivering his opened, GnRH1 neurons control the brain–pituitary–gonadal axis
brightly colored anal fin in front of the female. When a D regulating reproduction in all vertebrates via a projection to
male manages to lure a female into his territory, she will nor- the pituitary (Bushnik and Fernald, 1995). GnRH1 is the
mally eat by sifting the substrate in the territory. ND males signaling peptide sent from the brain to the pituitary to trigger
will mimic female behavior sufficiently well so that the D release of gonadotropins and ultimately regulate testes growth.
male allows NDs to enter the territories and feed before their Thus social control of maturation in A. burtoni is achieved by
deception is discovered. This ND male behavior occurs because changing the GnRH-containing cells in the brain.
only sites defended as territories contain food so that ND males Social status can regulate the physiology of the reproduc-
need to enter to eat. ND female impersonators are quickly tive state, even in adult A. burtoni as shown by converting
chased off, but for females it is different. The D male will males from D > ND or ND > D by moving them to new
lead her to his pit and continue courtship movements. D males communities. Specifically, when D males were moved to
swim vigorously in front of the female, quivering their entire communities containing larger D males, they rapidly became
body with spread anal fins. If appropriately stimulated, the ND (e.g., D > ND), and similarly when ND males were moved
female will lay her eggs in the pit and collect them in her mouth to communities with smaller conspecifics, they became D (e.g.,
immediately. After she has deposited several eggs, the male will ND > D). Following 4 weeks in the altered social setting,
swim in front of her displaying the egglike spots on his anal fin GnRH cell size was measured and showed that changing the
(ocelli). D males display this fin because the spots may seem to social status alone was sufficient to change GnRH neuron
the female like eggs not yet collected (Wickler, 1962). Thus, size in the brain (Figure 2). As expected, the gonadosomatic
while attempting to ‘collect’ the egg-spots, the female ingests index (GSI) changed correspondingly (Francis et al., 1993).
milt ejected near them by the male and ensures fertilization. Thus reproduction is socially controlled in adults, as well as
The spawning male may repeatedly interrupt his courtship in juveniles, via changes induced in the GnRH neurons in
and mating to chase off intruders into his territory. After several the brain.
bouts of egg laying and fertilization, the female departs with Although causing a large change in brain structure by
fertilized eggs that she broods in her mouth for 2 weeks changing social status is quite remarkable, the timescale of
(Fernald, 1977). this initial experiment did not reflect how rapidly behavioral
Even this abbreviated description of the natural behavior of and neural changes could occur. Indeed, the 4-week interval
A. burtoni shows the important role visual signals play in medi- tested was substantially longer than any observed changes in
ating social behavior. As is typical for this kind of rapid social behavior following status switches, which can occur in
interactions, each behavioral act influences the next, both in minutes. Analyzing socially induced changes in neural struc-
the individual and in other animals involved in the encounter. tures on a significantly shorter timescale revealed another
What do animals attend to during aggressive social interac- surprise.
tions? Using ethological methods, early workers identified Clearly social status sets both soma size of POA GnRH-
several fixed action patterns and key stimuli that mediate containing neurons and through this pathway, GSI, and these
social signaling in A. burtoni. Specifically, Leong (1969) effects are reversible. The relatively large testes and GnRH
analyzed the role of the black eyebar by testing how T males neurons characteristic of D males are a consequence of their
responded to A. burtoni dummies painted with various config- social dominance, and when this dominance advantage is
urations of the distinctive body patterns. When the eyebar was lost, both neurons and testes shrink, although as shown by
presented alone, T males increased their readiness to attack White et al. (2002), there is striking asymmetry in the
targets while presentation of the orange-red patch of humeral physiological responses. Thus social information about status
scales alone decreased attack readiness. Subsequent experi- causes the changes in the brain, but how this is achieved is
ments tested the importance of the orientation of the eyebar not known.
relative to the body and other visual stimuli (Heiligenberg It is important to emphasize that the effect of social status
and Kramer, 1972; Heiligenberg et al., 1972). All the work sup- on GnRH1 cell size and GnRH1 mRNA expression is limited
ported the hypothesis that the black eyebar and the red to the GnRH1-containing neurons of the POA. As we have
humeral patch influence the aggressiveness of T males in previously shown, A. burtoni has three distinct genes that
opposite directions. code for three distinct GnRH-like molecules (White et al.,
1994; White and Fernald, 1998) expressed at three distinct
sites in the brain (White et al., 1995). The GnRH forms not
2.02.2 How Does Social Status Regulate the HPG found in the POA are expressed in two other distinct cell
Axis? groups, one located in the terminal nerve region (GnRH3),
the other in the mesencephalon (GnRH2; see White et al.,
Being reared with broodmates suppresses early social and phys- 1995 for details). Neither of these other GnRH-containing
ical development, but in A. burtoni even more effective social cells showed any changes in size or mRNA production as
regulation occurs when older, larger animals are housed with a function of social status (Davis and Fernald, 1990; White
younger animals. Davis and Fernald (1990) raised animals et al., 2002). Males and females share the brain–pituitary–
from hatching in the presence of adult males and showed these gonadal axis used to control reproduction, but female
fish have suppressed gonadal maturation relative to fish reared A. burtoni have a strikingly different system that regulates
without the presence of adults. Suppressed animals not only reproduction. GnRH1-containing cells in the POA of females
had hypogonadal testes but also GnRH1-containing neurons also change size, but size change depends on reproductive
in the POA c.1/8 the volume of those in dominant males. status alone (White and Fernald, 1993).
22 Social Regulation of Sex: How the Brain Controls Reproductive Circuits

100
90
80
100
70
90
60
80 50
70 40
60 30
%
50 20
40 10
30 100
20
10
350
100 400
150 450
200
So

250
m

300
a
si

350 ND>D
ze

400

D>ND
m

450
2

D
)

ND

Figure 2 Three-dimensional plot of mean soma diameter of preoptic area (POA) gonadotropin-releasing hormone (GnRH1)-immunoreactive
neurons in males of different social status. There are significant differences between D and D/ND males and between ND and ND/D males.
Percentage of individuals with mean soma size in a given size bin are plotted for each treatment condition. Redrawn from Francis, R.C., Soma, K.,
Fernald, R.D., 1993. Social regulation of the brain-pituitary-gonadal axis. Proc. Natl. Acad. Sci. U.S.A. 90, 7794–7798.

White et al. (2002) also showed that the socially induced As expected, social control of the reproductive axis via
changes in status resulted in significant changes in gene expres- GnRH1 also influences important endocrine factors, including
sion. Measuring changes in mRNA from all three forms of androgen released from the gonads. Castrated A. burtoni T
GnRH found in A. burtoni, they found that only the POA males have hypertrophied GnRH neurons (Francis et al.,
GnRH1 mRNA was regulated corresponding to a change in 1992a; Soma et al., 1996) showing that androgen has a feed-
social status. The change in mRNA in the POA form of GnRH1 back effect on GnRH cell size. The important discovery is that
was evident at 3 days after a change in social status. Such social the set point for this feedback loop is the social status since
regulation demonstrates clearly that key social information is D males have larger GnRH neurons despite having higher
used to control cellular and molecular processes in the brain. androgen levels (Soma et al., 1996). D males that are castrated
Given the rapid macroscopic change in GnRH neurons are able to maintain their rank despite having lowered
following a change in status, we devised a novel behavioral androgen levels (Francis et al., 1992b). Possibly, prior domi-
paradigm to test the speed of this process. We placed a D nance experienced on the part of the D male and the size differ-
male in an aquarium with one ND male and three females, ence among animals contribute to this result.
with associated fish colonies on each side. The target D male Clearly, social status regulates the production and release of
was removed before dawn, so when the lights came on, the GnRH1 into the pituitary. Another potential site for regulation
ND male was the sole male in the tank with three females. is the GnRH1 receptor in the pituitary. Our laboratory has
We videotaped the response and subsequently recorded when shown that A. burtoni has genes that encode two distinct
the ND male first exhibited an eyebar and the first D behaviors. GnRH receptors (Robison et al., 2001; Flanagan et al., 2007).
Twenty minutes after the first expression of D behavior, Using real-time PCR, we have been able to show that the
we sacrificed the animal and subsequently mapped the mRNA of one of these receptor types is upregulated rapidly
expression of an activity-dependent immediate-early gene and dramatically in the pituitary of D males as compared
(egr-1; Burmeister et al., 2005). We found a rapid genomic with ND males (Au et al., 2006). It remains to be discovered
response in the GnRH1 neurons of the POA in response to whether this receptor regulation results solely from a change
the perception of social opportunity 20 min after the NT in social status or if other factors are also involved. In addition
male adopted dominant coloration and behaviors. This to its primary role, analysis of the distribution of GnRH recep-
suggests that these animals are fully primed to ascend, and tors (Chen and Fernald, 2006) revealed that this peptide must
based on their recognition of the new circumstances, they play a modulatory role in the animal as well as regulating
respond at both the behavioral and molecular levels. reproduction.
Social Regulation of Sex: How the Brain Controls Reproductive Circuits 23

While the changes in GnRH neuron size and concomitant change in social status is transduced into this molecular
changes in GnRH production and in hormone receptors are change in the cortisol receptor remains a mystery.
part and parcel of a response to new reproductive opportunity,
other brain changes caused by social status are more subtle. As
is typical of many social hierarchies, D males have high levels 2.02.3 How Is GnRH1 Released in a Pulsatile
of testosterone and low levels of the stress hormone cortisol, Fashion?
and these levels are reversed in ND males (Fox et al., 1997).
Among other changes caused by social subordination, the stress The development and function of the reproductive system in
response inhibits the reproductive axis in many species and vertebrates ultimately depends on the timing and levels of
results in chronic elevation of stress hormones, which, if sus- signaling by gonadal sex steroids (Phoenix et al., 1959). As
tained, is known to be detrimental (Webster et al., 2008; described above, production of these steroids is controlled by
Kaplan and Manuck, 2004; Kaplan, 2008). Thus, it is a puzzle neurons expressing GnRH1, which comprise the final output
how socially suppressed ND A. burtoni cope with sustained of the brain to the HPG axis. Over 30 years ago, Belchetz
levels of elevated cortisol levels. One possible mechanism for et al. (1978) showed that action of GnRH1 depends on coordi-
modulating the responsiveness of nondominant individuals nated, pulsatile GnRH1 release, not simply elevated levels of
to stress would be to change the sensitivity of the cortisol recep- the decapeptide, requiring some kind of synchronization in
tors that mediate its potentially damaging effects on the body the output of these neurons. Such episodic activation of the
(Avitsur et al., 2001) and brain (Sapolsky, 1996). pituitary gonadotropes has been observed in multiple verte-
We first showed that A. burtoni has three receptor genes for brate taxa, including mammals and fish (9–12); however,
cortisol: two glucocorticoid receptors (GR1 and GR2) and specific mechanisms that underlie this required coordinated
a mineralocorticoid receptor (MR) (Greenwood et al., 2003). activity of GnRH1 neurons are unknown in any system.
Of these genes, GR1 has two splice variants (GR1a and Synchrony could in principle derive from coincident input
GR1b), which differ by a nine-amino acid insertion in the from a ‘pacemaker’ neural population, from direct coupling
DNA-binding domain in GR1b that greatly reduces binding of GnRH1 neurons, or from some combination of
to cortisol, and has been shown to act as a dominant mechanisms.
negative inhibitor of transcription in zebrafish (Schaaf et al., Recording calcium signals from brain slices taken from
2008). We have recently shown that when A. burtoni males dominant and nondominant A. burtoni males, we found
become ND, they replace the high sensitive receptor with this a striking difference in the relationship among neuronal firing
less sensitive receptor in the hypothalamic/POA, site of the patterns inferred from records of calcium signaling (Figure 3).
GnRH neurons (Korzan et al., 2014). This modulation of Specifically, cross-correlograms of the time of occurrence of
cortisol receptor subtype expression could mitigate the calcium signals recorded from nondominant males show no
consequences of socially induced increases in cortisol levels correlation among GnRH1 neurons; that is, they are no
in ND males. Just how external social information about the different from random firing. In contrast, in dominant males,

Figure 3 Cross-correlograms of calcium signals of gonadotropin-releasing hormone (GnRH1) neurons for nondominant (a) and dominant
(b) animals recorded from in vitro slices taken through the preoptic area of Astatotilapia burtoni males. Recordings from the nondominant males
(a) show no significant correlation in their expression of calcium signals representing neuronal firing, while those from the dominant males (b) are
highly correlated. Red bar indicates the expected value calculated from the data generated by randomly reordering the recordings. Kagawa,
unpublished.
24 Social Regulation of Sex: How the Brain Controls Reproductive Circuits

the occurrence of calcium signals is significantly correlated. of large-diameter cells of the parenchyma of the POA, in
These data suggested that firing patterns of GnRH1 neurons a pattern similar to that of GnRH1 expression. To test whether
in A. burtoni, as in other vertebrates, are pulsatile in reproduc- Gjd2a is expressed in GnRH1 neurons, we colabeled brain
tively competent dominant males and not in reproductively sections and found that 96.3  1.8% of GnRH1þ neurons
incompetent nondominant males. also express Gjd2a mRNA (Figure 5; n ¼ 4 fish). These results
To identify possible origins of this synchrony among suggest that the electrical coupling we observe between
GnRH1 neurons, we used A. burtoni in which the GnRH1 GnRH1 neurons is mediated by the gap junction Gjd2a.
neurons had been labeled transgenically (EGFP; Juntti et al., Previously, gap junction-mediated coupling has been sus-
2013). We recorded from pairs of genetically labeled GnRH1 pected to play a role, as synchronous firing can be observed
neurons in the brains of reproductively active fish and showed in neurons mechanically isolated from brain slices, in cultures
that these cells are strongly and uniformly interconnected by of embryonic mouse and primate neurons, and immortalized
electrical synapses that can be reversibly blocked (Ma et al., mouse GnRH1 neurons express the connexin proteins that
2015, Figure 4). The electrical coupling we measure between constitute gap junctions (13–15). These data are the first to
GnRH1 neurons predicts that these neurons express connexins, confirm that gap junctions connect GnRH1 neurons in any
the proteins that comprise gap junctions. We searched the vertebrate and that they very likely contribute to the coordi-
A. burtoni genome for likely connexin genes and focused on nated firing of GnRH1 neurons to produce the pulsatile output
the neuronal connexin gap junction delta 2 (Gjd2; also known needed for reproduction, the most important act of any
as Cx36 in mammals and Cx35 in fish). We found three homo- organism.
logues most closely related to mammalian Gjd2, which we call
Gjd2a, Gjd2b, and Gjd2c (Ma et al., 2015). One of these, 2.02.4 Can Males Be Deceptive about Their
Gjd2a, appears to be a true ortholog, as the genes to the 50 Reproductive State?
end of mouse Gjd2 (Aqr, Dph6, Meis2) are also found flanking
A. burtoni Gjd2a. We generated antisense RNA probes against Can A. burtoni males be deceptive about their reproductive
each to determine whether any was expressed in the POA. We state? Deception in animals is an important question in animal
found that one of these, Gjd2a, was expressed in a population cognition because it is often taken as a hallmark of cognitive

Figure 4 Gonadotropin-releasing hormone (GnRH1) neurons are connected via electrical synapses. (a) Ventral view of the hypothalamus/preoptic
area (POA) of an Astatotilapia burtoni brain showing GnRH1þ cell bodies and fibers labeled with EGFP fluorescence at the top of the figure and termi-
nations of these neurons in the pituitary at the bottom of the figure (top: anterior; bottom: posterior). (b) Arrows indicate schematically patch clamp
electrophysiological recordings from pairs of GnRH1 neurons in the POA. Representative paired whole-cell recordings revealing postsynaptic spikelets
driven by full action potential in presynaptic GnRH1 cells V1 (red trace) in response to suprathreshold DC current injection to one cell (I1-red trace) or
the other cell (I2-blue trace). All paired recordings were similarly symmetric. Scale bar, 5 mV; time, 0.2 s. Modified from Ma, Y.Y., Juntti, S.A., Hu,
C.K., Huguenard, J.R., Fernald, R.D., 2015. Electrical synapses connect a network of gonadotropin releasing hormone neurons in a cichlid fish. Proc.
Natl. Acad. Sci. U.S.A. 112, 3805–3810.
Social Regulation of Sex: How the Brain Controls Reproductive Circuits 25

(a) (b) (c) (d)

Figure 5 Gonadotropin-releasing hormone (GnRH1) neurons express mRNA for the connexin Gjd2a. Preoptic area (POA) sections were fluo-
rescently colabeled for mRNA encoding GnRH1 (a) and Gjd2a (b); merged channels are shown in (c). The red square in the Nissl-stained section
highlights region displayed (d). Scale bar, 30 mm in a–c. Modified from Ma, Y.Y., Juntti, S.A., Hu, C.K., Huguenard, J.R., Fernald, R.D., 2015.
Electrical synapses connect a network of gonadotropin releasing hormone neurons in a cichlid fish. Proc. Natl. Acad. Sci. U.S.A. 112, 3805–3810.

skill. To ask this question, we used a novel paradigm in which although there was no physical or chemical contact possible,
two differently sized males share a tank, divided in half by the larger male made several ‘attacks’ toward the small male
a clear, watertight divider and a black removable divider by swimming rapidly toward the smaller male behind the
(Chen and Fernald, 2011). The general idea is to see how barrier. The smaller male quickly lost his coloration, including
a small D male responds to a larger D male when he can his eyebar in response upon seeing these ‘attacks.’ The behavior
only see the larger fish. Experimentally, a half terra-cotta pot, of the smaller male was typical for an animal losing his territo-
cut in half lengthwise, was placed so that it allowed the males rial status as confirmed by quantifying the behavior. Indeed,
on each side to occupy a ‘shared’ shelter, though with the black the smaller males essentially abandoned their part of the
divider in place, neither animal knew the other animal was shelter, digging a new pit remote from the site of the flowerpot.
present (Figure 6). On one side was a male that was about Interestingly, this suppression of dominance, based entirely on
four times larger than the male on the other side, and each visual signals, as reflected by the behavioral quantification
male had an appropriately sized female in their hemitank. (Chen and Fernald, 2011), also reduced the expression of
Both the small and large fish were habituated to the tank androgen hormones, but only for the first 3 days. Seven days
with the opaque barrier in place for 2 days during which time after the black barrier was lifted, the smaller animals recovered
each behaved like a normal dominant male in its territory: their hormone expression levels and other brain markers of
excavating gravel from the hemipot, courting the female in dominance while maintaining the coloration of an ND male.
their half of the tank, leading her back to the shelter, and per- Moreover, they could be seen courting their females when
forming typical courtship and territorial male behaviors, all out of view of the dominant male. So the effects of the visual
of which were quantified and matched the behavior of normal suppression resulted in changes in the expression of aggressive,
D males. On the third day, the opaque barrier was lifted, and territorial behavioral responses by the smaller male but not in

Figure 6 Front view of the aquarium (45 l) divided in half with a watertight, clear divider (gray midline) and a removable opaque barrier (black
midline). There is a small male fish (left compartment) and the large male fish (about four times larger in the right compartment). The half terra-cotta
pot (red curved line) was cut so that both fish ‘shared’ the same shelter, though they were not aware of each other’s presence. This ‘shared’ shelter
was hemisected by both center dividers. A layer of gravel covered the bottom of the tank.
26 Social Regulation of Sex: How the Brain Controls Reproductive Circuits

sustained physiological changes. This suggests that the Plexiglas barrier so the female received only visual informa-
smaller males uncoupled physiological changes in circulating tion from the males. We quantified the female’s preference
hormones from their effects on outward appearance, seemingly based on her interactions and proximity to that male over
presenting a false outward appearance not consonant with a 20-min period. Following this, we staged a fight between
internal changes. This appears to be an example of deceptive the two males in one of the male compartments chosen at
behavior on the part of the male, allowing him to continue random. Subsequently, the chosen male either won or lost
his courtship but not be influenced by the larger male. One that fight. Our control condition was for the female to
can assume that the smaller animal had learned that the clear choose between two males and not to see a subsequent fight.
barrier prevented the larger male from actual physical attacks, We hypothesized that different IEG expression patterns
and this recognition led the smaller animals to this novel would be generated by the condition of females seeing their
strategy. The smaller ND male attended carefully to the D chosen male win or lose a fight. We then mapped brain gene
male when carrying out his behavior consistent with the atten- expression patterns, comparing the mRNA levels of egr-1 and
tion hierarchy described in a later section. c-fos in six brain nuclei comprising the SBN (Desjardins et al.,
2010).
Surprisingly, females seeing their selected male win or lose
2.02.5 Genomic Consequences of Female Mate a fight produced dramatically different brain IEG expression
Choice patterns. Females who saw their preferred male win a fight
had activated brain nuclei associated with reproduction and
For females of many species, information about potential reproductive behavior. Specifically the anterior hypothalamus,
mates can change their behavior dramatically, which makes ventromedial hypothalamus, POA, and the periaqueductal gray
sense since female mate choice is extremely important for all had significantly more activation. In striking contrast,
the survival of their offspring. We wondered what cognitive females who saw their preferred male lose a fight had a much
activity might accompany behavioral and physiological higher expression of the IEGs that were in a part of the brain
changes in females in response to potential mates. Specifi- associated with anxiety, the lateral septum (Desjardins et al.,
cally, how does the female brain respond to social informa- 2010). A remarkable aspect of these results is that the females
tion about potential mates? This experimental question were responding to visual information alone, since they did
depends on deciding both how and where to look for a signal not mate with the male, suggesting that the social information
in the brain in response to female mate choice. We chose to had caused changes in activation of key brain areas. It is impor-
measure brain sites that were marked by changes in gene tant to remember that the IEG expression we measured is surely
expression using immediate-early (IEG) genes, egr-1 and only a very small part of the total brain response and hence is
c-fos, as a female chooses a mate. IEGs are inducible transcrip- just a glimpse of the genomic response to social information.
tion factors that comprise a part of the first wave of gene However, the differential responses in specific brain areas
expression induced in neurons being activated. Extensive show that females are activating their brains based on visual
experimental work has shown that a range of natural experi- information and may use this to guide decisions about what
ences including sensory stimuli can induce IEG expression, to do.
and consequently, it has been used in mammals and birds One additional question is how this information might
(e.g., Mello et al., 1992; Rusak et al., 1990) to identify social inform the female’s choice of a mate. In a separate experiment,
responses. More recently, we showed that egr-1 is highly we performed the same protocol but after the female had
conserved in A. burtoni and that functionally it responds chosen and seen the staged fight, she had to choose again. In
robustly within 30 min of stimulation (Burmeister and Fer- this second choice, if she had seen her preferred male lose,
nald, 2005) and that c-fos is also a valuable genetic signal indi- she almost always switched her choice, and if he won, she rarely
cating brain responses in A. burtoni. As for the brain location, switched her choice (Figure 7).
we hypothesized that the conserved vertebrate social behavior
network (SBN) would be a logical place to look. The SBN was
originally described by Newman (1999) as a collection of
brain nuclei implicated in a variety of social behaviors
including male mating behavior, female sexual behavior,
parental behavior, and aggressive behavior. Anatomical
homologues to this network have subsequently been identi-
fied in birds and fish (Goodson, 2005; Goodson and Bass,
2002) and are likely to exist in other vertebrates as well. It
was unknown, however, whether they might also respond to
social information as well as to behavioral actions. In
previous experiments on female mate choice, we showed
Figure 7 Tabulation of female Astatotilapia burtoni choices after she
that reproductively ready (i.e., gravid) females prefer to asso-
saw her preferred male either win or lose a fight (top row) as compared
ciate with dominant, reproductively active males while non- to her choices after not seeing a fight (bottom row). After seeing her
gravid females prefer nondominant, nonreproductive males preferred male win a fight, the female rarely switches her choice
(Clement et al., 2005). (p ¼ 0.0002; Fishers exact test), but after seeing her preferred male
Using a similar paradigm, we placed females in a tank lose, she nearly always switches her choice (p ¼ 0.0004; Fishers exact
with one male at each end, behind a clear, watertight test; Klausner et al., in preparation).
Social Regulation of Sex: How the Brain Controls Reproductive Circuits 27

2.02.6 What Social Information Influences Changes D male cannot see them, another behavior not possible when
in Status? a D male is present. These behaviors reflect a sophisticated
social calculus in which ND males are doing the most they
2.02.6.1 Attention Hierarchies in Male Astatotilapia burtoni
can to increase their chances of becoming dominant. It is
Animals in social groups monitor the behavior of conspecifics possible that they are also learning about being dominant
and use their observations to guide their ongoing behavior. For through watching, a skill we have also reported elsewhere
example, individuals may attend to male fighting, females (Alcazar et al., 2014).
choosing mates, or many other ongoing social interactions. In an analysis of eye movements in A. burtoni, we observed
In all social hierarchies, subordinate animals attend very that when a D male was in the presence of ND males and he
closely to the behavior of dominant animals in what has oriented his eyes toward ND males, they would typically
been named an ‘attention hierarchy’ (Chance and Larsen, move out of his way (Figure 9; Fernald, 1985). This
1976). Animals watch higher-ranking animals carefully
presumably to calibrate their potential opportunities of social
ascent. Attention hierarchies have been identified in humans,
particularly in groups of children (Boulton and Smith, 1990)
where individuals modulate their behavior depending on their
own status relative to that of others. Within a hierarchy, when
a higher-ranking individual threatens or attacks a lower-
ranking individual, that individual often then subsequently
attacks an individual of still lower in rank (Vaughn and Waters,
1981). In addition to humans, attention hierarchies have been
described in baboons and mandrills (Emory, 1976) as well as
in reptiles (Summers et al., 2005).
To understand attention hierarchies in A. burtoni, we video-
taped groups of individually marked animals (n ¼ 20 per
group; four replicates) and quantified interactions between
dominant and nondominant males (Desjardins et al., 2012).
We found that D and ND males never behaved aggressively
at the same time. Even more interesting was that when the D
male was out of view in a shelter, the ND males that were larger
and attempting to ascend, behaved aggressively, and even
courted females, behaviors that never occurred when the D
male could see the ND male (Figure 8).
In this example, each time the dominant animal is out of
view, the intermediate male attacks another ND male until Figure 9 An example of eye movements during social interactions.
the dominant male reappears. When the dominant male Fish with eye positions have been drawn from film images at 160 ms
returns to the scene, he attacks within a few seconds but does intervals with the 1st through 11th frames labeled. Relative eye loca-
tions of the dominant animal are shown by lines extending stalks
not specifically target fish that have been aggressive to others
attached to the eyes at an arbitrary angle behind the central axis of the
in his absence (Desjardins et al., 2012).
eye. Note that the nondominant (ND) animals move out of the region
Our data show that the ND males are attending to the D being approached by the dominant male well before he arrives.
males and altering their behavior by acting aggressively which Redrawn from Fernald, R.D., 1985. Eye-movements in the African
is not possible when the D male is present. In addition, these cichlid fish, Haplochromis burtoni. J. Comp. Physiol. A Sens. Neural
males on occasion will approach and court females when the Behav. Physiol. 156, 199–208.

Figure 8 Schematic illustration of typical dominant male behavior in the presence of an intermediate male attempting to ascend to dominance.
Large gray rectangles represent the dominant male in his shelter and small black bars show when an individual chases or attacks another fish. Note
that intermediate males only attack other animals when they cannot be seen because the dominant male is in his shelter. From data presented in
Desjardins, J.K., Hofmann, H.A., Fernald, R.D., 2012. Social context influences aggressive and courtship behavior in a cichlid fish. PLoS One 7,
e32781.
28 Social Regulation of Sex: How the Brain Controls Reproductive Circuits

anticipation of the actions of the D male contributes to the Bystanders were then tested in a forced choice paradigm to
attention hierarchy of males within the social hierarchy and demonstrate their preference for either A and E, or B and D.
suggests that the ND males attend to specific attributes of the Both of these pairings were novel to the bystanders and differed
male behavior. only in their position in the inferred dominance hierarchy. In
both tests (A vs E and B vs D) bystander fish spent more time
near rival males that were lower in the dominance ranking
2.02.6.2 Transitive Inference in Astatotilapia burtoni
(Figure 10). These results show that fish learned the implied
Social living requires more sophisticated cognitive abilities in hierarchy as bystanders and, importantly, that A. burtoni can
many vertebrates because the social environment offers indi- use transitive inference to understand the likely outcome of
viduals an opportunity to gather and use information about fights between dominant males.
the behavior of others (Brown and Laland, 2003). But how
do animals collect and use social information (for reviews
see McGregor, 2005; Brown et al., 2011)? Eavesdropping, the
2.02.7 Conclusions
collection of information via observation can provide benefits
to the bystander (McGregor, 2005). Eavesdropping behavior,
Clearly, social information influences hormones, brain
best documented in conflict situations, suggests that individ-
action, and behavior. In the cichlid model system presented
uals adjust their fighting behavior according to knowledge
here, social information via multiple sensory systems
gained by observing their opponent (Johnstone, 2001;
impacts complex behavioral patterns and the entire HPG
Johnstone and Bshary, 2004).
axis from the expression and secretion of GnRH in the brain
In A. burtoni, we have recently shown that males can infer
to the development of sperm in the gonads. In this species,
who will win fights solely from observing pairs of individuals
we have shown that numerous cognitive processes play
fighting. This ability, called ‘transitive inference,’ is tested
a role in the regulation. Specifically, males pay close atten-
when a subject is given information about a specific relation-
tion to one another, and they can predict who will be the
ship among items based on a property of each item. In social
dominant male in a fight. Females also make choices that
animals, the ability to make inferences about other individuals’
reflect their understanding of social dominance among
relative place in a dominance hierarchy and, therefore, predict
males. It has been a mystery how GnRH neurons are con-
the outcome of competition should be a useful skill (Emery,
nected to produce the requisite pulsatile secretion. We
2006). Transitive inference has been demonstrated in a number
suggest that the gap junctions we describe between these
of avian species including pigeons, jays, and corvids (for
neurons are likely central to that key function. While we
summary and review see Emery, 2006). However, we recently
now have a better understanding of the relationship between
showed that A. burtoni males show transitive inference about
social behavior, use of social information, and brain activa-
dominance among other animals Grosenick et al. (2007).
tion, we do not know how animals process and store social
Two groups of dominant bystander fish saw staged, pairwise
information and then retrieve it in the service of complex
fights between five pairs of conspecifics. One group of five
cognitive tasks. The cichlid fish, A. burtoni, because of its
had a dominance hierarchy of A > B > C > D > E while the
complex social organization and the wealth of background
other had no dominance hierarchy (e.g., A ¼ B ¼ C ¼ D ¼ E).
information into brain activation, is likely to be integral in
the study of dominance, social information, and their impact
on the brain.

Acknowledgments

I have been privileged to work with a superb collection of undergrad-


uates, graduate students, and postdoctoral fellows without whom this
work would not have been possible. Supported by NIH-NS 034950,
NIH MH101373, and NSF IOS-0923588 to R.D.F.

References
Figure 10 Time spent near rival males during preference testing. Bar
plots of the difference in time spent near the higher-ranking versus the Alcazar, R.M., Hilliard, A.T., Becker, L., Bernaba, M., Fernald, R.D., 2014. Brains over
lower-ranking rival dominant males in the AE (top) and BD (bottom) brawn: experience overcomes a size disadvantage in fish social hierarchies. J. Exp.
preference tasks by context. Filled bars, familiar context (e.g., Biol. 217, 1462–1468.
rearranged training aquarium); open bars, novel context (e.g., new Au, T.M., Greenwood, A.K., Fernald, R.D., 2006. Differential social regulation of two
pituitary gonadotropin-releasing hormone receptors. Behav. Brain Res. 170,
aquarium). One-sample permutation t-tests (50 000 replicates) showed
342–346.
that in all cases bystanders spent significantly more time near the
Avitsur, R., Stark, J.L., Sheridan, J.F., 2001. Social stress induces glucocorticoid
lower-ranking rival, namely D or E (AE familiar context, P50.01; AE resistance in subordinate animals. Horm. Behav. 39, 247–257.
novel context, P50.003; BD familiar context, P50.003; BD novel context, Belchetz, P.E., Plant, T.M., Nakai, Y., Keogh, E.J., Knobil, E., 1978. Hypophyseal
P50.008; where P50.01 etc. refers to 50 000 replicates with a 0.01 responses to continuous and intermittent delivery of hypothalamic gonadotropin-
probability of significance). Error bars indicate s.e.m. releasing hormone. Science 202, 631–633.
Social Regulation of Sex: How the Brain Controls Reproductive Circuits 29

Boulton, M., Smith, P.K., 1990. Affective bias in children’s perceptions of dominance Greenwood, A.K., Butler, P.C., White, R.B., Demarco, U., Pearce, D., Fernald, R.D.,
relationships. Child. Dev. 61, 221–229. 2003. Multiple corticosteroid receptors in a teleost fish: distinct sequences,
Brown, C., Laland, K.N., 2003. Social leaning in fishes: a review. Fish Fish. 4, expression patterns, and transcriptional activities. Endocrinology 144, 4226–4236.
280–288. Greenwood, A.K., Fernald, R.D., 2004. Social regulation of the electrical properties of
Brown, C., Laland, K.N., Krause, J., 2011. Fish cognition and behavior. In: Pitcher, T. gonadotropin-releasing hormone neurons in a cichlid fish (Astatotilapia burtoni).
(Ed.), Fish and Aquatic Resources Series. Wiley-Blackwell. Biol. Reprod. 71, 909–918.
Bshary, R., Würth, M., 2001. Cleaner fish Labroides dimidiatus manipulate client reef Grosenick, L., Clement, T.S., Fernald, R.D., 2007. Fish can infer social rank by
fish by providing tactile stimulation. Proc. Biol. Sci. 268, 1495–1501. observation alone. Nature 445, 429–432.
Bshary, R., Wickler, W., Fricke, H., 2002. Fish cognition: a primate’s eye view. Anim. Harbott, L.K., Burmeister, S.S., White, R.B., Vagell, M., Fernald, R.D., 2007. Androgen
Cogn. 5, 1–13. receptors in a cichlid fish, Astatotilapia burtoni: structure, localization, and
Burmeister, S.S., Fernald, R.D., 2005. Evolutionary conservation of the egr-1 expression levels. J. Comp. Neurol. 504, 57–73.
immediate-early gene response in a teleost. J. Comp. Neurol. 481, 220–232. Heiligenberg, W., Kramer, U., Schulz, V., 1972. The angular orientation of the black
Burmeister, S.S., Jarvis, E.D., Fernald, R.D., 2005. Rapid behavioral and genomic eye-bar in Haplochromis burtoni (Cichlidae, Pisces) and its relevance to aggres-
responses to social opportunity. PLoS Biol. 3, e363. sivity. Z. Tierpsychol. 76, 168–176.
Burmeister, S.S., Kailasanath, V., Fernald, R.D., 2007. Social dominance regulates Heiligenberg, W., K, U., 1972. Aggressiveness as a function of external stimulation.
androgen and estrogen receptor gene expression. Horm. Behav. 51, 164–170. J. Comp. Physiol. Psychol. 77, 332–340.
Bushnik, T.L., Fernald, R.D., 1995. The population of GnRH-containing neurons Johnstone, R.A., 2001. Eavesdropping and animal conflict. Proc. Natl. Acad. Sci.
showing socially mediated size changes project to the pituitary in a teleost, U.S.A. 98, 9177–9180.
Haplochromis burtoni. Brain Behav. Evol. 46, 371–377. Johnstone, R.A., Bshary, R., 2004. Evolution of spite through indirect reciprocity
Chance, M., Larsen, R.R., 1976. Social Structure of Attention. John Wiley & Sons Ltd. (vol. 271, p. 1917, 2004). Proc. R. Soc. B Biol. Sci. 271, 2637.
Chen, C.C., Fernald, R.D., 2006. Distributions of two gonadotropin-releasing hormone Juntti, S.A., Hu, C.K., Fernald, R.D., 2013. Tol2-mediated generation of a transgenic
receptor types in a cichlid fish suggest functional specialization. J. Comp. Neurol. haplochromine cichlid, Astatotilapia burtoni. PLoS One 8.
495, 314–323. Kaplan, J.R., 2008. Origins and health consequences of stress-induced ovarian
Chen, C.C., Fernald, R.D., 2011. Visual information alone changes behavior and dysfunction. Interdiscip. Top. Gerontol. 36, 162–185.
physiology during social interactions in a cichlid fish (Astatotilapia burtoni). PLoS Kaplan, J.R., Manuck, S.B., 2004. Ovarian dysfunction, stress, and disease: a primate
One 6, e20313. continuum. ILAR J. 45, 89–115.
Clement, T.S., Parikh, V., Schrumpf, M., Fernald, R.D., 2005. Behavioral coping Korzan, W.J., Grone, B.P., Fernald, R.D., 2014. Social regulation of cortisol receptor
strategies in a cichlid fish: the role of social status and acute stress response in gene expression. J. Exp. Biol. 217, 3221–3228.
direct and displaced aggression. Horm. Behav. 47, 336–342. Laland, K., Brown, C., Krause, J., 2003. Learning in fishes: from three-second
Coulter, G., 1991. Lake Tanganyika and Its Life. Oxford University Press, Oxford. memory to culture. Fish Fish. 4, 199–202.
Davis, M.R., Fernald, R.D., 1990. Social control of neuronal soma size. J. Neurobiol. Leong, C.-Y., 1969. The quantitative effect of releasers on the attack readiness of the
21, 1180–1188. fish Haplochromis burtoni. Z. Tierpsychol. 65, 29–50.
Desjardins, J.K., Hofmann, H.A., Fernald, R.D., 2012. Social context influences Lorenz, K., 1937. The companion in the bird’s world. Auk 54, 245–273.
aggressive and courtship behavior in a cichlid fish. PLoS One 7, e32781. Ma, Y.Y., Juntti, S.A., Hu, C.K., Huguenard, J.R., Fernald, R.D., 2015. Electrical
Desjardins, J.K., Klausner, J.Q., Fernald, R.D., 2010. Female genomic response to synapses connect a network of gonadotropin releasing hormone neurons in
mate information. Proc. Natl. Acad. Sci. U.S.A. 107, 21176–21180. a cichlid fish. Proc. Natl. Acad. Sci. U.S.A. 112, 3805–3810.
Emery, N.J., 2006. Cognitive ornithology: the evolution of avian intelligence. Philos. Maruska, K., Fernald, R.D., 2014. Social regulation of gene expression in the African
Trans. R. Soc. B Biol. Sci. 361, 23–43. cichlid fish Astatotilapia burtoni. In: Canli, T. (Ed.), Oxford Handbook of Molecular
Emory, G.R., 1976. Aspects of attention, orientation, and status hierarchy in mandrills Psychology. Oxford University Press.
(Mandrillus sphinx) and gelada baboons (Theropithecus gelada). Behaviour 59, McGregor, P., 1993. Signaling in territorial systems: a context for individual identifi-
70–87. cation, ranging and eavesdropping. Phil. Trans. 340, 237–244.
Fernald, R.D., 1977. Quantitative behavioral observations of Haplochromis burtoni McGregor, P., 2005. Animal Communication Networks. Cambridge University Press.
under semi-natural conditions. Anim. Behav. 25, 643–653. Mello, C.V., Vicario, D.S., Clayton, D.F., 1992. Song presentation induces gene-
Fernald, R.D., 1984. Vision and behavior in an African cichlid fish. Am. Sci. 72, expression in the songbird forebrain. Proc. Natl. Acad. Sci. U.S.A. 89,
58–65. 6818–6822.
Fernald, R.D., 1985. Eye-movements in the African cichlid fish, Haplochromis burtoni. Newman, S.W., 1999. The medial extended amygdala in male reproductive behavior –
J. Comp. Physiol. A Sens. Neural Behav. Physiol. 156, 199–208. a node in the mammalian social behavior network. Advancing from the Ventral
Fernald, R.D., 1997. The evolution of eyes. Brain Behav. Evol. 50, 253–259. Striatum to the Extended Amygdala Ann. N.Y. Acad. Sci. 877, 242–257.
Fernald, R.D., 2012. Social control of the brain. Annu. Rev. Neurosci. 35. Phoenix, C.H., Goy, R.W., Gerall, A.A., Young, W.C., 1959. Organizing action of
Fernald, R.D., Hirata, N.R., 1977a. Field study of Haplochromis burtoni: habitats and prenatally administered testosterone propionate on the tissues mediating mating
co habitants. Environ. Biol. Fish. 2, 299–308. behavior in the female guinea pig. Endocrinology 65, 369–382.
Fernald, R.D., Hirata, N.R., 1977b. Field study of Haplochromis burtoni: Quantitative Pitcher, T.J., Green, D.A., Magurran, A.E., 1986. Dicing with death – predator
behavioral observations. Anim. Behav. 25, 964–975. inspection behavior in minnow shoals. J. Fish Biol. 28, 439–448.
Flanagan, C.A., Chen, C.C., Coetsee, M., Mamputha, S., Whitlock, K.E., Robison, R.R., White, R.B., Illing, N., Troskie, B.E., Morley, M., Millar, R.P.,
Bredenkamp, N., Grosenick, L., Fernald, R.D., Illing, N., 2007. Expression, Fernald, R.D., 2001. Gonadotropin-releasing hormone receptor in the teleost
structure, function, and evolution of gonadotropin-releasing hormone (GnRH) Haplochromis burtoni: structure, location, and function. Endocrinology 142,
receptors GnRH-R1SHS and GnRH-R2PEY in the teleost, Astatotilapia burtoni. 1737–1743.
Endocrinology 148, 5060–5071. Rusak, B., Robertson, H.A., Wisden, W., Hunt, S.P., 1990. Light-pulses that shift
Fox, H.E., White, S.A., Kao, M.H., Fernald, R.D., 1997. Stress and dominance in rhythms induce gene-expression in the suprachiasmatic nucleus. Science 248,
a social fish. J. Neurosci. Off. J. Soc. Neurosci. 17, 6463–6469. 1237–1240.
Francis, R.C., Jacobson, B., Wingfield, J.C., Fernald, R.D., 1992a. Hypertrophy of Sapolsky, R.M., 1996. Why stress is bad for your brain. Science 273, 749–750.
gonadotropin releasing hormone-containing neurons after castration in the teleost, Schaaf, M.J.M., Champagne, D., Van Laanen, I.H.C., Van Wijk, D.C.W.A.,
Haplochromis burtoni. J. Neurobiol. 23, 1084–1093. Meijer, A.H., Meijer, O.C., Spaink, H.P., Richardson, M.K., 2008. Discovery of
Francis, R.C., Jacobson, B., Wingfield, J.C., Fernald, R.D., 1992b. Castration lowers a functional glucocorticoid receptor beta-isoform in zebrafish. Endocrinology
aggression but not social-dominance in male Haplochromis burtoni (Cichlidae). 149, 1591–1599.
Ethology 90, 247–255. Soma, K.K., Francis, R.C., Wingfield, J.C., Fernald, R.D., 1996. Androgen regulation of
Francis, R.C., Soma, K., Fernald, R.D., 1993. Social regulation of the brain-pituitary- hypothalamic neurons containing gonadotropin-releasing hormone in a cichlid fish:
gonadal axis. Proc. Natl. Acad. Sci. U.S.A. 90, 7794–7798. integration with social cues. Horm. Behav. 30, 216–226.
Goodson, J.L., 2005. The vertebrate social behavior network: evolutionary themes and Summers, C.H., Forster, G.L., Korzan, W.J., Watt, M.J., Larson, E., Overli, O.,
variations. Horm. Behav. 48, 11–22. Hoglund, E., Ronan, P.J., Summers, T.R., Renner, K.J., Greenberg, N., 2005.
Goodson, J.L., Bass, A.H., 2002. Vocal-acoustic circuitry and descending vocal Dynamics and mechanics of social rank reversal. J. Comp. Physiol. A Neuroethol.
pathways in teleost fish: convergence with terrestrial vertebrates reveals conserved Sens. Neural Behav. Physiol. 191, 241–252.
traits. J. Comp. Neurol. 448, 298–322. Uexküll, J.J.B.V., 1921. Umwelt und Innenwelt der Tiere. Julius Springer, Berlin.
30 Social Regulation of Sex: How the Brain Controls Reproductive Circuits

Vaughn, B.E., Waters, E., 1981. Attention structure, sociometric status, and domi- White, S.A., Fernald, R.D., 1993. Gonadotropin-releasing hormone-containing neurons
nance – interrelations, behavioral-correlates, and relationships to social compe- change size with reproductive state in female Haplochromis burtoni. J. Neurosci.
tence. Dev. Psychol. 17, 275–288. Off. J. Soc. Neurosci. 13, 434–441.
Webster, R.I., Majnemer, A., Platt, R.W., Shevell, M.I., 2008. Child health and parental White, S.A., Kasten, T.L., Bond, C.T., Adelman, J.P., Fernald, R.D., 1995. Three
stress in school-age children with a preschool diagnosis of developmental delay. gonadotropin-releasing hormone genes in one organism suggest novel roles for an
J. Child. Neurol. 23, 32–38. ancient peptide. Proc. Natl. Acad. Sci. U.S.A. 92, 8363–8367.
White, R.B., Fernald, R.D., 1998. Ontogeny of gonadotropin-releasing hormone (GnRH) White, S.A., Nguyen, T., Fernald, R.D., 2002. Social regulation of gonadotropin-
gene expression reveals a distinct origin for GnRH-containing neurons in the releasing hormone. J. Exp. Biol. 205, 2567–2581.
midbrain. Gen. Comp. Endocrinol. 112, 322–329. Wickler, W., 1962. Zur Stammesgeschichte funktionell korrelierter Organ-und Ver-
White, S.A., Bond, C.T., Francis, R.C., Kasten, T.L., Fernald, R.D., Adelman, J.P., haltensmerkmale: Eiattrappen und Maubrüten bei afrikanischen Cichliden.
1994. A second gene for gonadotropin-releasing hormone: cDNA and expression Z. Tierpsychol. 19, 129–164.
pattern in the brain. Proc. Natl. Acad. Sci. U.S.A. 91, 1423–1427.
2.03 Socially Controlled Sex Change in Fishes
John Godwin and Melissa Lamm, North Carolina State University, Raleigh, NC, USA
Ó 2017 Elsevier Inc. All rights reserved.

2.03.1 Introduction 31
2.03.1.1 Hermaphroditism in Fishes 32
2.03.1.2 Social Regulation of Sex Change 33
2.03.1.3 Gonadal Change in Sex-Changing Fishes 33
2.03.1.4 Steroid Hormones and Sex Change 34
2.03.1.4.1 Sex Steroids and Behavioral Change 36
2.03.1.4.2 Corticosteroids and Sex Change 38
2.03.2 Neural Signaling Systems and Sex Change 39
2.03.2.1 Gonadotropin-Releasing Hormone and Gonadotropins 39
2.03.2.2 Kisspeptins and Gonadotropin-Inhibitory Hormone 39
2.03.2.3 Monoamine Neurotransmitters and Sex Change 40
2.03.2.4 Nonapeptides and Sex Change 41
2.03.3 Conclusions and Future Directions 42
References 43

2.03.1 Introduction gobies (Gobiidae), and others (see Godwin, 2009, 2010;
Kobayashi et al., 2013; Lamm et al., 2015 for recent reviews).
Teleost fishes are the oldest and, in many respects, the most While the reasons why sex change in general and socially
diverse of the vertebrate lineages. This diversity takes a variety controlled sex change in particular are observed in fishes and
of forms, but one that particularly stands out is the variety of not in other groups are not clear, several possibilities have
sexual expression seen across teleosts. Teleosts exhibit the been suggested. These include reduced evolutionary constraints
full range of sexual development patterns observed in in fishes with external fertilization since the ducts for release of
vertebrates more generally (see Devlin and Nagahama,
2002; de Mitcheson and Liu, 2008; Kobayashi et al., 2013
for detailed reviews; Figure 1). The majority of fish
Gonochorism Adult female
species display the familiar pattern of genotypic sex
determination and gonochorism, where individuals mature Adult male
as one sex or the other and remain that sex. However, there
is wide variation in genotypic sex-determining mechanisms Gonochorism with alternate male phenotypes
across fishes including male heterogamety, female hetero- Adult female
gamety, and polyfactorial sex determination. Some groups Fixed expression
of AMP Parasitic male
of fishes display gonochorism that is characterized instead
by environmental sex determination in two major forms. Bourgeois male
Temperature-dependent sex determination (TSD) is seen in Adult female
Plastic expression
several groups of fishes and shows some strong similarities of AMP Bourgeois
Parasitic male male
in patterns and mechanisms to those described especially for
reptiles. Fishes also exhibit social determination of sex, Protandry
a phenomenon that is not seen in other vertebrate groups. Juvenile (female) Adult male Adult female
This can occur either before or after sexual maturation
and has been documented from 27 families of fishes Monandric protogyny)
(de Mitcheson and Liu, 2008). The behavioral and neuroen- Juvenile (female) Adult female Adult male
docrine aspects of sex change that occur postmaturationally
in response to social cues are the focus of this chapter. Diandric protogyny Adult female
Terminal phase
Socially controlled sex change was first documented in the Juvenile (female)
(TP) male
early 1970s for two coral reef dwelling species, the fairy basslet Adult initial phase
Pseudanthias squamipinnis and the Indo-Pacific cleaner wrasse (IP) male
Labroides dimidiatus (Fishelson, 1970; Robertson, 1972, respec- Bidirectional sex change Adult female
tively; Pseudanthias was formerly classified as Anthias). This Juvenile (female)
sexual pattern has subsequently been documented for a variety Adult male
of other species including other members of the families that
include wrasses (Labridae), parrotfishes (Scaridae), damsel- Figure 1 Range of sexual patterns in fishes for both gonochoristic
fishes (Pomacentridae), groupers and basslets (Serranidae), and hermaphroditic species. ‘AMP’ refers to alternate male phenotypes.

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00022-5 31


32 Socially Controlled Sex Change in Fishes

gametes are relatively similar between males and females, and promising directions for the study of this phenomenon in
less modification is therefore necessary with a change in sex future studies.
(Francis, 1992). There is also strong selection for sex change
in some species that can provide evolutionary advantages to
2.03.1.1 Hermaphroditism in Fishes
sex-changing individuals (Ghiselin, 1969; Warner et al., 1975;
Warner, 1984). Finally, it has been proposed that there is The diversity of sexual patterns in fishes exceeds that observed
a fundamental difference in the relative timing of differentia- in any other group of vertebrates. These patterns have been
tion for the brain and gonadal tissues between tetrapod verte- reviewed in detail elsewhere starting with Atz in 1964 and
brates and at least many species of fishes (Francis, 1992). with many excellent contributions in the years since (Rein-
Specifically, while the gonads develop relatively early and the both, 1980, 1988; Devlin and Nagahama, 2002; de Mitcheson
production of gonadal hormones plays an important role in and Liu, 2008), so only a relatively broad overview is
organizing the brain in well-studied mammalian models, the provided here. The diversity in sexual patterns displayed by
reverse is generally true for fishes. Gonadal differentiation teleosts includes several patterns that do not involve sex
occurs during a free-living and fully independent juvenile change, and most groups of fishes are gonochoristic, maturing
period where developing individuals are essentially ‘miniature as either males or females and remaining that sex through the
adults’ fully able to sense and integrate information about their life history. However, gonochoristic species can nevertheless
abiotic and social environments. This creates at least the possi- exhibit sexual determination that is wholly or partly deter-
bility of adaptively adjusting sex determination in the direction mined by environmental cues. While best characterized in
likely to be reproductively advantageous for the individual. reptiles, TSD is seen in several groups of fishes with the best
Consistent with this hypothesis, studies in cichlid and labrid studied examples being from the Atheriniforms (Conover,
fishes show that social dominance can significantly influence 2004) and flatfishes (Luckenbach et al., 2009). The key envi-
sexual differentiation with larger individuals disproportion- ronmental influence can also be social as demonstrated for
ately becoming males (Francis and Barlow, 1993; Munday Midas cichlids (Francis and Barlow, 1993) and initial sex
et al., 2006). Social influences on sex determination in adult differentiation in bluehead wrasses (Munday et al., 2006 –
functional sex change may act through similar physiological discussed further below). Many species that are gonochoristic
mechanisms. nevertheless display lability in the development of secondary
Sex-changing fishes provide attractive models for studying sexual characteristics and behavior to produce within-sex
the hormonal bases of sexual and aggressive behavior for dimorphism and alternate mating phenotypes (reviewed in
several reasons. The first is that, unlike most model species, Moore, 1991; Taborsky, 1998; Godwin, 2010). Alternate
sexual developmental processes can be studied in an adult male mating phenotypes are also seen in a number of sex-
animal in which behavioral changes take place as sexual devel- changing species including especially the wrasses and
opment is occurring. A second experimentally valuable feature parrotfishes.
of sex change is that it often occurs relatively quickly, particu- Forms of sexual lability seen in mature fishes include
larly with protogynous (female to male) species. Both cleaner sequential hermaphroditism, simultaneous hermaphroditism,
wrasses and bluehead wrasses complete gonadal sex change and some species that are simultaneous hermaphrodites before
in approximately 1–2 weeks following removal of dominant losing ovarian tissue to become males. Sequential hermaphro-
males from social groups in nature (Robertson, 1972; Warner ditism or sex change occurs in both protandrous (male-to-
and Swearer, 1991). Some anemonefish and gobies also female) and protogynous (female-to-male) directions. Protandry
show rapid behavioral changes during sex change (Godwin, (‘first male’) is characterized by male-to-female sex change.
1994a; Black et al., 2005, 2011). Another attractive feature of Protandry may also be a functional description for develop-
many sex-changing species is the presence of alternate male mental patterns that are essentially protogynous. For example,
mating phenotypes that exhibit within-sex dimorphisms in anemonefishes in the genera Amphiprion and Premnas mature as
morphology and behavior that are useful for exploring males and can become females on becoming socially dominant
hormonal mechanisms regulating these characteristics (Moore, members of groups resident on large tropical sea anemones.
1991; Crews, 1993; Godwin, 2010). In these species, one of However, examining development in several of these species
these phenotypes is typically large, often colorful, and exhibits shows that gonads are initially immature ovaries and only
strong courtship, territorial defense, and aggression. The second develop testicular tissue if an individual assumes the male
male type typically exhibits what Taborsky termed ‘parasitic’ breeding role (Fricke and Fricke, 1977; Fricke, 1983; Godwin,
male tactics where reproductive success is obtained through 1994b). Protogyny (‘first female’) is characterized by func-
what are termed ‘sneaker’ or ‘streaker’ tactics (Gross, 1980; tional, postmaturational female-to-male sex change. Monandric
Taborsky, 1998). protogynous species tend to first develop as immature females
This chapter primarily focuses on the hormonal regula- that can then first sexually mature as females in what is termed
tion of socially regulated sex change and both within- and the initial phase (IP) and then later change sex to become males
between-sex differences in behavior in species that exhibit in the terminal phase (TP males). Diandric protogynous
this sexual pattern. We first introduce the different forms species, by contrast, can mature as either females or males in
of socially regulated sex change observed in teleost fishes. the IP and then change to become TP males later. This develop-
Then, we review the roles of steroid hormones, neuropep- ment of TP males involves sex change for females and role
tide hormones and other neural signaling systems in regu- change for IP males. The transition to TP male by either females
lating sex differences in morphology and behavior as well or IP males also often involves the development of TP-typical
as sex change. This will be followed by a discussion of morphology and behaviors including defense of a breeding
Socially Controlled Sex Change in Fishes 33

territory and increased display of aggressive and courtship presumably olfactory, cues affected the incidence of sex change
behaviors. in isolated females of the goby Coryphopterus glaucofraenum,
Many species in the family Serranidae, which contains the although group size was also important. Females in tanks
groupers as well as basslets and hamlets, display simultaneous receiving water from tanks housing conspecific females were
hermaphroditism. This sexual pattern is characterized by the significantly more likely to change sex than females in tanks
presence of mature ovarian and testicular tissue within the receiving water from tanks holding conspecific males. Fishes
same gonad, and individuals can alternate between female can be extremely sensitive to pheromonal cues that affect the
and male mating roles over the course of a brief spawning reproductive axis (reviewed in Stacey, 1987). It is interesting
period in an elaborate mating behavior termed ‘egg trading’ to note that increases in close ‘inspection’ of the cloacal region
(Fischer, 1980). Other serranid fishes can display simultaneous of gravid females is one of the first clear behavioral changes
hermaphroditism when relatively small and then lose the seen in sex-changing female bluehead wrasses (Godwin and
ovarian tissue to become males on attaining large size and Lamm, pers. obs.). These ‘inspections’ could expose them to
social dominance (Petersen, 1987). important olfactory cues to help initiate sex change.
Sex change is typically unidirectional, being either protogy- The pattern of exposure to key sensory cues also seems likely
nous or protandrous. However, a number of goby species to be very important in regulating sex change. This is primarily
exhibit bidirectional sex change. This is hypothesized to be seen in experiments that vary the ratio of males and females
an adaptation in at least some cases to strong predation pres- that a sex change candidate is exposed to. Increasing the fema-
sure in their coral reef habitats (Hobbs et al., 2004). Gobies le:male ratio in experimental groups increases the likelihood of
are typically relatively small, bottom-dwelling fishes that are sex change in the fairy basslet P. squamipinnis (Shapiro, 1980),
not strong swimmers, and it would therefore be dangerous to the saddleback wrasse T. duperrey (Ross et al., 1990), the bridled
move from their densely branching coral homes to find mates. goby C. glaucofraenum (Cole and Shapiro, 1995), and Potter’s
Bidirectional sex change allows any two conspecific gobies on angelfish Centropyge potteri (Lutnesky, 1994). Lutnesky showed
a coral head to form a female–male pair for breeding. quantitatively that the encounter rate of female C. potteri with
males was negatively associated with the occurrence of
female-to-male sex change. It is also noteworthy that removal
2.03.1.2 Social Regulation of Sex Change
of dominant TP male bluehead wrasses from small reefs results
A full understanding of socially controlled sex change will in an approximate one-to-one replacement by large females
require understanding the sequence of physiological events who change sex, suggesting sex ratios are important in natural
that ‘transduce’ a social cue or cues into the neuroendocrine social groups as well (Warner and Swearer, 1991). It is
changes that regulate gonadal transformation. While we have intriguing to consider how encounter rates with larger and
clear evidence that social interactions can regulate sex change, smaller conspecifics might influence the neural systems regu-
the sensory modalities that mediate these social influences lating behavioral and gonadal sex change, but no studies
are not well understood. have yet directly examined this question.
The initial studies showing social control of sex change in
two coral reef fishes, the fairy basslet P. squamipinnis and cleaner
2.03.1.3 Gonadal Change in Sex-Changing Fishes
wrasse L. dimidiatus, showed that removal of large and socially
dominant males from social groups led to rapid sex change by Sex change involves a dramatic restructuring of the gonad that
the largest resident female. Later demonstrations of similar can occur very rapidly. For example, both bluehead and cleaner
inhibitory influences on sex change by dominant secondary wrasses can completely transform a functional and mature
sex individuals followed in a number of other species including ovary into a mature testes and become capable of fertilizing
both protogynous and protandrous sex changers (e.g., Fricke, eggs in as little as 7 to 10 days. A full review of gonadal changes
1983; Ross et al., 1983; Warner and Swearer, 1991; Godwin, in sex-changing fishes is beyond the scope of this chapter, so
1994b; Black et al., 2005). Precisely what are the critical inhib- just a few examples are provided here.
itory influences provided by these socially dominant individ- Gonadal change has been well studied in protogynous Tha-
uals remain poorly understood. There is evidence for both lassoma wrasses. Nakamura et al. (1989) described the process
visual and olfactory cues providing this inhibitory input, and of sex change in the saddleback wrasse T. duperrey at both the
auditory/vibrational cues are possible as well. light and electron microscopic level. Figure 2 illustrates the
One of the most in-depth studies of sensory cues regulating sequence of changes in both gonadal structure and body color-
sex change was conducted by Ross et al. (1983) in the Hawaiian ation over the course of sex change in bluehead wrasses (Thalas-
saddleback wrasse (Thalassoma duperrey). Working in experi- soma bifasciatum). In this species, females induced to change sex
mental pens, they were able to show that female-to-male sex by removal of the large TP males from their social groups on
change in this species involved both stimulation from small small reefs in the field exhibit very rapid changes in behavior,
conspecifics and inhibition from larger conspecifics. Visual gonadal structure, and body coloration. The ovaries exhibit
cues are of critical importance as allowing sex-change candi- breakdown in the first 2–3 days of sex change, exhibiting
dates to see, but not physically interact with a larger conspecific, atresia of large, mature follicles. This is followed by the begin-
prevented sex change. Blocking visual contact with a porous nings of spermatogenesis approximately 4–5 days after the
barrier did not allow females to undergo sex change despite still initiation of sex change (TP male removal). This new spermato-
presumably being exposed to olfactory and possibly auditory genic tissue arises within the lamellae containing degenerating
cues. By contrast, olfactory cues appear to be important in other ovarian tissue. Coloration changes depend on the teleost
species. Cole and Shapiro (1995) found that waterborne, androgen 11-ketotestosterone (11KT), analogous to the actions
34 Socially Controlled Sex Change in Fishes

(a) (b) (c)

(d) (e) (f)

Figure 2 Morphological and gonadal sex change in the bluehead wrasse. Left: color changes from female (top) to TP male (bottom) (image by J.
Godwin). Right: six gonadal sex change stages from ovary to functional testis (images by M. Lamm and J. Godwin). (a) Stage 1: mature ovary with
healthy vitellogenic oocytes (VO) and previtellogenic oocytes (PVO); (b) stage 2: atretic vitellogenic oocytes (AVO) with degraded zona pellucida;
(c) stage 3: atretic previtellogenic oocytes (APVO); (d) stage 4: proliferation of presumed spermatogonia (SG) and Leydig cells; (e) stage 5: onset of
spermatogenesis, indicated by spematocytes (SC) in spermatocysts; (f) stage 6: mature, tailed sperm (SP). Scale bar: 50 mm. Based on classification
by Nakamura, M., Hourigan, T.F., Yamauchi, K., Nagahama, Y., Grau, E.G., 1989. Histological and ultrastructural evidence for the role of gonadal
steroid hormones in sex change in the protogynous wrasse Thalassoma duperrey. Environ. Biol. Fishes 24 (2), 117–136.

of dihydrotestosterone in many tetrapods. The beginning of focused on these mediators since the earliest physiological
permanent coloration changes in sex-changing bluehead studies of sex change. The first studies examined the effects
wrasses coincides approximately with the onset of spermato- of exogenous hormone administration on the occurrence of
genesis (Lamm et al., 2015). These sex changers can then be sex change, beginning with Stoll (1955) manipulating andro-
capable of fertilizing eggs from female spawning partners in gens in the bluehead wrasse. However, as techniques for
as few as 8 days after the initiation of sex change. measurement developed there has been an increasing focus
Anemonefishes exhibit protandrous sex change and a social on examining patterns in circulating steroid hormone levels
system characterized by mating pairs on host anemones con- and biosynthesis across sexual phenotypes and sex change.
sisting of large, socially dominant females, mature males, and Attention has primarily focused on gonadal steroid
often a variable number of subadults. Removal of the domi- hormones, but increasing evidence suggests a role for gluco-
nant females stimulates male-to-female sex change in the corticoids in sex determination events both during develop-
mature males (Fricke and Fricke, 1977; Godwin, 1994a,b). ment in gonochoristic species and in adult sex change in
The gonads of these mature males consist of an immature hermaphroditic species.
ovarian portion and a distinct part of the gonad that is mature As with gonochoristic fishes, sex-changing species often
spermatogenic tissue. In the anemonefish Amphiprion melano- show strong sexual dimorphisms in circulating levels of sex
pus, this spermatogenic portion of the gonad rapidly matures steroid hormones (Table 1). As in tetrapods, estradiol 17b
in the 10 days after onset of sex change, but immature oocytes (E2) is the key circulating estrogen in fishes and tends to be
replace this tissue by 10 days later and go on to mature as the found at higher levels in females (Devlin and Nagahama,
entire gonad becomes a mature ovary. 2002). The key hormone responsible for inducing male
Perhaps the most interesting examples of gonadal transfor- secondary sexual characteristics in fishes is the nonaromatiz-
mation are seen in several genera of bidirectionally sex- able 11KT, which plays an analogous role in this process to
changing gobies. In these species, the tissues that form the that played by dihydrotestosterone in tetrapods. Expression
ovarian and spermatogenic portions of the gonad are physi- of these male secondary sexual characteristics, primarily color
cally delimited, and sex change involves activation of one changes and often elaboration of fins, coincides with the onset
portion and inactivation of the other. The goby Trimma okina- of spermatogenesis and rising 11KT levels (e.g., Nakamura
wae exhibits serial bidirectional sex changes that can be et al., 1989; Hourigan et al., 1991; Cardwell and Liley,
completed in as little as 4 days, and these involve rapid alter- 1991a,b; Warner and Swearer, 1991; Ohta et al., 2007;
ations in gonadotropin receptor mRNA expression (Kobayashi Figure 3). These changes can also be induced by exogenous
et al., 2009). We return to these rapid endocrine changes in androgens (Stoll, 1955; Cardwell and Liley, 1991a; Semsar
gobies in the next section. and Godwin, 2004). The role of testosterone in fishes is some-
what less clear (Borg, 1994; Frisch, 2005; Godwin, 2010) and
shows a less clear pattern in terms of female–male differences
2.03.1.4 Steroid Hormones and Sex Change
including in sex-changing species where testosterone levels
Sex steroid hormones play key roles in sex determination and may be higher in females (e.g., Godwin and Thomas, 1993;
differentiation across vertebrate animals, and so attention has Figure 3). Testosterone appears to be important primarily as
Socially Controlled Sex Change in Fishes 35

Table 1 Circulating steroid levels in sex-changing fishes

Direction of
Species, common name Family sex change E2 Testosterone 11-Ketotestosterone References

Thalassoma duperrey, Labridae Diandric \ > (⚥ ¼ TP_) \ ¼ ⚥ ¼ TP_ (\ ¼ ⚥) < TP_ Nakamura et al. (1989)
saddleback wrasse protogyny IP_ ¼ TP_ IP_ ¼ TP_ IP_ < TP_ and Hourigan et al.
(1991)
Halichoeres trimaculatus, Labridae
three-spot wrasse
Pseudolabrus sieboldi, Labridae Protogyny \¼⚥¼_ NA \<⚥<_ Ohta et al. (2008)
bambooleaf wrasse
Sparisoma viride, stoplight Scaridae Diandric \ > (⚥ ¼ TP_) \ ¼ ⚥, \ < TP_ \ << (⚥ ¼ TP_) Cardwell and Liley
parrotfish protogyny IP_ < TP_ (1991a,b)
Amphiprion melanopus, Pomacentridae Protandry \ > (_ ¼ ⚥) \>(_ ¼ ⚥) _>(⚥ ¼ \) Godwin and Thomas
Cinnamon anemonefish (1993)
Lythrypnus dalli, Gobiidae Protogyny \>_ Mixed results \¼_ Lorenzi et al. (2008)
bluebanded goby and Lorenzi et al.
(2012)
Gobiodon histrio, coral Gobiidae Bidirectional \>_ \¼_ Kroon et al. (2003)
goby
G. erythrospilus, coral goby Gobiidae Bidirectional \¼_ Kroon et al. (2009)
Coryphopterus nicholsii, Gobiidae Protogyny \>_ \<_ \<_ Kroon and Liley (2000)
blackeye goby
Plectropomus leopardus, Serranidae Protogyny \>_ \¼_ \<_
coral trout

IP, initial phase; TP, terminal phase; E2, estradiol 17b.

a ‘prohormone’ in many species of fishes, serving as a biochem- (degeneration) of yolky oocytes is one of the first histologically
ical precursor for either E2 or 11KT production. discernible signs of gonadal sex change in Thalassoma wrasses
A critical role for sex steroid hormone signaling in gonadal and other species (Sadovy and Shapiro, 1987). Several lines
sex change is supported by two types of studies where exoge- of evidence are consistent with a key role for changes in estro-
nous steroids are administered to IP individuals and where genic signaling in the initiation of sex change. These include
steroid hormone synthesis is manipulated pharmacologically. demonstrations that inhibiting estrogen production pharmaco-
A first and continued focus of these studies has been androgen logically using both steroidal and nonsteroidal aromatase
signaling, stimulated at least in part by the responsiveness of inhibitors is effective in inducing gonadal sex change in several
male sex determination to androgenic stimulation more gener- families of sex-changing fishes (Table 2). Estrogen synthesis
ally in fishes (extensively reviewed in Devlin and Nagahama, can decline very quickly at the initiation of sex change. The
2002). Administration of androgen in the form of either the California goby species Lythrypnus dalli shows rapid declines
testosterone-derivative methyltestosterone or 11KT successfully in aromatase activity in both the gonad and brain at the initia-
induced female-to-male sex change in species from several tion of protogynous sex change (Black et al., 2005, 2011)
families of fishes that display socially controlled sex change although reductions in estrogen production at least at the
including wrasses (Labridae), parrotfishes (Scaridae), the whole-brain level lag behind increases in aggressive behavior
basses (Serranidae), gobies (Gobiidae), and a variety of species in sex changers. Several other species also show declines in
where social control of sex change has not been demonstrated either estrogen production or expression of the gonadal form
(see Devlin and Nagahama, 2002; Frisch, 2005 for reviews). of aromatase with sex change (fishes have two aromatase genes,
Despite clear evidence that androgens can induce protogynous cyp191a1a and cyp19a1b, expressed predominantly in the
sex change, it is not clear that an increase or alteration in gonads and brain, respectively: Gelinas et al., 1998; Forlano
androgen production is the key initiating event in protogynous et al., 2006).
sex change. In one of the most comprehensive studies of steroid As with many gonochoristic fish species during develop-
profiles during naturally occurring sex change in wild fish, ment, sex change can be manipulated effectively with exoge-
Nakamura et al. (1989) did document increases in circulating nous estradiol in species across several families (Table 2).
11KT, but increases over female levels were not observed until The bidirectionally sex-changing coral goby Gobiodon erythrospi-
the animals were fully developed TP males – well after the initi- lus was the subject of a study notable both because sex change
ation of sex change. was manipulated in both directions and also because the study
Declines in estrogenic signaling are also reliably associated was performed on corals in the field (Kroon et al., 2005). This
with protogynous sex change. For example, E2 levels showed species typically lives in pairs that shelter in the branches of
immediate and significant declines with the initiation of sex corals and has limited movement ability across the reef as their
change in saddleback wrasses (Nakamura et al., 1989). This is small size and relatively weak swimming abilities are thought
consistent with the observation that widespread atresia to contribute a high predation risk when away from shelter.
36 Socially Controlled Sex Change in Fishes

block this transition in isolated females and induce male-


RelaƟve blood steroid levels to-female change in isolated males.
Protogynous sex change can also be manipulated in other
gobies, wrasses, and serranids through manipulations of estro-
genic signaling (Table 2), and one report indicates male-
to-female sex change can be induced in the three-spot wrasse
with estrogen implants (Kojima et al., 2008). Differences in
plasma hormone levels and gonadal structure indicate sex
change involves significant changes in steroidogenic pathways
in the gonad. This is supported by recent transcriptomic studies
in both sea bream (Sparidae) and bluehead wrasse (Manousaki
et al., 2014; Liu et al., 2015). Gonadal aromatase mRNA
(cyp19a1a) is expressed at higher levels in ovaries in both
species while mRNAs for key androgen synthetic enzymes
- were elevated in testes (11b-hydroxylase for both sea bream
and bluehead wrasses, a variety of other androgen synthetic
enzymes for wrasses as well). Both species showed a very large
number of differences in the ovarian transcriptome, but a rela-
RelaƟve blood steroid levels

tively modest number of differences overall when comparing


brain transcriptomes. This is consistent with other findings
indicating very modest sex differences in fish brain transcrip-
tomes, at least at an overall level (e.g., Wong et al., 2014). Of
note with respect to estrogenic regulation of sex change, the
brain form of aromatase showed a trend toward higher expres-
sion in the brains of female bluehead wrasses (Liu et al., 2015),
a result consistent with significant differences found in our
more recent quantitative PCR and in situ hybridization compar-
isons (Lamm et al., unpublished data).
-
2.03.1.4.1 Sex Steroids and Behavioral Change
The dramatic alterations in circulating sex steroid levels and
extensive literature supporting a role for these hormones in
Figure 3 Top panel: circulating steroid hormone levels in the anemo- mediating sexual and aggressive behavior in other vertebrates
nefish Amphiprion melanopus for males and females in natural habitat as suggest a critical role in regulating the behavioral changes
well as males undergoing protandrous sex change induced by experi- that occur with sex change. While there is certainly support
mental removals of dominant females from social groups. Bottom for this general hypothesis, there are also conflicting findings
panel: circulating steroid hormone levels in the saddleback wrasse and a lack of clarity about what the key initiating events are
(Thalassoma duperrey) shown for wild-caught females and terminal in the behavioral sex change process. A large body of research
phase (TP) males and in females induced to undergo sex change in in tetrapods going back to Berthold’s studies with capons and
experimental pen. Stages in blue boxes indicate progress in sex change
particularly in the modern era with work in Guinea pigs by Wil-
process when samples were taken. Relative levels of different hormones
liam Young’s group (Phoenix et al., 1959) has implicated
are depicted to approximately the same scale except for cortisol in top
figure, where levels were substantially higher. Figures redrawn from androgens of testicular origin as critical for organizing the
Godwin, J.R., Thomas, P., 1993. Sex change and steroid profiles in the neural substrates of male-typical sociosexual behaviors during
protandrous anemonefish Amphiprion melanopus (Pomacentridae, Tele- development and activating these behaviors in adulthood.
ostei). Gen. Comp. Endocrinol. 91, 144–157; Nakamura, M., Hourigan, Fishes, however, may be fundamentally different from tetra-
T.F., Yamauchi, K., Nagahama, Y., Grau, E.G., 1989. Histological and pods in terms of the hormonal bases of neural sexual dimor-
ultrastructural evidence for the role of gonadal steroid hormones in sex phisms, with some studies suggesting these differences are
change in the protogynous wrasse Thalassoma duperrey. Environ. Biol. purely activational in nature (Hiraki et al., 2012). Although
Fishes 24 (2), 117–136, respectively. there are good examples of critical roles for androgens in regu-
lating male-typical sociosexual behaviors in fishes (Borg, 1994;
Munakata and Kobayashi, 2010; Godwin, 2010), it has also
As noted above, bidirectional sex change is predicted to be been postulated that aromatization and effects on brain
adaptive under these conditions as it allows any two adults estrogen levels are the overriding steroid hormone influence
to form a breeding pair. Kroon and colleagues demonstrated on sexual differentiation of neural function in fishes (Le Page
estrogenic control over this process by implanting male et al., 2010).
G. erythrospilus with E2 to induce male-to-female change and As noted above, circulating E2 levels are typically higher in
implanting females with the nonsteroidal aromatase inhibitor females and 11KT levels are higher in males, and this is true of
fadrozole to induce female-to-male sex change. Additionally, both socially controlled protogyny and protandry (Table 1).
capitalizing on a pattern where unpaired individuals become However, relatively few studies have experimentally induced
male, these investigators were able to show that estrogen could sex change and followed the time course of both hormonal
Socially Controlled Sex Change in Fishes 37

Table 2 Experiments involving sex steroid hormones, steroidogenesis inhibitors, and neuropeptide hormone manipulations

Direction of sex Color


Species, common name Family change Manipulation Gonad change? change? References

Thalassoma bifasciatum Labridae Diandric protogyny Methyltestosterone Yes: \ ➔ _ Yes Stoll (1955)
administered to females
Halichoeres trimaculatus, Labridae Protogyny E2 administered in feed to Yes: _ ➔ \ Kojima et al. (2008)
three-spot wrasse IP males
H. trimaculatus, three-spot Labridae Protogyny 11KT or AI administered in Yes: \ ➔ _ Higa et al. (2003)
wrasse feed to females
T. bifasciatum Labridae Diandric protogyny Testosterone administered Yes: \ ➔ _ Kramer et al. (1997)
to females
T. bifasciatum Labridae Diandric protogyny 11KT or AI administered to Yes: \ ➔ _ Yes Godwin (2009)
females
Sparisoma viride, stoplight Scaridae Diandric protogyny KT injections Yes: \ ➔ _ Yes Cardwell and Liley
parrotfish (1991b)
Coryphopterus nicholsii, Gobiidae Protogyny Implants of 11KT, KA, or Yes: \ ➔ _ Kroon and Liley (2000)
blackeye goby AI
Gobiodon erythospilus, coral Gobiidae Bidirectional E2 implants Yes: _ ➔ \ Kroon et al. (2005)
goby AI implants Yes: \ ➔ _

AI, aromatase inhibitor; 11KT, 11-ketotestosterone; KA, 11-ketoadrenosterone; IP, initial phase; E2, estradiol 17b.

and behavioral changes. One system that has been well behavioral sex change is much faster, with sex changers being
studied in this way is the bluebanded goby (L. dalli). This is able to exhibit male-typical behaviors within minutes of
a small-bodied species and has facilitated studies in captive assuming social dominance and typically spawning in the
conditions that nevertheless can allow naturalistic social male role within 1–2 h during the spawning period (Warner
interactions. Bluebanded gobies form clear dominance hierar- and Swearer, 1991; Godwin et al., 1996). Behavioral change
chies, and removal of dominant males leads to rapid sex occurring very rapidly at the onset of sex change has been
change in the largest and most dominant (alpha) female observed in a variety of species now and is likely the usual
(Rodgers et al., 2005, 2007). Female-to-male sex change is situation under field conditions (e.g., Robertson, 1972). The
associated with significant decreases in gonadal aromatase time course of behavioral change is consistent with a model
activity, although significant reductions are not seen early where gonadal alterations and related changes in gonadal
on in the sex change process (Black et al., 2005). By contrast, steroid hormone profiles are not driving behavioral
reductions in brain aromatase activity coincide with and are changes (although steroid hormones can act rapidly through
significantly correlated to increases in dominance-related nongenomic mechanisms). Also consistent with gonadal
behaviors by alpha females undergoing sex change. These change not driving behavioral change is the finding that
findings are consistent with a reduction in neural estrogen gonadectomy does not block behavioral sex change in blue-
levels being necessary for male-typical dominance behavioral head wrasses, although it does prevent the development of
profiles and sex change to proceed, but a later study in L. dalli TP male-typical color change (Godwin et al., 1996). Likewise,
that profiled behavioral changes early in the initiation of sex castration of TP males does not reduce or discernibly alter the
change found a lack of concordance between brain aromatase expression of male-typical courtship and aggressive behavior
activity and dominance-related behavior, leading the authors under field conditions (Semsar and Godwin, 2003). While
to suggest behavioral changes were not caused by reductions these findings do not support a role for gonadal steroids in
in neural aromatization (Black et al., 2011). These were TP male-typical behavior and behavioral sex change, other
whole-brain measures of aromatase activity due to the small findings do indicate a role. Implanting ovariectomized
size of this fish, and more neuroanatomically localized effects females with 11KT induces the development of both TP
cannot be excluded, but these authors have also suggested the male-typical body coloration and alters behavior over the
intriguing hypothesis that decreases in aromatization could course of 10–12 days in the presence of dominant TP males
lead to behavioral effects by channeling more of the under field conditions (i.e., socially inhibitory conditions;
hormonal substrate (testosterone) into 11KT synthesis (Black Semsar and Godwin, 2004). Importantly with respect to
et al., 2005, 2011; Lorenzi et al., 2012). behavior, this manipulation did not increase aggression or
Bluehead wrasses also show significant increases in 11KT induce territorial behavior generally but did increase the like-
with female-to-male sex change, but several observations are lihood of showing courtship toward a gravid female. Also
inconsistent with these increases being the causal drivers of noteworthy in this context is that territorial TP male stoplight
increases in TP male sexual and aggressive behavior. Sex parrotfish (Sparisoma viride) had higher circulating 11KT than
change is very rapid in bluehead wrasses under natural field ‘bachelor’ TP males that did not hold territories on reefs
conditions, with the transition from functional female to (Cardwell and Liley, 1991b).
the capability to produce sperm and fertilize eggs occurring As with bluebanded gobies, estrogenic influences appear
in as few as 8 days (Warner and Swearer, 1991). Interestingly, to play a role in sex change in bluehead wrasses. Estradiol
38 Socially Controlled Sex Change in Fishes

implants administered to females did prevent behavioral sex Nozu and Nakamura (2015) report a similar effect for the pro-
change under socially permissive conditions (i.e., TP males togynous three-spot wrasse where a high dose of cortisol
removed from social groups) and also increased aromatase administered to females in feed for 6 weeks induced at least
mRNA abundances in the preoptic area (POA) of the hypo- the initiation of sex change in all individuals.
thalamus (Marsh-Hunkin et al., 2013). These observations Perry and Grober (2003) developed a model that postu-
along with higher aromatase mRNA levels in females relative lated chronically elevated cortisol levels prevented sex change
to TP males (Liu et al., 2015) and strong aromatase expression in social subordinates (‘release of social stress’ hypothesis).
in the POA (Marsh et al., 2006) suggest brain levels of E2 are One experimental test involving implanting dominant
higher in females than TP males and likely drop with behav- females of the protogynous sandperch (Parapercis cylindrica)
ioral sex change. However, consistent with the observation with cortisol did not support this model as this treatment
that declines in aromatase activity lag behind behavioral did not prevent sex change in a permissive environment
changes in the bluebanded gobies discussed above (Black (Frisch et al., 2007), but effects elsewhere in the HPI axis
et al., 2011), preliminary evidence suggests that declines in such as on corticotropin-releasing hormone activity remain
forebrain aromatase mRNA expression lag behind the initia- possible. Solomon-Lane et al. (2013) proposed an alternative
tion of behavioral sex change in bluehead wrasses (Cabral model they termed ‘classical facilitation of metamorphosis’
et al., unpublished results). However, aromatase activity can based on the evolutionarily conserved role of glucocorticoid
be altered very rapidly through phosphorylation (reviewed signaling in promoting metamorphic processes. This model
in Charlier et al., 2015), and this could be important in behav- proposes that it is instead an increase in glucocorticoid
ioral change. It will be critical to assess estrogen signaling at signaling that facilitates sex change at the onset of permissive
a detailed neuroanatomical level to more fully address conditions. This model is consistent with patterns observed
a potential role in behavioral sex change. It is noteworthy in following removal of dominant males from social groups
this context that brain aromatase in fishes is expressed in in bluebanded gobies, where dominant females (sex-
radial glia rather than neurons and this glial expression is changing individuals) show elevations in excreted cortisol
strong in the POA (see Forlano et al., 2001; Forlano and at 1 and 3 days following removal of dominant males
Bass, 2006). These aromatase-positive glia are found in very (Figure 4). This may be a common pattern as sex change in
close proximity to neurons of key neuroendocrine signaling many species is correlated with significant increases in
systems in bluehead wrasses, creating the likelihood of high activity including especially elevations in aggressive behavior
and potentially rapidly changing local estradiol levels (Marsh (e.g., bluehead wrasses show three- to fourfold increases in
et al., 2006). aggression early on in sex change). The direction of sex
change may not be important for potential glucocorticoid
2.03.1.4.2 Corticosteroids and Sex Change effects as the anemonefish A. melanopus exhibits protandry
Socially controlled sex change is mediated through social inter- with significant increases in both aggressive behavior and
actions and typically involves changes in individual domi- circulating cortisol during the sex change process, although
nance status within a social group. The behavioral males and females were not different (Godwin and Thomas,
mechanisms mediating these social status effects appear to be 1993; Godwin, 1994a). Likewise, social dominance in groups
primarily related to being the initiator or recipient of aggressive of the protandrous anemonefish Amphiprion ocellaris is associ-
dominance-related behaviors, and these typically change ated with elevated blood cortisol and expression of
rapidly at the onset of sex change (Robertson, 1972; Warner
and Swearer, 1991; Godwin et al., 1996; Black et al., 2005,
2011). Differences in social status are often correlated with
differences in activity of the glucocorticoid endocrine stress
axis, and these hormones can produce rapid changes in
behavior and endocrine function. Therefore, this endocrine
system is a logical candidate for ‘transducing’ changes in
dominance status into changes in reproductive function.
Solomon-Lane et al. (2013) make a detailed case that the hypo-
thalamic–pituitary–interrenal (HPI) axis is a likely regulator of
the sex change process due to its involvement across vertebrates
in social status, agonistic behavior, and important life history
transitions. In support of this hypothesis, these authors cite
(1) studies linking social status to glucocorticoid levels in
a range of fishes, (2) links to agonistic behavior in a range of
vertebrates including some teleost models, and (3) the role Figure 4 Changes in cortisol released, waterborne cortisol, by sex-
changing bluebanded gobies (Lythrypnus dalli). ‘Pre’ refers to dominant
the primary glucocorticoid in fishes, cortisol, plays in sex
females before sex change is initiated and the 2 h to 15 days times are
determination in distantly related species. This regulation of
sampling points following male removal, which produces the conditions
sex determination is seen in both gonochoristic species that induce sex change in females. Figure from Solomon-Lane, T.K.,
(medaka; Hayashi et al., 2010) and several species that Crespi, E.J., Grober, M.S., 2013. Stress and serial adult meta-
exhibit TSD (e.g., Japanese and southern flounders: Yamaguchi morphosis: multiple roles for the stress axis in socially regulated sex
et al., 2010; Mankiewicz et al., 2013; and South American change. Front. Neurosci. 7, 210, http://dx.doi.org/10.3389/fnins.2013.
pejerrey: Hattori et al., 2009; Fernandino et al., 2012). 00210.
Socially Controlled Sex Change in Fishes 39

glucocorticoid receptor mRNA in the brain (Iwata et al., of the other, and these changes take approximately 4 days.
2012). Interestingly, given that dominance induces female Kobayashi and colleagues found that mRNA expression for
differentiation in this species, the dominant fish also showed the two gonadotropin receptors (follicle-stimulating hormone
significantly higher neural expression of brain aromatase (FSH) receptor and luteinizing hormone (LH) receptor)
mRNA. showed significant changes within 8–12 h of the initiation of
sex change, increasing in testicular tissue and declining in
ovarian tissue with female-to-male change while showing the
2.03.2 Neural Signaling Systems and Sex Change opposite pattern with male-to-female change. These changes
appeared to be functionally important as they conferred
Social control of sex change implies the critical initiating events responsiveness to gonadotropic stimulation as measured by
of the process must involve neural signaling systems. These in vitro steroid hormone production.
events must induce both adaptive changes in sexual and aggres- Direct manipulative approaches also provide some support
sive behavior and a reorganization of the reproductive system. for a GnRH role in sex change. Also working with bluehead
Studies have primarily focused on components of the hypotha- wrasses, a series of studies by Kramer and colleagues showed
lamic–pituitary–gonadal (HPG) axis, but other neural that varying degrees of sex change could be induced in isolated
signaling systems have also been implicated in regulating captive female bluehead wrasses through manipulations that
behavioral and gonadal sex change. either directly altered gonadotropins or altered systems that
affect gonadotropin production and release. These experiments
involved injections of (1) human chorionic gonadotropin
2.03.2.1 Gonadotropin-Releasing Hormone and
(Koulish and Kramer, 1989), (2) a synthetic form of GnRH
Gonadotropins
with the dopamine receptor antagonist domperidone (Kramer
Gonadotropin-releasing hormone (GnRH) is generally et al., 1993), and (3) neuropeptide Y which was hypothesized
considered the key ‘gatekeeper’ hormone in vertebrate to affect gonadotropin signaling (Kramer and Imbriano, 1997).
reproduction with activation of GnRH neurons being crit- While these studies certainly do provide support for the
ical for such key life history events as puberty in mammals. hypothesis that socially controlled sex change involves changes
GnRH neurons are also critically involved in the activation in gonadotropic signaling and are generally consistent with
of the reproductive axis during assumption of social domi- results in another protogynous wrasse (Coris julis, Reinboth
nance in males of the gonochoristic cichlid fish Astatotilapia and Brusle-Sicard, 1997), a few points should be noted. These
burtoni (Davis and Fernald, 1990; Burmeister et al., 2005; studies involved female wrasses in captive conditions receiving
reviewed in Maruska and Fernald, 2011). It is noteworthy frequent injections (three injections/week). These conditions
in this context that cichlid fishes are in the suborder of could influence baseline activation of the HPA axis which, as
fishes, the Labroidei, that also includes well-studied sex- noted above (Section 2.03.1.2, glucocorticoids), could impor-
changing species in the wrasse, parrotfish, and damselfish tantly influence sex change. Second, the degree of sex change
families (Streelman and Karl, 1997). This relationship induced was relatively modest in terms of both progression
suggests that mechanisms by which social influences induce through the process in some cases and frequency, particularly
reproductive activation in A. burtoni can likely inform our for the synthetic GnRH and NPY studies. Finally, the socially
understanding of this phenomenon in socially controlled isolated conditions of the experimental females provided
sex change. a high degree of experimental control but do not mimic the
A number of correlative and manipulative studies point to inhibitory influence of larger individuals that is a key regulator
a role for gonadotropic signaling in socially controlled sex of sex change under natural conditions (Warner and Swearer,
change, and wrasses and gobies are well studied in this 1991; Ross et al., 1983 for the congeneric saddleback wrasse).
respect. Bluehead wrasses show differences in GnRH neuron A convincing demonstration that the neuroendocrine system
size in the POA across sexual phenotypes with TP males of interest is part of the ‘final common pathway’ regulating
having greater numbers than either females or IP males sex change would show that sex change could be induced by
(Grober and Bass, 1991). These numbers could be increased this putative mediator in the presence of a more dominant
to TP-male-typical levels by 11KT implants in females, individual and, ideally, in the natural environment.
although this treatment also induced sex change so it is
possible that 11KT influences were not directly causal in this
2.03.2.2 Kisspeptins and Gonadotropin-Inhibitory Hormone
change (Grober et al., 1991). Consistent with these findings
in bluehead wrasses, males of both Ballan wrasses and the The strong influences of kisspeptins and gonadotropin-inhibi-
protandrous anemonefish A. melanopus also show greater tory hormone (GnIH) on the HPG axis in other fishes and
numbers of POA GnRH neurons than females (Eloffson other vertebrates generally suggest these should be a productive
et al., 1999, 1997, respectively). focus in studies of socially controlled sex change. Kisspeptins,
Although focusing on gonadal tissue rather than the brain, a group of RY- or RFamide peptides encoded by the kiss1
a particularly intriguing finding regarding gonadotropin and/or kiss2 genes, typically stimulate expression and release
signaling and sex change comes from a study of the bidirection- of GnRH in the POA and gonadotropins in the pituitary of
ally sex-changing goby T. okinawae (Kobayashi et al., 2009). fishes and other vertebrates (reviewed by Elizur, 2009; Oakley
The gonads in T. okinawae have both ovarian and testicular et al., 2009; Tena-Sempere et al., 2012). However, effects can
portions, although only one of these is active at a given time. depend on the gonadal state of individuals and the partially
Sex change involves regression of one tissue type and activation divergent activities of kiss1 and kiss2 in fishes that possess
40 Socially Controlled Sex Change in Fishes

both genes (Zmora et al., 2012, 2014). For example, intramus- reviewed by Ogawa and Parhar, 2014). For example, goldfish
cular injections of kiss2 in gonochoristic female and male LPXRFa-1, -2, and -3 stimulated LH and FSH release from
hybrid bass (Morone saxatilis  Morone chrysops) induced release pituitary cultures of precocious male sockeye salmon (Amano
of LH at both puberty and recrudescence, whereas kiss1 was et al., 2006), and goldfish LPXRFa-1 increased mRNA levels
stimulatory only at recrudescence. Furthermore, kiss1 and for the beta subunits of both LH and FSH (lhb and fshb) in
kiss2 increased gnrh1 expression at pre-prepuberty, but kiss2 pituitary cultures of sexually mature grass puffers of both
downregulated gnrh1 expression during recrudescence (kiss1 sexes (Shahjahan et al., 2011). Similarly, in Nile tilapia,
had no effect) (Zmora et al., 2012). During the breeding LPXRFa-2 treatment increased LH and FSH release in sexually
season, kiss1 and kiss2 had mostly stimulatory but partially mature females injected intraperitoneally and in pituitary
divergent effects on the genes of the HPG axis and gonadotro- cells in culture from sexually mature males (Biran et al.,
pins in male brain and pituitary cultures. The kisspeptin 2014). However, studies in goldfish have also shown negative
receptor antagonists peptide 234, which blocks activation of effects of GnIH on gonadotropin synthesis and release
kissr2, and peptide 359, which blocks activation of kissr1 and (Zhang et al., 2010; Qi et al., 2013), which has been shown
kissr2, reversed these effects (Zmora et al., 2015). to be dependent on the spawning season, state of the gonads,
Interestingly, the same study by Zmora et al. (2015) found and circulating gonadal steroid levels (Moussavi et al., 2012),
that the kisspeptin receptor antagonists decreased avt mRNA and also suggested by Qi et al. (2013) to potentially be
levels in the POA. In both this study and a study in medaka socially dependent.
(Kanda et al., 2013), kisspeptins were found to directly regulate To our knowledge, the actions of GnIH on sex change or
arginine vasotocin (AVT) neurons. Studies in fishes have also sexual behavior in fishes have not yet been investigated. In
found direct connections between the AVT and GnRH1 male quail, intracerebroventricular (ICV) injection of GnIH
systems, e.g., in the sex-changing rock hind grouper (Kline inhibited male-typical sexual and aggressive behaviors and
et al., 2016), and studies in mammals found AVP innervation increased estrogen synthesis in the POA via aromatase activa-
of kisspeptin neurons in mice (Vida et al., 2010) and Syrian tion within 30 min of injection, whereas GnIH RNA interfer-
hamsters (Williams et al., 2011). Interactions among the AVT, ence increased these behaviors 1 day after injection (Ubuka
kisspeptin, and GnRH systems could be important connections et al., 2014). GnIH RNA interference also induced sexual and
regulating behavioral and gonadal changes during sex change aggressive behaviors 2 days after injection in male white-
in fishes. crowned sparrows (Ubuka et al., 2012). Effects are also seen
In the protogynous orange-spotted grouper (Epinephelus in male rats where RFRP-3 ICV injection reduced sexual behav-
coioides), mature females had higher expression of hypotha- iors during the photophase (light period), but not scotophase
lamic kiss2 than did males (Shi et al., 2010). During 4 weeks (dark period) (Johnson et al., 2007). In estradiol-implanted
of methyltestosterone treatment to induce sex change in female white-crowned sparrows, GnIH inhibited copulation
females, kiss2 and gnrh1 mRNA levels decreased during the first solicitation behavior upon male song playback within
3 weeks, but increased two- and eightfold, respectively, during 30 min of ICV injection and decreased LH secretion (Bentley
the fourth week. Shi et al. (2010) suggest that lower kiss2 and et al., 2006). Though the actions of GnIH peptides on sexual
gnrh1 expression during early sex change may be important behavior in fishes are unknown, these studies nevertheless
for ovarian atresia, and levels may increase during testicular suggest a potential for rapid modulation of behavior and
development. We found that implantation of kiss2 for gonadal function by GnIH in fishes that are partially dependent
9–10 days in female bluehead wrasses with either active or on social and abiotic cues.
regressed ovaries at sampling was not sufficient to override
social inhibition and induce sex change in the presence of TP
2.03.2.3 Monoamine Neurotransmitters and Sex Change
males (kiss1 was not tested; Lamm et al., unpublished). Effects
of blocking kisspeptin signaling have not been described in any Monoamine neurotransmitters are critical modulators of sexual
sex-changing species. and aggressive behaviors across vertebrate animals including in
GnIH (named in birds) and its related peptides, also known fishes (Summers and Winberg, 2006). To date, however, rela-
as RFamide-related peptides (RFRP) in mammals and LPXRFa- tively few studies have addressed a potential role for mono-
mides (LPXRFa) in fishes, are another recently discovered amine signaling in socially controlled sex change.
group of RFamides that also interact with GnRH neurons and Larson and colleagues examined monoamine involvement
gonadotropes to regulate the release of gonadotrophins in in protogynous sex change in saddleback wrasses using the
vertebrates (reviewed by Tsutsui et al., 2010; Ogawa and experimental pen approach that had previously established
Parhar, 2014). The gnih precursor transcript encodes a prepro- social control of the process in this species (Larson et al.,
peptide that is cleaved into two to four GnIH-related 2003a,b). Measures of monoaminergic activity using HPLC
RFamides. One of the mature RFamides was first discovered approaches demonstrated a complex set of changes in the
in quail and named GnIH due to its ability to inhibit LH brains of sex-changing females including indications of
release from cultured pituitaries (Tsutsui et al., 2000). The a decrease in serotonergic activity in the raphe nucleus during
gnih gene and its receptor(s), part of the neuropeptide the first week of social dominance. Importantly, these correla-
FF receptor family, have been found in a variety of fishes, but tive studies were followed by manipulative studies testing the
in vivo and in vitro studies in gonochoristic fishes have shown ability of pharmacological manipulations of monoamines to
complex and even conflicting results of GnIH on the influence sex change. Under social conditions that normally
synthesis and release of gonadotrophins (Zhang et al., 2010; inhibit sex change (i.e., a large TP male present in the group),
Moussavi et al., 2012; Qi et al., 2013; Biran et al., 2014; either putatively reducing serotonergic signaling with the
Socially Controlled Sex Change in Fishes 41

5-HT2 receptor antagonist ritanserin or increasing noradren-


ergic signaling with the drugs ephedrine or maprotiline
significantly increased the occurrence of ovary-to-testes differ-
entiation. Conversely, under socially permissive conditions Dominant & intact
(i.e., experimental female was largest individual in the group),
ovary-to-testes transformation could be significantly inhibited
through either putatively increasing serotonergic signaling
Dominant & gonads removed
using the serotonin-reuptake inhibitor sertraline or reducing
noradrenergic signaling with the a-receptor antagonist
phenoxybenzamine.
Support for a role of monoamines in regulating sex
Subordinant & intact
change in other species is lacking overall. Manipulating sero-
tonergic activity did not affect the likelihood of sex change
in bluebanded gobies, and brain levels of serotonin and
the key serotonin metabolite 5-hydroxyindoleacetic acid Subordinant &
(5-HIAA) were not correlated with aggressive or courtship gonads removed
behavior. These results were mirrored at a detailed neuroan-
atomical level with a similar lack of significant sex or social
status differences in serotonergic immunoreactive neurons in Relative levels of AVT mRNA
bluebanded gobies (Lorenzi and Grober, 2012). Although
not directly related to sex change, the serotonin-reuptake Figure 5 Social status influences AVT mRNA levels in the preoptic
inhibitor fluoxetine can inhibit aggressive behavior of TP area of the hypothalamus in bluehead wrasses while gonadal status
male bluehead wrasses with both acute administration does not. Dominant individuals were the largest in their social groups
under field conditions and chronic administration under while subordinants were not. Intact animals underwent ‘sham’ opera-
tions to control for the surgical procedure. Inset shows AVT mRNA in
captive conditions (Perreault et al., 2003). Interestingly
the preoptic area visualized by the technique of in situ hybridization.
and of potential relevance to behavioral sex change, chronic
AVT, arginine vasotocin. Figure depicts relative AVT mRNA levels in this
fluoxetine treatment also reduced expression of AVT mRNA comparison from Semsar, K., Godwin, J., 2003. Social influences on
in TP male bluehead wrasses (Semsar et al., 2004 and the arginine vasotocin system are independent of gonads in a sex-
Section 2.03.2.4 below). changing fish. J. Neurosci. 23, 4386–4393.

2.03.2.4 Nonapeptides and Sex Change


territorial behavior in nonterritory holding TP males and the
Perhaps no class of neural signaling molecules has generated AVP V1 receptor antagonist ‘Manning compound’ could
more attention as modulators of social behavior than the reduce it (Semsar et al., 2001) and (2) administration of
conserved nonapeptides in the arginine vasopressin and Manning compound could prevent establishment on open
oxytocin families (Goodson and Thompson, 2010). Fishes territories by nonterritorial TP males and prevent behavioral
express these peptides as AVT and isotocin (IST), respectively, sex change by large females after removal of TP males from
and provide intriguing models for studying nonapeptide their social groups on small reefs (Semsar and Godwin, 2004).
modulation of sociosexual behavior because of the diversity Bluehead wrasses also exhibit differences in AVT receptor
of social systems and sexual differentiation patterns they mRNA expression across sexual phenotypes (Lema et al.,
exhibit. The first demonstration of a nonapeptide effect on 2012). As with many other gene families, a genome duplication
sociosexual behavior was in a teleost (Wilhelmi et al., 1955; in the teleost lineage led to multiple forms of vasotocin recep-
Pickford and Strecker, 1977), and work in teleost models has tors (Lema, 2010), and bluehead wrasses express two forms of
provided some critical advances in our understanding of these the V1a receptor subtype: V1a1 and V1a2. Both receptor
hormones (e.g., Goodson and Bass, 2000; Godwin and subtypes are expressed in some key brain regions for socio-
Thompson, 2012 for review). Most attention in sex-changing sexual behavior including the ventral telencephalon, POA,
fishes has focused on the AVT system, but it increasingly lateral recess, and periventricular portion of the posterior
appears IST actions may also be important. tuberculum (Lema et al., 2012). These include some critical
Bluehead wrasses show rapid behavioral changes at the regions of what has been called the ‘Social Behavior Network’
onset of sex change (Warner and Swearer, 1991; Godwin or ‘Social Decision-Making Network’ in vertebrate animals
et al., 1996), and these changes are correlated with increases (Newman, 1999; O’Connell and Hofmann, 2012). As with
in AVT mRNA abundances in the POA in field experiments AVT mRNA expression, sexual phenotype differences are seen
(Godwin et al., 2000). As noted above, behavioral sex change with AVT receptor mRNAs with higher V1a2 mRNA abundance
does not depend on the presence of gonads in this species in the hypothalamus of TP males than females and evidence of
and neither do dominance-related increases in AVT mRNA increases during sex change. Interestingly, these receptor
abundances, occurring even in dominant ovariectomized mRNAs are also expressed in the gonads where V1a1 mRNA
females exhibiting behavioral sex change (Semsar and Godwin, shows higher abundances in testes than ovaries and increases
2003; Figure 5). Evidence for a causal role of AVT in TP-male- during sex change while V1a2 mRNA shows significant eleva-
typical behavior and behavioral sex change comes from field tions in sex-changing gonads relative to the ovary (Lema
experiments where (1) administration of AVT could induce et al., 2012).
42 Socially Controlled Sex Change in Fishes

Information on AVT signaling in sex-changing fishes is AVT and AVP are implicated in activation of the endocrine
otherwise relatively limited but does include a few intriguing stress axis (Engelmann et al., 2004; Huffman et al., 2015)
studies. The goby T. okinawae is a bidirectional sex changer, and, as noted above, there is evidence for activation of the
and females that transformed into males under experimental HPI axis during behavioral sex change in bluebanded gobies
conditions showed larger AVT-immunoreactive neurons than and the anemonefish A. melanopus (Solomon-Lane et al.,
fish that transitioned from male to female (Grober and 2013; Godwin and Thomas, 1993; fishes have interrenal
Sunobe, 1996). The protogynous rock hind grouper glands rather than adrenal glands, and the interrenals are
(Epinephelus adscensionis) exhibits a haremic social structure in the key source of cortisol). Increases in AVT signaling leading
the wild, and sex change can be induced in captive groups of to activation of the HPI axis could be consistent with the
females (Kline et al., 2011a). As with bluehead wrasses, rock ‘classical facilitation of metamorphosis’ model described by
hinds express a V1a-type AVT receptor in a variety of brain Solomon-Lane and colleagues, but this remains to be rigor-
areas implicated in sociosexual behaviors and reproduction ously tested.
including those described above for bluehead wrasses (Kline
et al., 2011b). AVT signaling may also be important in
protrandrous sex-changing species. Administration of 2.03.3 Conclusions and Future Directions
Manning compound reduced aggressive behaviors in both
juveniles and mature males in the protandrous anemonefish In his 1988 review of mechanisms by which the endocrine
A. ocellaris in staged laboratory encounters (Yaeger et al., system might direct the process of sex change in fishes, Rudolf
2014). These investigators also examined neural activation Reinboth regretfully noted that “It is hard to admit that till now
through use of c-Fos immunohistochemistry and showed that neither my own efforts nor those of other research groups have
mature males receiving Manning compound also showed succeeded in generating promising concepts.” Dr Reinboth
reduced c-Fos protein expression in the POA and the underestimated his early and sustained contributions to this
posterior tuberculum (putative homolog of the ventral area of study that proved both informative and inspiring for
tegmental area in tetrapods), although not the anterior the researchers who followed. While many important ques-
tuberal nucleus (putative homolog of the ventromedial tions remain, our understanding of this fascinating reproduc-
hypothalamus of tetrapods). tive phenomenon has moved well ahead in the ensuing
A potential role for IST in sex change and sex-changing decades and this progress appears likely to accelerate. This is
species is much less studied. Sex differences in IST expression due in part to advances that allow matching sophisticated tech-
are observed in one very well-studied gonochoristic teleost nical approaches with the heuristic advantages of nontradi-
model, with female medaka exhibiting higher IST mRNA abun- tional model systems and the ‘experiments of nature’ that
dances than males (Kawabata et al., 2012), and at least two sex- these systems provide.
changing species. Female bluebanded gobies showed greater Gonadal steroid hormones play a prominent and important
numbers of IST-immunoreactive cells than either control, role in control of sex change at the gonadal level. There is also
established males or fish in the late stages of female-to-male evidence for a role of these hormones in behavioral sex change
sex change (Black et al., 2004). By contrast, RNA sequencing and effects on the behavior of sex-changing species more gener-
showed a trend toward TP male bluehead wrasses having ally, but it is not yet clear whether changes in steroid hormone
higher levels of IST mRNA at the level of the forebrain and signaling play a role in the key initiating events of sex change
midbrain transcriptome (Liu et al., 2015) and IST mRNA because behavioral sex change can occur in at least one model
levels were significantly higher in TP males in the POA when system in the absence of gonads (bluehead wrasses), and signif-
measured by in situ hybridization (Thompson et al., icant declines in estrogen production by brain aromatase in the
unpublished). brain lag behind the beginnings of behavioral sex change in
While more studies are needed, the available information another (bluebanded gobies). Important next steps in address-
is consistent with an important role for AVT signaling in ing the potential role of steroid signaling in behavioral sex
mediating important events in sex change. It is less clear change will be more neuroanatomically focused to assess
precisely what the role of this signaling may be. Behavioral changes in the key brain areas for sexual and aggressive
sex change is typically associated with the assumption of behavior, primarily those in the social behavior network.
social dominance and increases in aggressive behavior, and Our understanding of the neural circuitry subserving social
these actions are consistent with known effects of AVT and behavior has advanced significantly in recent years. A major
related peptides in vertebrates including fishes, although advance in this area was the identification of what was termed
the precise nature of AVT effects can vary across species the ‘Social Behavior Network’ in mammals by Newman
(Godwin and Thompson, 2012). There are also intriguing (1999). This concept has more recently been extended to verte-
possibilities for AVT signaling in the ‘transduction’ of social brates more generally and into a ‘Social Decision-Making
cues into reproductive responses. Hamsters and mice show Network’ that includes both the Social Behavior Network and
direct projections of AVP neurons onto kisspeptin neurons the mesolimbic reward system (O’Connell and Hofmann,
(Vida et al., 2010; Williams et al., 2011), and so AVT 2011, 2012). There have been no systematic studies of this
signaling could act through kisspeptin neurons to influence network in sex-changing fishes, but these animals should
the HPG axis. Kline (2010) also found evidence of AVT provide valuable models. This is because the rapid behavioral
V1a receptor expression in POA GnRH neurons in the rock and neurodevelopmental events of sex change take place in
hind grouper, providing evidence for potential direct interac- observable and manipulable adult animals. The immediate
tions of AVT signaling with the reproductive axis. Lastly, both early gene approaches that have been used so productively to
Socially Controlled Sex Change in Fishes 43

explore neural activity in mammals and birds are becoming Elizur, A., 2009. The KiSS1/GPR54 system in fish. Peptides 30, 164–170.
increasingly well established in fishes, making this a very acces- Elofsson, U.O., Winberg, S., Francis, R.C., 1997. Number of preoptic GnRH-
immunoreactive cells correlates with sexual phase in a protandrously
sible approach (e.g., Burmeister et al., 2005; Yaeger et al.,
hermaphroditic fish. The dusky anemonefish (Amphiprion melanopus). J. Comp.
2014). Physiol. A Sens. Neural Behav. Physiol. 181, 484–492.
Lastly, rapid advances in genomics are making these Elofsson, U.O., Winberg, S., Nilsson, G.E., 1999. Relationships between sex and the
approaches feasible in a range of species beyond traditional size and number of forebrain gonadotropin-releasing hormone immunoreactive
models, including sex-changing fishes (e.g., Manousaki et al., neurones in the ballan wrasse (Labrus berggylta), a protogynous hermaphrodite.
J. Comp. Neurol. 410, 158–170.
2014; Liu et al., 2015). Combining transcriptional approaches Engelmann, M., Landgraf, R., Wotjak, C.T., 2004. The hypothalamic-neurohypophysial
such as RNA sequencing at the level of brain areas implicated in system regulates the hypothalamic–pituitary–adrenal axis under stress: an old
rapid behavioral change should be particularly useful for concept revisited. Front. Neuroendocrinol. 25, 132–149.
understanding the initiating events in the sex change process. Fernandino, J.I., Hattori, R.S., Kishii, A., Strussmann, C.A., Somoza, G.M., 2012. The
cortisol and androgen pathways cross talk in high temperature-induced mascu-
While transcriptional approaches such as RNAseq are inher-
linization: the 11b-hydroxysteroid dehydrogenase as a key enzyme. Endocrinology
ently descriptive and manipulative tests have until recently 153, 6003–6011.
been challenging in nonmodel species, recent methodological Fischer, E.A., 1980. The relationship between mating system and simultaneous
advances including siRNA and vivo morpholinos (e.g., Notch hermaphroditism in the coral reef fish Hypoplectrus nigricans (Serranidae). Anim.
et al., 2011) should facilitate these important functional tests Behav. 28, 620–634.
Fishelson, L., 1970. Protogynous sex reversal in the fish Anthias squamipinnis
of key gene expression events in sex change. (Teleostei, Anthiidae) regulated by the presence or absence of a male fish. Nature
227, 90–91.
Forlano, P.M., Deitcher, D.L., Myers, D.A., Bass, A.H., 2001. Anatomical distribution
and cellular basis for high levels of aromatase activity in the brain of teleost Wsh:
References aromatase enzyme and mRNA expression identify glia as source. J. Neurosci. 21,
8943–8955.
Amano, M., Moriyama, S., Iigo, M., et al., 2006. Novel fish hypothalamic neuropep- Forlano, P.M., Schlinger, B.A., Bass, A.H., 2006. Brain aromatase: new lessons from
tides stimulate the release of gonadotrophins and growth hormone from the non-mammalian model systems. Front. Neuroendocrinol. 27 (3), 247–274. http://
pituitary of sockeye salmon. J. Endocrinol. 188, 417–423. dx.doi.org/10.1016/j.yfrne.2006.05.002.
Bentley, G.E., Jensen, J.P., Kaur, G.J., Wacker, D.W., Tsutsui, K., Wingfield, J.C., Francis, R.C., 1992. Sexual lability in teleosts – developmental factors. Q. Rev. Biol.
2006. Rapid inhibition of female sexual behavior by gonadotropin-inhibitory 67, 1–18.
hormone (GnIH). Horm. Behav. 49 (4), 550–555. Francis, R.C., Barlow, G.W., 1993. Social-control of primary sex-differentiation in the
Biran, J., Golan, M., Mizrahi, N., Ogawa, S., Parhar, I.S., Levavi-Sivan, B., 2014. Midas cichlid. Proc. Natl. Acad. Sci. U.S.A. 90, 10673–10675.
LPXRFa, the piscine ortholog of GnIH, and LPXRF receptor positively regulate Fricke, H.W., 1983. Social control of sex: field experiments with the anemonefish
gonadotropin secretion in Tilapia (Oreochromis niloticus). Endocrinology 155, Amphiprion bicinctus. Z. Teirpsychol. 61, 71–77.
4391–4401. Fricke, H.W., Fricke, S., 1977. Monogamy and sex change by aggressive dominance in
Black, M.P., Reavis, R.H., Grober, M.S., 2004. Socially induced sex change regulates coral reef fish. Nature 266, 830–832.
forebrain isotocin in Lythrypnus dalli. Neuroreport 15 (1), 185–189. http:// Frisch, A., 2005. Sex-change and gonadal steroids in sequentially-hermaphroditic
dx.doi.org/10.1097/01.wnr.0000103273.91211.7a. teleost fish. Rev. Fish Biol. Fish. 14 (4), 481–499. http://dx.doi.org/10.1007/
Black, M.P., Balthazart, J., Baillien, M., Grober, M.S., 2005. Socially induced and rapid s11160-005-3586-8.
increases in aggression are inversely related to brain aromatase activity in a sex- Frisch, A., Walker, S., McCormick, M., Solomon-Lane, T., 2007. Regulation of pro-
changing fish, Lythrypnus dalli. Proc. R. Soc. B Biol. Sci. 272 (1579), 2435– togynous sex change by competition between corticosteroids and androgens: an
2440. http://dx.doi.org/10.1098/rspb.2005.3210. experimental test using sandperch, Parapercis cylindrica. Horm. Behav. 52 (4),
Black, M.P., Balthazart, J., Baillien, M., Grober, M.S., 2011. Rapid increase in 540–545. http://dx.doi.org/10.1016/j.yhbeh.2007.07.008.
aggressive behavior precedes the decrease in brain aromatase activity during Gelinas, D., Pitoc, G.A., Callard, G.V., 1998. Isolation of a goldfish brain cytochrome
socially mediated sex change in Lythrypnus dalli. Gen. Comp. Endocrinol. 170 (1), P450 aromatase cDNA: mRNA expression during the seasonal cycle and after
119–124. http://dx.doi.org/10.1016/j.ygcen.2010.09.019. steroid treatment. Mol. Cell. Endocrinol. 138, 81–93.
Borg, B., 1994. Androgens in teleost fishes. Comp. Biochem. Physiol. C Pharmacol. Ghiselin, M.T., 1969. The evolution of hermaphroditism among animals. Quart. Rev.
Toxicol. Endocrinol. 109, 219–245. Biol. 44, 189–208.
Burmeister, S.S., Jarvis, E.D., Fernald, R.D., 2005. Rapid behavioral and genomic Godwin, J., 1994a. Behavioural aspects of protandrous sex change in the anemonefish,
responses to social opportunity. PLoS Biol. 3 (11), e363. http://dx.doi.org/ Amphiprion melanopus, and endocrine correlates. Anim. Behav. 48, 551–567.
10.1371/journal.pbio.0030363. Godwin, J.R., 1994b. Histological aspects of protandrous sex change in the anemo-
Cardwell, J.R., Liley, N.R., 1991a. Hormonal control of sex and color change in the nefish Amphiprion melanopus. J. Zool. (Lond.) 232, 199–213.
stoplight parrotfish, Sparisoma viride. General Comp. Endocrinol. 81 (1), 7–20. Godwin, J., 2009. Social determination of sex in reef fishes. Sem. Cell Dev. Biol. 20
Cardwell, J.R., Liley, N.R., 1991b. Androgen control of social status in males of a wild (3), 264–270.
population of stoplight parrotfish, Sparisoma viride (Scaridae). Horm. Behav. 25 (1), Godwin, J.R., Thomas, P., 1993. Sex change and steroid profiles in the protandrous
1–18. http://doi.org/10.1016/0018-506x(91)90035-g. anemonefish Amphiprion melanopus (Pomacentridae, Teleostei). Gen. Comp.
Charlier, T.D., Cornil, C.A., Patte-Mensah, C., Meyer, L., Mensah-Nyagan, A.G., Endocrinol. 91, 144–157.
Balthazart, J., 2015. Local modulation of steroid action: rapid control of enzymatic Godwin, J., Crews, D., Warner, R.R., 1996. Behavioural sex change in the absence
activity. Front. Neurosci. 9, 83. http://dx.doi.org/10.3389/fnins.2015.00083. of gonads in a coral reef fish. Proc. Roy. Soc. Lond. B Biol. Sci. 263,
Cole, K.S., Shapiro, D.Y., 1995. Social facilitation and sensory mediation of adult 1683–1688.
sex change in a cryptic, benthic marine goby. J. Exp. Mar. Biol. Ecol. 186, Godwin, J., Sawby, R., Warner, R.R., Crews, D., Grober, M.S., 2000. Hypothalamic
65–75. arginine vasotocin mRNA abundance variation across sexes and with sex change in
Conover, D.O., 2004. Temperature-dependent sex determination in fishes. In: Valen- a coral reef fish. Brain Behav. Evol. 55, 77–84.
zuela, N., Lance, V. (Eds.), Temperature-dependent Sex Determination in Verte- Godwin, J., 2010. Neuroendocrinology of sexual plasticity in teleost fishes. Front.
brates. Smithsonian Institution Press, WA, pp. 11–20. Neuroendocrinol. 31, 203–216.
Crews, D., 1993. The organizational concept and vertebrates without sex chromo- Godwin, J., Thompson, R.R., 2012. Nonapeptides and social behavior in fishes. Horm.
somes. Brain Behav. Evol. 42, 202–214. Behav. 61, 230–238.
Davis, M.R., Fernald, R.D., 1990. Social control of neuronal soma size. J. Neurobiol. Goodson, J.L., Bass, A.H., 2000. Forebrain peptides modulate sexually polymorphic
21, 1180–1188. vocal circuitry. Nature 403, 769–772.
Devlin, R.H., Nagahama, Y., 2002. Sex determination and sex differentiation in fish: an Goodson, J.L., Thompson, R.R., 2010. Nonapeptide mechanisms of social cognition,
overview of genetic, physiological, and environmental influences. Aquaculture 208, behavior and species-specific social systems. Curr. Opin. Neurobiol. 20 (6),
191–364. 784–794. http://dx.doi.org/10.1016/j.conb.2010.08.020.
44 Socially Controlled Sex Change in Fishes

Grober, M.S., Bass, A.H., 1991. Neuronal correlates of sex/role change in labrid fishes: Kobayashi, Y., Nagahama, Y., Nakamura, M., 2013. Diversity and plasticity of sex
LHRH-like immunoreactivity. Brain Behav. Evol. 38, 302–312. determination and differentiation in fishes. Sex. Dev. 7 (1–3), 115–125. http://
Grober, M.S., Jackson, I., Bass, A.H., 1991. Gonadal steroids affect LHRH preoptic cell dx.doi.org/10.1159/000342009.
number in a sex/role changing fish. J. Neurobiol. 22 (7), 734–741. Kojima, Y., Bhandari, R.K., Kobayashi, Y., Nakamura, M., 2008. Sex change of adult
Grober, M.S., Sunobe, T., 1996. Serial adult sex change involves rapid and reversible initial-phase male wrasse, Halichoeres trimaculatus by estradiol-17b treatment.
changes in forebrain neurochemistry. Neuroreport 7 (18), 2945–2949. General Comp. Endocrinol. 156 (3), 628–632. http://dx.doi.org/10.1016/
Gross, M.R., Charnov, E.L., 1980. Alternative male life histories in bluegill sunfish. j.ygcen.2008.02.003.
Proc. Natl. Acad. Sci. U.S.A. 77, 6937–6940. Koulish, S., Kramer, C.R., 1989. Human chorionic gonadotropin (hCG) induces gonad
Guiguen, Y., Fostier, A., Piferrer, F., Chang, C.-F., 2010. Ovarian aromatase reversal in a protogynous fish, the bluehead wrasse, Thalassoma bifasciatum
and estrogens: a pivotal role for gonadal sex differentiation and sex change (Teleostei, Labridae). J. Exp. Zool. Part A Ecol. Genet. Physiol. 252 (2), 156–168.
in fish. Gen. Comp. Endocrinol. 165, 352–366. http://dx.doi.org/10.1016/ http://dx.doi.org/10.1002/jez.1402520207.
j.ygcen.2009.03.002. Kramer, C.R., Koulish, S., Bertacchi, P.L., 1988. The effects of testosterone implants
Hattori, R.S., Fernandino, J.I., Kishii, A., Kimura, H., Kinno, T., Oura, M., on ovarian morphology in the bluehead wrasse, Thalassoma bifasciatum (Bloch)
Somoza, G.M., Yokota, M., Strüssmann, C.A., Watanabe, S., 2009. Cortisol- (Teleostei: Labridae). J. Fish. Biol. 32, 397–407.
induced masculinization: does thermal stress affect gonadal fate in pejerrey, Kramer, C., Caddell, M., Bubenheimer Livolsi, L., 1993. sGnRH-A [(d-Arg6, Pro9,
a teleost fish with temperature-dependent sex determination? PLoS One 4, e6548. NEt-) LHRH] in combination with domperidone induces gonad reversal in
http://dx.doi.org/10.1371/journal.pone.0006548. a protogynous fish, the bluehead wrasse, Thalassoma bifasciatum. J. Fish
Hayashi, Y., Kobira, H., Yamaguchi, T., Shiraishi, E., Yazawa, T., Hirai, T., Kamei, Y., Biol. 42 (2), 185–195.
Kitano, T., 2010. High temperature causes masculinization of genetically female Kramer, C.R., Imbriano, M.A., 1997. Neuropeptide Y (NPY) induces gonad reversal in
medaka by elevation of cortisol. Mol. Reprod. Dev. 77, 679–686. http://dx.doi.org/ the protogynous bluehead wrasse, Thalassoma bifasciatum (Teleostei: Labridae).
10.1002/mrd.21203. J. Exp. Zool. Part A Ecol. Genet. Physiol. 279 (2), 133–144.
Higa, M., Ogasawara, K., Sakaguchi, A., Nagahama, Y., Nakamura, M., 2003. Role of Kroon, F.J., Liley, N.R., 2000. The role of steroid hormones in protogynous sex change
steroid hormones in sex change of protogynous wrasse. Fish Physiol. Biochem. 28 in the Blackeye Goby, Coryphopterus nicholsii (Teleostei: Gobiidae). Gen. Comp.
(1), 149–150. Endocrinol. 118 (2), 273–283. http://dx.doi.org/10.1006/gcen.2000.7459.
Hiraki, T., Takeuchi, A., Tsumaki, T., Zempo, B., Kanda, S., Oka, Y., Nagahama, Y., Kroon, F.J., Munday, P.L., Pankhurst, N.W., 2003. Steroid hormone levels and bi-
Okubo, K., 2012. Female-specific target sites for both oestrogen and androgen in directional sex change in Gobiodon histrio. J. Fish Biol. 62, 153–167.
the teleost brain. Proc. R. Soc. B Biol. Sci. 279 (1749), 5014–5023. http:// Kroon, F.J., Munday, P.L., Westcott, D.A., Hobbs, J.P.A., Liley, N.R., 2005. Aromatase
dx.doi.org/10.1098/rspb.2012.2011. pathway mediates sex change in each direction. Proc. Biol. Sci. R. Soc. 272
Hobbs, J.A., Munday, P.L., Jones, G.P., 2004. Social induction of maturation and sex (1570), 1399–1405. http://dx.doi.org/10.1098/rspb.2005.3097.
determination in a coral reef fish. Proc. R. Soc. B Biol. Sci. 271 (1553), 2109–2114. Kroon, F.J., Munday, P.L., Westcott, D.A., 2009. Equivalent whole-body concentrations of
http://dx.doi.org/10.1098/rspb.2004.2845. 11-ketotestosterone in female and male coral goby Gobiodon erythrospilus, a bidi-
Hourigan, T.F., Nakamura, M., Nagahama, Y., Yamauchi, K., Grau, E.G., 1991. rectional sex-changing fish. J. Fish Biol. 75 (3), 685–692. http://dx.doi.org/10.1111/
Histology, ultrastructure, and in vitro steroidogenesis of the testes of two male j.1095-8649.2009.02274.x.
phenotypes of the protogynous fish, Thalassoma duperrey (Labridae). Gen. Comp. Lamm, M.S., Liu, H., Gemmell, N.J., Godwin, J.R., 2015. The Need for Speed:
Endocrinol. 83, 193–217. neuroendocrine regulation of socially-controlled sex change. Am. Zool. 55 (2),
Huffman, L.S., Hinz, F.I., Wojcik, S., Aubin-Horth, N., Hofmann, H.A., 2015. Arginine 307–322. http://dx.doi.org/10.1093/icb/icv041.
vasotocin regulates social ascent in the African cichlid fish Astatotilapia burtoni. Larson, E.T., Norris, D.O., Summers, C.H., 2003a. Monoaminergic changes associated
General Comp. Endocrinol. 212 (C), 106–113. http://dx.doi.org/10.1016/ with socially induced sex reversal in the saddleback wrasse. Neuroscience 119 (1),
j.ygcen.2014.03.004. 251–263. http://dx.doi.org/10.1016/S0306-4522(03)00119-2.
Iwata, E., Mikami, K., Manbo, J., Moriya-Ito, K., Sasaki, H., 2012. Social interaction Larson, E.T., Norris, D.O., Gordon Grau, E., Summers, C.H., 2003b. Monoamines
influences blood cortisol values and brain aromatase genes in the protandrous false stimulate sex reversal in the saddleback wrasse. General Comp. Endocrinol. 130
clown anemonefish, Amphiprion ocellaris. Zool. Sci. 29, 849–855. http://dx.doi.org/ (3), 289–298. http://dx.doi.org/10.1016/S0016-6480(02)00622-6.
10.2108/zsj.29.849. Le Page, Y., Diotel, N., Vaillant, C., Pellegrini, E., Anglade, I., Mérot, Y., Kah, O., 2010.
Johnson, M.A., Tsutsui, K., Fraley, G.S., 2007. Rat RFamide-related peptide-3 stim- Aromatase, brain sexualization and plasticity: the fish paradigm. Eur. J. Neurosci.
ulates GH secretion, inhibits LH secretion, and has variable effects on sex behavior 32 (12), 2105–2115. http://dx.doi.org/10.1111/j.1460-9568.2010.07519.x.
in the adult male rat. Horm. Behav. 51 (1), 171–180. Lema, S.C., 2010. Identification of multiple vasotocin receptor cDNAs in teleost fish:
Kaewwongse, M., Takayanagi, Y., Onaka, T., 2011. Effects of RFamide-related peptide sequences, phylogenetic analysis, sites of expression, and regulation in the
(RFRP)-1 and RFRP-3 on oxytocin release and anxiety-related behaviour in rats. hypothalamus and gill in response to hyperosmotic challenge. Mol. Cell. Endocrinol.
J. Neuroendocrinol. 23 (1), 20–27. 321, 215–230.
Kanda, S., Akazome, Y., Mitani, Y., Okubo, K., Oka, Y., 2013. Neuroanatomical Lema, S.C., Slane, M.A., Salvesen, K.E., Godwin, J., 2012. Variation in gene transcript
evidence that kisspeptin directly regulates isotocin and vasotocin neurons. PLoS profiles of two V1a-type arginine vasotocin receptors among sexual phases of
One 8 (4), e62776. bluehead wrasse (Thalassoma bifasciatum). General Comp. Endocrinol. 179 (3),
Kawabata, Y., Hiraki, T., Takeuchi, A., Okubo, K., 2012. Sex differences in the 451–464. http://dx.doi.org/10.1016/j.ygcen.2012.10.001.
expression of vasotocin/isotocin, gonadotropin-releasing hormone, and tyrosine Liu, H., Lamm, M.S., Rutherford, K., Black, M.A., Godwin, J.R., Gemmell, N.J., 2015.
and tryptophan hydroxylase family genes in the medaka brain. Neuroscience 218 Large-scale transcriptome sequencing reveals novel expression patterns for key
(C), 65–77. http://dx.doi.org/10.1016/j.neuroscience.2012.05.021. sex-related genes in a sex-changing fish. Biol. Sex Differ. 6, 26. http://dx.doi.org/
Kline, R.J., 2010. Hormonal Correlates of Coloration and Sexual Change in the 10.1186/s13293-015-0044-8.
Hermaphroditic Grouper, Epinephelus adscensionis (Ph.D. thesis). The University of Lorenzi, V., Earley, R.L., Rodgers, E.W., Pepper, D.R., Grober, M.S., 2008. Diurnal
Texas at Austin. patterns and sex differences in cortisol, 11-ketotestosterone, testosterone, and
Kline, R.J., Khan, I.A., Holt, G.J., 2011a. Behavior, color change and time for sexual 17beta-estradiol in the bluebanded goby (Lythrypnus dalli). Gen. Comp. Endocrinol.
inversion in the protogynous grouper (Epinephelus adscensionis). PLoS One 6 (5), 155 (2), 438–446.
e19576. http://dx.doi.org/10.1371/journal.pone.0019576.g005. Lorenzi, V., Earley, R., Grober, M.S., 2012. Differential responses of brain, gonad and
Kline, R.J., O’Connell, L.A., Hofmann, H.A., Holt, G.J., Khan, I.A., 2011b. The Muscle steroid levels to changes in social status and sex in a sequential and
distribution of an AVT V1a receptor in the brain of a sex changing fish, Epinephelus bidirectional hermaphroditic fish. PLoS One 7 (12), e51158.
adscensionis. J. Chem. Neuroanat. 42 (1), 72–88. http://dx.doi.org/10.1016/ Luckenbach, J.A., Borski, R.J., Daniels, H.V., Godwin, J., 2009. Sex determination in
j.jchemneu.2011.06.005. flatfishes: mechanisms and environmental influences. Semin. Cell Dev. Biol. 20,
Kline, R.J., Holt, G.J., Khan, I.A., 2016. Arginine vasotocin V1a2 receptor and GnRH-I 256–263. http://dx.doi.org/10.1016/j.semcdb.2008.12.002.
co-localize in preoptic neurons of the sex changing grouper, Epinephelus Lorenzi, V., Grober, M.S., 2012. Immunohistochemical localization of serotonin in the
adscensionis. Gen. Comp. Endocrinol. 225, 33–44. brain during natural sex change in the hermaphroditic goby Lythrypnus dalli.
Kobayashi, Y., Nakamura, M., Sunobe, T., Usami, T., Kobayashi, T., Manabe, H., Paul- General Comp. Endocrinol. 175 (3), 527–536. http://dx.doi.org/10.1016/
Prasanth, B., Suzuki, N., Nagahama, Y., 2009. Sex change in the gobiid fish is j.ygcen.2011.12.018.
mediated through rapid switching of gonadotropin receptors from ovarian to Lutnesky, M.M.F., 1994. Density-dependent protogynous sex-change in territorial
testicular portion or vice versa. Endocrinology 150, 1503–1511. haremic fishes - models and evidence. Behav. Ecol. 5, 375–383.
Socially Controlled Sex Change in Fishes 45

de Mitcheson, Y.S., de Mitcheson, S., Liu, M., 2008. Functional hermaphroditism in Phoenix, C.H., Goy, R.W., Gerall, A.A., Young, W.C., 1959. Organizing action of
teleosts. Fish Fish. 9, 1–43. prenatally administered testosterone propionate on the tissues mediating mating
Manousaki, T., Tsakogiannis, A., Lagnel, J., Sarropoulou, E., Xiang, J.Z., behavior in the female guinea pig. Endocrinology 65, 369–382.
Papandroulakis, N., Mylonas, C.C., Tsigenopoulos, C.S., 2014. The sex-specific Pickford, G.E., Strecker, E.L., 1977. The spawning reflex response of the killifish,
transcriptome of the hermaphrodite sparid sharpsnout seabream (Diplodus pun- Fundulus heteroclitus: isotocin is relatively inactive in comparison with arginine
tazzo). BMC Genomics 15 (1), 655. http://dx.doi.org/10.1186/1471-2164-15-655. vasotocin. Gen. Comp. Endocrinol. 32, 132–137.
Mankiewicz, J., Godwin, J., Holler, B., Turner, P., Murashige, R., Shamey, R., Qi, X., Zhou, W., Li, S., Lu, D., Yi, S., Xie, R., Liu, X., Zhang, Y., Lin, H., 2013.
Daniels, H., Borski, R., 2013. Tank color and stress steroids affect sex in a flatfish Evidences for the regulation of GnRH and GTH expression by GnIH in the goldfish,
with temperature-dependent sex determination. Integr. Comp. Biol. 53, 755–765. Carassius auratus. Mol. Cell Endocrinol. 366, 9–20.
Marsh, K.E., Creutz, L.M., Hawkins, M.B., Godwin, J., 2006. Aromatase immuno- Reinboth, R., Brusle-Sicard, S., 1997. Histological and ultrastructural studies on the
reactivity in the bluehead wrasse brain, Thalassoma bifasciatum: immunolocali- effects of hCG on sex inversion in the protogynous teleost Coris julis. J. Fish. Biol.
zation and co-regionalization with arginine vasotocin and tyrosine hydroxylase. 51, 738–749.
Brain Res. 1126 (1), 91–101. http://dx.doi.org/10.1016/j.brainres.2006.09.017. Reinboth, R., 1980. Can sex inversion be environmentally induced? Biol. Reprod. 22
Marsh-Hunkin, K.E., Heinz, H.M., Hawkins, M.B., Godwin, J., 2013. Estrogenic control (1), 49–59.
of behavioral sex change in the bluehead wrasse, Thalassoma bifasciatum. Am. Reinboth, R., 1988. Physiological problems of teleost ambisexuality. Environ. Biol.
Zool. 53 (6), 951–959. http://dx.doi.org/10.1093/icb/ict096. Fishes 22 (4), 249–259. http://dx.doi.org/10.1007/BF00004891.
Maruska, K.P., Fernald, R.D., 2011. Social regulation of gene expression in the Robertson, D.R., 1972. Social control of sex reversal in a coral-reef fish. Science 177
hypothalamic-pituitary-gonadal Axis. Physiology 26 (6), 412–423. http:// (4053), 1007–1009.
dx.doi.org/10.1152/physiol.00032.2011. Rodgers, E.W., Drane, S., Grober, M.S., 2005. Sex reversal in pairs of Lythrypnus
Moore, M.C., 1991. Application of organization activation theory to alternative male dalli: behavioral and morphological changes. Biol. Bull. 208 (2), 120–126.
reproductive strategies – a review. Horm. Behav. 25, 154–179. Rodgers, E.W., Earley, R.L., Grober, M.S., 2007. Social status determines sexual
Moussavi, M., Wlasichuk, M., Chang, J.P., Habibi, H.R., 2012. Seasonal effect of GnIH phenotype in the bi-directional sex changing bluebanded goby Lythrypnus
on gonadotrope functions in the pituitary of goldfish. Mol. Cell. Endocrinol. 350, dalli. J. Fish Biol. 70 (6), 1660–1668. http://dx.doi.org/10.1111/j.1095-
53–60. 8649.2007.01427.x.
Munakata, A., Kobayashi, M., 2010. Endocrine control of sexual behavior in teleost Ross, R.M., Losey, G.S., Diamond, M., 1983. Sex change in a coral-reef fish –
fish. General Comp. Endocrinol. 165 (3), 456–468. http://dx.doi.org/10.1016/ dependence of stimulation and inhibition on relative size. Science 221,
j.ygcen.2009.04.011. 574–575.
Munday, P.L., White, J.W., Warner, R.R., 2006. A social basis for the development of Ross, R.M., Hourigan, T.F., Lutnesky, M.M.F., Singh, I., 1990. Multiple simultaneous
primary males in a sex-changing fish. Proc. R. Soc. Lond. B Biol. Sci. 273, sex-changes in social-groups of a coral-reef fish. Copeia 2, 427–433.
2845–2851. Sadovy, Y., Shapiro, D.Y., 1987. Criteria for the diagnosis of hermaphroditism in fishes.
Nakamura, M., Hourigan, T.F., Yamauchi, K., Nagahama, Y., Grau, E.G., 1989. Copeia 1, 136. http://dx.doi.org/10.2307/1446046.
Histological and ultrastructural evidence for the role of gonadal steroid hormones in Semsar, K., Kandel, F.L.M., Godwin, J., 2001. Manipulations of the AVT system shift
sex change in the protogynous wrasse Thalassoma duperrey. Environ. Biol. Fishes social status and related courtship and aggressive behavior in the bluehead
24 (2), 117–136. wrasse. Horm. Behav. 40, 21–31.
Newman, S.W., 1999. The medial extended amygdala in male reproductive behavior: Semsar, K., Godwin, J., 2003. Social influences on the arginine vasotocin system
a node in the mammalian social behavior network. Ann. N.Y. Acad. Sci. 877, are independent of gonads in a sex-changing fish. J. Neurosci. 23,
242–257. 4386–4393.
Notch, E.G., Shaw, J.R., Coutermarsh, B.A., Dzioba, M., Stanton, B.A., 2011. Mor- Semsar, K., Godwin, J., 2004. Multiple mechanisms of phenotype development in the
pholino gene Knockdown in adult Fundulus heteroclitus: role of SGK1 in bluehead wrasse. Horm. Behav. 45, 345–353.
Seawater Acclimation. PLos One 6 (12), e29462. http://dx.doi.org/10.1371/ Semsar, K., Perreault, H.A., Godwin, J., 2004. Fluoxetine-treated male wrasses exhibit
journal.pone.0029462.t001. low AVT expression. Brain Res. 1029, 141–147.
Nozu, R., Nakamura, M., 2015. Cortisol administration induces sex change from ovary Shahjahan, M., Ikegami, T., Osugi, T., et al., 2011. Synchronised expressions of
to testis in the protogynous wrasse, Halichoeres trimaculatus. Sex. Dev. 9 (2), LPXRFamide peptide and its receptor genes: seasonal, diurnal and circadian
118–124. http://dx.doi.org/10.1159/000373902. changes during spawning period in grass puffer. J. Neuroendocrinol. 23,
O’Connell, L.A., Hofmann, H.A., 2011. The Vertebrate mesolimbic reward system and 39–51.
social behavior network: a comparative synthesis. J. Comp. Neurol. 519 (18), Shapiro, D.Y., 1980. Serial female sex changes after simultaneous removal of males
3599–3639. http://dx.doi.org/10.1002/cne.22735. from social groups of a coral reef fish. Science 209, 1136–1137.
O’Connell, L.A., Hofmann, H.A., 2012. Evolution of a vertebrate social decision- Shi, Y., Zhang, Y., Li, S., Liu, Q., Lu, D., Liu, M., Meng, Z., Cheng, C.H.,
making network. Science 336 (6085), 1154–1157. http://dx.doi.org/10.1126/ Liu, X., Lin, H., 2010. Molecular identification of the Kiss2/Kiss1ra system
science.1218889. and its potential function during 17alpha-methyltestosterone-induced sex
Oakley, A.E., Clifton, D.K., Steiner, R.A., 2009. Kisspeptin signaling in the brain. reversal in the orange-spotted grouper, Epinephelus coioides. Biol. Reprod.
Endocr. Rev. 30, 713–743. 83, 63–74.
Ogawa, S., Parhar, I.S., 2014. Structural and functional divergence of gonadotropin- Solomon-Lane, T.K., Crespi, E.J., Grober, M.S., 2013. Stress and serial adult meta-
inhibitory hormone from jawless fish to mammals. Front. Endocrinol. (Lausanne) morphosis: multiple roles for the stress axis in socially regulated sex change. Front.
5, 117. Neurosci. 7, 210. http://dx.doi.org/10.3389/fnins.2013.00210.
Ohta, K., Hirano, M., Mine, T., Mizutani, H., Yamaguchi, A., Matsuyama, M., 2007. Stacey, N.E., 1987. Roles of hormones and Pheromones in fish reproductive behavior.
Body color change and serum steroid hormone levels throughout the process of sex In: Crews, D. (Ed.), Psychobiology of Reproductive Behavior: An Evolutionary
change in the adult wrasse, Pseudolabrus sieboldi. Mar. Biol. 153 (5), 843–852. Perspective. Englewood Cliffs, Prentice-Hall, NJ, pp. 28–60.
http://dx.doi.org/10.1007/s00227-007-0856-0. Stoll, L.M., 1955. Hormonal control of the sexually dimorphic pigmentation of Tha-
Ohta, K., Hirano, M., Mine, T., Mizutani, H., Yamaguchi, A., Matsuyama, M., 2008. lassoma bifasciatum. Zoologica 40, 125–131.
Body color change and serum steroid hormone levels throughout the process of sex Streelman, J.T., Karl, S.A., 1997. Reconstructing labroid evolution with single-copy
change in the adult wrasse, Pseudolabrus sieboldi. Mar. Biol. 153 (5), 843–852. nuclear DNA. Proc. Biol. Sci. R. Soc. 264 (1384), 1011–1020. http://
Oka, Y., 2009. Three types of gonadotrophin-releasing hormone neurones and steroid- dx.doi.org/10.1098/rspb.1997.0140.
sensitive sexually dimorphic kisspeptin neurones in teleosts. J. Neuroendocrinol. Summers, C.H., Winberg, S., 2006. Interactions between the neural regulation of
21, 334–338. stress and aggression. J. Exp. Biol. 209, 4581–4589.
Perreault, H., semsar, K., Godwin, J., 2003. Fluoxetine treatment decreases territorial Taborsky, M., 1998. Sperm competition in fish: bourgeois males and parasitic
aggression in a coral reef fish. Physiol. Behav. 79 (4–5), 719–724. http:// spawning. Trends Ecol. Evol. 13, 222–227.
dx.doi.org/10.1016/S0031-9384(03)00211-7. Tena-Sempere, M., Felip, A., Gómez, A., Zanuy, S., Carrillo, M., 2012. Comparative
Perry, A.N., Grober, M.S., 2003. A model for social control of sex change: interactions insights of the kisspeptin/kisspeptin receptor system: lessons from non-
of behavior, neuropeptides, glucocorticoids, and sex steroids. Horm. Behav. 43, mammalian vertebrates. Gen. Comp. Endocrinol. 175 (2), 234–243.
31–38. Tsutsui, K., Saigoh, E., Ukena, K., Teranishi, H., Fujisawa, Y., Kikuchi, M., Ishii, S.,
Petersen, C.W., 1987. Reproductive behaviour and gender allocation in Serranus Sharp, P.J., 2000. A novel avian hypothalamic peptide inhibiting gonadotropin
fasciatus, a hermaphroditic reef fish. Anim. Behav. 351, 601–1614. release. Biochem. Biophys. Res. Commun. 275 (2), 661–667.
46 Socially Controlled Sex Change in Fishes

Tsutsui, K., Bentley, G.E., Kriegsfeld, L.J., Osugi, T., Seong, J.Y., Vaudry, H., 2010. Wong, R.Y., McLeod, M.M., Godwin, J., 2014. Limited sex-biased neural gene
Discovery and evolutionary history of gonadotrophin-inhibitory hormone and kiss- expression patterns across strains in Zebrafish (Danio rerio). BMC Genomics 15
peptin: new key neuropeptides controlling reproduction. J. Neuroendocrinol. 22 (7), (905). http://www.biomedcentral.com/1471-2164/15/905.
716–727. Yaeger, C., Ros, A.M., Cross, V., DeAngelis, R.S., Stobaugh, D.J., Rhodes, J.S., 2014.
Ubuka, T., Mukai, M., Wolfe, J., Beverly, R., Clegg, S., Wang, A., Hsia, S., Li, M., Blockade of arginine vasotocin signaling reduces aggressive behavior and c-fos
Krause, J.S., Mizuno, T., Fukuda, Y., Tsutsui, K., Bentley, G.E., Wingfield, J.C., expression in the preoptic area and periventricular nucleus of the posterior
2012. RNA interference of gonadotropin-inhibitory hormone gene induces arousal tuberculum in male Amphiprion ocellaris. Neuroscience 267 (C), 205–218. http://
in songbirds. PLoS One 7, e30202. dx.doi.org/10.1016/j.neuroscience.2014.02.045.
Ubuka, T., Haraguchi, S., Tobari, Y., Narihiro, M., Ishikawa, K., Hayashi, T., Harada, N., Yamaguchi, T., Yoshinaga, N., Yazawa, T., Gen, K., Kitano, T., 2010. Cortisol is
Tsutsui, K., 2014. Hypothalamic inhibition of socio-sexual behaviour by increasing involved in temperature-dependent sex determination in the Japanese flounder.
neuroestrogen synthesis. Nat. Commun. 5, 3061. Endocrinology 151, 3900–3908. http://dx.doi.org/10.1210/en.2010-0228.
Vida, B., Deli, L., Hrabovszky, E., Kalamatianos, T., Caraty, A., Coen, C.W., Liposits, Z., Zhang, Y., Li, S., Liu, Y., Lu, D., Chen, H., Huang, X., Liu, X., Meng, Z., Lin, H.,
Kallo, I., 2010. Evidence for Suprachiasmatic vasopressin neurones innervating Cheng, C.H., 2010. Structural diversity of the GnIH/GnIH receptor system in teleost:
kisspeptin neurones in the Rostral periventricular area of the Mouse brain: regu- its involvement in early development and the negative control of LH release.
lation by oestrogen. J. Neuroendocrinol. 22 (9), 1032–1039. http://dx.doi.org/ Peptides 31, 1034–1043.
10.1111/j.1365-2826.2010.02045.x. Zmora, N., Stubblefield, J., Zulperi, Z., Biran, J., Levavi-Sivan, B., Muñoz-Cueto, J.A.,
Warner, R.R., Robertson, D.R., Leigh, E.G., 1975. Sex change and sexual selection. Zohar, Y., 2012. Differential and gonad stage-dependent roles of kisspeptin1 and
Science 190, 633–638. kisspeptin2 in reproduction in the modern teleosts, Morone species. Biol. Reprod.
Warner, R.R., 1984. Mating behavior and hermaphroditism in coral-reef fishes. Am. 86, 177.
Sci. 72, 128–136. Zmora, N., Stubblefield, J., Golan, M., Servili, A., Levavi-Sivan, B., Zohar, Y., 2014.
Warner, R.R., Swearer, S.E., 1991. Social-control of sex change in the bluehead The medio-basal hypothalamus as a dynamic and plastic reproduction-related
wrasse, Thalassoma bifasciatum (Pisces, Labridae). Biol. Bull. 181, 199–204. kisspeptin-gnrh-pituitary center in fish. Endocrinology 155 (5), 1874–1886.
Wilhemi, A.E., Pickford, G.E., Sawyer, W.H., 1955. Initiation of the spawning reflex Zmora, N., Stubblefield, J.D., Wong, T.T., Levavi-Sivan, B., Millar, R.P., Zohar, Y.,
response in Fundulus by the administration of fish and mammalian neurohypo- 2015. Kisspeptin antagonists reveal kisspeptin 1 and kisspeptin 2 differential
physeal preparations and synthetic oxytocin. Endocrinology 57, 243–325. regulation of reproduction in the teleost, Morone saxatilis. Biol. Reprod. 93
Williams, W.P., Jarjisian, S.G., Mikkelsen, J.D., Kriegsfeld, L.J., 2011. Circadian (3), 76.
control of kisspeptin and a gated GnRH response mediate the Preovulatory lutei-
nizing hormone Surge. Endocrinology 152 (2), 595–606. http://dx.doi.org/
10.1210/en.2010-0943.
2.04 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive
Tactics: Vocal Fish as Model Systems
Ni Y Feng and Andrew H Bass, Cornell University, Ithaca, NY, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by A.H. Bass, M.S. Grober, volume 1, pp. 579–610, Ó 2009, Elsevier Inc.

2.04.1 Introduction 47
2.04.1.1 Plasticity in a Suite of Characters 48
2.04.2 Alternative Male Reproductive Morphs in Midshipman Fish – A Case Study 48
2.04.2.1 Reproductive and Vocal–Acoustic Behaviors 48
2.04.2.2 Somatic, Gonadal, and Endocrine Traits 51
2.04.2.3 Central Vocal Network 51
2.04.2.3.1 Central Vocal Network, Structure and Function 51
2.04.2.3.2 Intra- and Intersexual Divergence in Vocal Network 53
2.04.3 Hormones, Vocal Behavior, and Vocal Central Pattern Generator 53
2.04.3.1 Divergent Steroid Hormone Profiles and Action between Sex and Male Morphs 54
2.04.3.1.1 Long-Term and Seasonal Steroid Action 54
2.04.3.1.2 Rapid Steroid Action 54
2.04.3.2 Sex- and Male Morph-Specific Patterns of Nonapeptide Actions on Vocal Neurophysiology 56
2.04.4 Hormonal Modulation of Hearing 56
2.04.4.1 Seasonal Auditory Plasticity 58
2.04.4.2 Steroid-Dependent Auditory Plasticity 59
2.04.5 Sites of Hormone Synthesis and Action 59
2.04.5.1 Neuropeptides 59
2.04.5.2 Aromatase 60
2.04.5.3 Androgen and ERs 61
2.04.5.4 Glucocorticoids 61
2.04.5.5 Catecholamines 62
2.04.6 Daily and Seasonal Rhythms in Vocal–Acoustic Mechanisms – Genetic Insights 62
2.04.6.1 VMN Transcriptome 63
2.04.6.2 Saccular Epithelium Transcriptome 65
2.04.7 Summary 65
Acknowledgments 65
References 65

Glossary
Advertisement calls Vocalizations made by males to attract Saccule Division of the vertebrate inner ear that
females. primarily functions as an auditory organ in many species
Arginine vasotocin Nonapeptide in nonmammalian of teleost fish.
vertebrates, including fish, that is homologue of Satellite-spawning A mating tactic whereby a male
vasopressin. attempts to steal fertilizations by spawning immediately
Aromatase (estrogen synthase) Converts testosterone to outside of the nests of other nest-guarding males when
estradiol. a female is present.
Gonosomatic index (GSI) Ratio of gonad mass/body Sneak-spawning A mating tactic whereby a male attempts
mass. to steal fertilizations by spawning inside the nests of other
Isotocin Nonapeptide homologue of mammalian oxytocin nest-guarding males when a female is present.
found in fish.

2.04.1 Introduction reproductive phenotypes that present the opportunity to inves-


tigate how natural variation in neural and hormonal mecha-
Teleost fish, which comprise nearly half of all living species nisms drive plasticity in behavior. In order to causally link
of vertebrates (Nelson, 2006), exhibit a wide range of neural and hormonal events to variation in behavioral output,

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00018-3 47


48 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

it is beneficial to study behaviors that are readily quantifiable, (Bass, 1992). Here, we largely focus on the last two character
amenable to experimental manipulation, relatively simple sets with respect to acoustic communication in a family of
in their biomechanical basis, yet naturally variable. Vocal– highly vocal fish, known as the toadfishes (Batrachoididae),
acoustic behaviors exhibit these characteristics, as they are vari- which includes the plainfin midshipman fish (Porichthys notatus
able in several quantifiable characteristics such as duration, Girard) (e.g., Greenfield et al., 2008; Rice and Bass, 2009).
frequency, and amplitude. Furthermore, vocalizations are often Section 2.04.2 provides an overview of investigations of vocal
reliable readouts of motor command signals from the central and reproductive behaviors in the midshipman fish to demon-
nervous system (CNS) and reflect an animal’s internal state strate that ARTs can be manifested at multiple levels of biolog-
and hormonal milieu (Bradbury and Vehrencamp, 2011). ical organization. Sections 2.04.3 and 2.04.4 assess hormonal
For many species of vertebrates, including teleost fish, vocal- modulation of neurophysiological mechanisms, namely vocal
ization is a key feature of social communication (Bradbury and (Section 2.04.3) and auditory (Section 2.04.4) that underlie
Vehrencamp, 2011; Fine and Parmentier, 2015). Fish vocal acoustic courtship behaviors crucial for toadfish reproduction.
behaviors exhibit natural variation across social contexts Section 2.04.5 reviews sexual polymorphisms in neuroendo-
(e.g., Brantley and Bass, 1994), daily and seasonal cycles crine anatomical phenotypes. This includes the distribution
(e.g., Locascio and Mann, 2008; Forlano et al., 2015b), and of neuropeptides that have been extensively studied in the
reproductive phenotypes (see below). Furthermore, compared context of reproductive plasticity among species exhibiting
to other vertebrates such as birds that utilize complex neural a wide range of life history patterns (see Section 2.04.1) –
circuitry to control multiple groups of sonic muscles, fish gonadotropin-releasing hormone (GnRH) and the nonapepti-
have a relatively simple and dedicated central pattern generator des arginine vasotocin (AVT) and isotocin that are the fish
(CPG) network that controls as few as one pair of sonic muscles homologues of mammalian arginine vasopressin and oxytocin,
(see recent reviews Bass et al., 2015; Fine and Parmentier, respectively (Bentley, 1998). Also reviewed are the expression
2015). Thus, the simple yet naturally variable vocalizations of patterns of steroid receptors and steroid synthesizing enzymes
fishes are excellent models for understanding the neural and such as aromatase that catalyzes the conversion of testosterone
hormonal basis of vertebrate behavior. to estrogen. Section 2.04.6 reports on the most recent investiga-
While most species of teleost fish exhibit a single reproduc- tions using transcriptomic RNA sequencing methods to
tive phenotype within a sex, at least seven life history patterns identify genetic and molecular mechanisms of reproductive-
of sexual polymorphisms in behavioral and neurobiological related plasticity in vocalization and audition. Section 2.04.7
characters have arisen in teleosts. Specifically, these include presents summary comments on reproductive and behavioral
species exhibiting (1) alternative reproductive tactics (ARTs) plasticity among teleost fishes.
with two distinct phenotypes within a sex that utilize different
behavioral tactics to acquire mates; (2) singular, unidirectional
adult sex change and within-sex morph change; (3) singular,
2.04.2 Alternative Male Reproductive Morphs
unidirectional sex change; (4) singular, unidirectional intrasex-
in Midshipman Fish – A Case Study
ual (male) morph change; (5) serial, back-and-forth, sex
change; (6) bisexual, simultaneous hermaphrodite; and
The mechanisms underlying the expression of ARTs, from
(7) serial, back-and-forth shift by males between reproductive
behavioral ecology and life history strategies to neuroendocri-
and nonreproductive states (for further discussion, see Foran
nology and central motor control, have been extensively
and Bass, 1999). Elucidating how this staggering diversity in
studied in the plainfin midshipman fish (Figure 1; Table 1).
teleost mating strategies is reflected in and determined by the
We first describe reproductive and vocal–acoustic behaviors
CNS is therefore a challenging task that requires a comparative
and then review the current understanding of the underlying
approach. Here, we review insights on the neural and
anatomical and physiological mechanisms.
hormonal mechanisms driving inter- and intrasexual dimor-
phisms in vocal–acoustic behavior gained from fish models
for acoustic communication. Though we focus on species
2.04.2.1 Reproductive and Vocal–Acoustic Behaviors
with life history pattern 1, the reviewed studies show how
neurobiological characters that lead to the performance of The plainfin midshipman is a highly vocal species of fish that
a social communication behavior, in this case vocalization, mainly breeds at night during the late spring and summer in
can vary between reproductive morphs. Hence, investigations the rocky intertidal zone along the western United States and
of vocal teleosts can inform us about potential mechanisms Canada (Figure 1(a); Bass, 1996). Successful reproduction in
in other species (patterns 2–7) that underlie short- and long- midshipman and other toadfishes involves a suite of courtship
term phenotypic plasticity both within and between the sexes. and spawning behaviors and relies on species and sex-typical
vocalizations (Figure 1(b) and 1(c); Brantley and Bass, 1994;
McIver et al., 2014). The plainfin midshipman has two male
2.04.1.1 Plasticity in a Suite of Characters
reproductive morphs, types I and II, that diverge in courtship
A suite of characters can often be used to define reproductive and spawning behaviors as well as a range of morphological
phenotypes in teleost fishes, including body size and colora- and hormonal characters (Section 2.04.2.2; Figure 1, Table 1;
tion, gonad anatomy and physiology, genital papilla shape, Bass, 1996). Comparable tactics and morphs have been
sex-specific accessory gonadal structure, courtship display described in at least one other species of toadfish, the Lusita-
behaviors and related structures (e.g., vocal organs), as well nian toadfish, Halobatrachus didactylus (e.g., Modesto and
as neuroendocrine and associated endocrine mechanisms Canário, 2003; Vasconcelos et al., 2010).
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 49

(a) Midshipman’s rocky intertidal habitat

(b) Midshipman spawning behavior

(c) Type I male vocal repertoire


Advertisement hum

Continous for mins to hrs

30 ms
Grunt train
Agonistic growl

100 ms

40 ms Also produced by type II males and females


(d) Somatic traits
I II

Figure 1 Midshipman ecology, spawning behavior, and alternative reproductive tactics. (a) Left: During the summer breeding season, type I males
defend nests under rocks along the rocky intertidal zone. White arrows in left panel point to the residential type I males whose rocks have been over-
turned in the picture. Right: Each midshipman nest can contain one or more resident type I males, visiting females, sneak/satellite-spawning type II
males, and newly fertilized eggs, embryos and hatched larvae attached to the underside of the rock. (b) Schematic drawings depicting spawning
scenarios, one where there is no sneaker type II male present (left) and one where a type II male satellite spawns by positioning his body adjacent to
the nest while fanning sperm into the nest of the spawning pair. (c) Midshipman vocal repertoire used under different social contexts. Type I males
produce long duration, ‘hum’ advertisement calls, and agonistic ‘growls’ and ‘grunt trains.’ Type II males and females have only been observed to
produce isolated grunts under agonistic contexts. (d) Somatic characters that diverge across and within sexes include the size and morphology of the
gonads and the vocal organ. Type I males have larger swim bladder and vocal muscles, while type II males and females have larger gonads relative
to body size. Photos by Margaret Marchaterre, line drawings by Margaret Nelson.
50 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

Table 1 Inter- and intrasexual polymorphic characters in the plainfin midshipman fish

Type I male Type II male Female

Somatic traits
Body size Large Small Intermediate
Gonad/body size ratio Small Large Large (gravid); small (spent)
Ventral coloration Olive-gray Mottled yellow Bronze/golden (gravid);
mottled (spent)
Behavioral traits
Nest building Yes No No
Egg guarding Yes No No
Vocal repertoire Hums, grunts, Grunts Grunts
grunt trains, growls
Call duration Long Short Short
Call fundamental frequency High Low Low
Vocal motor traits
Vocal muscle mass, fiber number, Large Small Small
and diameter
Vocal motoneuron size Large Small Small
Fictive call discharge frequency High Low Low
Fictive call duration Long Short Short
Endocrine traits
Predominant circulating steroids 11-Ketotestosterone Testosterone Testosterone, estradiol
Neuroendocrine traits
Aromatase enzyme activity, mRNA, protein Low High High
GnRH-POA neuron size Large Small Small
GnRH-POA neuron number/body size ratio Low High Low
AVT-POA neuron size Large Small Large
AVT-POA neuron number/body size ratio Low High Low
Fictive call response to exogenous hormones
11-ketotestosterone Facilitation No effect No effect
Testosterone No effect AR-dependent facilitation ER-dependent facilitation
Estradiol Facilitation Facilitation Facilitation
Cortisol Facilitation Suppression Suppression
Arginine vasotocin Suppression No effect No effect
Isotocin No effect Suppression Suppression

AVT, arginine-vasotocin; GnRH, gondotropin-releasing hormone; POA, preoptic area.


Modified from previous version of chapter Bass, A.H., Grober, M.S., 2009. Reproductive plasticity in fish: evolutionary lability in the patterning of
neuroendocrine and behavioral traits underlying divergent sexual phenotypes. In: Pfaff, D.W., Arnold, A.P., Moss, R., Etgen, A.M., Fahrbach, S.E.,
Rubin, R.T., (Eds.), Hormones, Brain and Behavior. Academic Press, New York, pp. 579–610.

Type I males were those already known in the literature tactics, suggesting an additional level of plasticity within the
to build nests under rocky shelters in the intertidal and sub- type I morph (Lee and Bass, 2004, 2006).
tidal environments where they fertilize the eggs of females Ibara et al. (1983) first recognized that nesting males
that are deposited on the roof of the nest (Figure 1(a) produce an unusually long duration, tonelike acoustic signal
and 1(b)) (e.g., Greene, 1924; Arora, 1948). Females depart that they called ‘hums’ (Figure 1(c); Ibara et al., 1983),
after releasing all of their eggs and type I males remain to perhaps the same ‘peculiar humming sounds’ reported even
care for the eggs. Midshipman are polygynous in that a single earlier by Hubbs (1920). Brantley and Bass (1994) showed
nest may have several thousand eggs from different females; that hums function to attract females to nests and character-
a gravid female produces at most close to 200 eggs each ized their spectral and temporal properties along with the
breeding season (DeMartini, 1988; Bass, 1996; McIver properties of agonistic grunt vocalizations (Figure 1(c); see
et al., 2014). Juvenile males and females ranging in age McIver et al., 2014 for extensive sound analysis). Individual
from 5 to 12 months are not found in nests, but rather in hums may continue without cessation for an hour or more
eel grass beds in close proximity to nesting sites (Brantley and then continue after a brief pause so that males will
et al., 1993a). produce them throughout an entire evening (McIver et al.,
Type II males do not acoustically court females, do not 2014; Feng et al., 2015).
build nests, and do not guard eggs. Rather, type II males Underwater playbacks show that females exhibit positive
attempt to steal fertilizations from type I males by satellite- phonotaxis to hums or tones that mimic the spectral and
spawning from positions immediately outside, or by sneak- temporal properties of hums (Ibara et al., 1983; McKibben
spawning inside, the nest of a type I male (Figure 1(b); Brantley and Bass, 1998, 2001). Individual grunts, on average about
and Bass, 1994). Lee and Bass (2004) and Lee and Bass (2006) 200 ms in duration, may be produced singly or repetitively
later showed that small type I males also exhibit sneaklike at intervals of about 400 ms (‘grunt trains’) during agonistic
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 51

interactions (Figure 1(c); Brantley and Bass, 1994; McIver 2004b). 11-Ketotestosterone is a nonaromatizable (i.e., cannot
et al., 2014). The fundamental frequency (F0) of the multi- be converted to an estrogen by aromatase) form of testosterone
harmonic hum and the pulse repetition rate (PRR) of the found among teleost fishes. Plasma 11-ketotestosterone levels
grunt are temperature-dependent, but hover close to 90– are several times greater than testosterone in reproductively
100 Hz in the midshipman’s natural habitat (F0 is equivalent active type I males, but essentially nondetectable in type II
to the PRR of hums) (Brantley and Bass, 1994; McIver et al., males and females (Brantley et al., 1993b; Knapp et al.,
2014). Type I males also produce growls with physical attri- 1999b; Sisneros et al., 2004b). Within type I males, hormone
butes that are an amalgam between grunts and hums levels are correlated with courtship activity as actively
(Figure 1(c); McIver et al., 2014). Grunts do not evoke posi- humming type I males have higher 11-ketotestosterone levels
tive phonotaxis (McKibben and Bass, 1998). Neither type II than nonhumming males (Genova et al., 2012). Regardless
males nor females are known to either growl or hum, of sex, the levels of predominant steroids tend to peak in the
although they do generate low-amplitude, isolated grunts prenesting and early nesting season (Sisneros et al., 2004b).
in nonnesting contexts (Figure 1(c); Brantley and Bass, The decline in 11-ketotestosterone levels over the breeding
1994). season in nesting type I males also corresponds to increased
investment in parental care of fertilized eggs and larvae (Knapp
et al., 1999b). The circulating steroid profiles of type I male and
2.04.2.2 Somatic, Gonadal, and Endocrine Traits
female midshipman resemble those in closely related toad-
Like other species exhibiting multiple reproductive phenotypes fishes that exhibit two male morphs (Modesto and Canário,
(see Section 2.04.1; Bass and Grober, 2009), type I and type II 2003) and, more generally, teleosts with ARTs (Brantley et al.,
male midshipman can be distinguished by a suite of gonadal, 1993b; Oliveira, 2006; Knapp and Neff, 2007). In summary,
somatic, and endocrine characters (Bass, 1996). The two male seasonal profiles of circulating steroids diverge between sexes
morphs show a dramatic dimorphism in body, gonad and and within male morphs, with type II males aligning with
vocal muscle size (Bass and Marchaterre, 1989a,b). On average, females instead of type I males.
type II males are about 50% smaller in body size than type I
males. The GSI is close to ninefold greater in type II males
2.04.2.3 Central Vocal Network
who may invest close to 20% of their body mass in testes
(Figure 1(d)). Gravid females with mature eggs have a large We first present an overview of the organization of the vocal
GSI like that of type II males, although the female GSI is greater motor network in the brain of midshipman fish and, more
(Figure 1(d)). generally, in toadfishes. This provides the framework for
Midshipman, such as other toadfishes, generate sound by a more complete review in Section 2.04.4 of the hormonal
contracting a pair of superfast muscles attached to the wall of modulation of the neurophysiological mechanisms of vocali-
the swim bladder (Figure 1(d); Cohen and Winn, 1967; zation. The vocal network of midshipman also provides an
Rome, 2006). The vocal muscles are more massive in type I exemplar of how a suite of neurobiological characters that
males, due in part to dramatic dimorphisms ranging from lead to the performance of a social communication behavior,
muscle fiber number to the dimensions of muscle fibers, sarco- in this case vocalization, can vary between reproductive
meres, and Z-lines (zone of overlap between thin, actin morphs.
filaments of adjacent sarcomeres), to the number of
mitochondria, all of which are greater in type I males 2.04.2.3.1 Central Vocal Network, Structure and Function
compared to type II males and females that are similar to each The initial demonstration that low molecular weight biotin
other (Bass and Marchaterre, 1989a; Brantley et al., 1993a). compounds (biocytin and neurobiotin) could be transneuro-
Both male morphs and females have olive-gray, dorsal body nally transported across multiple synapses in midshipman
coloration. The ventral body coloration of type I males in fish allowed the visualization of the entire complement of
reproductive condition is light to dark gray, while that of type vocal hindbrain neurons (Figure 2(a); Bass et al., 1994). Subse-
II males is mottled yellow, and of gravid females is bronze to quent studies revealed forebrain and midbrain vocal sites that
golden (Table 1). Females that have released their mature eggs together with more focal studies of hindbrain sites delineated
are more like type II males in their ventral coloration. a central vocal network that was highly interconnected with
The two male midshipman morphs diverge from each both the auditory and neuroendocrine systems (e.g., Bass
other, and from females, in plasma levels of androgens (testos- et al., 2000; Goodson and Bass, 2002; Kittelberger, 2006;
terone and 11-ketotestosterone), estrogens (17b-estradiol), and Chagnaud et al., 2011; Kittelberger and Bass, 2013; Chagnaud
cortisol that is the main glucocorticoid present in teleost fish and Bass, 2013). Comparable patterns of connectivity were
(Table 1; see Bury and Sturm, 2007 for review of steroid observed in closely related toadfish (Chagnaud and Bass,
biosynthesis in teleosts). Type II males have two- to threefold 2014) and in more distantly related species of vocal teleosts
higher cortisol levels compared to type I males (Arterbery (Bass et al., 2015).
et al., 2010a). Circulating testosterone levels are detectable in Intracellular recording and staining identified the func-
both male and females, though found to be the highest in tional properties of a vocal CPG in the caudal hindbrain
type II males and the lowest in type I males, while intermediate and the rostral spinal cord that included three anatomically
in females (Brantley et al., 1993b; Sisneros et al., 2004b). and physiologically distinct neuronal populations: vocal
17b-Estradiol is the predominant circulating steroid in females, motor nucleus (VMN), vocal pacemaker nucleus (VPN),
although some males also show detectable but much lower and vocal prepacemaker nucleus (VPP) (Figure 2(a)–2(c);
levels than testosterone (Brantley et al., 1993b; Sisneros et al., Bass and Baker, 1990, 1991; Chagnaud et al., 2011, 2012;
52 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

(a) (d) Natural call


VPP

VPP Vocal nerve fictive call

VPN

.1mV
1s

VMN VMN

vTr .1mV
25 ms
VPN Frequency
(b)
VOC duration
Spinal cord Hindbrain Midbrain
VMB Forebrain
VMN
VPN VPP POA
VN

(c)

Figure 2 Hindbrain central pattern generator (CPG) for vocalization in the plainfin midshipman fish, Porichthys notatus. (a) Applying biocytin to
the cut end of the nerve that innervates a single vocal muscle at the level of the swim bladder leads to bilateral, transneuronal labeling of the vocal
CPG that includes three topographically separate populations of neurons. Vocal muscles are innervated by a vocal motor nucleus (VMN) whose
axons make up the vocal tract (vTr) and vocal nerve (VN). The VMN is innervated by an adjacent column of vocal pacemaker nucleus (VPN) that
receives input from a more rostral vocal prepacemaker nucleus (VPP). Bar scale represents 200 mm in A and B (25 mm in the insets). (b) The
vocal CPG, mapped onto a lateral view of a midshipman brain, receives input from a vocal midbrain region (VMB) that is driven, in turn, by the
forebrain’s preoptic area (POA) and anterior hypothalamus. (c) Shown here are representative intracellular records from vocal motor, pacemaker,
and prepacemaker neurons (see text for more complete description). Vocal prepacemaker neurons project to the auditory system, thereby
providing a vocal corollary discharge that relays information from the vocal CPG to the auditory system about the temporal properties of natural
vocalizations. A descending forebrain–midbrain pathway drives the vocal CPG. (d) Spontaneous neurophysiological responses from the VN,
a fictive call, are aligned with a naturally occurring ‘grunt train,’ highlighting the correspondence between the temporal properties of the VN motor
volley and natural vocalizations. The lower trace is an expansion of a single vocal nerve response (VOC) that corresponds to a single grunt;
duration is the time between first and last pulse and frequency is the pulse repetition rate. Timescale and amplitude for all records are indicated.
Modified from Chagnaud, B.P., Baker, R., Bass, A.H., 2011. Vocalization frequency and duration are coded in separate hindbrain nuclei. Nat.
Commun. 2, 346–11; Chagnaud, B.P., Zee, M.C., Baker, R., Bass, A.H., 2012. Innovations in motoneuron synchrony drive rapid temporal modula-
tions in vertebrate acoustic signaling. J. Neurophysiol. 107, 3528–3542.
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 53

Chagnaud and Bass, 2013, 2014). Bilateral and contiguous and Baker, 1990; Bass et al., 1996), axon diameters (Bass and
VMNs lie along the midline, extending from the caudal hind- Andersen, 1991), and neuromuscular junctions (Fluet and
brain into the rostral spinal cord (Figure 2(a)–2(c)). Each Bass, 1990) than type II males and females. The volume of the
VMN gives rise to axons, coalescing into a vocal tract that VMN follows a similar pattern (Bass et al., 1996). Unlike VMN
exits the brain via the paired occipital nerve roots to form dimensions, there is no significant sexual dimorphism in the
a vocal nerve that innervates the ipsilateral vocal muscle number of vocal motoneurons as determined by the number
(Figure 2(a)–2(c)). The VPN neurons form a column that of myelinated axons in the vocal nerve, although type II males
extends along the ventrolateral border of each VMN have fewer myelinated fibers compared to type I males and
(Figure 2(b) and 2(c)). Individual pacemaker neurons bilater- females (Bass and Andersen, 1991). Because type I males have
ally innervate both VMNs. The more rostral VPP (originally three- to fivefold more muscle fibers, but a similar number of
designated the ventral medullary nucleus (Bass et al., 1994)) motoneurons compared to females and type IIs, it is predicted
innervates the paired VPN columns (Figure 2(a)–2(c)). that type I males have larger motor units, i.e., each motoneuron
Each neuronal compartment of the vocal CPG (VMN, VPN, innervates a larger number of muscle fibers (Bass and Baker,
VPP) has distinct neurophysiological attributes (Figure 2(c)). 1990; Bass and Andersen, 1991). Along with peripheral vocal
The VMN is the last node that relays the output of the CPG muscle morphology (see above), these central traits likely
directly to vocal muscles. The highly synchronous output of contribute to the inter- and intrasexual differences in the vocal
each VMN generates a vocal motor volley that is readily repertoire (Brantley and Bass, 1994; McIver et al., 2014).
recorded, intracranially, from each vocal nerve root as it exits Developmental studies of the vocal motor system support
the brain (Figure 2(c) and 2(d)). Bilaterally, the vocal motor the hypothesis that the two male reproductive morphs in the
volleys fire in phase, leading to the simultaneous contraction plainfin midshipman fish adopt nonsequential, mutually
of both vocal muscles (Cohen and Winn, 1967). VMN moto- exclusive growth trajectories during their first year of life
neurons fire action potentials with a highly stereotyped repeti- (Bass, 1996; Bass et al., 1996; Knapp et al., 1999a). Sexual
tion rate/frequency that determines muscle contraction rate in maturation of the type I male’s vocal CPG parallels that of
a 1:1 manner (Figure 2(d)). Furthermore, the differential size- the vocal muscle (Brantley et al., 1993a; Bass et al., 1996).
dependent recruitment of VMN neurons determines call ampli- Type II males and females retain a juvenile phenotype, whereas
tude (Chagnaud et al., 2012). a subset of juvenile males exhibits a growth trajectory that leads
Afferent input to the VMN arises from VPNs that generate to the type I male vocal CPG phenotype. Both type II male and
pacemaker-like (hence their name) action potentials that are female midshipman reach sexual maturity at an earlier age and
matched 1:1 in frequency with the highly synchronous output smaller body size than type I males (Bass et al., 1996).
of the VMN (Chagnaud et al., 2011). Finally, upstream to the Together, the results indicate a life history trade-off between
VPN, the VPP generates sustained depolarizations whose dura- investment in larger body size and an expansive vocal motor
tion correlates with the duration of VPN-VMN activity system by type I males versus earlier reproduction and gonad
(Chagnaud et al., 2011). In summary, VMN, VPN, and VPP investment by type II males and females. The adoption of
separately code for distinct properties of natural vocalizations, one developmental trajectory versus the other involves trade-
namely amplitude (VMN), PRR that sets the fundamental offs between the age and size at sexual maturity and a suite
frequency (VPN), and duration (VPP) (Figure 2(c); Bass and of secondary sex characters. Though both male morphs share
Baker, 1990; Chagnaud et al., 2011, 2012; Chagnaud and Bass, gonadal sex, they are highly divergent in a suite of other char-
2014). acters that the type II male morph shares with females, reflect-
In the sections below, we refer to the neurophysiologically ing a common pattern of developmental trade-offs (Table 1).
recorded vocal motor volley (Figure 2(d)) as a fictive call because
it is a patterned motor command that directly determines the
properties of a natural behavior (see Marder and Calabrese, 2.04.3 Hormones, Vocal Behavior, and Vocal
1996), in this case vocalization. The ability to quantify changes Central Pattern Generator
in fictive calls induced by pharmacological perturbations has
allowed us to elucidate causal roles of hormones and neuromo- Given that vertebrate vocalizations reflect an interaction between
dulators in the patterning of vocalization (Section 2.04.3). internal states and external social/reproductive contexts, it is not
surprising that steroid hormones, known to regulate other
2.04.2.3.2 Intra- and Intersexual Divergence in Vocal Network aspects of reproductive behavior and physiology, also directly
As summarized in Section 2.04.2.1, type I male midshipman fish control vocalizations by acting upon central vocal control path-
have a more extensive vocal repertoire than type II males and ways (e.g., Remage-Healey and Bass, 2006; Bass and Remage-
females, which are similar to each other. The divergence in vocal Healey, 2008). Aside from early qualitative reports of testoster-
behavior is paralleled by a divergence in the anatomy of both the one’s potentiating effects on sound production in the satinfin
sonic muscles (Section 2.04.2.2, Figure 1(d)) and of the vocal shiner, Cyprinella analostana (formerly Notropis analostanus)
network. Centrally, inter- and intrasexual polymorphisms in (Winn and Stout, 1960), steroid regulation of fish vocalization
midshipman fish range from the dimensions of motoneurons has not been well studied outside of the toadfish family. In the
(VMN) and premotoneurons (VPN, VPP) to hormone-specific last decade, comprehensive studies in toadfishes have established
influences on the output of the vocal network (see Section rapid and robust steroid effects on vocal behavior and steroid
2.04.3; Table 1). Specifically, inter- and intrasexual dimorphisms sensitivity of the underlying central vocal motor network
are found in the size of vocal motoneurons, such that type I (Remage-Healey and Bass, 2004, 2005, 2006, 2007). Further-
males have larger somata (Bass and Marchaterre, 1989b; Bass more, inter- and intrasexual divergence in circulating hormone
54 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

profiles (see Section 2.04.2.2) are paralleled by sex and male toward levels seen in adult type I males (Brantley et al., 1993a).
morph patterns of neural sensitivity to specific steroid hormones Although circulating steroid levels were not measured by
(Brantley et al., 1993b; Sisneros et al., 2004b; Remage-Healey Brantley et al. (1993a), a later study that induced 11-KT levels
and Bass, 2007; Arterbery et al., 2010a; Fergus and Bass, 2013). in type II males to physiological levels found in type I males
Steroid action is mediated by ligands binding with specific also showed an expansion of the vocal muscle (Lee and Bass,
intracellular cytosolic receptors, forming a steroid–receptor 2005).
complex that translocates into the nucleus and targets Long-term (8–9 weeks) testosterone-propionate treatment
steroid-specific DNA response elements to regulate gene expres- also enlarged the soma size of VPP, VPN, and VMN neurons
sion (Lösel and Wehling, 2003). This mode of action initiates in juvenile males compared to size-matched controls (Bass,
a genomic response that could take 30 min to days. Steroids 1995; Bass and Forlano, 2008). Neurophysiologically, this
can also exert rapid, nongenomic effects that occur on the order treatment increased the PRR of the vocal motor volley, or fictive
of seconds to less than 30 min. Rapid steroid action may calls (Section 2.04.2.3.1), evoked from juvenile males (Bass,
involve activation of nonclassical membrane receptors in addi- 1995). In adult type II males, this treatment had no effects
tion to classical intracellular receptors that lead to changes in on VMN soma size (Bass and Forlano, 2008). Furthermore,
secondary messenger pathways (Lösel and Wehling, 2003). In increasing 11-KT plasma levels in type II males to those of
toadfish, both long-term and rapid modes of steroid action type I males did not induce an increase in overall VMN volume,
have been observed, including seasonal changes in reproduc- nor the expression of type I courtship behaviors (Lee and Bass,
tive states and rapid changes in context- and stimulus-driven 2005). Instead, exogenous 11-KT intensified type II cuckholdry
vocalization (see Table 1 for summary). These steroid effects behaviors (Lee and Bass, 2005). Thus, after committing to the
on vocal behavior are likely mediated by steroid receptors type II trajectory, the vocal CPG is no longer sensitive to the
expressed throughout the vocal control network that patterns masculinizing effects of androgens leading to a type I male
vocalizations (see Section 2.04.5). phenotype, suggesting that male morph differences in
midshipman are organized during an earlier critical period.
Finally, the rise in androgen levels at the beginning of the
2.04.3.1 Divergent Steroid Hormone Profiles and Action
breeding season likely activates and prepares the central and
between Sex and Male Morphs
peripheral nervous systems structurally and metabolically for
It is well established that steroid hormones act over a dynamic acoustic courtship activity, especially in type I males.
range of timescales. During developmental critical periods
among mammals (e.g., perinatal and puberty), steroids exert 2.04.3.1.2 Rapid Steroid Action
long-lasting organizational effects on peripheral and central In the midshipman and the closely related Gulf toadfish (Opsa-
tissues that lay down the foundation for sex differences in nus beta), field and in vivo neurophysiology studies show that
adulthood (see review by Arnold, 2009). In adulthood, steroids steroids act rapidly on the order of minutes to modulate vocal
exert reversible activational effects that control sexually dimor- behavior and vocal CPG output (Table 1). Steroid hormones
phic phenotypes (Arnold, 2009). In this section, we review the are likely acting via specific receptors within neuroendocrine
current knowledge of steroid hormone regulation of toadfish and vocal pathways (Figure 3(a)). The neuroanatomical loca-
vocalization at vocal motor and behavioral levels over multiple tion of steroid receptors is reviewed in more detail below in
timescales. Because this topic has been recently extensively Section 2.04.5. Rapid steroid modulation of the vocal CPG
reviewed (Bass et al., 2015), we focus discussions here on sex was first demonstrated by in vivo neurophysiological investiga-
and morph differences. tions in type I male midshipman (Remage-Healey and Bass,
2004) and subsequently in male and female Gulf toadfish
2.04.3.1.1 Long-Term and Seasonal Steroid Action (Remage-Healey and Bass, 2006) as well as type II male and
A prominent effect of steroid action over the developmental and female midshipman (Remage-Healey and Bass, 2007).
seasonal timescale is the growth of primary and secondary sexual Steroid action in the vocal neural network in midshipman is
characteristics. In the context of midshipman courtship vocal both sex- and male morph-specific (Figure 3(b); Remage-
behavior, 11-ketotestosterone (11-KT) has been shown to act Healey and Bass, 2007). In both type I male midshipman
peripherally to increase the size of the vocal muscle and and male Gulf toadfish, which is only known to have one
testosterone-propionate centrally to increase the size of vocal male morph, systemic injections of 11-KT, but not testosterone,
motoneurons (Brantley et al., 1993a; Bass, 1995; Bass and increased the duration of electrically evoked fictive calls within
Forlano, 2008). As mentioned above, the marked sexual and 5 min (Figure 3(b)) (Remage-Healey and Bass, 2004, 2006). In
morph dimorphism in vocal muscle mass parallels their respec- contrast, systemic injections of testosterone, but not 11-KT,
tive vocal repertoires: type I males who produce long duration rapidly increased fictive call duration in type II male and female
courtship hums have sixfold larger vocal muscle mass (normal- midshipman (Figure 3(b)) (Remage-Healey and Bass, 2007).
ized to body weight) when compared to type II males or females These results are consistent with the divergent profiles of
who can only produce isolated grunts (Figure 1(d); Brantley and predominant circulating androgens found between the male
Bass, 1994). The vocal muscle hypertrophy in type I males is morphs and sexes of midshipman (see Section 2.04.2.2).
androgen-dependent (Brantley et al., 1993a). Long-term Although testosterone potentiated vocal CPG output in both
(8–9 weeks) treatment with androgen implants, including type II male and females, its aromatization to 17b-estradiol
11-KT and testosterone-propionate, increased the vocal muscle was necessary for potentiating the female vocal CPG, while
weight and myofibril and mitochondrial-filled sarcoplasm ultra- androgen receptor activation was necessary for potentiation
structure in juvenile type I males, type II males, and females, in type II males (Remage-Healey and Bass, 2007).
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 55

(a) Steroid receptor and aromatase distribution within vocal network

Cerebellum Midbrain
Forebrain
Hindbrain 1,2,4,5
Spinal cord PAG
1-5 OB
VMN PL 1-5
1 POA On
VPN VPP 1,5
1,2,4,5 AT Opn
vT
Vocal nerve 1 mm
POA-AH
Pituitary
α 3:ERβ1 4:ERβ2 5:ARO
1:AR 2:ERα

(b) Rapid steroid effects on vocal physiology


Type I male
200 11-KT 200 T
Type II male
180 180
Female
160 160
140 140
120 120
100 100
80 80
% Baseline duration

60 60
40 40
0 15 30 45 60 75 90 105120 0 15 30 45 60 75 90 105120

200 Cortisol 200 Estradiol


180 180
160 160
140 140
120 120
100 100
80 80
60 60
40 40
0 15 30 45 60 75 90 105120 0 15 30 45 60 75 90 105120
Time (min)

Figure 3 Rapid steroid action in the vocal network reveals sex and morph differences. (a) Neuroanatomical distribution of steroid receptors and
aromatase (estrogen synthase). Abbreviations: AR, androgen receptor; ER, estrogen receptor; ARO, aromatase; OB, olfactory bulb; On, olfactory nerve;
Opn, optic nerve; POA-AH, preoptic area–anterior hypothalamus; vT, ventral tuberal nucleus; AT, anterior tuberal nucleus; PL, paralemniscal nucleus;
PAG, periaqueductal gray; VPP, vocal prepacemaker nucleus; VPN, vocal pacemaker nucleus; VMN, vocal motor nucleus. (b) Summary of results from
in vivo neurophysiology studies demonstrating rapid (within 5 min) effects of peripheral steroid injection on the duration of electrically evoked fictive
calls (% change with respect to baseline). Plotted are means for each time point taken from Remage-Healey and Bass (2004, 2007). When multiple
doses were used, the means from the most effective dose were plotted. Abbreviations: 11-KT, 11-ketotestosterone; T, testosterone. Modified from
Bass, A.H., Chagnaud, B.P., Feng, N.Y., 2015. Comparative neurobiology of sound production in fishes. In: Ladich, F. (Ed.), Sound Communication in
Fishes, Animal Signals and Communication. Springer Vienna, Vienna, pp. 35–75, with kind permission from Springer Science þ Business Media.

The rapid effects of cortisol in midshipman also diverged cortisol on fictive call duration did not last more than
across phenotypic sex, as cortisol injections increased fictive 30 min, compared to 2 h for 11-KT and 45 min for cortisol in
call duration in type I males but decreased duration in type II intact preparations (Remage-Healey and Bass, 2004, 2006).
males and females (Figure 3(b); Remage-Healey and Bass, Thus, the results suggest that steroid action within upstream
2007). However, no sex differences were observed in Gulf toad- vocal–acoustic centers mediates sustained stimulation of vocal
fish, as cortisol induced a rapid increase in fictive call duration behavior, while steroid action within the hindbrain alone is
in both males and females (Remage-Healey and Bass, 2006). sufficient to elicit short-term increases in vocalization
Interestingly, when the hindbrain vocal CPG was isolated (Remage-Healey and Bass, 2004).
from descending forebrain and midbrain input in type I male Unlike 11-KT, testosterone, and cortisol, the rapid effects of
midshipman and male Gulf toadfish, the effects of 11-KT and 17b-estradiol did not differ dramatically between the male
56 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

morphs or sexes (Figure 3(b)). In midshipman, 17b-estradiol family of peptides (Bentley, 1998) that modulates a range of
increased fictive call duration in type I males, type II males, vertebrate social behaviors (reviewed in Goodson and
and females (Remage-Healey and Bass, 2004, 2007). Thus, Bass, 2001; Godwin and Thompson, 2012; Goodson, 2013).
regardless of within-male comparisons, type II males always In teleosts, AVT (homologue to mammalian vasopressin)
aligned with females in their neurophysiological responses to and isotocin (homologue to mammalian oxytocin) are
steroids, including 17b-estradiol (Figure 3(b)). In the Gulf produced exclusively within the POA (Figure 4(a); see Section
toadfish, 17b-estradiol had no effect in either males or females 2.04.5.1 for anatomical distribution) (Goodson and Bass,
(Remage-Healey and Bass, 2006), revealing an unexpected 2001; Goodson, 2008). Although nonapeptide regulation of
divergence in hormone sensitivity between closely related toad- vocal behavior is investigated in far less detail compared to
fish species. In summary, in vivo neurophysiology studies have steroids, complementary in vivo neurophysiology studies have
uncovered sex, morph, and species differences of rapid steroid demonstrated sex and male morph-specific actions within
action within the vocal CPG. the midshipman vocal network (Goodson and Bass,
Rapid steroid effects on natural vocal behavior have also 2000a,b). Furthermore, in addition to fish (Goodson and
been investigated in the Gulf toadfish by field experiments, Bass, 2000a,b), nonapeptide modulation of vocalization is
which largely support in vivo neurophysiology results summa- widespread among vertebrates, including amphibians and
rized above (Table 1) (Remage-Healey and Bass, 2005, birds (reviewed in Goodson and Bass, 2001).
2006). First, it was observed that focal nesting males began In type I male midshipman, AVT injection into the region
vocalizing within 48 h of exposure to an actively calling natural of the caudal POA and anterior hypothalamus (AH) induced
population, and this exposure increased plasma 11-KT levels in a dose-dependent inhibition of fictive call duration and the
exposed males compared to males not exposed to calling number of calls elicited from the POA-AH (Figure 4(b);
neighbors (Remage-Healey and Bass, 2005). Furthermore, Goodson and Bass, 2000a). Supporting vocal inhibition by
males not exposed to calling neighbors did not begin to AVT, delivery of an arginine-vasopressin V1a receptor
vocalize. Finally, nesting males exposed to acoustic playbacks antagonist significantly increased call duration (Figure 4(b);
of conspecific calls with durations that approximated the pop- Goodson and Bass, 2000a). Additionally, AVT injection into
ulation mean increased call rate and duration as well as plasma a vocally active midbrain site in the tegmentum inhibited
levels of 11-KT within 20 min. These results suggested that the number of fictive calls elicited from the same area, as
a social stimulation of 11-KT levels potentiates vocal behavior. well as increased the latency of fictive call initiation, but did
A causal relationship between 11-KT levels and expression not affect call duration (Goodson and Bass, 2000b). These
of natural vocal behaviors was established by a follow-up study results show that AVT inhibits vocal network excitability at
of Gulf toadfish showing that exogenous delivery of food con- both forebrain and midbrain levels in type I males. In
taining 11-KT increased calling rate in nesting males within contrast to AVT, isotocin was ineffective in changing vocal
10–20 min (Remage-Healey and Bass, 2006). Interestingly, output parameters in type I males, suggesting that despite
there was no change in call duration, suggesting an influence the high conservation in amino acid sequence and synthesis
of sound playbacks and the sense of hearing on the results of within the POA, AVT and isotocin bind to receptors with
the earlier study showing increases in call duration as well high specificity.
(Remage-Healey and Bass, 2005). Together, these field experi- In type II males and females, however, AVT had no effects
ments support the hypothesis that 11-KT rapidly facilitates on the output of the vocal network (Figure 4(b); Goodson
male vocal behavior, likely by acting via classical intracellular and Bass, 2000b). Instead, isotocin injection in the POA-
receptors, which was supported by receptor-specific antagonist AH of type II males and females exerted dose-dependent
treatments during fictive calling (Remage-Healey and Bass, inhibition of vocal output, while an oxytocin receptor
2004). 11-KT presumably acts within the vocal network antagonist facilitated vocal output (Figure 4(b)). These
expressing androgen receptors (see Figure 3(a); Forlano et al., results suggest that in species with ARTs such as the
2010), though the effects of focal steroid injections directly midshipman, vasotocin-like peptides are dissociated from
into vocal nuclei remain to be investigated. gonadal sex and align instead with phenotypic sex. More
In summary, complementary field behavior and laboratory broadly, these results along with studies in other
neurophysiological studies in toadfishes demonstrate steroid vertebrates show that nonapeptides belonging to the
regulation of dynamic social behaviors that fluctuate on the vasotocin family are evolutionarily labile in their sex-,
minute-by-minute timescale. Neurophysiology revealed sex, morph-, and species-dependent actions on reproductive
male morph, and species-specific steroid action in the vocal and courtship behaviors (Goodson, 2008; Bass and Grober,
network (Table 1; Figure 3(b)). The accessibility to monitor 2009).
and manipulate natural vocal behavior as well as the output
of the vocal network makes toadfishes excellent models for
studying hormone regulation of the neural substrates of a verte- 2.04.4 Hormonal Modulation of Hearing
brate behavior.
Sound-producing teleost fishes provide important model
systems for investigating peripheral and central mechanisms
2.04.3.2 Sex- and Male Morph-Specific Patterns
encoding acoustic stimuli (Bass and McKibben, 2003; Edds-
of Nonapeptide Actions on Vocal Neurophysiology
Walton and Fay, 2008). Most studies of hormonal influ-
AVT and isotocin are nine amino acid peptides, known as non- ences on audition in fishes have focused on plasticity at
apeptides, that belong to the arginine vasopressin-oxytocin the level of the saccule, the main auditory end organ of
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 57

(a)
Cerebellum Midbrain
Forebrain
Spinal cord Hindbrain
PAG OB
VMN PL
VPN POA On
VPP
AT Opn
vT
Vocal nerve 1 mm
POA-AH
Pituitary
(b)
Summed fictive call duration
150 Type I males
(% baseline)
100

50

0
1,000ng 0ng 0.1ng 10ng 1,000ng 1,000ng
Anti-AVP Control AVT IT

325
Females
Summed fictive call duration

300

100
(% baseline)

50

0
100ng 0ng 0.1ng 10ng 1,000ng 1,000ng
Anti-OXY Control IT AVT

300 Type II males


Summed fictive call duration
(% baseline)

100

50

0
100ng 0ng 0.1ng 10ng 1,000ng 1,000ng
Anti-OXY Control IT AVT

Figure 4 Arginine vasotocin neural expression and regulation of vocal central pattern generator output. (a) Simplified representation of arginine–
vasotocin (AVT) pathways (red arrows) mapped onto a sagittal view of the plainfin midshipman fish vocal network (black arrows) based on immuno-
cytochemistry (Goodson and Bass, 2000b). Arrow thickness denotes projection density. AVT-immunoreactive (AVT-ir) pathways originate almost
exclusively from the preoptic area (POA) and innervate vocally active brain sites in the POA, anterior tuberal nucleus (AT), ventral tuberal
hypothalamus (vT), paralemniscal midbrain tegmentum (PL), and midbrain periaqueductal gray (PAG). Other abbreviations: OB, olfactory bulb; On,
olfactory nerve; Opn, optic nerve; POA-AH, preoptic area–anterior hypothalamus; VMN, vocal motor nucleus; VPN, vocal pacemaker nucleus; VPP,
vocal prepacemaker nucleus. (b) Vocal motor output of the midshipman in response to POA-AH stimulation 10 min after delivery of AVT, isotocin
(IT), or their respective mammalian antagonists (Anti-AVP; Anti-OXY). Top panel, type I males; middle panel, females; bottom panel, type II males.
Total fictive call duration of type I males is inhibited in a dose-dependent manner by AVT, disinhibited by anti-AVP, and not affected by IT. By
contrast, fictive calls of females and type II males are not affected by AVT, but inhibited by IT and disinhibited by anti-OXY. From Bass, A.H.,
Chagnaud, B.P., Feng, N.Y., 2015. Comparative neurobiology of sound production in fishes. In: Ladich, F. (Ed.), Sound Communication in Fishes,
Animal Signals and Communication. Springer Vienna, Vienna, pp. 35–75, with kind permission from Springer Science þ Business Media.

the inner ear in toadfishes and many other teleost species distinct branches of the eighth (VIII) cranial nerve. We first
(Cohen and Winn, 1967; Bass and McKibben, 2003; discuss patterns of seasonal plasticity that provided the
Edds-Walton and Fay, 2008). The saccule’s hair cell epithe- foundation for subsequent studies looking at hormonal
lium is innervated by ganglion cell afferents that form influences. A recent and more detailed review of hormonal
58 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

mechanisms and hearing in fishes is available elsewhere Adopting methods first used in goldfish to record a saccular
(Forlano et al., 2016). evoked potential (Furukawa and Ishii, 1967), the analog of
a cochlear microphonic, Sisneros (2009) showed that the
seasonal changes in frequency encoding in the midshipman
2.04.4.1 Seasonal Auditory Plasticity
female saccule could be explained by plasticity at the level of
Sisneros and Bass (2003) provided the first example of natu- the hair cell epithelium (Sisneros, 2009). Subsequent studies
rally occurring, seasonal changes in peripheral frequency sensi- showed the same seasonal phenotype for type I males, with
tivity in a teleost fish, in this case by saccular, auditory afferents pilot studies supporting this pattern for type II males
in female midshipman fish (Figure 5(a); Sisneros and Bass, (Sisneros, 2009; Whitchurch and Sisneros, 2011; Rohmann
2003). The saccular afferents of female midshipman that are and Bass, 2011). Thus, seasonal changes in peripheral hearing
in nonreproductive condition show maximal frequency sensi- sensitivity show neither a sex- nor male morph-specific
tivity in the range of approximately 60–100 Hz, whereas pattern. Interestingly, seasonal shifts in peripheral frequency
females in reproductive condition during the breeding season sensitivity may not be shared with other toadfish species, at
show peak sensitivity extending from approximately 60 to least based on a study of the Lusitanian toadfish that has
350 Hz. This broader range of sensitivity encompasses the ARTs (Section 2.04.2.1) but does not show the seasonal migra-
fundamental frequency and upper harmonics of male adver- tion to deeper water habitats observed for midshipman
tisement calls that can have as much and sometimes even (Vasconcelos et al., 2011).
more energy than the fundamental (Figure 5(a)). From More recent studies have investigated the molecular basis
a neurophysiological perspective, the higher harmonics might for peripheral auditory plasticity. The frequency tuning of
improve the neurophysiological encoding of a call’s funda- hair cells in reptiles and birds can be explained by electrical
mental frequency (McKibben and Bass, 1999). From a behav- resonance, with the abundance of large conductance,
ioral perspective, one advantage of enhanced detection of the calcium-activated potassium (BK) channels increasing as reso-
higher harmonics for all members of the species would be an nant frequency increases (Fettiplace and Fuchs, 1999). Saccular
improvement in the ability to detect the calls of nesting type hair cells in teleosts also exhibit electrical resonance (Steinacker
I males from a greater distance since the upper harmonics and Romero, 1992; Sugihara and Furukawa, 1995). Rohmann
would propagate farther in the shallow water environment et al. (2013) showed that seasonal changes in saccular encod-
where midshipman spawn (Fine and Lenhardt, 1983; Bass ing in midshipman could largely be explained by changes in
and Clark, 2003). BK mRNA abundance (Rohmann et al., 2013). This study

(a) (b) Apical


Control Summer
Estradiol Testosterone
1.0 F1 0
F0
F2 -20
0.8
Power spectrum (dB/Hz)

-40

-60
0.6
VS

-80
Apical
0.4 -100

-120
0.2
-140

0.0 -160
0 100 200 300 400 500 600 700 800
Frequency (Hz)

Figure 5 Neurophysiological and anatomical evidence for steroid- and season-dependent auditory encoding of higher call frequencies. (a) The
power spectrum of frequency components of the multiharmonic hum is plotted in gray (right y-axis). The phase-locking precision of saccular affer-
ents to different frequencies is plotted as a function of the vector strength of synchronization (VS, left y-axis). In summer breeding females and
winter nonbreeding females implanted with estradiol or testosterone, the saccular afferents are better able to encode the upper harmonics of the hum
(F1 ¼ 200 Hz; F2 ¼ 300 Hz) that contain as much or more power than the fundamental frequency (F0 ¼ 100 Hz). (Adapted from Sisneros, J.A.,
Forlano, P.A., Deitcher, D.A., Bass, A.H., 2004a. Steroid-dependent auditory plasticity leads to adaptive coupling of sender and receiver. Science 305,
404–407.) (b) Localization of estrogen receptor (ER) hair cells of the saccular epithelium, ERb1 (green in top panel) is at apical end of hair cells
(white arrows), while ERb2 (magenta in bottom panel) is distributed throughout the body of the hair cells with varied levels of expression along the
epithelium. Hair cells are labeled with a hair cell-specific antibody (magenta in top panel; green in bottom panel). Scale bar represents 20 mm.
Adapted from Fergus, D.J., Bass, A.H., 2013. Localization and divergent profiles of estrogen receptors and aromatase in the vocal and auditory
networks of a fish with alternative mating tactics. J. Comp. Neurol. 521, 2850–2869.
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 59

also showed that BK channels were localized to the somata of including humans (e.g., Chong and Liu, 2016; Frisina and
saccular hair cells and were not detectable in other cells within Frisina, 2016). Peripheral action of estradiol is likely mediated
the epithelium or in nearby auditory ganglion cells. Together, by ERs located in auditory hair cells in midshipman (see
the results strongly support the proposal that seasonal changes above), as well as in birds and mammals (Hultcrantz et al.,
in peripheral frequency encoding depend on BK channels in 2006; Noirot et al., 2009). There has been extensive neurophys-
saccular hair cells (Rohmann et al., 2013). Morphological iological study in midshipman and other teleost fishes of the
changes complementing these physiological and molecular main auditory nucleus in the midbrain (Bass and McKibben,
changes in saccular hair cell function include seasonal increases 2003; Edds-Walton and Fay, 2008) that expresses steroid recep-
in the numbers of saccular hair cells in reproductive compared tors (Forlano et al., 2010; Fergus and Bass, 2013). However, the
to nonreproductive females (Coffin et al., 2012). potential for steroid- and/or seasonal-dependent plasticity in
auditory physiology in the midbrain or other central auditory
sites that express steroid receptors (Forlano et al., 2010; Fergus
2.04.4.2 Steroid-Dependent Auditory Plasticity
and Bass, 2013) has yet to be investigated as it has been in other
As reviewed in Section 2.04.2.2, midshipman fish, like other vertebrates (see Caras, 2013).
seasonally breeding teleosts, show seasonal changes in
plasma levels of circulating steroid hormones (Brantley
et al., 1993b; Oliveira, 2009). In particular, levels of both 2.04.5 Sites of Hormone Synthesis and Action
17b-estradiol and testosterone begin to peak during the
spring, just before males and females move into the intertidal Studies mapping the distribution and/or abundance of
zone for spawning. Treatment of nonreproductive females neuropeptide-synthesizing neurons along with steroid recep-
with an intraperitoneal implant of either 17b-estradiol or tors provide support for the action of these hormones in vocal
testosterone for 3–4 weeks leads to plasma levels of these and auditory systems. These investigations also provide a more
steroids comparable to those of females in reproductive general roadmap for future studies investigating the neural
condition and induces an improved auditory phenotype basis for the actions of these hormones on reproductive
nearly identical to that of the naturally occurring one of repro- behaviors.
ductive females (Figure 5(a); Sisneros et al., 2004a). The
improvements are gradual, as nonreproductive females with
2.04.5.1 Neuropeptides
testosterone implants for up to 2 weeks still resemble controls
(Sisneros et al., 2004a). Supporting evidence for the periph- The CNS distribution of neurons synthesizing GnRH and AVT
eral effects of estrogen comes from the anatomical demonstra- has been extensively studied in teleost fish. The POA is a major
tion for estrogen receptors (ERs) in the hair cell epithelium site of synthesis for both of these peptides. Prior reviews have
(Figure 5(b); Fergus and Bass, 2013), as well as aromatase extensively considered dimorphisms and polymorphisms in
synthesis by saccular ganglion cells that could provide for the numbers and dimensions of GnRH and AVT neurons across
the local conversion of testosterone to estrogen (Forlano several teleost species exhibiting reproductive plasticity (e.g.,
et al., 2005; Fergus and Bass, 2013). The most recent studies Bass and Grober, 2009). Since the first studies that identified
show two types of ERs, ERb1 and ERb2, localized to saccule polymorphisms in the POA-GnRH phenotype of a sex
hair cells (Fergus and Bass, 2013). Consistent with the neuro- changing fish (Grober et al., 1991; Grober and Bass, 1991),
physiological results showing shared patterns of seasonal comparable investigations of such species have largely
plasticity in frequency sensitivity between male morphs and examined AVT mechanisms (see Godwin and Thompson,
the sexes, quantitative PCR shows no morph- or sex-specific 2012; Almeida and Oliveira, 2015). The reader is referred to
patterns in the abundance of mRNA transcripts for both forms other chapters in this volume for more detailed coverage of
of ERb and aromatase (Fergus and Bass, 2013). ERa, shown in these species and those that exhibit serial changes in social
regions of the epithelium adjacent to hair cells (Forlano et al., status from reproductive to nonreproductive states (Godwin,
2005), also shows similar levels of abundance across morphs 2016; Fernald, 2016). For the purposes of this chapter, more
and the sexes (Fergus and Bass, 2013). detailed remarks are confined to species like midshipman fish
In sum, while the influence of circulating steroids on encod- that exhibit ARTs.
ing mechanisms by the hair cell epithelium of the auditory GnRH containing cells in the POA were an early focus of
saccule remains correlative (see Rohmann and Bass, 2011), studies of reproductive plasticity in fishes because these
the available studies for the plainfin midshipman fish show neurons directly innervate the teleost pituitary (Oka and
that steroid-dependent auditory plasticity leads to seasonal Ichikawa, 1990) and thus can potentially exert rapid changes
improvements in the sense of hearing that inform both mate in gonadotropin release from the anterior pituitary and, in
identification and localization during the breeding season. turn, affect the secretion of gonadal steroids. The first such
The findings of seasonal shifts in the sense of hearing in studies were in a poeciliid, the southern platyfish (Xiphophorus
midshipman fish are complemented by more recent studies maculatus) (Halpern-Sebold et al., 1986). Though ARTs were
in songbirds showing seasonal changes in hearing (e.g., see not described for poeciliids until later (Zimmerer and Kallman,
Caras, 2013; Vélez et al., 2015; and references therein). These 1989; Ryan et al., 1990), the investigators showed that the
studies, together with those showing the influence of steroids number of GnRH neurons in the POA diverged between small
on seasonal peripheral auditory function in midshipman, and large male morphs of platyfish. Later studies documenting
complement studies in other teleosts (e.g., Maruska et al., POA GnRH polymorphisms in the adult reproductive morphs
2012; Zeyl et al., 2013), birds (Gall et al., 2013) and mammals, of midshipman fish, together with studies of several species
60 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

showing sex and role change, provided neuroanatomical testosterone into estrogens (Simpson et al., 1994). Neural aro-
support for the hypothesis that these neurons play a central matase activity can therefore modulate steroid-dependent
role in triggering events that lead to age- and male morph- behaviors by controlling the local ratio of testosterone and
related changes in reproductive status (Grober et al., 1994; estrogens (Bass and Remage-Healey, 2008; Adkins-Regan,
Bass and Grober, 2009). 2013). Importantly, the more potent androgens, such as
Like GnRH neurons in teleosts, AVT-containing neurons are 11-KT in teleosts and 5a-dihydrotestosterone in tetrapods, are
found in the POA and show both intra- and intersexual not aromatizable and cannot be shunted by aromatase from
dimorphisms in size and number in midshipman fish that exerting androgen action (Bentley, 1998; Adkins-Regan, 2013).
are comparable to polymorphisms found in teleosts Compared to other taxa, teleosts exhibit a remarkably high
exhibiting adult sex and role change (Foran and Bass, 1998; level of neural aromatase activity (Callard et al., 1990; Forlano
Bass and Grober, 2009). The actions of AVT on behavior, like et al., 2001, 2006). In the plainfin midshipman, the rate of aro-
the neuroanatomical patterns, appear to be species-specific matase activity, as measured by enzymatic conversion of radio-
(see Godwin and Thompson, 2012; also see discussion in active ligand, was higher in the forebrain compared to the
Santangelo and Bass, 2006). In the midshipman, AVT is midbrain and hindbrain (Schlinger et al., 1999). Furthermore,
expressed in the parvocellular and magnocellular nuclei of hindbrain aromatase activity was higher in females and type II
the POA (Goodson and Bass, 2000b). These AVT-containing males than in type I males (Schlinger et al., 1999). This male
POA neurons project locally to the immediate surrounding morph difference was likely a constitutive property of the
neuropil, as well as throughout diverse regions of the anterior CNS and independent of circulating gonadal testosterone,
hypothalamus and midbrain (Figure 4(a)). Forebrain targets because type II male hindbrain had higher aromatase activity
include the pituitary and hypothalamic regions such as the than type I males even after castration (Schlinger et al.,
lateral hypothalamus and the anterior tuberal nucleus, 1999). During the nesting season, type II males had the highest
a vocal active region that is also a part of the ascending aromatase mRNA expression in the peripheral regions of the
auditory system, which showed a greater number of VMN (Forlano and Bass, 2005a). Sex and morph differences
AVT-positive somata in nonreproductive fish collected during were also observed by quantitative PCR, which showed that
the winter compared to reproductive fish collected during the in hindbrain tissue that had the VMN removed, type II and
summer (Goodson and Bass, 2000b; Goodson et al., 2003). females had significantly higher aromatase expression than
Dense terminal fields are also found in vocally active areas in type I males (Fergus and Bass, 2013). Furthermore, within
the midbrain tegmentum such as the paralemniscal nucleus the isolated VMN, type II males had the highest levels of aroma-
(Goodson and Bass, 2000b; Feng and Bass, 2014). Although tase expression, though this was not statistically significant
little AVT immunoreactivity was found in the hindbrain vocal (Fergus and Bass, 2013). This constitutive high level of hind-
CPG, dense staining was observed in the area postrema brain aromatase activity in females and type II males could
above the VMN (Goodson and Bass, 2000b). shunt testosterone from exerting androgenic and masculiniza-
The distribution of putative isotocin matched closely to that tion effects on the vocal network.
observed for AVT, especially concerning the vocal circuitry Both females and type I males showed seasonal expression
(Goodson et al., 2003). Briefly, isotocin-immunoreactive of aromatase in several brain areas (Forlano and Bass, 2005a).
somata were found in the POA and projected to vocal regions During the prenesting season in early spring, females exhibited
in the hypothalamus and midbrain (Goodson et al., 2003). At higher aromatase mRNA levels in the central and peripheral-
hindbrain levels, isotocin-immunoreactive terminals could be glial containing regions of VMN, the midbrain PAG, and the
found within the vicinity of the vocal CPG, but there was no magnocellular nucleus of the POA that includes the GnRH
evidence of a direct projection within the vocal nuclei and AVT synthesizing neurons described in Section 2.04.5.1
(Goodson et al., 2003). However, isotocin showed expression (type II males were not available for seasonal comparisons)
in many regions associated with auditory processing, such as (Forlano and Bass, 2005a). These brain regions in type I males,
the central posterior nucleus of the thalamus (CP) that is on the other hand, expressed the highest aromatase during the
a major recipient of ascending input from the auditory nesting season (Forlano and Bass, 2005a). A companion study
midbrain, as well as in CP’s telencephalic targets (Bass et al., of nonreproductive females showed that aromatase mRNA
2000; Goodson et al., 2003). Since no sex or morph differences expression was increased to prenesting levels after 20–30 days
were observed in AVT or isotocin distributions, the differential of testosterone or estrogen implants (Forlano and Bass,
effects of these peptides on vocal motor output could be medi- 2005b). Thus, the seasonal changes in steroid levels could
ated by sex- or morph-specific expression of AVT or isotocin modulate neural aromatase expression in a sex-dependent
receptors within individual vocal nuclei, which remain to be manner.
characterized in midshipman. Aromatase distribution within the midshipman vocal
network has been characterized using midshipman-specific
mRNA probes and antibodies (Figure 3(a); Forlano et al.,
2.04.5.2 Aromatase
2001). Aromatase transcript and protein expression were abun-
It has been well established that the brain itself is capable of de dant throughout critical nodes of the vocal network, including
novo steroid synthesis in addition to being sensitive to and the forebrain POA, midbrain PAG, and hindbrain VMN
metabolizing circulating steroids derived from peripheral sour- (Forlano et al., 2001). Double labeling with glial and neuronal
ces (Baulieu and Robel, 1990; Forlano et al., 2006; Saldanha specific markers revealed that aromatase is mostly expressed
and Schlinger, 2008). Aromatase is an important steroidogenic within glial, but not neuronal, cell populations (Forlano
enzyme that converts aromatizable androgens such as et al., 2001). For example, the highest density of aromatase
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 61

was found in telencephalic radial glia that surrounded the midshipman brain, as have been shown in other teleosts
surface of the telencephalon as well as the medial ventricular (Harbott et al., 2007). Furthermore, it remains to be tested
surface (the telencephalon of teleosts has a single central whether divergent sex- or morph-specific AR subtype expres-
ventricle). Furthermore, dense aromatase-containing glial cells sion in vocal nuclei could account for differences in neural
were found in periventricular cell layers in the PAG and androgen sensitivity.
surrounding the dorsolateral margin of VMN and extended Several ERs, including ERa, ERb1, and ERb2, have been
their processes throughout the VMN in a radial-like pattern. sequenced and localized in the midshipman brain
Thus, aromatase is found within every major midbrain–hind- (Figure 3(a); Forlano et al., 2005; Fergus and Bass, 2013).
brain node of the midshipman vocal control network, likely Similar to neuropeptides, AR, and aromatase, protein expres-
providing sources of locally converted estradiol that vary in sion of all three ERs were found in the POA (Forlano et al.,
levels across seasons and reproductive morphs, which was 2001). In the hypothalamus, ERb2 showed robust expression
shown to rapidly increase fictive call duration (Table 1; in the vocally active AT (Goodson and Bass, 2000a; Fergus
Figure 3(b); Remage-Healey and Bass, 2004, 2007). and Bass, 2013). Within midbrain vocal centers, the PAG
expressed both ERb1 and ERb2, though ERb2 showed a broader
distribution (Fergus and Bass, 2013). ERb2 was also expressed
2.04.5.3 Androgen and ERs
in the periventricular cell layer of the main auditory nucleus in
Neuroanatomical studies in midshipman have localized tran- the midbrain, the torus semicircularis (Fergus and Bass, 2013).
script and protein expression of both androgen and ERs. These In the hindbrain vocal pattern generator, ERa, ERb1, and ERb2
receptors are expressed within neuroendocrine regions impor- were all strongly expressed within VMN neurons (Forlano et al.,
tant for reproductive behavior as well as within vocal regions 2005; Fergus and Bass, 2013). Interestingly, ERb2 staining in
dedicated to controlling vocal behavior. Figure 3(a) summa- VMN neurons was stronger in females and type II males
rizes steroid receptor expression within the midshipman vocal compared to type I males, which was confirmed by quantitative
network. PCR (qPCR) of transcript levels in the isolated VMN (Fergus
Teleosts have two paralogous copies of androgen recep- and Bass, 2013). These qPCR studies also revealed a significant
tors, ARa and ARb, that likely arose due to a teleost-specific elevation of ERa within type II male VMN compared to female
whole genome duplication (Ogino et al., 2009, 2016). Phylo- VMN. These sex and morph differences in ER expression were
genetic analyses indicate that ARb is closer to the ancestral AR specific to the VMN, as qPCR of ER expression in surrounding
sequence, with ARa having accumulated novel substitutions hindbrain tissue did not show any significant differences.
at a faster rate than ARb (Ogino et al., 2009, 2016). Character-
ization of ARa and ARb in medaka (Oryzias latipes) and the
2.04.5.4 Glucocorticoids
western mosquitofish (Gambusia affinis) indicated that ARa
is a more potent transcription factor and is constitutively As reviewed in Section 2.04.3.1.2, neurophysiological studies
expressed in the nucleus, whereas ARb exhibits cytosolic in midshipman fish identified male morph-specific profiles
expression with ligand-inducible nuclear translocation for the influences of cortisol, the main circulating glucocorti-
(Ogino et al., 2009, 2016). For both ARs, 11-KT tends to be coid in teleost fish (Section 2.04.2.2), on vocal CPG activity.
a more potent ligand than testosterone (Bentley, 1998; Ogino Two enzymes are integral to the biosynthetic pathway
et al., 2009, 2016). for cortisol, 11-beta(b)-hydroxysteroid dehydrogenase
In type I male midshipman, relative expression of the two (11bHSD), and 11b-hydroxylase (11bH) (Bury and Sturm,
AR subtypes in vocal muscle was dependent on recent 2007). Especially interesting for teleosts is that 11bHSD and
humming activity (Genova et al., 2012). Vocal muscle ARa 11bH are also integral to the biosynthesis of 11-KT. Given
expression was significantly higher in humming males than the duality of enzyme function, it is perhaps not surprising
in nonhumming males, whereas ARb was higher in nonhum- that the mRNA expression patterns for 11bHSD and 11bH in
ming males (Genova et al., 2012). Perhaps the highly active, the brain of midshipman fish exhibit no single, morph-
type I vocal muscles require the more efficient functional typical pattern. For example, 11bHSD mRNA abundance was
properties of ARa, such as its higher transcriptional activity highest in type II males in the vocal hindbrain, a region
(Ogino et al., 2009, 2016). extending between the cerebellum and rostral spinal cord
In the CNS, only ARb expression has been characterized in that houses the vocal CPG, but highest in type I males in the
the midshipman (Forlano et al., 2010). ARb mRNA was highly forebrain (Arterbery et al., 2010b). Significant differences in
expressed in the POA, anterior and ventral tuberal nuclei of the 11bH mRNA levels were only found in the forebrain with
hypothalamus, vocal active sites from which fictive calls can be type II males having the highest levels. Given the absence of
evoked (Goodson and Bass, 2000a; Kittelberger, 2006; Forlano detectable circulating levels of 11-KT in type II males (Brantley
et al., 2010). ARb mRNA was also found in midbrain vocal– et al., 1993b), we presume that regulation of the expression
acoustic regions, such as the PAG, as well as in hindbrain vocal levels of 11bHSD and 11bH in the CNS is related to shifts in
pattern generator, including the VPP and VMN (Forlano et al., regional cortisol levels. A major caveat to these studies is
2010). An early report in the oyster toadfish (Opsanus tau) using that the analysis included large brain regions, e.g., the fore-
steroid autoradiography reported a sparse distribution of brain included the olfactory bulb, telencephalon, and the
androgen receptors in the medulla at the level of VMN, POA. This sampling method could have masked local varia-
including a region that likely corresponds to VPP (Fine et al., tion in individual regions (e.g., POA) or nuclei (e.g., VMN).
1996). It is unclear if ARa and ARb receptor subtypes exhibit The expression patterns for 11bHSD and 11bH mRNA in
divergent localization or expression patterns in the the vocal muscle were less complex and thus more
62 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

amenable to functional interpretations (Arterbery et al., coeruleus, and the area postrema (Forlano et al., 2014).
2010b). Compared to type II males and females, type I Specific to midshipman was a dense population of CA neurons
males had several fold higher transcript levels for both along the rostral, dorsolateral margin of VMN that provided
enzymes that could provide a local source of 11-KT, which dense CA input to the paired VMN. Catecholaminergic
has been linked to seasonal increases in vocal muscle neurons in the diencephalon had dense input to the central
size (Brantley et al., 1993a; Walsh et al., 1995; Sisneros and peripheral auditory systems. This included the hindbrain
et al., 2009). octavolateralis efferent nucleus that innervates the hair cell
The abovementioned transcript levels of 11bHSD and epithelium of the inner ear’s auditory saccule, as well as the
11bH likely represented constitutive expression levels as saccular epithelium itself. The pattern of CA input varied
the tissues were collected from animals not engaged in vocal between the breeding and nonbreeding seasons: input to the
activity at the time of sacrifice (Arterbery et al., 2010b). efferent nucleus was denser in reproductive females compared
A follow-up study of the vocal muscle in type I males showed to nonreproductive females, whereas the saccular epithelium
that the transcript expression levels of 11bHSD varied with showed the opposite pattern, namely denser input in nonre-
the behavioral states of humming versus nonhumming productive females (Forlano et al., 2015a). The CA input to
(Genova et al., 2012). There was a strong trend for higher VMN was also greater in type II males compared to type I
11bHSD levels in nonhumming males that may help to males during the reproductive summer months (Ghahramani
protect the vocal muscle from the potentially catabolic effects et al., 2015). Thus, CA action in auditory and vocal areas of the
of higher circulating cortisol levels by converting cortisol to midshipman brain could underlie seasonal and morph-
its inactive metabolite, cortisone (see Joëls et al., 2007; dependent differences in auditory processing and vocal
Arterbery et al., 2010b). In this study, it was also shown function.
that the higher circulating 11-KT levels in humming males Support for the role of catecholamines in mediating social
were matched by higher levels of 11-KT within the testis stimulation of audition comes from behavioral studies that
(Genova et al., 2012). There was also a positive correlation quantified activation of CA neurons in response to underwater
between the levels of circulating 11-KT and of 11bHSD playback of advertisement hums (Petersen et al., 2013). Activa-
mRNA in the testis, consistent with the testis being a major tion of CA neurons was measured by quantifying the number
site of synthesis for systemically available 11-KT (Genova of CA neurons showing co-label with the immediate early
et al., 2012). gene c-fos, a proxy for neural activation (Petersen et al.,
Teleosts also have two glucocorticoid receptors (GR), GR1 2013). Type I males who were exposed to hums had increased
and GR2, that may differ in their sensitivity to corticosteroids activation of CA neurons in the dopaminergic periventricular
(see Arterbery et al., 2010a and references therein). In general, posterior tuberculum and the noradrenergic locus coeruleus
the expression pattern of GR resembles that of 11bHSD with when compared to those exposed to ambient noise (Petersen
one exception – GR1 was highest in all brain regions in type et al., 2013). Furthermore, the activation of CA neurons in
II males and not just in the vocal hindbrain (Arterbery et al., these two brain regions showed positive correlations with
2010a). In the vocal muscle of type I males, GR1 and GR2 neuronal activation of the ascending auditory pathway,
mRNA levels were higher in nonhummers (Genova et al., including auditory regions of the midbrain (torus semicircula-
2012). Perhaps together with elevated ARb (Section 2.04.5.3) ris) and thalamus (central posterior nucleus) (Petersen et al.,
and 11bHSD (see above) transcript levels, the two GRs help 2013). Together, this recent body of work suggests that CA
maintain the muscle in a state of preparedness for the onset actions within auditory and vocal brain regions likely impact
of long duration humming. seasonal and social regulation of midshipman acoustic
In sum, the complexity of the expression profiles for communication. The neurophysiological consequences of CA
mRNA transcript abundance of receptors and biosynthetic inputs to the vocal and auditory systems remain to be
enzymes involved in both glucocorticoid and androgen investigated.
signaling pathways likely relates to the duality in biosyn-
thetic enzyme function and of duplicate receptor subtypes.
It also remains important to assess the impacts of changing 2.04.6 Daily and Seasonal Rhythms in Vocal–
enzyme transcript levels on protein expression and local Acoustic Mechanisms – Genetic Insights
abundance of the steroids themselves in brain and
peripheral tissues, such as the vocal muscle (e.g., see Pradhan Thus far, as reviewed in the preceding sections, we have
et al., 2014). employed a hypothesis-driven approach to study in detail
several hormonal pathways and their effects on vocal behavior
across multiple levels, including behavior, neurophysiology,
2.04.5.5 Catecholamines
and gene expression. Recently, we used next-generation RNA
Recent studies in midshipman have characterized the CNS sequencing (RNA-seq) to characterize global gene expression
distribution, seasonal and social regulation, as well as intrasex- patterns in crucial nodes within vocal motor and acoustic
ual differences in expression of tyrosine hydroxylase (TH), the pathways: the VMN (Feng et al., 2015) and the hair cell
rate-limiting enzyme for catecholamine synthesis (Petersen epithelium of the auditory saccule (Fergus et al., 2015).
et al., 2013; Forlano et al., 2014, 2015a; Ghahramani et al., From this approach, we not only gained insights into
2015). Like in other teleosts, dense clusters of catecholamin- the general molecular processes important for vocal
ergic (CA)-containing neurons were found in the olfactory (Section 2.04.6.1) and auditory (Section 2.04.6.2) function,
bulb, the telencephalon, the POA, the diencephalon, the locus but also expanded our list of functionally important
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 63

candidate genes, including specific ion channels and (Table 2), from the large data set generated from differential
hormone/neuromodulators, to be validated and studied in expression and downstream data analyses. Furthermore, the
future hypothesis-driven research. wealth of neurophysiological, anatomical, and behavioral
data in the midshipman model system provided validation
for transcriptome-identified candidate molecular mechanisms
2.04.6.1 VMN Transcriptome
underlying vocal characters such as duration and frequency
Within the broader aim of linking genes to behavior, we asked (Figure 2). Here, we focus our discussion on hormone-
whether the VMN, housing a homogeneous population of related insights gained from transcriptome analyses.
vocal motoneurons (alongside glia) (Figures 2 and 6), By comparing gene expression in all VMN samples to all H
expresses a unique molecular profile that supports the samples, we identified close to 3000 transcripts whose expres-
patterning of temporally precise vocal behavior. Furthermore, sion levels were upregulated in the VMN (Figure 6(a)). We first
we wondered whether VMN gene expression changes over subjected these VMN-upregulated transcripts to Gene Ontology
daily and seasonal cycles to drive variation in neural excit- (GO) term enrichment analysis, i.e., enrichment of number of
ability and in turn, natural vocal behavior. To answer these genes belonging to a certain GO term/molecular function.
questions, we compared gene expression in surgically isolated There were a large number of cellular respiration-related GO
VMN to the surrounding hindbrain tissue (H) at three time terms that were enriched in the VMN, consistent with the
points corresponding to minimal or maximal vocal activity: VMN’s ability to maintain firing at 100 Hz for minutes to
summer night (high vocal activity), summer morning (low hours repetitively throughout a single night of courtship
vocal activity), and winter night (low vocal activity) (Feng activity in type I males. There were also several hormone-
et al., 2015). Because we used brain tissue from type I males related GO terms that were enriched in the VMN, including
who were not vocally active at the time of tissue collection, ‘gonadotropin secretion,’ ‘endocrine hormone secretion,’ and
our results represent baseline changes in gene expression ‘sterol biosynthetic process.’ Additionally, many transcripts
with implications in changes in neuronal excitability, and upregulated in the VMN, but no transcripts upregulated in H,
not activity-induced changes. mapped to KEGG (Kyoto Encyclopedia of Genes and
Drawing from previously accumulated knowledge of the Genomes) pathways related to steroid hormone synthesis
midshipman vocal and auditory circuits as well as relevant liter- and degradation (Feng et al., 2015). Together, these results
ature, we were able to identify potentially functionally impor- indicate that VMN is both an energetically and hormonally
tant candidate genes, many of which were hormone related active nucleus, supporting previous studies.

(a) Differential transcript expression (b) VMN neuroanatomy and candidate genes
1e5 4
VMN VMN_summer
3
1e4
2
VMN mean

1e3 1
0
1e2
−1

1e1 −2
Centered log2(FPKM+1)

−3
1e0
−4
1

5
3
0

WNH SNH SMH WNV SNV SMV


1e

1e
1e
1e

1e

1e

H mean
4 4
All_summer Peak_SNV
Centered log2(FPKM+1)

3 3 VN
2 2
1 1
Motoneuron somata
0 0
Glial AR beta mRNA (ar-a)
−1 −1
Glial aromatase-ir and mRNA (cyp19a1a)
−2 −2
ER alpha mRNA & ER beta-ir (esr1, esr2)
−3 −3
GABA-ir (gabra3, gabra5)
−4 −4
Gap junction-coupled motoneurons (cx30, cx43)
WNH SNH SMH WNV SNV SMV WNH SNH SMH WNV SNV SMV

Figure 6 Differential expression identifies functionally important candidate genes. (a) Two methods were used to determine tissue (upper left) and
daily/seasonal (remaining panels) differential expression patterns (see Feng et al., 2015 for method details). Pattern names are shown in the upper
left of each panel and used in Table 2. Sample group names are acronyms indicating the tissue type and time of collection. W, winter; S, summer; N,
night; M, morning; V, VMN; H, surrounding hindbrain; FPKM, fragments per kilobase of transcript per million mapped reads. (b) Schematic of
a cross section of the VMN showing known anatomical features (also see Figure 2(a)). Transcriptome-identified candidate genes correlating to known
VMN characteristics are listed in parentheses. Adapted from Feng, N.Y., Fergus, D.J., Bass, A.H., 2015. Neural transcriptome reveals molecular mech-
anisms for temporal control of vocalization across multiple timescales. BMC Genomics 16, 417.
64 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

Table 2 Select hormone-related candidate genes identified by Table 2 Select hormone-related candidate genes identified by
differential expression in the VMN (Feng et al., 2015) and saccular differential expression in the VMN (Feng et al., 2015) and saccular
epithelium (inner ear) transcriptomes (Fergus et al., 2015). Candidate epithelium (inner ear) transcriptomes (Fergus et al., 2015). Candidate
genes are categorized by function; expression patterns are listed in the genes are categorized by function; expression patterns are listed in the
right column and correspond to Figure 6(a) right column and correspond to Figure 6(a)dcont'd

Transcript name Expression pattern Transcript name Expression pattern

Steroids Insulin receptor-like All-Summer


17b-Hydroxysteroid Ear-Summer Neuropeptide b precursor VMN
dehydrogenase 14 Opioid growth factor receptor-like VMN-Summer
3-Keto-steroid reductase-like VMN, VMN-Summer Parathyroid hormone/parathyroid VMN-Summer
Androgen receptor alpha VMN, All-Summer hormone-related peptide
Androgen-induced gene 1 VMN, VMN-Summer receptor-like
Cholesterol 25-hydroxylase-like All-Summer, Ear-Summer Peptide yy-like VMN
protein member 1-like Pituitary adenylate cyclase-activating VMN
Dihydroxyvitamin VMN polypeptide type i receptor-like
d 24-mitochondrial-like isoform 1 Vasoactive intestinal polypeptide VMN
Estrogen receptor alpha Peak-SNV receptor 1-like
Estrogen receptor beta 2 VMN Thyroid hormone
Estrogen-related receptor alpha VMN Thyroid hormone receptor alpha VMN and Ear
Estrogen-related receptor b type 1 Ear-Summer Thyroid hormone receptor- VMN
Estrogen-related receptor g Ear-Summer associated protein 3-like
Glucocorticoid receptor VMN-Summer Thyroid receptor-interacting protein VMN
Hydroxysteroid 11b-dehydrogenase VMN, Ear-Summer 6-like
1-like Other neuromodulators
Hydroxysteroid dehydrogenase-like VMN-Summer Adenosine receptor a1-like VMN
protein 2 D-like dopamine receptor-like VMN
Lanosterol 14a-demethylase-like VMN, Ear-Summer Melatonin receptor type 1a-like VMN
Liver x receptor VMN-Summer,a Ear-Summer Prostaglandin e synthase 2-like VMN
Neutral cholesterol ester hydrolase VMN-Summer Prostaglandin f2-alpha receptor-like VMN
1-like Acetylserotonin o-methyltransferase- All-Summer
Non-specific lipid-transfer Ear-Summer like
Non-specific lipid-transfer protein Ear-Summer
Nuclear receptor ror-a VMN,a Ear-Summer Note the subset of candidates shared by both VMN and inner ear.
Nuclear receptor subfamily 2 group f Ear-Summer
a
Indicates transcripts that were significantly upregulated in the VMN but not
reported in candidate lists reported in Feng et al. (2015).
member 6 Modified from Feng, N.Y., Fergus, D.J., Bass, A.H., 2015. Neural transcriptome
Ovarian aromatase VMN reveals molecular mechanisms for temporal control of vocalization across multiple
Oxysterol-binding protein 10 VMN-Summer timescales. BMC Genomics 16, 417; Fergus, D.J., Feng, N.Y., Bass, A.H., 2015. Gene
isoform 1 expression underlying enhanced, steroid-dependent auditory sensitivity of hair cell
epithelium in a vocal fish. BMC Genomics 16, 782.
Oxysterol-binding protein 8-like VMN-Summer
Oxysterol-binding protein 9-like Peak-SNV
isoform 3
Retinoid x receptor g Ear-Summer To further identify genes and pathways important for VMN
Sterol 26-mitochondrial-like Peak-SNV
function, we looked for genes whose expression levels were
Sterol regulatory element-binding VMN
increased during time points corresponding to natural levels of
protein 1
Sterol regulatory element-binding VMN high vocal activity. The focus was on candidate genes showing
protein 2 biologically relevant expression patterns, mainly those with
Sterol-c4-methyl oxidase Ear-Summer elevated expression during: the summer night only in the
Neuropeptides/peptide hormones VMN (‘Peak-SNV’); both the summer morning and night in
Atrial natriuretic peptide-converting VMN VMN (‘VMN-Summer’); the summer in both H and VMN
enzyme-like (‘All-Summer’) (Table 2; Figure 6(a)). These results suggest
Calcitonin gene-related peptide VMN, VMN-Summer, All-VMN that hormone synthesis and receptor levels within the VMN
precursor vary dynamically across behaviorally relevant timescales. The
Cholecystokinin type a receptor VMN
expression patterns of several candidate genes and gene
Growth hormone receptor VMN, Peak-SNV
products in the VMN, such as steroid receptors and aromatase,
Growth hormone-regulated tbc All-Summer
protein 1-a-like were consistent with results from prior studies that employed
Inhibin beta b chain-like VMN a variety of quantitative and anatomical methods (see Section
Insulin gene enhancer protein isl-1 VMN, Summer ear 2.04.5 and Figure 6(b) for summary). Although many
Insulin gene enhancer protein VMN-Summer candidates have been previously studied, thus providing
isl-2a-like corroborating evidence for the transcriptome results, most
Insulin-like growth factor binding VMN, VMN-Summer candidates were unstudied in the context of vocal behavior,
protein 1 such as those listed under ‘Neuropeptides/peptide hormones,’
Insulin-like growth factor i VMN ‘Thyroid hormone,’ and ‘Other neuromodulators’ (Table 2;
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 65

see Feng et al., 2015 for more in-depth discussions). Many of with the hypothesis that the type II male phenotype is the more
these candidates would be ideal candidates for future recent innovation.
behavioral, neurophysiological, and anatomical studies in the Although types I and II male midshipman express diver-
midshipman vocal CPG. gent suites of morph-specific characters (Table 1), some
characters are shared between the two morphs such as the
sneak-spawning tactic (Lee and Bass, 2006). The uncoupling
2.04.6.2 Saccular Epithelium Transcriptome
of gonadal phenotype (male or female) from secondary sex
We sequenced the transcriptome of the saccular epithelium characters, like those witnessed for the vocal motor system,
(SE) collected from summer reproductive and winter nonre- leads to the evolution of conserved suites of behavioral
productive type I males to identify candidate genes and path- and neuroendocrine characters for each reproductive morph
ways underlying the well-documented seasonal plasticity in between species and unique traits expressed within species,
auditory sensitivity (Rohmann and Bass, 2011; Fergus et al., e.g., vocal traits (Bass and Grober, 2009). Midshipman fish
2015). Like in the VMN, several candidate genes were upregu- and other teleosts with comparable ARTs (e.g., sunfish, see
lated in the summer reproductive SE related to steroid action, Neff and Svensson, 2013) may represent an intermediate
including estrogen signaling (Table 2), previously shown to condition between species with genetic morph determina-
enhance auditory sensitivity (see Section 2.04.4; Sisneros tion, such as some poeciliids (Zimmerer and Kallman,
et al., 2004a; Fergus et al., 2015). Because this experiment 1989; Ryan et al., 1990) and those in which males and/or
was performed in parallel with the VMN study, and sequences females can change sex during their lifetime (see Godwin,
from both studies were pooled during de novo transcriptome 2016).
assembly, SE and VMN results were directly comparable. Taken together, the available evidence suggests that the
Compared to the VMN and H, the SE contained the highest brain of toadfishes can be characterized as an exquisitely sensi-
number of unique transcripts. Furthermore, seasonal changes tive endocrine organ that directs the expression of sexually
in gene expression were more dramatic in the SE than in the dimorphic and reproductive strategy-specific vocal behavior
CNS tissues. Nevertheless, we identified a suite of candidate under multiple social and environmental contexts. Recent
genes that were shared between the SE and VMN (Table 2), large-scale gene expression studies have enlarged the cast of
suggesting that audio-vocal coupling may have a genetic basis, hormonal and molecular pathways that can regulate vocal
though further studies are needed to test this hypothesis acoustic communication over daily and seasonal cycles (Fergus
(Fergus et al., 2015). et al., 2015; Feng et al., 2015). These candidates will inform
future experimental investigations in the toadfish model
system to validate and characterize their function. The
2.04.7 Summary hormone sensitivity and relative simplicity of the vocal
behavior and its underlying neural circuit make toadfishes
The adaptive radiation of teleost fishes, which comprise nearly excellent models for elucidating the hormonal and neural basis
half of all living species of vertebrates (Nelson, 2006), exhibits of vocalization in vertebrates and how these mechanisms
a wide range of sexual phenotypes. Teleost models thus allow diverge between and within sexes.
us to link natural variation in brain and hormonal mechanisms
to patterns of adult reproductive plasticity. As discussed in this
chapter, behavioral phenotypes within a single species and sex
that follow different reproductive tactics can often exhibit Acknowledgments
equally divergent characters ranging from body coloration
and signaling structures (e.g., those used in acoustic communi- We offer a very special thanks to all of our collaborators for their input
cation) to circulating hormone levels, body and gonad size, into multiple aspects of the work reviewed here and to Margaret
Marchaterre and Margaret Nelson for the photographs and line draw-
and a range of neurobiological traits. Many characters are
ings in Figure 1, respectively. Thanks to the editors for the opportunity
expressed as a correlated suite, with shifts between reproductive
to submit this review and especially to Jacques Balthazart for his helpful
morphs involving divergence in a ‘covariant character set’ (after edits. The authors’ research was supported by NSF (IOS 1457108)
West-Eberhard, 1989). during the preparation of this chapter.
ARTs have been especially well studied at multiple levels of
biological organization in the highly vocal, plainfin
midshipman fish that has two male morphs with one of the References
morphs resembling females in vocal behavior and underlying
vocal mechanisms (Table 1). Each of two male morphs, type Adkins-Regan, E., 2013. Hormones and Animal Social Behavior. Princeton University
I courting and type II satellite/sneak-spawning, has a distinct Press.
Almeida, O., Oliveira, R.F., 2015. Social status and arginine vasotocin neuronal
suite of phenotypic characters (Table 1). Like other closely
phenotypes in a cichlid fish. Brain Behav. Evol. 85, 203–213.
related toadfishes (family Batrachoididae), type I midshipman Amorim, M.C.P., Vasconcelos, R.O., Fonseca, P.J., 2015. Fish sounds and mate
exhibit vocal courtship, nest building and defense, and parental choice. In: Ladich, F. (Ed.), Sound Communication in Fishes, Animal Signals and
care (Amorim et al., 2015; Bass et al., 2015). Type II males Communication. Springer Vienna, Vienna, pp. 1–33.
neither build nests nor guard eggs (Brantley and Bass, 1994). Arnold, A.P., 2009. The organizational-activational hypothesis as the foundation for
a unified theory of sexual differentiation of all mammalian tissues. Horm. Behav.
This led to the proposal that type II males are a derived char- 55, 570–578.
acter state (Bass, 1992). Small type I males also exhibit Arora, H.L., 1948. Observations on the habits and early life history of the batrachoid
sneak-spawning tactics (Lee and Bass, 2006), further consistent fish, Porichthys notatus Girard. Copeia 1948, 89.
66 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

Arterbery, A.S., Deitcher, D.L., Bass, A.H., 2010a. Corticosteroid receptor expression Caras, M.L., 2013. Estrogenic modulation of auditory processing: a vertebrate
in a teleost fish that displays alternative male reproductive tactics. Gen. Comp. comparison. Front. Neuroendocrinol. 34, 285–299.
Endocrinol. 165, 83–90. Chagnaud, B.P., Bass, A.H., 2013. Vocal corollary discharge communicates call
Arterbery, A.S., Deitcher, D.L., Bass, A.H., 2010b. Divergent expression of 11beta- duration to vertebrate auditory system. J. Neurosci. 33, 18775–18780.
hydroxysteroid dehydrogenase and 11beta-hydroxylase genes between male Chagnaud, B.P., Bass, A.H., 2014. Vocal behavior and vocal central pattern generator
morphs in the central nervous system, sonic muscle and testis of a vocal fish. Gen. organization diverge among toadfishes. Brain Behav. Evol. 84, 51–65.
Comp. Endocrinol. 167, 44–50. Chagnaud, B.P., Baker, R., Bass, A.H., 2011. Vocalization frequency and duration are
Bass, A., 1992. Dimorphic male brains and alternative reproductive tactics in coded in separate hindbrain nuclei. Nat. Commun. 2 (346). http://dx.doi.org/
a vocalizing fish. Trends Neurosci. 15, 139–145. 10.1038/ncomms1349.
Bass, A., Andersen, K., 1991. Inter- and intrasexual dimorphisms in the vocal control Chagnaud, B.P., Zee, M.C., Baker, R., Bass, A.H., 2012. Innovations in motoneuron
system of a teleost fish: motor axon number and size. Brain Behav. Evol. 37, synchrony drive rapid temporal modulations in vertebrate acoustic signaling.
204–214. J. Neurophysiol. 107, 3528–3542.
Bass, A., Baker, R., 1991. Evolution of homologous vocal control traits. Brain Behav. Chong, K.K., Liu, R.C., 2016. Hormone-dependent and experience-dependent
Evol. 38, 240–254. auditory plasticity for social communication. In: Bass, A.H., Sisneros, J.A.,
Bass, A.H., 1995. Alternative life history strategies and dimorphic males in an acoustic Fay, R.R., Popper, A.N. (Eds.), Hearing and Hormones. Springer International
communication system. In: Symposium on Reproductive Physiology of Fish. Publishing.
Bass, A.H., 1996. Shaping brain sexuality. Am. Sci. 84, 352–363. Coffin, A.B., Mohr, R.A., Sisneros, J.A., 2012. Saccular-specific hair cell addition
Bass, A.H., Baker, R., 1990. Sexual dimorphisms in the vocal control system of correlates with reproductive state-dependent changes in the auditory saccular
a teleost fish: morphology of physiologically identified neurons. J. Neurobiol. 21, sensitivity of a vocal fish. J. Neurosci. 32, 1366–1376.
1155–1168. Cohen, M.J., Winn, H.E., 1967. Electrophysiological observations on hearing and
Bass, A.H., Clark, C.W., 2003. The physical acoustics of underwater sound communi- sound production in the fish, Porichthys notatus. J. Exp. Zool. 165, 355–369.
cation. Springer Handbook of Auditory Research. In: Simmons, A., Fay, R.R., DeMartini, E.E., 1988. Spawning success of the male plainfin midshipman. I. Influ-
Popper, A. (Eds.), Acoustic Communication. Springer, New York, NY, pp. 15–64. ences of male body size and area of spawning site. J. Exp. Mar. Biol. Ecol. 121,
Bass, A.H., Forlano, P.M., 2008. Neuroendocrine mechanisms of alternative repro- 177–192.
ductive tactics: the chemical language of reproductive and social plasticity. In: Edds-Walton, P.L., Fay, R.R., 2008. Directional and frequency response characteristics
Oliveira, R.F., Taborsky, M., Brockmann, H.J. (Eds.), Alternative Reproductive in the descending octaval nucleus of the toadfish (Opsanus tau). J. Comp. Physiol.
Tactics, an Integrative Approach. Cambridge University Press, Cambridge, A 194, 1013–1029.
pp. 109–131. Feng, N.Y., Bass, A.H., 2014. Melatonin action in a midbrain vocal-acoustic network.
Bass, A.H., Grober, M.S., 2009. Reproductive plasticity in fish: evolutionary lability in J. Exp. Biol. 217, 1046–1057.
the patterning of neuroendocrine and behavioral traits underlying divergent sexual Feng, N.Y., Fergus, D.J., Bass, A.H., 2015. Neural transcriptome reveals molecular
phenotypes. In: Pfaff, D.W., Arnold, A.P., Moss, R., Etgen, A.M., Fahrbach, S.E., mechanisms for temporal control of vocalization across multiple timescales. BMC
Rubin, R.T. (Eds.), Hormones, Brain and Behavior. Academic Press, New York, Genomics 16, 417.
pp. 579–610. Fergus, D.J., Bass, A.H., 2013. Localization and divergent profiles of estrogen
Bass, A.H., Marchaterre, M.A., 1989a. Sound-generating (sonic) motor system in receptors and aromatase in the vocal and auditory networks of a fish with alter-
a teleost fish (Porichthys notatus): sexual polymorphism in the ultrastructure of native mating tactics. J. Comp. Neurol. 521, 2850–2869.
myofibrils. J. Comp. Neurol. 286, 141–153. Fergus, D.J., Feng, N.Y., Bass, A.H., 2015. Gene expression underlying enhanced,
Bass, A.H., Marchaterre, M.A., 1989b. Sound-generating (sonic) motor system in steroid-dependent auditory sensitivity of hair cell epithelium in a vocal fish. BMC
a teleost fish (Porichthys notatus): sexual polymorphisms and general synaptology Genomics 16, 782.
of sonic motor nucleus. J. Comp. Neurol. 286, 154–169. Fernald, R., 2016. Social regulation of sex: how the brain controls reproductive
Bass, A.H., McKibben, J.R., 2003. Neural mechanisms and behaviors for acoustic circuits. In: Pfaff, D.W., Joëls, M. (Eds.), Hormones, Brain, and Behavior, third ed.
communication in teleost fish. Prog. Neurobiol. 69, 1–26. Academic Press, New York.
Bass, A.H., Remage-Healey, L., 2008. Central pattern generators for social vocaliza- Fettiplace, R., Fuchs, P.A., 1999. Mechanisms of hair cell tuning. Annu. Rev. Physiol.
tion: androgen-dependent neurophysiological mechanisms. Horm. Behav. 53, 61, 809.
659–672. Fine, M.L., Lenhardt, M.L., 1983. Shallow-water propagation of the toadfish mating
Bass, A.H., Bodnar, D.A., Marchaterre, M.A., 2000. Midbrain acoustic circuitry in call. Comp. Biochem. Physiol. Part A Physiol. 76, 225–231.
a vocalizing fish. J. Comp. Neurol. 419, 505–531. Fine, M.L., Parmentier, E., 2015. Mechanisms of fish sound production. In: Ladich, F.
Bass, A.H., Chagnaud, B.P., Feng, N.Y., 2015. Comparative neurobiology of sound (Ed.), Sound Communication in Fishes, Animal Signals and Communication.
production in fishes. In: Ladich, F. (Ed.), Sound Communication in Fishes, Animal Springer Vienna, Vienna, pp. 77–126.
Signals and Communication. Springer Vienna, Vienna, pp. 35–75. Fine, M.L., Chen, F.A., Keefer, D.A., 1996. Autoradiographic localization of dihy-
Bass, A.H., Horvath, B.J., Brothers, E.B., 1996. Nonsequential developmental drotestosterone and testosterone concentrating neurons in the brain of the oyster
trajectories lead to dimorphic vocal circuitry for males with alternative reproductive toadfish. Brain Res. 709, 65–80.
tactics. J. Neurobiol. 30, 493–504. Fluet, A., Bass, A., 1990. Sexual dimorphisms in the vocal control-system of a teleost
Bass, A.H., Marchaterre, M.A., Baker, R., 1994. Vocal-acoustic pathways in a teleost Fish – ultrastructure of neuromuscular-junctions. Brain Res. 531, 312–317.
fish. J. Neurosci. 14, 4025–4039. Foran, C.M., Bass, A.H., 1998. Preoptic AVT immunoreactive neurons of a teleost fish
Baulieu, E.E., Robel, P., 1990. Neurosteroids - a new brain-function. J. Steroid Bio- with alternative reproductive tactics. Gen. Comp. Endocrinol. 111, 271–282.
chem. Mol. Biol. 37, 395–403. Foran, C.M., Bass, A.H., 1999. Preoptic GnRH and AVT: axes for sexual plasticity in
Bentley, P.J., 1998. Comparative Vertebrate Endocrinology, third ed. Cambridge teleost fish. Gen. Comp. Endocrinol. 116, 141–152.
University Press, Cambridge. Forlano, P.M., Bass, A.H., 2005a. Seasonal plasticity of brain aromatase mRNA
Bradbury, J.W., Vehrencamp, S.L., 2011. Principles of Animal Communication. expression in glia: divergence across sex and vocal phenotypes. J. Neurobiol. 65,
Sinauer Associates Incorporated. 37–49.
Brantley, R.K., Bass, A.H., 1994. Alternative male spawning tactics and acoustic Forlano, P.M., Bass, A.H., 2005b. Steroid regulation of brain aromatase expression in
signals in the plainfin midshipman fish Porichthys notatus Girard (Teleostei, glia: female preoptic and vocal motor nuclei. J. Neurobiol. 65, 50–58.
Batrachoididae). Ethology 96, 213–232. Forlano, P.M., Deitcher, D.L., Bass, A.H., 2005. Distribution of estrogen receptor alpha
Brantley, R.K., Marchaterre, M.A., Bass, A.H., 1993a. Androgen effects on vocal mRNA in the brain and inner ear of a vocal fish with comparisons to sites of
muscle structure in a teleost fish with inter-sexual and intra-sexual dimorphism. aromatase expression. J. Comp. Neurol. 483, 91–113.
J. Morphol. 216, 305–318. Forlano, P.M., Deitcher, D.L., Myers, D.A., Bass, A.H., 2001. Anatomical distribution
Brantley, R.K., Wingfield, J.C., Bass, A.H., 1993b. Sex steroid levels in Porichthys and cellular basis for high levels of aromatase activity in the brain of teleost fish:
notatus, a fish with alternative reproductive tactics, and a review of the hormonal aromatase enzyme and mRNA expression identify glia as source. J. Neurosci. 21,
bases for male dimorphism among teleost fishes. Horm. Behav. 27, 332–347. 8943–8955.
Bury, N.R., Sturm, A., 2007. Evolution of the corticosteroid receptor signalling pathway Forlano, P.M., Ghahramani, Z.N., Monestime, C.M., Kurochkin, P., Chernenko, A.,
in fish. Gen. Comp. Endocrinol. 153, 47–56. Milkis, D., 2015a. Catecholaminergic innervation of central and peripheral auditory
Callard, G., Schlinger, B., Pasmanik, M., 1990. Nonmammalian vertebrate models in circuitry varies with reproductive state in female midshipman fish, Porichthys
studies of brain-steroid interactions. J. Exp. Zool. 4, 6–16. notatus. PLoS One 10, e0121914.
Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems 67

Forlano, P.M., Kim, S.D., Krzyminska, Z.M., Sisneros, J.A., 2014. Catecholamin- Hultcrantz, M., Simonoska, R., Stenberg, A.E., 2006. Estrogen and hearing:
ergic connectivity to the inner ear, central auditory, and vocal motor circuitry in a summary of recent investigations. Acta Oto-Laryngol. 126, 10–14.
the plainfin midshipman fish Porichthys notatus. J. Comp. Neurol. 522, Ibara, R.M., Penny, L.T., Ebeling, A.W., van Dykhuizen, G., Cailliet, G., 1983. The
2887–2927. Mating Call of the Plainfin Midshipman Fish, Porichthys notatus. Predators and
Forlano, P.M., Marchaterre, M., Deitcher, D.L., Bass, A.H., 2010. Distribution of Prey in Fishes, Developments in Environmental Biology of Fishes. Springer,
androgen receptor mRNA expression in vocal, auditory, and neuroendocrine circuits Dordrecht, Netherlands, pp. 205–212.
in a teleost fish. J. Comp. Neurol. 518, 493–512. Joëls, M., Karst, H., Krugers, H.J., Lucassen, P.J., 2007. Chronic stress: implications
Forlano, P.M., Maruska, K.P., Sisneros, J.A., Bass, A.H., 2016. Hormone-dependent for neuronal morphology, function and neurogenesis. Front. Neuroendocrinol. 28,
plasticity of auditory systems in fishes. In: Bass, A.H., Sisneros, J.A., Fay, R.R., 72–96.
Popper, A.N. (Eds.), Hearing and Hormones. Springer International Publishing. Kittelberger, J.M., 2006. Midbrain periaqueductal gray and vocal patterning in a teleost
Forlano, P.M., Schlinger, B.A., Bass, A.H., 2006. Brain aromatase: new lessons from fish. J. Neurophysiol. 96, 71–85.
non-mammalian model systems. Front. Neuroendocrinol. 27, 247–274. Kittelberger, J.M., Bass, A.H., 2013. Vocal-motor and auditory connectivity of
Forlano, P.M., Sisneros, J.A., Rohmann, K.N., Bass, A.H., 2015b. Neuroendocrine the midbrain periaqueductal gray in a teleost fish. J. Comp. Neurol. 521,
control of seasonal plasticity in the auditory and vocal systems of fish. Front. 791–812.
Neuroendocrinol. 37, 129–145. Knapp, R., Neff, B.D., 2007. Steroid hormones in bluegill, a species with male
Frisina, R.D., Frisina, D.R., 2016. Hormone replacement therapy and its effects on alternative reproductive tactics including female mimicry. Biol. Lett. 3, 628–631.
human hearing. In: Bass, A.H., Sisneros, J.A., Fay, R.R., Popper, A.N. (Eds.), Knapp, R., Marchaterre, M.A., Bass, A.H., 1999a. Early development of the motor and
Hearing and Hormones. Springer International Publishing. premotor circuitry of a sexually dimorphic vocal pathway in a teleost fish. J. Neu-
Furukawa, T., Ishii, Y., 1967. Neurophysiological studies on hearing in goldfish. robiol. 38, 475–490.
J. Neurophysiol. 30, 1377–1403. Knapp, R., Wingfield, J.C., Bass, A.H., 1999b. Steroid hormones and paternal care in
Gall, M.D., Salameh, T.S., Lucas, J.R., 2013. Songbird frequency selectivity and the plainfin midshipman fish (Porichthys notatus). Horm. Behav. 35, 81–89.
temporal resolution vary with sex and season. Proc. R. Soc. B Biol. Sci. 280, Lee, J.S.F., Bass, A.H., 2004. Does exaggerated morphology preclude plasticity
20122296. to cuckoldry in the midshipman fish (Porichthys notatus)? Natur-
Genova, R.M., Marchaterre, M.A., Knapp, R., Fergus, D., Bass, A.H., 2012. Gluco- wissenschaften 91.
corticoid and androgen signaling pathways diverge between advertisement calling Lee, J.S.F., Bass, A.H., 2006. Dimorphic male midshipman fish: reduced sexual
and non-calling fish. Horm. Behav. 62, 426–432. selection or sexual selection for reduced characters? Behav. Ecol. 17, 670–675.
Ghahramani, Z.N., Timothy, M., Kaur, G., Gorbonosov, M., Chernenko, A., Lee, J.S.F., Bass, A.H., 2005. Differential effects of 11-ketotestosterone on dimorphic
Forlano, P.M., 2015. Catecholaminergic fiber innervation of the vocal motor system traits in a teleost with alternative male reproductive morphs. Horm. Behav. 47,
is intrasexually dimorphic in a teleost with alternative reproductive tactics. Brain 523–531.
Behav. Evol. 86, 131–144. Locascio, J.V., Mann, D.A., 2008. Diel periodicity of fish sound production in Charlotte
Godwin, J., Thompson, R., 2012. Nonapeptides and social behavior in fishes. Horm. Harbor, Florida. Trans. Am. Fish. Soc. 137, 606–615.
Behav. 61, 230–238. Lösel, R., Wehling, M., 2003. Nongenomic actions of steroid hormones. Nat. Rev. Mol.
Godwin, J.R., 2016. Sex changing fishes. In: Pfaff, D.W., Joëls, M. (Eds.), Hormones, Cell Biol. 4, 46–56.
Brain, and Behavior, third ed. Academic Press, New York. Marder, E., Calabrese, R.L., 1996. Principles of rhythmic motor pattern generation.
Goodson, J.L., 2008. Nonapeptides and the evolutionary patterning of sociality. Prog. Physiol. Rev. 76, 687–717.
Brain Res. 3–15. Maruska, K.P., Ung, U.S., Fernald, R.D., 2012. The African cichlid fish Astatotilapia
Goodson, J.L., 2013. Deconstructing sociality, social evolution and relevant non- burtoni uses acoustic communication for reproduction: sound production, hearing,
apeptide functions. Psychoneuroendocrinology 38, 465–478. and behavioral significance. PLoS One 7, e37612.
Goodson, J.L., Bass, A.H., 2000a. Forebrain peptides modulate sexually polymorphic McIver, E.L., Marchaterre, M.A., Rice, A.N., Bass, A.H., 2014. Novel underwater
vocal circuitry. Nature 403, 769–772. soundscape: acoustic repertoire of plainfin midshipman fish. J. Exp. Biol. 217,
Goodson, J.L., Bass, A.H., 2000b. Vasotocin innervation and modulation of vocal- 2377–2389.
acoustic circuitry in the teleost Porichthys notatus. J. Comp. Neurol. 422, McKibben, J., Bass, A., 2001. Peripheral encoding of behaviorally relevant acoustic
363–379. signals in a vocal fish: harmonic and beat stimuli. J. Comp. Physiol. A 187,
Goodson, J.L., Bass, A.H., 2001. Social behavior functions and related anatomical 271–285.
characteristics of vasotocin/vasopressin systems in vertebrates. Brain Res. Rev. McKibben, J.R., Bass, A.H., 1998. Behavioral assessment of acoustic parameters
35, 246–265. relevant to signal recognition and preference in a vocal fish. J. Acoust. Soc. Am.
Goodson, J.L., Bass, A.H., 2002. Vocal–acoustic circuitry and descending vocal 104, 3520–3533.
pathways in teleost fish: convergence with terrestrial vertebrates reveals conserved McKibben, J.R., Bass, A.H., 1999. Peripheral encoding of behaviorally relevant
traits. J. Comp. Neurol. 448, 298–322. acoustic signals in a vocal fish: single tones. J. Comp. Physiol. A 184, 563–576.
Goodson, J.L., Evans, A.K., Bass, A.H., 2003. Putative isotocin distributions in sonic Modesto, T., Canário, A., 2003. Morphometric changes and sex steroid levels during
fish: relation to vasotocin and vocal-acoustic circuitry. J. Comp. Neurol. 462, the annual reproductive cycle of the Lusitanian toadfish, Halobatrachus didactylus.
1–14. Gen. Comp. Endocrinol. 131, 220–231.
Greene, C.W., 1924. Analysis of the gases of the air-bladder of the California singing Neff, B.D., Svensson, E.I., 2013. Polyandry and alternative mating tactics. Philos.
fish, Porichthys notatus. J. Biol. Chem. 59, 615–621. Trans. R. Soc. Lond. Ser. B Biol. Sci. 368, 20120045.
Greenfield, D.W., Winterbottom, R., Collette, B.B., 2008. Review of the toadfish genera Nelson, J.S., 2006. Fishes of the World, fourth ed. John Wiley & Sons, Hoboken.
(Teleostei: batrachoididae). Proc. Calif. Acad. Sci. 59, 665–710. Noirot, I.C., Adler, H.J., Cornil, C.A., Harada, N., Dooling, R.J., Balthazart, J., Ball, G.F.,
Grober, M.S., Bass, A.H., 1991. Neuronal correlates of sex-role change in Labrid 2009. Presence of aromatase and estrogen receptor alpha in the inner ear of zebra
fishes – LHRH-like immunoreactivity. Brain Behav. Evol. 38, 302–312. finches. Hear. Res. 252, 49–55.
Grober, M.S., Fox, S.H., Laughlin, C., Bass, A.H., 1994. GnRH cell size and number in Ogino, Y., Katoh, H., Kuraku, S., Yamada, G., 2009. Evolutionary history and functional
a teleost fish with two male reproductive morphs: sexual maturation, final sexual characterization of androgen receptor genes in jawed vertebrates. Endocrinology
status and body size allometry. Brain Behav. Evol. 43, 61–78. 150, 5415–5427.
Grober, M.S., Jackson, I., Bass, A.H., 1991. Gonadal-steroids affect LHRH preoptic Ogino, Y., Kuraku, S., Ishibashi, H., Miyakawa, H., Sumiya, E., Miyagawa, S., et al.,
cell number in a sex-role changing fish. J. Neurobiol. 22, 734–741. 2016. Neofunctionalization of androgen receptor by gain-of-function mutations in
Halpern-Sebold, L.R., Schreibman, M.P., Margolis-Nunno, H., 1986. Differences teleost fish lineage. Mol. Biol. Evol. 33, 228–244.
between early-maturing and late-maturing genotypes of the platyfish (Xiphophorus Oka, Y., Ichikawa, M., 1990. Gonadotropin-releasing hormone (GnRH) immunoreactive
maculatus) in the morphometry of their immunoreactive luteinizing-hormone- system in the brain of the dwarf gourami (Colisa lalia) as revealed by light
releasing hormone-containing cells – a developmental-study. J. Exp. Zool. 240, microscopic immunocytochemistry using a monoclonal antibody to common amino
245–257. acid sequence of GnRH. J. Comp. Neurol. 300, 511–522.
Harbott, L.K., Burmeister, S.S., White, R.B., Vagell, M., Fernald, R.D., 2007. Androgen Oliveira, R.F., 2006. Neuroendocrine mechanisms of alternative reproductive tactics in
receptors in a cichlid fish, Astatotilapia burtoni: structure, localization, and fish. In: Sloman, K.A., Wilson, R.W., Balshine, S. (Eds.), Behaviour and Physiology
expression levels. J. Comp. Neurol. 504, 57–73. of Fish. Elsevier, pp. 297–357.
Hubbs, C.L., 1920. The bionomics of Porichthys notatus Girard. Am. Nat. 54, Oliveira, R.F., 2009. Social behavior in context: hormonal modulation of behavioral
380–384. plasticity and social competence. Integr. Comp. Biol. 49, 423–440.
68 Neural, Hormonal, and Genetic Mechanisms of Alternative Reproductive Tactics: Vocal Fish as Model Systems

Petersen, C.L., Timothy, M., Kim, D.S., Bhandiwad, A.A., Mohr, R.A., Sisneros, J.A., Sisneros, J.A., 2009. Seasonal plasticity of auditory saccular sensitivity in the
Forlano, P.M., 2013. Exposure to advertisement calls of reproductive competitors vocal plainfin midshipman fish, Porichthys notatus. J. Neurophysiol. 102,
activates vocal-acoustic and catecholaminergic neurons in the plainfin midshipman 1121–1131.
fish, Porichthys notatus. PLoS One 8, e70474. Sisneros, J.A., Bass, A.H., 2003. Seasonal plasticity of peripheral auditory frequency
Pradhan, D.S., Solomon-Lane, T.K., Willis, M.C., Grober, M.S., 2014. A mechanism sensitivity. J. Neurosci. 23, 1049–1058.
for rapid neurosteroidal regulation of parenting behaviour. Proc. R. Soc. B Biol. Sisneros, J.A., Alderks, P.W., Leon, K., Sniffen, B., 2009. Morphometric changes
Sci. 281. associated with the reproductive cycle and behaviour of the intertidal-nesting, male
Remage-Healey, L., Bass, A.H., 2004. Rapid, hierarchical modulation of vocal plainfin midshipman Porichthys notatus. J. Fish Biol. 74, 18–36.
patterning by steroid hormones. J. Neurosci. 24, 5892–5900. Sisneros, J.A., Forlano, P.A., Deitcher, D.A., Bass, A.H., 2004a. Steroid-dependent
Remage-Healey, L., Bass, A.H., 2005. Rapid elevations in both steroid hormones and auditory plasticity leads to adaptive coupling of sender and receiver. Science 305,
vocal signaling during playback challenge: a field experiment in Gulf toadfish. 404–407.
Horm. Behav. 47, 297–305. Sisneros, J.A., Forlano, P.M., Knapp, R., Bass, A.H., 2004b. Seasonal variation of
Remage-Healey, L., Bass, A.H., 2006. From social behavior to neural circuitry: steroid steroid hormone levels in an intertidal-nesting fish, the vocal plainfin midshipman.
hormones rapidly modulate advertisement calling via a vocal pattern generator. Gen. Comp. Endocrinol. 136, 101–116.
Horm. Behav. 50, 432–441. Steinacker, A., Romero, A., 1992. Voltage-gated potassium current and resonance in
Remage-Healey, L., Bass, A.H., 2007. Plasticity in brain sexuality is revealed by the the toadfish saccular hair cell. Brain Res. 574, 229–236.
rapid actions of steroid hormones. J. Neurosci. 27, 1114–1122. Sugihara, I., Furukawa, T., 1995. Potassium currents underlying the oscillatory
Rice, A.N., Bass, A.H., 2009. Novel vocal repertoire and paired swimbladders of the response in hair cells of the goldfish sacculus. J. Physiol. 489, 443–453.
three-spined toadfish, Batrachomoeus trispinosus: insights into the diversity of the Vasconcelos, R.O., Simõest, J.M., Almada, V.C., Fonseca, P.J., Amorim, M.C.P.,
Batrachoididae. J. Exp. Biol. 212, 1377–1391. 2010. Vocal behavior during territorial intrusions in the Lusitanian toadfish: boat-
Rohmann, K.N., Bass, A.H., 2011. Seasonal plasticity of auditory hair cell frequency whistles also function as territorial “keep-out” signals. Ethology 116, 155–165.
sensitivity correlates with plasma steroid levels in vocal fish. J. Exp. Biol. 214, Vasconcelos, R.O., Sisneros, J.A., Amorim, M.C.P., Fonseca, P.J., 2011. Auditory
1931–1942. saccular sensitivity of the vocal Lusitanian toadfish: low frequency tuning allows
Rohmann, K.N., Fergus, D.J., Bass, A.H., 2013. Plasticity in ion channel expression acoustic communication throughout the year. Z. Vgl. Physiol. 197, 903–913.
underlies variation in hearing during reproductive cycles. Curr. Biol. 23, Vélez, A., Gall, M.D., Lucas, J.R., 2015. Seasonal plasticity in auditory processing of
678–683. the envelope and temporal fine structure of sounds in three songbirds. Anim.
Rome, L.C., 2006. Design and function of superfast muscles: new insights into the Behav. 103, 53–63.
physiology of skeletal muscle. Annu. Rev. Physiol. 68, 193–221. Walsh, P.J., Mommsen, T.P., Bass, A.H., 1995. Biochemical and molecular aspects of
Ryan, M.J., Hews, D.K., Wagner Jr., William E., 1990. Sexual selection on alleles that singing in batrachoidid fishes. In: Hochachka, P.W., Mommsen, T.P. (Eds.),
determine body size in the swordtail Xiphophorus nigrensis. Behav. Ecol. Sociobiol. Biochemistry and Molecular Biology of Fishes, pp. 279–289.
26, 231–237. West-Eberhard, M.J., 1989. Phenotypic plasticity and the origins of diversity. Annu.
Saldanha, C.J., Schlinger, B.A., 2008. Steroidogenesis and neuroplasticity in the Rev. Ecol. Syst. 20, 249–278.
songbird brain. In: Neuroactive Steroids in Brain Function, Behavior and Neuro- Whitchurch, E.A., Sisneros, J.A., 2011. Seasonal plasticity of saccular sensitivity in the
psychiatric Disorders. Springer, Netherlands, Dordrecht, pp. 201–216. type II sneaker-male plainfin midshipman fish (Porichthys notatus). J. Acoust. Soc.
Santangelo, N., Bass, A.H., 2006. New insights into neuropeptide modulation of Am. 129, 2471.
aggression: field studies of arginine vasotocin in a territorial tropical damselfish. Winn, H.E., Stout, J.F., 1960. Sound production by the satinfin shiner, Notropis
Proc. R. Soc. B Biol. Sci. 273, 3085–3092. analostanus, and related fishes. Science 132, 222–223.
Schlinger, B.A., Greco, C., Bass, A.H., 1999. Aromatase activity in hindbrain vocal Zeyl, J.N., Love, O.P., Higgs, D.M., 2013. Condition-dependent auditory processing in
control region of a teleost fish: divergence among males with alternative repro- the round goby (Neogobius melanostomus): links to sex, reproductive condition and
ductive tactics. Proc. R. Soc. B Biol. Sci. 266, 131–136. female estrogen levels. J. Exp. Biol. 216, 1075–1084.
Simpson, E.R., Mahendroo, M.S., Means, G.D., Kilgore, M.W., Hinshelwood, M.M., Zimmerer, E.J., Kallman, K.D., 1989. Genetic-basis for alternative reproductive tactics
Graham-Lorence, S., et al., 1994. Aromatase cytochrome P450, the enzyme in the pygmy swordtail, Xiphophorus nigrensis. Evolution 43, 1298–1307.
responsible for estrogen biosynthesis. Endocr. Rev. 15, 342–355.
2.05 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology
Kent D Dunlap, Trinity College, Hartford, CT, USA
Ana C Silva, Unidad Bases Neurales de la Conducta, Instituto de Investigaciones Biologicas Clemente Estable, Montevideo, Uruguay;
and Universidad de la Republica, Montevideo, Uruguay
G Troy Smith, Indiana University, Bloomington, IN, USA
Harold H Zakon, University of Texas, Austin, TX, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by H.H. Zakon, G.T. Smith, volume 1, pp. 611–639, Ó 2009, Elsevier Inc.

2.05.1 Introduction 69
2.05.1.1 Electric Fish as a Model in Behavioral Neuroendocrinology 70
2.05.1.2 The EOD as a Communication Signal 71
2.05.1.3 Breeding Cycle and Endogenous Levels of Hormones 71
2.05.2 EOD Waveform and Amplitude and Their Control by Electrocytes 72
2.05.2.1 Generation of the EOD Waveform 72
2.05.2.1.1 Pulse Fish 72
2.05.2.1.2 Wave Fish 73
2.05.2.2 EOD Waveform: Communication Function 73
2.05.2.3 EOD Waveform and Electrocytes: Sexual Dimorphism 74
2.05.2.4 EOD Waveform and EOD Amplitude: Hormonal Regulation 74
2.05.2.4.1 Effects of Exogenous Steroids on EOD Waveform and Electric Organ Morphology 74
2.05.2.4.2 Effects of Exogenous Steroids on Electric Organ Ion Currents 75
2.05.2.4.3 EOD Waveform: Seasonal Modulation and Its Hormonal Regulation 76
2.05.2.4.4 EOD Waveform: Diel, Social, and Stress-Induced Modulation and Its Hormonal Regulation 76
2.05.3 EOD Rate/Frequency and Its Regulation by the Pacemaker Nucleus 78
2.05.3.1 EOD Rate in Pulse Fish: Social and Diel Influences and Their Hormonal Regulation 79
2.05.3.1.1 Social Influences 79
2.05.3.1.2 Diel Influence 79
2.05.3.2 EOD Frequency in Wave Fish: Role in Social Behavior and Its Hormonal Regulation 80
2.05.3.2.1 Social Behavior 80
2.05.3.2.2 Hormonal Regulation 80
2.05.4 EOD Modulations and Their Regulation by Prepacemaker Nuclei 81
2.05.4.1 EOD Modulations: Communication Function 81
2.05.4.1.1 Pulse Fish 81
2.05.4.1.2 Wave Fish 83
2.05.4.2 EOD Modulations: Neural Control by Prepacemaker Nuclei 86
2.05.4.3 EOD Modulations: Species Diversity and Their Hormonal Control in Apteronotidae 87
2.05.4.3.1 Species Diversity 87
2.05.4.3.2 Neuroendocrine Regulation of Chirping in Apteronotus 87
2.05.4.4 EOD Modulations: Seasonality and Neuroendocrine Regulation in Brachyhypopomus 88
2.05.4.5 EOD Modulations: Steroidal Regulation of Nonsex-Biased Territorial Aggression in Gymnotus 89
2.05.5 EOD Reception through the Electrosensory System 89
2.05.5.1 Electroreceptor Types, Function, and Tuning 89
2.05.5.2 Electroreceptor Coding of EOD Waveforms 91
2.05.5.2.1 Pulse Fish 91
2.05.5.2.2 Wave Fish 91
2.05.5.3 Sensory Coding of Chirps 92
2.05.5.3.1 Electroreceptors 92
2.05.5.3.2 Central Nervous System 92
2.05.5.4 Electroreception: Sex Differences and Neuroendocrine Regulation 92
2.05.6 Conclusions 93
References 94

2.05.1 Introduction organ discharges (EODs) and those of other fish using special-
ized sensory receptors called electroreceptors. There are two
Weakly electric fish generate weak electric fields from modified clades of weakly electric fish: the Gymnotiformes from South
muscle cells in the electric organ. They sense their own electric America and the Mormyroidea from Africa. These groups are

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00019-5 69


70 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

so phylogenetically distant that electric organs and electrosen- EOD signal parameters and electroreception differ between
sory capabilities must have evolved independently in each the sexes, participate in reproductive and agonistic communica-
group. Electric fish use this extraordinary electric sense to locate tion, and respond to hormones. Because each process in electro-
and identify nearby objects (electrolocation) and to communi- communication is regulated by a discrete set of cells, it is
cate with each other (electrocommunication) (Hopkins, relatively easy to pinpoint the locus and cellular mechanisms
1974a,b). EODs enable fish to communicate about their of hormone action. Most cells in the circuit are accessible for elec-
species identity, sex, social status, and motivational state. trophysiological recordings, so their biophysical properties can
be compared before and after hormone treatment. Moreover,
EODs are generated by current flow across ion channel proteins,
2.05.1.1 Electric Fish as a Model in Behavioral enabling us to identify candidate genes for molecular analysis of
Neuroendocrinology sex differences in behavior and their hormonal regulation.
Electric fish are especially tractable experimental subjects for Another attractive aspect of these fish is the richness and
studying the neural and hormonal mechanisms underlying diversity of their behavior and ecology. For example, while
behavior. Electric signals are stereotyped and easily quantifi- most species show a strong sexual dimorphism in the EOD
able, allowing detailed analysis of signal structure. They vary (Figures 2 and 3), a few show a ‘reverse’ sexual dimorphism,
by waveform, amplitude, rate (or frequency), and transient and some seem to be monomorphic (Westby, 1988;
modulations of rate and amplitude. Each of these signal Alves-Gomes and Hopkins, 1997; Zhou and Smith, 2006; Ho
parameters can be traced to three cell types in the underlying et al., 2007; Turner et al., 2007; Smith, 2013). Some species
electrocommunication circuitry: (1) EOD waveform and lay eggs on vegetation or in the leaf litter and abandon them,
amplitude are determined by the electrocytes of the electric whereas others construct nests and tend to their young
organ located primarily in the tail, (2) baseline EOD rate or (Hagedorn and Heiligenberg, 1985; Crampton and Hopkins,
frequency is determined by neurons in the hindbrain pace- 2005). Species also differ in habitat choice and degrees of soci-
maker nucleus (PN), and (3) EOD modulations are deter- ality: in some, individuals are solitary and establish territories
mined by neurons in the thalamic and midbrain in shallow streams; in others, fish school in open water or
prepacemaker nuclei (Figure 1). The detection of the EOD even gather in large multispecies assemblages (Hopkins,
occurs through a fourth cell type, the electroreceptors, distrib- 1981; Oestreich and Zakon, 2005; Tan et al., 2005). Because
uted all over the body surface and concentrated near the head. phylogenies of both clades of weakly electric fish are available
(Alves-Gomes et al., 1995; Alves-Gomes, 1999; Lavoue and
Sullivan, 2004; Crampton and Albert, 2006; Tagliacollo et al.,
Diencephalon 2016), it is possible to make reasonable speculations about
how sexual dimorphisms and the underlying neuroendocrine
Telencephalon Electromotor neurons
mechanisms have evolved. Molecular phylogenetics has shown
that signals can evolve rapidly within clades to yield a remark-
able diversity of communication behaviors even within closely
related species (Arnegard et al., 2005, 2010; Feulner et al.,
EO
2006, 2007; Turner et al., 2007).
SPPn PN In this chapter, we review the role of electrocommunication
PPn
in the social behavior of electric fish and its regulation by
Figure 1 Pacemaker nucleus of a South American fish and its inputs. gonadal steroid hormones and neuromodulators. After intro-
Schematic illustration of the two inputs to the pacemaker nucleus: the ducing the general function of the EOD in communication
prepacemaker nucleus (PPn) and the sublemniscal prepacemaker and patterns of endogenous hormone secretion, we organize
nucleus (SPPn). These inputs modulate the firing frequency of the the chapter into four sections according to EOD signal param-
pacemaker neurons in the pacemaker nucleus (PN) that synapse on the eter and its cellular substrate: first, the EOD waveform and
electromotor neurons in the spinal cord and ultimately drive the electric amplitude and their control by electrocytes; second, EOD
organ (EO). rate/frequency and its control by the PN; third, EOD

Figure 2 Pulse-type electric organ discharges (EODs) from the South American fish, Brachyhypopomus gauderio. Males (above) possess thicker
longer tails than females (below) and produce EOD pulses that are longer in duration, especially the second (P2, negative) phase. The EODs are from
representative mature fish of each sex, scaled in amplitude and overlain. Modified from Perrone, R., 2012. La vasotocina modula el comportamiento
social de dos especies de peces eléctricos con diferente socialidad. PEDECIBA, Universidad de la República, Montevideo.
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 71

Brienomyrus brachyistius (t.p.) both groups of electric fish to evolve signals that are less detect-
able to these predators (i.e., minimize the DC and low-
frequency components of their EODs) (Stoddard, 1999;
Hanika and Kramer, 1999).
Brienomyrus brachyistius (I.bp.)
2.05.1.3 Breeding Cycle and Endogenous Levels of Hormones
Both mormyroid and gymnotiform electric fish breed primarily
during the rainy season (Kirschbaum, 1975, 1995), although
Ivindomyrus opdenboschi there are a few exceptions to this rule such as the electric eel
which reportedly breeds in the dry season (Assuncao and
juv Schwassman, 1995) and B. occidentalis, reported to be an acycl-
ad
ical breeder (Hagedorn, 1992). Environmental cues that trigger
gonadal recrudescence and reproduction in these species
Stomathorhinus corneti include the pH and conductivity of the water, water level, the
physical stimulation of rainfall, and food availability (reviewed
in Kirschbaum, 1995). In general, electric fish are transequato-
rial in their distribution so that day length and temperature are
Mormyrus rume
not reliable zeitgebers for them. Some species of electric fish,
however, also inhabit the temperate zone. For example, in
the southern temperate zones of Uruguay, Brachyhypopomus
gauderio (formerly Brachyhypopomus pinnicaudatus) times its
1 ms spawning season based on seasonal temperature fluctuations
(Silva et al., 2002, 2003; Quintana et al., 2004).
Figure 3 Pulse-type electric organ discharges (EODs) from five African
Electric fish show a pattern of steroid secretion typical of
electric fish. Note the sexual dimorphism and/or age difference (Ivindomy-
rus opdenboschi) in EOD pulse shape. Courtesy of C.D. Hopkins. many teleost fish. Sex steroids are highest in mature adults,
with 11-ketotestosterone (11kT) as the predominant gonadal
steroid in males, 17b estradiol (E2) prominent in females,
modulations and their control by prepacemaker nuclei; and and testosterone of similar values in males and females. For
finally, EOD detection and processing by electroreceptors and both sexes, plasma steroids concentrations are greatest during
central electrosensory circuits. the breeding season.
In several cases, plasma steroid concentrations have been
measured directly from fish in their natural habitat. In male
2.05.1.2 The EOD as a Communication Signal
B. gauderio in Uruguay, 11kT is higher in males during the
Just as olfactory, visual, or acoustic signaling in other groups of breeding season (January) than in the nonbreeding season
animals, electric signaling coordinates social interaction in (June) (Dunlap et al., 2011b) and correlates positively with
weakly electric fish. Electric signals are restricted to aquatic testicular size, plasma levels of testosterone, and masculine
environments, and they attenuate with distance more rapidly features of the EOD (Gavassa et al., 2011). E2 is higher in
than acoustic or visual stimuli (Hopkins, 1999). Consequently, females than in males and correlates positively with testos-
electric signals are used for communication only over short terone levels and feminine EOD features, but is unrelated to
distances of a few body-lengths. However, they can be used gonadal size. In field-captured Sternopygus macrurus, androgen
at night or in light-restricted habitats such as silty streams or levels of males correlate positively with the degree of testicular
the bottoms of large rivers, and they can be delivered from development and body size, and masculine EOD parameters
within a protected location such as a clump of weeds. Impor- (Zakon et al., 1991). However, while all large adult males
tantly, the temporal fine structure of electric signals is not dis- have high androgen levels and low EOD frequencies, immature
torted by the medium allowing slight variations of EOD males, which have low androgen levels, have either high or low
waveforms to carry important information (Hopkins and EOD frequencies. Thus, some factor other than androgen level
Bass, 1981; McGregor and Westby, 1992; Hopkins, 1999). influences EOD frequency in immature males (Zakon et al.,
Electric signals provide a private communication channel 1991). Female Sternopygus possess little or no 11kT and have
since most predators cannot sense them. Nevertheless, some testosterone levels comparable to males. E2 levels in immature
predators are electroreceptive. For example catfish, which are females are similar to those of mature or immature males
abundant in the rivers inhabited by electric fish, possess ampul- (<0.5 ng ml1), and gravid females with well-yolked eggs
lary electroreceptors (see 2.05.5.1) allowing them to detect have higher levels (2 ng ml1) (Zakon et al., 1991).
direct current (DC) and low-frequency components of EODs. In the laboratory, individual variation in androgens among
In streams with the highest density of catfish, as many as males is associated with body size, EODs, and social status.
45% of electric fish (Brachyhypopomus occidentalis) have injured Male brown ghosts (Apteronotus leptorhynchus) show a positive
tails, mostly likely due to predation attempts by catfish correlation between plasma 11kT levels, body size, and two
(Dunlap et al., 2016). Electric eels also hunt weakly electric EOD parameters (EOD frequency and chirp rate) (Dunlap,
fish by tracking their EODs (Westby, 1988). Electroreceptive 2002). In the mormyrid Brienomyrus brachyistius, males housed
predators have exerted a formidable selection pressure on in captive social groups (three males and three females) in
72 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

which the males establish a dominance hierarchy, 11kT levels is low resting rate during the day when they are inactive. Wave-
highest (16 ng ml1) in the dominant male, next highest type EODs are continuously produced and, therefore, energeti-
(10 ng ml1) in the next highest-ranked male, and least cally demanding (Lewis et al., 2014). However, some wave-type
(6 ng ml1) in the subordinate male (Carlson et al., 2000). fish overcome this problem by dramatically decreasing EOD
amplitude in the daytime (see Section 2.05.2.4.4). With one
exception (Gymnarchus niloticus), all mormyroids produce
2.05.2 EOD Waveform and Amplitude and Their pulse-type EODs; about half of gymnotiform species have
Control by Electrocytes pulse-type EODs and the other half have wave-type EODs.
The shape of the EOD waveform is determined by the
In most electric fish, the EOD is generated from muscle-derived morphological and electrophysiological properties of the elec-
cells in the electric organ termed electrocytes. Like muscle cells, trocytes. The number of phases and their polarities depend on
they generate action potentials, but unlike muscle cells, they fixed features of the electric organ such as the site(s) of innerva-
fire in near-synchrony and do not contract. Because the electro- tion on the electrocytes, the number of electrically excitable
cytes lie in columns in series, the voltage of the action poten- faces, the geometry of the electrocytes, the location of electro-
tials sum to create a signal strong enough to form an electric cytes across the body, and the coordination of neural inputs
field that extends several body-lengths into the water. along the length of the electric organ (Rodríguez-Cattáneo
The shape of the EOD is species-specific and is categorized et al., 2013). Variation within a species, such as sex differences
into two groups. Pulse-type EODs are generally short- or individual differences, depends on more subtle variation in
duration (0.2–4 ms and rarely up to 20ms) electrical waves electrophysiological properties of the action potentials gener-
that are emitted by a fish at variable and relatively low rates ated by each electrocyte face (see Section 2.05.2.4.2).
(5–50 Hz) (Figures 2 and 3). Wave-type EODs are generally
longer in duration but, within an individual fish, are emitted
2.05.2.1 Generation of the EOD Waveform
at a very constant baseline rate (Figure 4). However, across
species with wave-type EODs, the baseline EOD frequency 2.05.2.1.1 Pulse Fish
varies broadly (60–1600 Hz). (For historical reasons, the Among species with pulse-type EODs, the EOD differs in the
pace of the EOD in pulse fish is usually termed ‘EOD rate’ while number and polarity of their phases and by the amplitude
that of wave fish is termed ‘EOD frequency.’ We keep that and duration of each phase (Figures 2 and 3). The simplest
convention below, though these terms are essentially synony- EOD is a monophasic pulse. This is generated by electrocytes
mous, given that both are controlled by the rhythmic firing with one innervated face that generates an action potential
of the same hindbrain PN (see Section 2.05.3).) and another face that is electrically unexcitable (Bennett, 1961;
Pulse and wave EODs each have their own advantages and Figure 5(a)). Upon depolarization by synaptic input, the inner-
disadvantages. On one hand, wave-type EODs allow a more vated face fires an action potential during which positive current
rapid sampling rate – up to 1600 Hz in some apteronotids – (Naþ ions) enters the innervated face. This causes positive
than pulse-type fish. On the other hand, pulse-type fish can
discharge at low baseline rates and rapidly increase EOD rate (a) Monophasic EOD pulse
when they need information faster. This also lets them regulate
energy expenditure on an as-needed basis and maintain a very

Sternopygus macrurus EOD range

50−200 Hz (b)
1 Diphasic EOD pulse 1
Eigenmannia virescens

250−500 Hz

Apteronotus lephorhynchus
2

600−1100 Hz

Apteronotus albifrons 2

800−1250 Hz Figure 5 Generation of electric organ discharge (EOD) pulses.


(a) A monophasic EOD pulse is generated by current flow into the
10 ms innervated face of an electrocyte and out the opposite face. This
generates a pulse as in the right-hand side of the figure. (b) The first
Figure 4 Wave-type electric organ discharges (EODs) from four phase of a diphasic EOD pulse is generated similarly. However, if the
South American species. From Smith, G.T., 1999. Ionic currents that uninnervated face is excitable, inward current may then flow into it
contribute to a sexually dimorphic communication signal in weakly elec- and out the innervated face. This results in the second phase of the
tric fish. J. Comp. Physiol. A 185, 379–387. EOD as indicated on the right-hand side of the figure.
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 73

current (probably Kþ ions) to exit the opposite face. If an electro- (a) Pacemaker nucleus
cyte is innervated on its posterior face, then current flows in that Electric organ
face and out the anterior face. The net positive current from all
the simultaneously active electrocytes flows toward the head
along the length of the electric organ into the water, and back
(b)
into the fish’s tail, resulting in a head-positive pulse.
In species that produce a biphasic discharge, both faces of Pacemaker
nucleus
the electrocyte are excitable. First, the innervated face fires an Spinal electromotoneurons
action potential during which current flows in one direction
Electrocytes
(Figure 5(b)). The flow of current through the other membrane
depolarizes it, causing an action potential; current then flows in (c)
the opposite direction in the organ. The alternate flow of High EOD frequency
current headward and then tailward gives the EOD its biphasic
shape (Bennett, 1961). Pacemaker
The waveform of the pulse EOD can even be tri- or quadri-
phasic (Figure 3). Multiphasic EOD pulses occur in both gym-
EOD
notiform and mormyrid fishes, although the cellular
mechanisms responsible for their generation differ in the two 3 ms
Low EOD frequency
groups (Hopkins, 1999; Pedraja et al., 2014; Pereira et al.,
2007; Rodriguez-Cattaneo et al., 2008). Mormyrid electrocytes Pacemaker
are uniform, flattened, pancake-like discs restricted to and
tightly stacked in the tail peduncle (the part of the tail just ante-
rior to the anal fin). Distinct phases of a multiphasic EOD are
EOD
generated as current propagates along stalks that penetrate (tri-
phasic) and, in some species, repenetrate (quadriphasic) the 12 ms
flattened electrocyte. Thus, the mormyrid electric organ acts
Figure 6 Relationship between electric organ discharge (EOD)
as a localized current source approximating a dipole producing
frequency and electric organ (EO) pulse duration in gymnotiform wave
a simple field that is essentially the same close to and far away
fish. (a) The location of the pacemaker nucleus and the electric organ.
from the fish (i.e., in near and far fields). (b) The output neurons of the pacemaker nucleus (the relay cells)
In gymnotiform pulse fish, the electrocytes are boxlike cells synapse on electromotor neurons and these synapse on the cells of the
surrounded by ample extracellular space that run in long tubes electric organ, the electrocytes. (c) Pacemaker firing frequency and
of connective tissue along the whole length of the long knife- electric organ pulse duration must be precisely correlated to produce
like body. These tubes or groups of tubes span different lengths, a sinusoidal EOD as in the gold-lined black knifefish (Sternopygus
the electrocytes in them may vary locally in their properties macrurus). Note the difference between an individual with a high EOD
(e.g., size or innervation pattern), and the neural inputs to frequency (such as a female) and one with a low EOD frequency (such
them may arrive at different times to stagger the discharge of as a male). From McAnelly, L.M., Zakon, H.H., 2000. Coregulation of
voltage-dependent kinetics of Naþ and Kþ currents in electric organ.
different EOD components. These complexities cannot be
J. Neurosci. 20, 3408–3414.
modeled as a simple dipole; they produce an EOD that differs
in its waveshape with time and along the body, and in the
near and far field (Pedraja et al., 2014). discharges. Some apteronotids produce EOD frequencies in
excess of 1 K Hz. Their electromotor system is electrotonically
2.05.2.1.2 Wave Fish coupled from the PN to the electromotor neurons (Pappas
A wave-type EOD is essentially a pulse-type EOD with an et al., 1975; Waxman et al., 1972). Apteronotus larvae begin
extremely regular discharge (Figure 4). If the duration of the life with a myogenic electric organ, but it degenerates within
EOD pulses matches the interpulse interval, the EOD will be 20 days of hatching, concurrent with a developmental increase
a periodic and nearly sinusoidal wave. In wave-type fish that in EOD frequency (Kirschbaum, 1977; Kirschbaum and
possess myogenic electric organs, the EOD pulse is monopha- Westby, 1975). The loss of the myogenic electric organ elimi-
sic, and the electrocytes have a single active face. The frequency nates the one obligatory chemical synapse in the electromotor
of a wave-type EOD is determined by the firing rate of the PN. circuit that might fail at these high operating frequencies.
However, in order for the waveform to approximate a sine Although the EOD frequency ranges of some apteronotids
wave, the electric organ pulse duration must also vary. So, for overlap, the waveform of each EOD cycle may differ among
example, the EOD frequency of Sternopygus sp. varies from 50 species, with some having a number of phases (Kramer et al.,
to 200 Hz while electric organ pulse duration varies inversely 1981; Turner et al., 2007).
from 12 to 3 ms (Figure 6). These two parameters covary so
that the fish with the lowest EOD frequency has the longest
2.05.2.2 EOD Waveform: Communication Function
pulse (Mills and Zakon, 1987, 1991). Both independent
parameters are sensitive to hormonal modulation (see Sections One important communication function of the waveform is to
2.05.2.4.1 and 2.05.2.4.2). convey information about species identity. EOD waveforms are
Fish in the family Apteronotidae are the only electric fish species-specific, and behavioral tests show that they are suffi-
with a neurogenic electric organ, and they all emit wave-type cient for species recognition (Hopkins and Bass, 1981;
74 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

Hopkins, 1976, 1981). Species-specific EODs are important for of the pattern in which the discharges are emitted. The EOD
reproductive isolation. Among the gymnotiformes, each wave pulse is exceptionally brief in this species (85–90 ms in both
species may ‘broadcast’ over a specific range thereby enjoying sexes), perhaps too brief to reliably resolve sex differences.
a ‘window’ with minimal interference from other species The correct pattern of interpulse intervals in a discharge may
(Hopkins, 1976, 1988). In areas of diverse species assemblages, be necessary for females to treat the pulse as a courtship signal
some species (especially apteronotids) may utilize similar EOD (Crawford, 1991, 1992; Machnik and Kramer, 2008; Wong and
frequency ranges. Nevertheless, the EODs are potentially Hopkins, 2007).
discriminable by the shape of the pulse of each cycle (Turner What is the function of sex differences in EOD waveform?
et al., 2007). Pulse-type species easily discriminate waveform EODs from males are likely more ‘stimulatory’ to females
differences between conspecific and heterospecific EODs. than those of females or juveniles. For example, the longer
In addition to these interspecific differences in waveform, duration of the second phase of the EOD pulse in male pintail
most electric fish species also show individual and sexual differ- knifefish (B. gauderio) makes the signal waveform asymmet-
ences in EOD waveform within the species (Figures 2 and 3). rical. This results in a brief DC component to the signal that
They have an exquisite capacity for detecting fine waveform stimulates the ampullary electroreceptors of a female (Metzner
differences as shown by laboratory conditioning experiments and Heiligenberg, 1991). Presumably the more dimorphic is
using real or synthesized EOD stimuli (Kramer and Otto, the second phase of the EOD, the more DC is generated, and
1991; McGregor and Westby, 1992). this may be attractive to females. Unfortunately, it also makes
Intraspecific variation in waveform is used in the context of the EOD pulse more detectable by electroreceptive predators
social communication (McGregor and Westby, 1992; Carlson, such as catfish. Indeed, male pintail knifefish are more likely
2000) and may be the basis for mate choice. When female than females to have a wounded or regenerated tail, presum-
pintail knifefish (B. gauderio) are presented males of different ably lost to predation (Hopkins et al., 1990). In addition,
sizes, they preferentially associate with larger, sexually mature sexual dimorphism of EOD waveform, as any other secondary
males, whose EOD pulses are longer in duration than those sexual character, likely acts as an honest proxy of male quality.
of smaller, immature males. This choice indicates a sexual Indeed, in B. gauderio, pulse duration is a good indicator of
attraction since the females often spawn with them. Interest- circulating androgen levels and gonad size in males (Gavassa
ingly, females spend less time near sexually immature males et al., 2011).
than the empty chamber (Curtis and Stoddard, 2003). This is
not surprising given the propensity of gymnotiform pulse fish
2.05.2.4 EOD Waveform and EOD Amplitude: Hormonal
to spread out by a few meters in their natural habitat (Hopkins,
Regulation
1974a,b; Westby, 1988; Crampton, 1998).
2.05.2.4.1 Effects of Exogenous Steroids on EOD Waveform
and Electric Organ Morphology
2.05.2.3 EOD Waveform and Electrocytes: Sexual
Many studies have demonstrated that exogenous sex steroids
Dimorphism
modify the EOD waveform in ways that are consistent with
Most pulse fish possess sexually dimorphic EODs, although sexual dimorphism in waveform. In all species of pulse fish
some are monomorphic (Crampton et al., 2013). In a case of tested to date, including both gymnotiforms and mormyrids,
apparent convergence, the males of species with myogenic androgen (testosterone or nonaromatizable androgen) treat-
EODs (i.e., not apteronotidae) produce pulses that are longer ment to juveniles of both sexes and castrated males broadens
in duration than those of females. These patterns occur in the pulse, mimicking the naturally occurring longer pulse dura-
both Gymnotiformes and Mormyrids and likely arose through tion in males (Bass and Hopkins, 1983; Freedman et al., 1989;
evolutionary convergence (Figures 2 and 3; Carlson et al., Hagedorn and Carr, 1985; Landsman and Moller, 1988; Mills
2000; Hanika and Kramer, 2005; Hopkins et al., 1990; and Zakon, 1991; Silva et al., 1999; Figure 7). Similar results
Hopkins, 1974a,b; Mills and Zakon, 1987). occur when females are treated with the aromatizable androgen
The ability of electric fish to perceive and respond to these testosterone, indicating that the effects are activational. Since
sex differences in the EOD waveform has been amply demon- estrogens do not affect pulse-type EODs when given alone or
strated in numerous species. For example, male fish court in combination with androgens, testosterone likely acts
female, but not male, EODs or EOD mimics in playback exper- through androgen receptors rather than binding to estrogen
iments (Hopkins and Bass, 1981). Conversely, females respond receptors after aromatization (Hagedorn and Carr, 1985;
preferentially to male versus female EOD pulses (Shumway Landsman and Moller, 1988).
and Zelick, 1988). In some pulse fish, the long-term actions of sex steroids
Species differ in which components of the EOD are used for affect only a portion of the EOD waveform. For example, in
sex recognition. For example, in many mormyroids, the EOD B. gauderio and other species, androgen treatment broadens
pulse is longer in males than in females, and playback experi- only the second phase of the EOD pulse (Figure 7). This
ments show that EOD pulse duration is the critical variable suggests that changes in ion currents are restricted to the ante-
for sex recognition (Hopkins and Bass, 1981; Machnik and rior face of the electrocyte since this is the face that generates the
Kramer, 2008). By contrast, male Pollimyrus adspersus (formerly second phase of the EOD (Hagedorn and Carr, 1985; Goldina
classified as Pollimyrus isidori; Crawford, 1997), another mor- et al., 2011; Markham and Stoddard, 2013).
myrid, ignore the EOD waveform, even though it possesses In most wave-type gymnotiforms, males have a longer EOD
a subtle sexual dimorphism in pulse amplitude (Crawford, pulse duration than females, and pulse duration is closely tied
1992). Instead, it attends to sex differences in the regularity to EOD frequency. In these species (e.g., Eigenmannia and
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 75

(a)
2.05.2.4.2 Effects of Exogenous Steroids on Electric Organ Ion
Currents
The cellular mechanisms underlying steroid action on EOD
waveform are best understood in the wave-type gymnotiform
Sternopygus macrurus, the gold-lined knifefish (Hopkins,
1974a,b). As discussed above, the electrocytes of Sternopygus
generate action potentials that vary from 3 ms, in a fish with
Pretreatment
a high EOD frequency such as a sexually mature female, to
Posttreatment 12 ms, in a fish with a low EOD frequency, such as a mature
male (Figure 6). Action potential duration is lengthened
(b)
(masculinized) over days to weeks by chronic androgen treat-
Juvenile/DHT
ment (Mills and Zakon, 1991; Figure 8).
At the cellular level, androgens broaden action potentials by
influencing the ionic currents of the electrocyte. Three different
ion currents are present in the Sternopygus electrocyte: a voltage-
0d 10 d dependent Naþ current, a delayed rectifying Kþ current, and an
inward rectifying Kþ current (Ferrari and Zakon, 1993). The
inward rectifying Kþ current sets the resting potential while
the Naþ current and delayed rectifying Kþ current shape the
EOD pulse. The rate at which the Naþ current turns off (inacti-
Figure 7 Androgens broaden the electric organ discharge (EOD) pulse
of both gymnotiform and mormyroid pulse-type fish. (a) The EOD pulse vates) and the delayed rectifying Kþ current turns on (activates)
of a male pintail knifefish (Brachyhypopomus pinnicaudatus) before and varies systematically with EOD frequency and pulse duration.
15 days after implantation with a capsule containing testosterone (Silva Individuals with low EOD frequencies have currents that acti-
et al., 1999). (b) Broadening of the EOD pulse of a juvenile mormyrid vate and inactivate slowly, while fish with the highest EOD
Brienomyrus brachyistius 10 days after implantation with DHT. Adapted frequencies have currents with the fastest rates (Ferrari et al.,
from Bass, A.H., Hopkins, C.D., 1985. Hormonal control of sex differ- 1995; McAnelly and Zakon, 2000; Figure 8). In males, the
ence in the electric organ discharge (EOD) of mormyrid fishes. slow inactivation of the Naþ current and slow activation of
J. Comp. Physiol. A 156, 587–604. the Kþ current means that Naþ flows into the cell unopposed
by an efflux of Kþ thereby producing a long duration action
Sternopygus), systemic androgen treatment broadens EOD dura- potential.
tion over a 2-week period while simultaneously lowering EOD Treatment of fish with the nonaromatizable androgen 5-a
frequency (Meyer, 1983; Mills and Zakon, 1987; Dunlap and dihydrotestosterone (DHT) slows inactivation rate of the Naþ
Zakon, 1998). However, in Sternopygus, localized androgen current and the activation rate of the delayed rectifying Kþ
treatment of the electric organ broadens pulse duration without current (McAnelly and Zakon, 2007). As described above, these
influencing EOD frequency (Few and Zakon, 2001). Thus, changes lengthen electrocyte action potential duration and
androgens exert a concerted but independent influence on broaden the EOD pulse. Conversely, estradiol speeds up the
the electric organ and PN. How androgens coordinate their inactivation of the Naþ current, which should shorten EOD
effects on two different target cell types of the electromotor pulse duration (Dunlap et al., 1997). It is not known if estra-
system raises the general question of how a hormone that diol also acts on Kþ currents. The actions of steroid hormones
acts on multiple sites in a neural network exerts a coordinated on these currents are genomic, as they take days to manifest.
action. The most likely targets for these hormonal influences on
In Sternopygus, estrogen implants in juvenile fish of both waveform are the ion channel genes themselves. Both Naþ
sexes raise (feminize) EOD frequency (Meyer, 1983; Dunlap and Kþ channel genes have been cloned from the electric organ.
et al., 1997). In addition, treatment of intact females or gonad- Two Naþ channel genes (Nav1.4a and Nav1.4b) and an acces-
ectomized fish of both sexes with human chorionic gonado- sory Naþ channel subunit (b1) are expressed in the electric
tropin (hCG) powerfully increases EOD frequency (Zakon organ (Liu et al., 2007, 2008). One gene, Nav1.4a, is expressed
et al., 1990). It is not known whether hCG acts directly on uniformly across the range of electrocyte action potential dura-
the pacemaker and electrocytes or whether it acts indirectly tions. The other two genes, Nav1.4b and the b1 subunit, are
by stimulating the release of another hormone from a nongona- expressed at higher levels in fish with narrow EOD pulses
dal source. (females) than in those with long EOD pulses (males);
The size and shape of the electrocytes is sexually dimorphic Nav1.4b and the b1 subunit are downregulated by androgens.
in some species of pulse fish in both orders (Hagedorn and Compared to expression of Nav1.4b alone, coexpression of the
Carr, 1985; Bass et al., 1986; Bass, 1986b; Freedman et al., Nav1.4b Naþ channel with the b1 subunit in Xenopus oocytes
1989). In some cases (e.g., B. gauderio), this is so pronounced increases the inactivation rate of the Naþ current. (Nav1.4a
that sexes differ in the length or thickness of the tail (Hopkins has not yet been expressed in oocytes.) In the current model
et al., 1990). In mormyrids, electrocytes in males are thicker (Figure 9), Nav1.4a, which is expressed equally in all electro-
and larger, although this does not result in a visible dimor- cytes and is the dominant Naþ channel gene expressed in
phism in the tail. As expected, treatment of females or sexually males, produces a Naþ current with a slow inactivation rate.
immature males with androgens induces these morphological As the level of Nav1.4b increases, relatively more of the Naþ
changes in electrocytes. current will be transmitted through a channel with a rapidly
76 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

(a) (b) 140 Hz (c)


π = 0.87 ms
145 Hz Long spike
21%

Change in duration (ms)


2
15% pulse
60 nA 15%
1
105 Hz
π = 0.87 ms
101 Hz 0

−4% −3%
200 nA −1
DHT Control
65 Hz 55 Hz
π = 0.87 ms
20 mV
5 ms
60 nA
2 ms

þ
Figure 8 Electrocyte action potential duration and Na current are individually distinct and influenced by hormones. (a) Intracellular recordings
made in electrocytes of high, mid, and low EOD (electric organ discharge) frequency juvenile Sternopygus. Current was injected with one electrode,
and the voltage responses of the membrane were recorded with another. The number to the right of each trace gives each fish’s EOD frequency. Note
the systematic individual variation in action potential duration. (b) Voltage clamp recordings from electrocytes from fish with comparable EOD
frequencies as in ‘a’ show that the Naþ current activates and inactivates at different rates. EOD frequency and the exponential time constant of decay
of the current (its rate of inactivation) is given to the right of each family of current traces. (c) Androgens broaden the EOD pulse and the electrocyte
action potential (labeled ‘long spike’). In this experiment recordings of these two parameters are made before and after treatment with DHT or empty
capsules. Note that the control fish show no change in either parameter, but the hormone-treated fish show an increase in both. From Zakon, H.H.,
1999. Sex steroids and weakly electric fish: a model system for activational mechanisms of hormone action. In: Matsumoto, A. (Ed.), Sexual Differen-
tiation of the Brain. CRC Press, Boca Raton, FL, pp. 95–112.

inactivating current. As levels of the b1 subunit increase in tails) and electrophysiological (longer EOD second phase)
tandem with Nav1.4b, the b1 subunit interacts with and speeds traits that coincide with gonadal maturation (Silva et al.,
up the inactivation rate of both Naþ channels and further 2002; Quintana et al., 2004) and elevated plasma 11kT
increases the inactivation rate and narrows the duration of (Dunlap et al., 2011b). An experimental elevation in water
each EOD pulse. temperature causes gonadal recrudescence and increased
Less is known about the molecular control of Sternopygus Kþ androgen levels in nonbreeding males, and experimental
current. Levels of mRNA for certain members of the Kv1 family androgen treatment enhances morphological and electrophys-
(Kv1.1a, Kv1.2a) are abundant in the electric organ of fish with iological traits characteristic of males during the breeding
short pulses and low in the electric organ of fish with long season (Silva et al., 1999; Quintana et al., 2004).
pulses (Few and Zakon, 2007). The level of Kv1.2b mRNA
does not vary with EOD pulse duration. Furthermore, the tran-
script abundance of Kv1.1a and Kv1.2a is suppressed by DHT 2.05.2.4.4 EOD Waveform: Diel, Social, and Stress-Induced
while that of Kv1.2b is unaffected. These genes have not yet Modulation and Its Hormonal Regulation
been expressed in oocytes, so the biophysical properties of the The changes in EOD waveform induced by exogenous steroids
channel proteins are currently unknown. However, the current described above occur over days to weeks. However, changes in
model suggests that the androgen-dependent subunits have EOD waveforms may also occur within minutes in freely
faster activation rates, so Kþ currents will be slower in males. moving fish. Hagedorn and Heiligenberg (1985) first reported
that the EOD amplitude of the dominant male Eigenmannia in
a community tank increases in amplitude (i.e., gets very loud
2.05.2.4.3 EOD Waveform: Seasonal Modulation and Its on an audio monitor) on the night of breeding, so much so
Hormonal Regulation that it drowns out the signals of other fish. This was a radical
Sexual dimorphism of EOD waveform cycles seasonally suggestion at a time when it was believed that the characteris-
because it is regulated by seasonal changes in gonadal steroid tics of the EOD were determined by fixed aspects of the
secretion. This was demonstrated by sampling a natural popu- anatomy and physiology of the electromotor system.
lation of B. gauderio at the subtropical edge of gymnotiform Under conditions where accurate EOD recordings can be
distribution in South America, where electric fish and most made in swimming fish, the sexually dimorphic EOD phase of
other freshwater teleosts breed during the austral spring to male B. gauderio increases rapidly in duration and amplitude after
summer (November to February) in association with warm the lights turn off (Franchina and Stoddard, 1998; Figure 10).
temperature and long photoperiod (Silva et al., 2003). In the Such nocturnal excursions occur in isolated males, but are further
nonbreeding season, males and females are indistinguishable enhanced in males exposed to conspecifics behind a mesh
externally. However, during the breeding seasons, males barrier. Stimulus males elicit a greater effect on EOD parameters
exhibit large differences from females in morphological (broad than stimulus females (Franchina et al., 2001). The hormonal
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 77

(a) Total β1 (a)

Expression levels (low to high) smNav1.4bL

Day

smNav1.4a Night

1 ms
(b)
8

Amplitude (mV/cm@10 cm)


2 6

Duration (ms)
(b) EOD frequency (low to high)
Long Short 4
EOD pulse duration
1
Amplitude 2
Electrocyte Na+ current Duration
Light Dark Light
0 0
smNav1.4a smNav1.4bL
Noon Midnight Noon
Time of day
(c)

Figure 9 Model for androgen action in the electric organ of a wave-


type gymnotiform (Sternopygus macrurus). (a) Two sodium channels
(smNav1.4bL; smNav1.4a) and a sodium channel auxiliary subunit
(beta1) are expressed in the electric organ (EO). smNav1.4a is 200 nA
expressed equally in all individuals and is hypothesized to inactivate
(i.e., close after opening) slowly (b). Nav1.4bL and b1 are expressed at 3 ms
high levels in fish (females) with high electric organ discharge (EOD) 8Br-cAM
frequencies and short EOD pulses (right side of the graph); these fish
also have sodium currents that inactivate rapidly. Androgens suppress
expression of Nav1.4bL and beta1 (which act on sodium channels to
further speed inactivation). Thus, males have low levels of these genes
(left side of the graph) but relatively higher levels of smNav1.4a that Figure 10 Diurnal changes in the electric organ discharges (EODs) of
would generate a current that inactivates slowly and lengthens the EO a pulse fish (Brachyhypopomus pinnicaudatus) and its possible biophys-
action potential. ical basis. (a) Traces of EOD waveform during the day and night illus-
trating the nocturnal increases in amplitude of positive and negative
phases of the EOD and duration of the negative phase of the EOD.
action underlying this effect on waveform must be only on the (b) Measurements of peak-to-peak amplitude and the duration of the
anterior membrane of the electrocyte since this is the membrane second phase of the EOD pulse of a male over 24 h. (c) Voltage clamp
that generates the second phase of the EOD pulse. Daily fluctua- recordings of the peak Naþ current from an electrocyte of a wave fish
tions of EOD amplitude in this pulse species suggest that fish (Sternopygus) before and after treatment with 8-bromo-cyclic AMP. Note
optimize their energy expenditure by generating the largest that the amplitude of the Naþ current is enhanced. Such an increase in
amplitude discharge only during periods of significant social the Naþ current likely increases the current output of each electrocyte
interaction. Two wave-type species, Sternopygus (Markham thereby increasing the EOD amplitude. From (a) and (b) Franchina,
et al., 2009) and Eigenmannia (Markham et al., 2013), also C.R., Stoddard, P., 1998. Plasticity of the electric organ discharge
produce rapid changes in EOD amplitude in response to social waveform of the electric fish Brachyhypopomus pinnicaudatus: I. Quantifi-
cation of day-night changes. J. Comp. Physiol. A 183, 759–768;
or circadian stimuli, indicating that such modulation may be
(c) from McAnelly, L.M., Zakon, H.H., 1996. Protein kinase A activation
widespread among gymnotiforms. increases sodium current magnitude in the electric organ of Sternopy-
The hormonal link between environmental stimuli and gus. J. Neurosci. 16, 4383–4388.
electrocyte excitability are the melanocortin peptides (alpha
melanocyte stimulating hormone (a-MSH) and/or adrenocor-
ticotropic hormone (ACTH)) that act via melanocortin recep- result of its activation is to increase the exocytosis of constitu-
tors on electrocytes. On the cellular level, the melanocortin tively trafficked Naþ channels, which increases electrocyte
receptor obligatorily activates a G-protein-coupled pathway Naþ current and, therefore, EOD amplitude (Markham and
that increases cAMP thereby activating protein kinase A. Stoddard, 2005; Markham et al., 2009; McAnelly et al., 2003;
The target of this kinase is not yet identified. However, the McAnelly and Zakon, 1996; Figure 11). In B. gauderio, females
78 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

Figure 11 Summary diagram of how activation of melanocortin receptors results in trafficking of Naþ channels into the electrocyte membrane to
increase electric organ discharge (EOD) amplitude. From Markham, M.R., McAnelly, L.M., Stoddard, P.K., Zakon, H.H., 2009. Circadian and social
cues regulate ion channel trafficking. PLoS Biol. 7, e1000203.

given androgen pretreatment show a stronger melanocortin- These two species (E. virescens and S. macrurus) also have
induced EOD modulation (Allee et al., 2009; Goldina et al., different endocrine responses to metabolic and social stress.
2011). This is an example of a long-term steroidal process In E. virescens, cortisol, an important stress hormone in many
enhancing a short-term process. fish, does not appear to play a role in the response to food
In addition to the rapid daily changes in EOD amplitude, deprivation. Endogenous cortisol levels do not change with
social and metabolic stress can modify EOD waveform or food deprivation, and cortisol treatment does not influence
amplitude. For example, socially stressing mature male B. occi- EOD amplitude. Instead, the response appears mediated by
dentalis causes the sexually dimorphic portion of the EOD pulse the peptide leptin: systemic leptin treatment increases EOD
to decrease in duration. When two males of comparable size amplitude in starved, but not fed fish (Sinnett and Markham,
are placed together in a tank and compete for a single hiding 2015). By contrast, in B. gauderio, exogenous cortisol adminis-
place, the winner shows a similar decrease of its EOD pulse tration reduces EOD amplitude and duration, but endogenous
duration and its EOD amplitude falls. Both EOD changes occur cortisol levels do not rise under food limitation or social chal-
within 2 h of the interaction (Hagedorn and Zelick, 1989). lenge. When starved, males respond to social challenge by
During the annual rain cycles of the tropics, fish are increasing androgen levels and enhancing EOD amplitude
commonly exposed to periods of low oxygen and food avail- and duration (Gavassa and Stoddard, 2012). Socially chal-
ability and thereby experience metabolic stress (Crampton, lenged males simultaneously elevate testosterone and cortisol
1998). To conserve energy during hypoxic stress, electric fish in proportion to signal amplitude. Thus, B. gauderio appears
decrease EOD amplitude (Crampton, 1998; Reardon et al., to protect its cortisol-sensitive electric advertisement signal by
2011). Presently, there is little information whether this is increasing food intake, limiting cortisol release and offsetting
hormonally driven or a direct effect of oxygen or ATP concen- signal reduction from cortisol with signal-enhancing andro-
tration on the ion channels of the electrocytes (Markham gens. The role of leptin in this species is not yet known.
et al., 2013).
However, researchers have examined the hormonal basis
of the EOD response to another metabolic stress, food 2.05.3 EOD Rate/Frequency and Its Regulation
shortage. One expected energy-conserving response to food by the Pacemaker Nucleus
deprivation is also a decrease in EOD amplitude. Indeed,
this occurs in the wave fish Eigenmannia virescens (Sinnett (By convention, we refer to the pace of the EOD as ‘EOD rate’
and Markham, 2015). Surprisingly, under similar condi- in pulse fish and ‘EOD frequency’ in wave fish. However, these
tions, the pulse species B. gauderio increases EOD amplitude terms refer to the same phenomenon and are essentially
(Gavassa and Stoddard, 2012). This species difference seems synonymous.)
paradoxical until the life histories of the two species are EOD rate/frequency conveys the full range of information
compared. Eigenmannia virescens may live for a few years encoded by the electric channel of communication, including
and breed each year (iteroparous), whereas, under some species identity, sexual identity, arousal, social status, and
circumstances, it appears that some B. gauderio may live motivational state. In addition, a broad range of environ-
less than a year and breed only once (semelparous). In mental, social, and physiological factors impact EOD rate/
principle, iteroparous species save their energy ‘for a better frequency (reviewed in Silva et al., 2007). For example, in all
day’ while semelparous species ‘go for broke’; further gymnotiform species tested, EOD rate/frequency is temperature
research is necessary to test this hypothesis broadly among dependent (Enger and Szabo, 1968; Boudinot, 1970; Dunlap
in electric fish. et al., 2000; Ardanaz et al., 2001). In many pulse-type species
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 79

of both orders, EOD rate increases during the night when fish When tested in the most basal conditions (isolated and
become more active. In pulse type fish, sudden or novel stimuli immobile during daytime), AVT treatment increases EOD rate
in the environment also cause changes in EOD rate of pulse- in B. gauderio and G. omarorum (Perrone et al., 2010, 2014).
type fish, a reaction known as the novelty response. Stimuli However, the effect of AVT on EOD rate differs between species,
activating Mauthner cells elicit an abrupt and prolonged depending on the degree of sociality. The effect is more
increase in EOD rate (Falconi et al., 1995). Finally, in the pres- pronounced and persistent in the gregarious B. gauderio than
ence of a nearby interfering fish, most gymnotiform wave-type in the solitary G. omarorum. In vitro electrophysiological record-
species shift their EOD frequency (jamming avoidance ings of the PN demonstrate that these different sensitivities
response; (Heiligenberg, 1986). Thus, given all these influences result from differential effects of AVT on the PN itself (Perrone
on EOD rate, it is probably rare that a fish discharges at its EOD et al., 2010, 2014). Furthermore, immunolabeling of preoptic
at basal rate. AVT neurons confirms the presence of hindbrain AVT projec-
During breeding, complex and precise information is tions close to the PN that might constitute the anatomical
encoded in the EOD rate, and different kinds of rate changes substrate for AVT influences on PN activity. Differences in the
occur in the context of reproduction. Several wave-type species spatial distribution of AVT projections between species might
show sexually dimorphic EOD frequencies used for mate recog- account for interspecific differences in AVT modulation
nition and selection (see Section 2.05.3.2.1). Furthermore, in (Perrone et al., 2014).
both wave- and pulse-type species, males and females modu-
late EOD rate to produce transient social signals that participate 2.05.3.1.2 Diel Influence
in courtship, spawning, and aggression (see Section 2.05.4.1). EOD rate increases during nocturnal arousal have been re-
In all electric fish, EOD rate/frequency is controlled by the ported in several species of the genera Brachyhypopomus and
medullary PN (Figure 1). Indeed, electric fish are one of the Gymnotus (Silva et al., 2007). Although the circadian system
few vertebrate groups in which the control of specific behav- of weakly electric fish has not yet been fully characterized,
ioral displays can be traced reliably to a single conspicuous these daily variations in EOD rate probably constitute a circa-
hindbrain nucleus (Bennett, 1971; Ellis and Szabo, 1980). dian rhythm of electric behavior. This is based on the
In this section, we review examples of how EOD rate varies following evidence reported recently in both B. gauderio and
by season and sex, how hormones modify EOD rate, and G. omarorum: strong daily variations of EOD rate (1) can be
how rate variations are generated at the cellular level in the replicated in isolated fish in laboratory controlled conditions
PN. These rate changes are particularly meaningful in species under a 12:12 light/dark photoperiod; (2) persist, albeit
with regular discharge rates; otherwise, the information they shifted, when placed in constant darkness; (3) depend on
carry could be masked if the background rate varied widely. the timekeeping hormone melatonin; and (4) do not depend
Since all mormyroids (except Gymnarchus) emit pulse-type on fish locomotion, which is more frequent during the
EODs with an irregular pattern of emission, we limit this nocturnal active phase (Migliaro and Silva, 2016a,b). Interest-
section to studies on Gymnotiformes. In addition, we focus ingly, this rhythm of EOD rate occurs in the wild despite the
on variation in EOD basal rate, and not on the emission of fact that fish are in constant darkness underneath aquatic
transient EOD rate modulations, which is covered in Section plants during the 24-h cycle (Migliaro and Silva, 2016a,b).
2.05.4. The important conclusion emerging from these studies This suggests that other environmental cues, e.g., temperature,
is that the PN is an extremely plastic medullary central pattern social interaction, might be crucial for entraining circadian
generator that responds to higher brain regions and a broad rhythmicity in weakly electric fish.
set of neuromodulators to adapt its function to diverse The control of circadian rhythms of electric behavior is
contexts. embedded in the complex integrative control of the brain.
In Brachyhypopomus, seasonal changes and social stimuli
modulate the nocturnal increase of EOD rate, which is
2.05.3.1 EOD Rate in Pulse Fish: Social and Diel Influences
more pronounced in breeding than in nonbreeding fish (Silva
and Their Hormonal Regulation
et al., 2007) and more in socially interacting fish than in iso-
2.05.3.1.1 Social Influences lated fish (Silva et al., 2007; Stoddard et al., 2007). Also inter-
In pulse fish, changes in EOD rate signal dominance status in esting, species differ in the nocturnal increase of EOD rate in
agonistic interactions. In B. gauderio dominant males show parallel with their different social structure. The nocturnal
higher EOD rates than subordinate males after reproductive increase is mild and transient in the solitary G. omarorum,
aggression, while subordinates of Gymnotus omarorum reduce whose individuals rarely interact, and strong and persistent
their EOD rate after nonbreeding inter- and intrasexual in the gregarious B. gauderio, in which social stimuli further
agonistic encounters (Silva et al., 2013). Interestingly, the enhances the nocturnal increase (Migliaro and Silva,
hypothalamic neuropeptide arginine vasotocin (AVT), an inte- 2016a,b). AVT, known to support species-, phenotype- and
grative mediator of social behavior, regulates this phenotype- context-dependent actions, has been postulated to mediate
dependent variation in EOD rate. In B. gauderio, the electrical this species-specific effect. Although AVT acts directly on the
dominance is abolished when a competitive antagonist of PN of both species to enhance EOD rate in a species-
AVT V1a receptors (Manning Compound) is administered to specific manner (Perrone et al., 2010, 2014), it is not respon-
the potential dominant fish prior to the encounter (Perrone, sible for the nocturnal increase of EOD rate in isolated
2012). In G. omarorum, AVT treatment to potential subordi- individuals (Migliaro and Silva, 2016a,b). However, AVT
nates before the contest enhances their electric submission mediates the social component of EOD rate nocturnal
(Perrone, 2012). increase in B. gauderio (Perrone et al., 2014), suggesting
80 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

intricate relationships between the AVTergic and the melato- (a) 1100
nergic system in the coupling of timekeeping and social
behavior in electric fish.

1000

EOD frequency (Hz)


2.05.3.2 EOD Frequency in Wave Fish: Role in Social Behavior
and Its Hormonal Regulation
2.05.3.2.1 Social Behavior 900
EOD frequency signals sex among several gymnotiform wave-
type fish. In most species (e.g., Sternopygus macrurus, E. virescens,
and Apteronotus albifrons), EOD frequency is lower in males 800
than in females (Figures 6(c) and 12(b); Hopkins, 1972;
Dunlap et al., 1998). Some species have sexually monomor-
phic EOD frequencies (e.g., Adontosternarchus devenanzii, Para- 700
pteronotus hasemani, and Apteronotus bonapartii; Zhou and 0 10 20 30 40
Smith, 2006, Ho et al., 2010; Petzold and Smith, 2016), and Body mass (g)
in a few species (e.g., A. leptorhynchus and Apteronotus rostratus),
males have higher EOD frequencies than females (b)
(Figure 12(a); Hagedorn and Heiligenberg, 1985). The ances- 1200
tral and common pattern is for males to have lower EOD
frequencies than females, with the alternative (males with
higher EOD frequencies) evolving as a case of reversed sexual

EOD frequency (Hz)


1100
dimorphism (Dunlap et al., 1998; Dunlap and Zakon, 1998).
In addition to signaling sexual identity, EOD frequency can
also convey information about social status. In a tank commu-
nity of Eigenmannia, the largest male possessed the lowest EOD 1000
frequency among the males, and the largest female had the
highest EOD frequency among the females (Hagedorn and
Heiligenberg, 1985). The largest fish of both sexes became 900
dominant; they bred with each other and prevented other
fish from breeding. When a playback recording of the high-
frequency EOD of the dominant male was substituted for 800
him during courtship, the female continued to respond and 0 20 40 60 80 100 120 140
lay her eggs. Body mass (g)
Similar relationships between EOD frequency and domi-
nance have been found in studies of brown ghost knifefish Figure 12 Electric organ discharge (EOD) frequencies of two related
gymnotiform wave-fish with sex differences in EOD frequency in
(A. leptorhynchus). In small groups (three males, one female)
opposite directions. (a) Brown ghost females are lower in frequency
the largest male with the highest EOD frequency (this species
than males. (b) Black ghost females are higher in frequency than
has a reversed sexual dimorphism) bred with the female males. From Zakon, H.H., Dunlap, K.D., 1999. Sex steroids and
(Hagedorn and Heiligenberg, 1985). In larger groups communication signals in electric fish: a tale of two species. Brain
(12 males, 12 females) housed with a limited number of Behav. Evol. 54, 61–69.
shelter tubes, males with the highest EOD frequency controlled
the most desired shelter tubes (Dunlap and Oliveri, 2002).
Without playback experiments it is not possible to know treatment with exogenous steroids mimics the endogenous
whether the EOD frequency was the basis for differences in sexual dimorphism. In Sternopygus and Eigenmannia, androgen
social status. treatment lowers EOD frequency over a 2-week period (Meyer,
EOD frequency can also signal within-sex variation in 1983; Mills and Zakon, 1987; Dunlap and Zakon, 1998). While
a species with different male morphs (Sternarchogiton nattereri; decreasing EOD frequency, androgens also lengthen EOD dura-
Fernandes et al., 2010). In this species, toothed males, tion, and with this combination, the waveform stays the same
compared to toothless males, have larger body size, testes shape. In the brown ghost, A. leptorhynchus, in which males
growth, and higher EOD frequencies in association with higher have higher EOD frequency than females (Figure 12(a)),
levels of 11-kT, even though both male types are reproductively 11kT raises EOD frequency, whereas E2 or T lowers it (Meyer
mature. The association of all these factors not only impact et al., 1987; Schafer and Zakon, 1996). Interestingly, in the
reproductive performance but also may vary in their expression closely related species, A. albifrons, in which males have lower
across seasons in concert with the highly variable environ- EOD frequencies than females (Figure 12(b)), 11kT or dihy-
mental conditions. drotestosterone (DHT) treatment lowers EOD frequency
(Dunlap et al., 1998). The close phylogenetic relationships
2.05.3.2.2 Hormonal Regulation between these two species with opposite androgenic actions
Sexual dimorphism in EOD frequency arises from the differen- upon EOD frequency make them good subjects for studying
tial action of gonadal steroid hormones. In many species, how sexual dimorphisms evolve.
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 81

In many cases (e.g. Sternopygus and Eigenmannia), estrogen cortisol levels do not correlate with EOD frequency, and
implants in juvenile fish of both sexes raise (feminize) EOD exogenous cortisol treatment does not alter EOD frequency.
frequency (Meyer, 1983; Dunlap et al., 1997; Dunlap and However, thyroxine treatment increases EOD frequency,
Zakon, 1998). However, Schaefer and Zakon (1996) found consistent with the role of thyroxine in many rate-
that estradiol reduced EOD frequency in female A. leptorhynchus dependent processes in neurons (Dunlap and Ragazzi,
which is consistent with the observation that females 2015).
have lower EOD frequency in this species. Treatment of intact
females or gonadectomized fish of either sex with
hCG potently increases EOD frequency in Sternopygus (Zakon 2.05.4 EOD Modulations and Their Regulation
et al., 1990), in which females have higher EOD frequencies. by Prepacemaker Nuclei
By contrast, male Sternopygus injected with hCG tandemly
increased 11kT plasma levels and decreased EOD frequency As discussed above, features of the EOD that convey informa-
(Zakon et al., 1990). It is not known whether hCG acts directly tion on species, sex, individual identity, or sexual maturity
on the pacemaker and electrocytes or whether it acts indirectly are stable or change slowly. By contrast, information on moti-
by releasing another hormone from a nongonadal source. vational states that change quickly, particularly during
In several studies, EOD frequency and plasma steroid agonistic and courtship interactions, are signaled by rapid
concentrations have been correlated in natural populations. EOD changes termed EOD modulations. Both wave and pulse
In the field, androgen levels of male S. macrurus correlate posi- fish transiently modify EOD frequency (wave fish) or pulse rate
tively with the degree of testicular development and body (pulse fish) and amplitude (Black-Cleworth, 1970; Kramer,
size and correlate negatively with EOD frequency (Zakon 1979; Zakon et al., 2002) during aggression and courtship.
et al., 1991). However, while all large adult males have high Such EOD modulations occur at greatest rates during the
androgen levels and low EOD frequencies, immature males, breeding season (however, see Gymnotus below).
which have low androgen levels, have either high or low In male–male interactions, aggression can be rather intense,
EOD frequencies. Thus, some factor other than androgen level with fish often chasing and biting each other while they modu-
influences EOD frequency in immature males (Zakon et al., late their EOD to signal dominance or submission. The expres-
1991). Population differences in sexual dimorphism have sion of EOD modulations commonly depends on the stage of
been reported in A. albifrons (Ho et al., 2013). Interestingly, the fight, with modulations changing frequency or form
EOD frequency in a Colombian population is more sexually between the assessment, contest, and resolution phases of the
dimorphic and more sensitive to androgen treatment than encounter (Hopkins, 1974a,b; Hupe and Lewis, 2008; Perrone
the EOD frequency of a Brazilian population. Thus, the evolu- et al., 2009; Batista et al., 2012).
tion of sexual dimorphism in electric behavior at the popula- In all electric fish studied, courtship occurs over a period of
tion level appears related to changes in androgen sensitivity a few nights. Typically courtship activity intensifies each night
across populations. with the greatest activity on the night of spawning. Courtship
Male brown ghosts in the laboratory also show a positive is accompanied by a variety of stereotyped body postures and
correlation between plasma 11kT levels, body size, and EOD movements and variations in the rhythm of the EOD, with
frequency; males with low levels (<2.5 ng ml1) have low each sex usually displaying a different pattern of EOD modula-
EOD frequencies, whereas those with levels from 5 to tion. Spawning occurs in a single bout or, more frequently, in
15 ng ml1 have the highest EOD frequencies (Zucker, 1997; a series of bouts (Hagedorn and Heiligenberg, 1985;
Dunlap, 2002). The role of endogenous sex steroids in control- Henninger, 2015). Below we give an overview of EOD modu-
ling EOD parameters is further supported by the fact that the lations in a broad range of electric fish, and then focus on the
EOD changes in the expected direction following gonadectomy hormonal control of EOD modulations in fish in the family
(Meyer, 1983; Zucker, 1997). Furthermore, in A. bonapartii, Apteronotidae, and in the genera Brachyhypopomus and
EOD frequency is not sexually dimorphic and is not correlated Gymnotus, where the neuroendocrine control has been studied
with plasma levels of androgens in either sex (Ho et al., 2010). in depth.
Because EOD frequency is ultimately set by the activity of
the hindbrain PN, sexual dimorphism of EOD frequency
2.05.4.1 EOD Modulations: Communication Function
most likely depends on the action of steroid hormones on
the firing properties of PN neurons. Androgen receptors have 2.05.4.1.1 Pulse Fish
been identified in PN neurons (Pouso et al., 2010), and andro- During social interactions, the pulse-type gymnotiform B. gau-
gens and estrogens change EOD frequency by directly derio displays three broad classes EOD modulations collectively
changing the rhythmic firing rates of neurons in the PN and/ termed social electric signals (SESs) (Perrone et al., 2009; Silva
or of the spinal electromotor neurons whose axons innervate et al., 2008). Interruptions are near-complete cessations of the
or form the electric organ. The rhythmic firing of the PN and EOD for approximately 100–200 ms. Accelerations are rela-
electromotor neurons is regulated primarily by transient tively small increases in pulse rate with little change in EOD
and persistent sodium and potassium currents (Schaefer and amplitude that last 900ms. Chirps are brief periods (approx-
Zakon, 1996; Smith and Zakon, 2000; Smith, 2006), but the imately 50–300ms) when EOD pulses occur at much higher
effects of gonadal steroids on these conductances have not rates and lower amplitudes than the basal EOD pulse rate.
been clarified so far. Chirps can be classified into four chirp types (A, B, C, and M)
Glucocorticoids do not appear to influence EOD based on the duration, amplitude modulation, and degree of
frequency in Apteronotus (Dunlap et al., 2006). Endogenous symmetry (Figure 13).
82 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

Figure 13 Context-dependent chirping behavior in Brachyhypopomus gauderio. (a) Dyadic male–female electric organ discharge (EOD) recordings
during courtship; the male emits chirps Types A, B, C, while the female maintains its regular discharge (indicated by dots). Chirps exhibited differ-
ences in duration, relative amplitude, time to minimal amplitude, and symmetry among chirp types. (b) Dyadic male–male EOD recordings during
agonistic interactions, the subordinate male turns off its discharge and emits short chirps Type S, while the dominant emits Type M chirps. Dots
indicate the occurrence of subordinates’ EODs. Modified from Perrone, R., 2012. La vasotocina modula el comportamiento social de dos especies de
peces eléctricos con diferente socialidad. PEDECIBA, Universidad de la República, Montevideo.

The production SESs varies by social context, sex, time of swims near his potential nest site next to a plant and emits
day, and season (Perrone et al., 2009; Silva et al., 2008). The pulses at rates of approximately 50–70 pulses/s. When a female
overall rate of SES production is about four times greater in approaches, the pair may engage in duets in which their EOD
male–male dyads than in male–female dyads. Virtually all rates sometimes shift in parallel, and other times mirror each
SESs occur at night, and some are produced differentially other. The male prods the female with his snout or chases
according to the phase of the dark period. For example, type her as his pulse rate increases dramatically in half-second bouts
A chirps are emitted early in the dark phase while types B of up to 200 pulses/s. Eventually the female moves toward
and C chirps are emitted just before sunrise. Finally, fish string the nest plant, and the male begins making courtship chirps
together long bouts and reciprocal exchanges of SESs that toward her (these are distinct from the chirps given in aggres-
might be analogous to the complex songs and duets of birds; sive interactions, called decrement bursts by; (Hagedorn,
however, the communication value of these higher-order 1988). A typical spawning chirp consists of approximately
patterns has not been examined. 25–200 low-amplitude pulses in rapid succession lasting
Male–male aggressive interactions in Brachyhypopomus only 100–200 ms. Males give long trains of chirps during spawning.
occur among breeding males; that is, only males that display The chirping male enters the plant and backs out. Then the
electric courtship toward females engage in agonistic competi- female enters, lays her eggs, and backs out. Finally, the male
tive encounters with other males. The larger resident male in reenters the plants, probably fertilizing the eggs. This cycle
the courtship arena struggles aggressively toward the smaller may be repeated until the female leaves.
intruder male. Fish adopt an antiparallel position and tend to In Brachyhypopomus, aggression and courtship are linked;
bite each other’s tail. They emit primarily interruptions (prob- both occur at greatest rates during the breeding season.
ably signaling submission) and chirps (probably signaling However, in G. omarorum, aggression also occurs outside the
dominance), with one chirp type (M) emitted exclusively breeding season when it serves mostly in territorial interactions
during male–male interactions (Figure 13). On average, males (Batista et al., 2012; Zubizarreta et al., 2015). Both male and
produce chirps every 2s, but these chirps are emitted in female Gymnotus are very aggressive, launching open mouth
‘bouts’ interspersed with periods without chirps. The contest attacks within seconds of the encounter. A clear sequence of
ends with the retreat of the intruder fish and the emergence EOD modulations follows. In the contest phase, ‘offs’ or
of a clear dominance–subordination status signaled by EOD complete interruptions of the EOD are emitted as submissive
rate, in which the dominant fish discharges at a higher signals. The fights themselves are very short (approximately
frequency than the subordinate (Perrone et al., 2009). 2–3 min). After the contest resolution, the submissive fish
In opposite-sex interactions, males emit chirp types A, B, increases the duration of the interruption and adds character-
and C and accelerations while females produce interruptions istic chirps of 400ms in duration. Males and females display
(Perrone et al., 2009). In this context, chirping likely conveys indistinguishable EOD modulations.
information about male quality, while interruptions likely Analogous modulations of the EOD have convergently
signal acceptance by the female. During courtship, the male evolved in the pulse-type mormyrid fishes. Brienomyrus
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 83

brachyistius produces three distinct types of EOD modulations agonistic interactions, but also produced at lower rates by
in different social contexts (Carlson and Hopkins, 2004). females (Dunlap, 2002) and by both sexes during courtship
Eleven different types of EOD modulations have been (Hagedorn and Heiligenberg, 1985). In many other apterono-
described in this species and have been correlated with social tids, there is no sex difference in chirp rate (Smith, 2013)
behaviors (Wong and Hopkins, 2007). Scallops are highly (Figures 14 and 16).
stereotyped, short (0.5 s), abrupt increases in EOD rate that
are hypothesized to function as advertisement signals by domi- 2.05.4.1.2.1 Agonistic Interactions
nant fish. Accelerations are more variable, longer-duration The best-studied example of agonistic chirping is in the brown
increases in EOD rate and are thought to be agonistic signals. ghost (A. leptorhynchus). Most chirps during aggression have
Rasps combine the abrupt increase in EOD rate of scallops relatively low-frequency excursion. These small chirps, which
with the variable, longer-duration rate increase of an accelera- have also been called ‘low-frequency’ or ‘type-II’ chirps, are
tion. Rasps are produced primarily by males prior to spawning produced much more commonly by males than by females
and are likely courtship signals. Receptive females respond to (Zupanc and Maler, 1993, 1997; Dulka and Maler, 1994; Dun-
rasps by producing fast bursts, in which they increase their lap et al., 1998; Figures 14 and 16). This sex difference
EOD rate to 30–100 Hz for less than 1 s. Courting males and depends, in part, on the circumstances under which the fish
females often ‘duet’ with alternating rasps and fast bursts are observed. Chirping is often studied by placing a fish in
(Wong and Hopkins, 2007). a perforated plastic tube to constrain its movements – a chirp
During aggressive encounters, the mormyrid P. isidori chamber – and presenting an electrical sine wave stimulus
increases pulse repetition rate from a baseline of tens of pulses across the tube. This produces a well-controlled stimulus but
per second up to 125 pulses/s for hundreds of milliseconds one with an unnatural geometry. Under these conditions,
(Kramer, 1978). Each barrage is followed by a brief silent males chirp robustly, whereas females seldom chirp when stim-
period. The losing fish either discharges at a low and regular ulus presentations last a minute or less. Females chirp moder-
rate of about 10 pulses/s or shuts off its EOD completely. ately after a longer exposure to a playback stimulus
Both sexes behave similarly. (Triefenbach and Zakon, 2008), and even more intensely in
During courtship and spawning, P. isidori communicates real social encounters (Tallarovic and Zakon, 2002).
with both electric and acoustic signals (Crawford et al., 1986; Chirp rate depends heavily on the specific parameters of the
Bratton and Kramer, 1989; Crawford, 1991). Early in the stimulus. For example, when males are studied in a chirp
spawning cycle, a male establishes a territory and chases away chamber with an EOD mimic near their own EOD frequency,
other fish, even females. Eventually he builds one or two nests chirp rate is directly proportional to stimulus intensity
out of plant material and begins vocalizing. The resting EOD (Figure 16; Dunlap et al., 1998) and varies by stimulus
pattern during this phase is irregular. On the night of spawning, frequency. Stimulus frequencies near the fish’s own EOD
the female emits EODs very regularly at about 10 pulses/s, frequency, which would mimic a same-sex encounter, elicit
starting on her own territory and continuing throughout the far more chirps than those mimicking an opposite-sex
night. During her visits, the male also changes his EOD pattern encounter (Engler and Zupanc, 2001; Hupe et al., 2008; Smith,
to a similar regular low pulse rate and vocalizes profusely 2013). Similarly, in staged encounters between two fish sepa-
(grunt-moan-growl calls); he is electrically silent when he rated by a mesh barrier, males chirp at other males at twice
vocalizes. As courtship intensifies, both fish engage in a series the rate as they do to females (Dunlap, 2002). In addition,
of stereotyped acrobatic swimming movements. In this phase, males respond at different rates toward stimulus frequencies
they maintain the regular rhythm of EOD pulses except for that might mimic males of different social status (Triefenbach
a brief moment when they are vent to vent and the male shuts and Zakon, 2003). When freely swimming fish are presented
off his EOD. The male ceases vocalizing for the rest of the night, with EOD mimic stimuli, males with low EOD frequencies
and the pair undergoes a series of spawnings. Following each (which tend to be the smallest and least mature) chirp more
spawning, the male picks up the eggs in his mouth and trans- at stimulus frequencies near or below their own EOD frequen-
fers them to a nest. After a night of spawning, the male chases cies but less at stimulus frequencies higher than their own
the female away. In playback experiments, Crawford (1991) (which is the range of the larger dominant males). Conversely,
showed that the regular EOD pulse rate by females during males with high EOD frequencies (larger dominant fish) chirp
the night of spawning elicits courtship vocalizations from the at EOD frequencies across the male range.
males, whereas the normal irregular rhythm given by the Within conspecific male interactions, chirping by one fish
male does not. affects chirp rate of another fish. This influence also depends
on the stimulus context. While confined in a chirp chamber,
2.05.4.1.2 Wave Fish fish chirp less toward stimuli containing chirps than identical
In wave-type fish, most EOD modulations fall into three two stimuli without chirps (Dunlap and Larkins-Ford, 2003).
categories based on the duration and degree of frequency excur- However, in dyadic interactions, the overall chirp rate by one
sion (Zakon et al., 2002). Chirps are large frequency increases fish correlates positively with the other fish (Dunlap, 2002; Sal-
(20–300 Hz) of short duration (10–200 ms) and rises are rela- gado and Zupanc, 2011), and chirps commonly occur in an
tively small increases (10–30 Hz) that last longer (1–10 s) “echo” pattern, with one fish producing a chirp with a latency
(Figures 14 and 15(a)). Chirps are further categorized by their of 600ms after the other fish’s chirp (Salgado and Zupanc,
duration and magnitude of frequency and amplitude change. 2011). This reciprocity can also change even within a particular
In several species (e.g., A. leptorhynchus) chirping is sexually encounter, with positive correlation in some phases and nega-
dimorphic, with chirps emitted at greatest rates by males during tive correlation in other phases. During the actual attack phase,
84
(a) (c) 1040

Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology


High-frequency chirp Gradual frequency rise
965
1300

EOD frequency (Hz)


1020
EOD frequency (Hz)

1200
1000 960

1100 980
955
1000 960

900 940
EOD EOD
(head–tail (head–tail
voltage) voltage)

0 20 40 60 80 100 0 200 400 600 800 1000


(b) Time (ms) (d) 500 Time (ms)

Low-frequency chirp 450


1300
EOD frequency (Hz)

400 High-frequency

Frequency modulation (Hz)


chirps
1200
350

1100 300
Male
250 Female
1000
200
900
150 Low-frequency
chirps
EOD
(head–tail 100
Gradual frequency
voltage) 50 rises

0 20 40 60 80 100 0
0.005 0.05 0.5 5 50
Time (ms)
Duration (s)

Figure 14 Types of electric organ discharge (EOD) modulations in brown ghost knifefish (Apteronotus leptorhynchus). (a–c) Blue traces show head–tail voltage and red traces illustrate EOD frequency during
chirps (a, b) and gradual frequency rises (GFRs, c). (a) During high-frequency chirps EOD frequency increases more than 200 Hz above its baseline, and there is a pronounced frequency undershoot at the
end of the chirp (red trace). EOD amplitude is also significantly reduced during high-frequency chirps (blue trace). (b) A low-frequency chirp plotted on the same frequency and timescale as in (a). Low-
frequency chirps are characterized by substantially less frequency modulation, the reduction or absence of a frequency undershoot at the end of the chirp, and the reduction or absence of amplitude modula-
tion. (c) A GFR. Note the different frequency and timescales compared to (a) and (b). GFRs typically have much less frequency modulation and last longer than chirps. Inset shows the GFR on an expanded
frequency scale. (d) Scatter plot illustrating frequency modulation and duration of >10 000 chirps and GFRs produced by male () and female (þ) Apteronotus leptorhynchus. Note that the duration scale is
logarithmic. High-frequency chirps, low-frequency chirps, and GFRs cluster in largely discrete parameter ranges. Both sexes produce low-frequency chirps and GFRs, but males produce most of the high-
frequency chirps.
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 85

(a) Chirps chirp rates decrease and rises are produced in greater rates
(Hupe and Lewis, 2008).
While chirp rates are very high during short-term (<20 min)
social interactions and stimulus presentation, the chirping
10 s
response to male stimuli habituates at longer timescales.
(b)
When presented with repeated artificial stimuli, chirp produc-
140 tion is almost extinguished after 90 min (Harvey-Girard et al.,
120 2010). This habituation occurs only when the stimulus
frequency is constant, and it can be reversed by presentation
Chirp rate (chirps min–1)

100 of a novel stimulus frequency. This may indicate a form of


long-term learning. When two males are housed together but
80
R2 = 0.7508 separated by a mesh barrier, chirp rate is high, 10 times back-
60 ground level, for the first 36h, but then decreases to about 3
times background level over the next 5 days (Dunlap et al.,
40 2011a). However, this long-term social interaction potentiates
the chirp response to artificial stimuli, indicating that fish
20
become sensitized to novel chirp-eliciting stimuli (Dunlap
0 et al., 2002).
0 5 10 15 20 The exact function of EOD modulations during male–male
Plasma 11KT (ng ml–1) interaction is not entirely clear (Smith, 2013; Zakon et al.,
2002). Because they are emitted at highest rates by the largest
Figure 15 Agonistic chirping in brown ghost wave fish. (a) Males and most dominant males, they have commonly been consid-
maintain a steady electric organ discharge (EOD) frequency, but when ered aggressive signals. However, in laboratory encounters,
presented with an electrical sine wave mimicking an EOD, they make
chirp rates are actually lower during the attack phase than in
a series of brief (20 ms) upward frequency modulations called chirps
(from Dulka and Maler, 1994). (b) Chirp rate in males is positively
the periods just preceding and following the attack (Hupe
correlated with their levels of 11-ketotestosterone From Dunlap, K.D., and Lewis, 2008). Moreover, chirp rates of dominant fish do
2002. Hormonal and body size correlates of electrocommunication not change during aggression, but they increase in subordinate
behavior during dyadic interactions in a weakly electric fish, Apteronotus fish. Similarly, recent field data show that when an intruding
leptorhynchus. Horm. Behav. 41, 187–194. male approaches a spawning pair, he is chased by the breeding

Apteronotus albifrons Apteronotus leptorhynchus


100 100

75 75
% Chirping
% Chirping

50 50

25 25

0 0
−60 −50 −40 −30 −20 −10 −60 −50 −40 −30 −20 −10

30
100
Chirps min–1

75
Chirps min–1

20

50
10 Males
25 Females

0 0
−60 −50 −40 −30 −20 −10 −60 −50 −40 −30 −20 −10
Stimulus strength (dB) Stimulus strength (dB)

Figure 16 Species differences in sex differences in chirping. Brown ghosts (Apteronotus leptorhynchus, right-hand side) show a large sex difference
in the percent of individuals that chirped in a minute long test period, as well as the number of chirps made. Black ghosts (Apteronotus albifrons, left-
hand side), on the other hand, show no sex difference in these parameters. From Dunlap, K.D., Thomas, P., Zakon, H.H., 1998. Diversity of sexual
dimorphism in electrocommunication signals and its androgen regulation in a genus of electric fish, Apteronotus. J. Comp. Physiol. A 183, 77–86.
86 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

male and emits chirps at high rates as he flees (Henninger, (Triefenbach and Zakon, 2008). This suggests that these chirps
2015). This suggests chirps serve as submissive signals or as are courtship signals. However, subordinate males tend to
a deterrents to further aggression. produce big chirps toward dominant males, indicating that
Rises are commonly considered submissive signals. In social they may also serve as submissive signals (Cuddy et al., 2012;
encounters, submissive fish produce long slow rises (Hopkins, Hupe et al., 2008).
1974b; Hagedorn and Heiligenberg, 1985). When presented
with EOD mimic stimuli, females with the highest EOD 2.05.4.1.2.3 Interspecific Interactions
frequencies (the smallest, least mature females) make more In A. leptorhynchus, conspecific fish elicit chirp production at
rises than females with the lowest EOD frequencies (which higher rates than heterospecific fish (Sternopygus and Brachyhy-
are larger and more mature). Conversely, male brown ghost popomus) that live sympatrically with Apteronotus. In addition,
with the lowest EOD frequencies (smallest, least mature males) artificial stimuli at frequencies within the species-specific range
make more rises than those with higher EOD frequencies cause more chirping than stimuli in the frequency ranges of
(larger and more mature males) (Triefenbach and Zakon, these heterospecifics (Dunlap et al., 2010). The fact that A. lep-
2008). However, in aggressive interactions, rises are emitted torhynchus chirps even at low rates to heterospecific stimuli
at greatest rates during an attack by both the attacking fish suggests that chirps may serve in interspecific interactions or
and the fish receiving the attack. This indicates that rises may communicate to conspecifics about the presence of heterospe-
also serve as an aggressive signal (Cuddy et al., 2012; Hupe cifics. When EODs of closely related fish that have different
et al., 2008). waveforms are presented all at the same frequency, male A. lep-
torhynchus show no differential response, indicating that EOD
2.05.4.1.2.2 Courtship Interactions waveform is not a salient feature of chirp-eliciting stimuli
The courtship of wave fish (Eigenmannia, A. leptorhynchus) in (Fugere and Krahe, 2010).
a laboratory setting was described thoroughly by Hagedorn
and Heiligenberg (1985). Males and females modulate their
2.05.4.2 EOD Modulations: Neural Control by Prepacemaker
EOD in different ways and at different phases. In the early
Nuclei
phase of Apteronotus courtship, the male chirps using small
chirps characteristic of aggressive encounters, and the female The neural mechanisms underlying the generation of EOD
responds with ‘long rises,’ which are typically submissive modulations are best understood in gymnotiform fish.
signals. Courtship then intensifies over a few nights. On the Inputs from two premotor nuclei called the prepacemaker
night of spawning, the male chirps incessantly (60–80 nucleus (PPn) and the sublemniscal prepacemaker nucleus
chirps/s), but then, as the chirping switches to big chirps (SPPn) cause transient changes in the output of the
(also known as type-I or high-frequency chirps), chirp rate PN (Figures 1 and 17). Chirps are generated when neurons
diminishes. In the presence of a male, the female continues from the PPn that synapse on relay cells in the PN release
making long rises, which now last tens of seconds. The male glutamate, which binds to AMPA (kainate/quisqualate)
often responds to the long rises of the female with renewed receptors to accelerate firing rates (Figure 17). Interruptions
bouts of courtship chirping. Within these episodes the female occur when the glutamatergic input is so strong or long-
repeatedly deposits her eggs. Females make a series of brief (5– lasting that it depolarizes the relay cells enough to inactivate
10 ms) chirps during ovulation. This might signal to the male, their sodium channels and cause them to cease firing (Spiro,
who may be patrolling the territory, that ovulation has 1997). When this happens, no command signals are sent to
occurred (Hagedorn and Heiligenberg, 1985). Such periovula- the spinal cord motor neurons, and the EOD transiently
tory chirping was also found in a naturally occurring spawn- ceases as well (Kawasaki and Heiligenberg, 1989). Different
ing, but the chirps have a different character. In a field chirp types (i.e., small vs big chirps) may result either from
population in Panama, Henninger (2015) observed that the activating more inputs from the PPn or by recruiting inputs
female produced long-duration and high-frequency chirps from the SPPn. Rises are likely regulated through inputs
just when she releases an egg. At that point, the male actually from an additional midbrain nucleus, the PPn-G. These
stops chirping for approximately 10–60 s and then resumes inputs, which cause more gradual changes in EOD frequency,
his high rate of chirping. Such EOD modulations produced use glutamate to activate NMDA receptors, which have much
by the male may contribute to the induction of ovulation. In slower activation kinetics than the AMPA receptors used in
Eigenmannia, playing signals of a courting male to gravid chirp production.
females induces them to spawn. The question of how EOD While production of EOD modulations is regulated by
signals influence the neuroendocrine axis or, conversely, how changes in prepacemaker nuclei, long-term habituation
various hormones initiate the various components of court- appears regulated by the forebrain (Harvey-Girard et al.,
ship electrical displays, is ripe for future investigation. 2010). Repeated chirp-eliciting stimuli cause a reduction in
The proportion of chirp types differs considerably between chirp rate and decreases immediate early gene expression
opposite- and same-sex encounters. Males produce far more big (Egr-1) in a specific telencephalic region (DDmg). This expres-
chirps (type I, III, and IV) when interacting with females or sion then increases when novel stimuli reverse this habituation.
when presented with stimuli in the female frequency range These observations suggest that Egr-1 expression in forebrain
than when presented with another male or its EOD. Males contributes to learning electrocommunication signals in
occasionally also emit big chirps in aggressive contexts to stim- a manner similar to that observed in bird song learning.
ulus frequencies across the species range, but they tend to make Steroids and neuromodulators modify neurotransmission
them more often to stimuli in the female EOD frequency range between the PPn and the PN (see Section 2.05.4.3), and such
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 87

techniques are not yet available to verify whether this relation-


PPnC PPnG SPPn ship is causal (Dunlap et al., 2013).

2.05.4.3 EOD Modulations: Species Diversity and Their


NMDA Hormonal Control in Apteronotidae
or Pacemaker
AMPA cells 2.05.4.3.1 Species Diversity
Fish in the largest family of gymnotiforms, Apteronotidae,
display a remarkable diversity of EOD modulations (Turner
Relay cells et al., 2007; Smith, 2013). Species variation is much higher
for chirps than for rises. Rises are not sexually dimorphic,
appear less important in social interaction, and thus may
AMPA NMDA
have evolved relatively slowly. By contrast, chirps are central
to courtship and aggression, which likely puts them under
(a) To neurogenic electric organ strong diversifying selection. Interspecifically, chirps vary in
almost every possible way, including chirp rate, duration, the
Relay cells magnitude of frequency and amplitude modulation, and the
degree of sexual dimorphism. Discriminant analysis indicates
that interspecific variation in chirp parameters could be used
V
by fish for species identification. This may be particularly
important in Apteronotidae, in which species’ EOD frequency
ranges overlap. Despite this broad diversification in chirps,
(b some chirp parameters appear to have evolved in a concerted
way. For example, species with the largest frequency modula-
Figure 17 (a) Social signals such as chirps and rises result from glu- tion during chirps also show the largest amplitude modulation,
tamatergic modulation of the pacemaker neurons. This figure illustrates suggesting biophysical constraints within the chirp-generating
the cell types (pacemaker and relay) on which the inputs from the SPPn neurons (Turner et al., 2007).
and the two divisions of the PPn synapse. NMDA and AMPA refer to Most species show sex differences in chirping consistent
two classes of glutamate receptors. (b) Fish (in this case a pulse fish of with their role in sexual and aggressive behavior. However,
the genus Hypopomus) can be induced to make social signals in an
species vary considerably in which specific chirp parameter
electrophysiological experiment. Relay cells (above) fire a barrage of
differs by sex. For example, in A. leptorhynchus, chirp rate is
action potentials as glutamatergic input from the prepacemaker nucleus
depolarizes them. The electric organ discharge (EOD) (below) shows highly sexually dimorphic (Figure 16) and dimorphic chirp
a 1:1 relationship with each pacemaker action potential. During the types vary primarily in frequency excursion. In the congeneric
barrage of action potentials, the EOD show a burst of pulses of A. albifrons, chirp rate is monomorphic and dimorphic chirp
decreasing amplitude (due to the inactivation of the Naþ channels in types vary primarily in duration (Dunlap et al., 1998;
the electrocytes). From Kawasaki, M., Heiligenberg, W., 1989. Distinct Kolodziejski et al., 2005). In yet another closely related species
mechanisms of modulation in a neuronal oscillator generate different (A. devenanzii and A. bonapartii), sexes differ only in chirp
socialsignals in the electric fish Hypopomus. J. Comp. Physiol. A 165, complexity (single vs multipeaked chirps) and no other chirp
731–741. parameter (Ho et al., 2010; Zhou and Smith, 2006).

2.05.4.3.2 Neuroendocrine Regulation of Chirping


changes underlie sex and seasonal differences in chirping. in Apteronotus
Several studies also indicate that socially induced changes in 2.05.4.3.2.1 Steroids
neurogenesis in the PPn contribute to long-term changes in In general, sex differences in chirping are regulated by activa-
chirping behavior (Dunlap et al., 2006, 2013). Throughout tional effects of androgens. In dyadic male interactions, chirp
adulthood, the PPn receives cells that are born in the adjacent rate correlates positively with plasma 11kT levels
periventricular region. Compared to other brain regions, the (Figure 15(b)). Gonadectomy in adult male A. leptorhynchus
PPn adds new cells at a particularly high rate (Zupanc and eliminates sexual dimorphism in chirp rate (Dunlap et al.,
Horschke, 1995), and about two-thirds of these cells differen- 1998), and androgen receptor blockade with flutamide in
tiate into neurons (Dunlap et al., 2008). Long-term social inter- P. hasemani eliminates sexual dimorphism in chirp duration
action increases the addition of new cells and the density of (Petzold and Smith, 2016). Androgen treatment increases
radial glial fibers that guide the migration of new cells to the the expression of sexually dimorphic chirping. Interestingly,
PPn while also enhancing chirping behavior. Social manipula- species differences in androgen sensitivity parallel the degree
tions that increase chirping behavior, such as presentation with of sexual dimorphism across species. For example, 11kT
EOD stimuli (Dunlap et al., 2008), novel social partners (Dun- treatment increases chirp rate in A. leptorhynchus, which is
lap and Chung, 2012), or complex social groups (Dunlap et al., dimorphic in chirp rate, but not in A. albifrons, which is mono-
2011b), also increase cell proliferation in the periventricular morphic in chirp rate. This parallel between degree of sexual
region that contributes cells to the PPn. These studies suggest dimorphism and androgen sensitivity is borne out at the pop-
that social enhancement of neurogenesis contributes to ulation level as well. Apteronotus albifrons from the Orinoco
changes in chirping behavior; however, experimental and Amazon, which differ in sexual dimorphism of EOD
88 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

frequency and its androgen sensitivity, do not differ in sexual compounds, serotonin, substance P, and AVT, as modulators
dimorphism of chirp rate, and their chirp rates are similarly of chirping behavior and its sexual dimorphism. In brown
insensitive to androgen (Ho et al., 2013). ghosts (A. leptorhynchus) substance P-like immunoreactivity in
Many studies show clearly that androgens play a large role in the PPn is stronger in males than in females, and the levels of
generating sexually dimorphic chirping, but several studies indi- immunoreactivity in females can be brought almost to the
cate that other factors are involved as well. Females treated with level of a sexually mature male by T implants (Weld and
androgen (T or DHT) chirp at higher rates (10–20 chirps/min) Maler, 1992; Dulka and Maler, 1994; Dulka et al., 1995). The
than females treated with empty capsules (2–4 chirps/min) but effects of androgens on the substance P-like system is likely
less than normal males under the same conditions entirely due to its upregulation of the neuropeptide since
(>40–60 chirps/min) (Dulka et al., 1995; Dunlap et al., there are no sex differences in the number of tachykinin
1998). This partial effect of androgen in females may be due receptors in the PPn (Weld et al., 1994). Interestingly, the
to organizational differences in the brains of the two sexes expression of substance P in the PPn is also highly sexually
that make them differentially responsive to androgen. Alterna- dimorphic in A. albifrons, a species in which chirp structure,
tively, some testicular compound other than androgen might but not chirp rate, is sexually dimorphic (Kolodziejski et al.,
be necessary for full expression of chirping behavior or 2005). This finding suggests that substance P might regulate
a female-specific compound (e.g., estrogen) might inhibit chirp- sex differences in the structure of chirps across species.
ing. Other studies indicate that within-sex variation in chirp rate In contrast to the stimulatory effects of substance P, sero-
depends crucially on the sensory stimuli. In dyadic male interac- tonin inhibits chirping. Serotonin injections in the brain
tions, chirp rates correlate positively with plasma levels of 11kT suppress chirping (Maler and Ellis, 1987), and, consistent
(Dunlap, 2002; Figure 15(b)), but when fish are presented with with their lower chirp rates, females have more serotonin-
synthetic EODs in a chirp chamber, 11kT levels (as inferred containing fibers in the PPn than males (Telgkamp et al.,
from excretion rates) are unrelated to chirp production (Cuddy 2007). Serotonin may also contribute to individual variation
et al., 2012). in chirping behavior associated with social rank. However,
In addition to steroidal effects on chirping responses to studies using pharmacological manipulation of serotonin
acute presentation of social stimuli, steroids also modify chirp- receptor subtypes show that seratonergic regulation of chirping
ing responses to long-term social interaction. The effect of is complex and may involve both direct and indirect pathways.
social housing on chirping is likely mediated by glucocorti- While 5HT2 receptor agonists decrease chirp rate, 5HT1A
coids (Dunlap et al., 2011a, 2013). Compared to isolated agonists increase chirp production, specifically of high-
males, males housed in pairs (but separated by mesh that pre- frequency chirps (Smith and Combs, 2008).
vented physical contact) show an increase in plasma cortisol Another neuromodulator, AVT, which modulates sexual
associated with an elevation of chirp rate in their dyadic inter- and agonistic behavior in a variety of other vertebrates, affects
actions and potentiation of chirp rate to synthetic stimuli. chirping in males by altering the proportion of chirp types
Plasma 11kT levels are not affected. Cortisol treatment to iso- (Bastian et al., 2001). It elevates the production of high-
lated fish causes a similar potentiation of chirping, and gluco- frequency chirps but decreases production of low-frequency
corticoid receptor blockade with RU486 suppresses chirp chirps. This suggests that AVT may serve to shift male behavior
production in dyadic interactions (Dunlap et al., 2011a). away from aggression to courtship. However, because AVT is
Several lines of evidence show that this effect of long-term not present in the PPn, it likely acts via indirect inputs to
social interaction on chirp production is mediated, in part, by the PPn.
a cortisol-dependent effect on neurogenesis. Social interaction
simultaneously increases plasma cortisol and cell proliferation
2.05.4.4 EOD Modulations: Seasonality and Neuroendocrine
in the periventricular zone that contributes new cells to the
Regulation in Brachyhypopomus
PPn. Many of these cells then differentiate into neurons in
the PPn within 7 days, coinciding with the onset of changes In Brachyhypopomus, the rate of EOD modulations (SESs)
in chirping behavior. Cortisol treatment causes similar increases dramatically (10-fold) during the breeding season,
increases in cell addition (Dunlap et al., 2006). However treat- with all three SESs increasing during male–female pairings
ment with a glucocorticoid receptor antagonist only partially and just chirps showing a seasonal increase in male–male pair-
blocked this socially induced increase in cell addition, indi- ings (Perrone et al., 2009). Sexual and seasonal variations in
cating that cortisol may also act via mineralocorticoid receptors the production of SESs are likely driven, at least in part, by regu-
or that some other pathway contributes to social effects on neu- lation of glutamatergic transmission and androgen binding in
rogenesis (Dunlap et al., 2011a). the PN (Quintana et al., 2010). Glutamate injection into the
ventral PN causes interruptions in females and in nonbreeding
2.05.4.3.2.2 Neuromodulators: Peptides and Amines males but induces chirping in breeding males by acting on
Steroid hormones modulate EOD properties presumably at the AMPA receptors. These observations suggest that the default
transcriptional level (see Section 2.05.2.4.2). To date, no rapid state of the ventral PN is to generate interruptions, but that it
nongenomic actions of steroids have been reported. Instead, can be modified in breeding males. During the breeding
short-term modulation of the EOD is mediated by peptides season, plasma 11kT levels rise, and androgen receptor expres-
and amines. The PPn receives input from neurons containing sion increases in many parts of the brain, including in pace-
many neuromodulators, including dopamine, norepinephrine, maker and relay cells of the PN (Pouso et al., 2010). The
serotonin, AVT, somatostatin, galanin, substance P, and meten- seasonal upregulation in androgen receptor is itself likely regu-
kephalin. Most research has focused on three of these lated by androgens, since androgen receptor expression in
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 89

testosterone-treated, nonbreeding fish rise to levels found in Ampullary-type receptors are a relatively ancestral trait in
breeding fish. Together, these results suggest that androgens vertebrates and are found in many aquatic animals that lack
cause seasonal activation of chirping in males by increasing electric organs (e.g., elasmobranchs, lampreys, catfishes, some
the expression of AMPA receptors in the PN. amphibians, platypuses) as well as in both clades of weakly
Another possible mechanism of behavioral change in electric fishes (Bodznick and Northcutt, 1981). Ampullary
B. gauderio is the socially induced and seasonal addition of receptors are similar in gymnotiform and mormyroid fish.
new cells to brain regions that contribute to electrocommunica- They are broadly tuned and behave as low-pass filters
tion (Dunlap et al., 2011b). Fish housed in mixed-sex groups (30–40 Hz to virtually DC). This low-frequency tuning
under seminatural captive conditions that promote gonadal allows ampullary receptors to integrate inputs over time,
maturation and reproductive behavior show elevated rates of which makes them exquisitely sensitive to very weak electrical
cell proliferation in the PN, in the periventricular zone adjacent fields (approximately 10–20 mV cm1 in freshwater fish,
to the PPn, and in electrosensory regions (electrosensory lateral and <1 mV cm1 in marine fish), but largely unable to detect
line lobe, ELL). This effect of social interaction on brain cell the high-frequency components present in most EODs
proliferation occurs only in the breeding season, when fish (Bodznick and Montgomery, 2005). Ampullary receptors func-
emit abundant SESs. This suggests that perception of social tion primarily in passive electrolocation – i.e., in the ability of
stimuli promotes neurogenesis in the electrocommunication fish to passively detect the bioelectric fields generated even by
circuitry and may contribute to seasonal changes in its output. animals that do not have electric organs. However, in weakly
However, it is still unknown whether or how new neurons electric fishes, ampullary receptors may also contribute to the
modify the activity of the electrocommunication circuitry. detection of low-frequency components of electrocommunica-
tion signals (see below).
Tuberous receptors are much more variable across species,
2.05.4.5 EOD Modulations: Steroidal Regulation of Nonsex-
and their filtering properties are often plastic. Unlike ampullary
Biased Territorial Aggression in Gymnotus
receptors, tuberous receptors act as high-pass filters, and they
In most electric fish species, males are larger and exhibit more are typically two to four orders of magnitude less sensitive
aggression than females, particularly in the breeding season. than ampullary receptors. Consequently, tuberous receptors
However, in Gymnotus, aggression is exhibited equally by males cannot passively detect bioelectric fields, but are well adapted
and females both in and outside of the breeding seasons to detect the stronger, high-frequency components of EODs
(Batista et al., 2012; Zubizarreta et al., 2015). In such nonsex- for active electrolocation and communication. Tuberous recep-
biased aggression, interruptions are emitted early in the contest tors are usually tuned to the frequency components of each
as a form of ‘electrical hiding.’ After the resolution of the fight, fish’s EOD. The receptors of pulse fish are broadly tuned to
individuals losing the contest produce chirps to signal capture the wide frequency spectrum of an EOD pulse (Bass,
subordination. 1986; Bastian, 1977; Hopkins and Heiligenberg, 1978;
Across many vertebrates, elevated male aggression during Kawasaki, 1985; Watson and Bastian, 1979). The tuberous
the breeding season is regulated by testicular androgens. receptors of wave fish are usually more sharply tuned to a fish’s
However, Gymnotus displays aggression outside the breeding own EOD frequency (Hopkins, 1976). Since EOD frequency
season, and several experiments indicate that extragonadal differs between sexes, receptor tuning is sexually dimorphic
steroids regulate this aggression (Jalabert et al., 2015). Circu- as well (see below; Zakon and Meyer, 1983).
lating 11-kT levels in dominant (aggressive) and isolated There are two types of tuberous receptors in the pulse-type
(nonaggressive) males are similar, and castration fails to affect mormyroids (Table 1). Mormyromasts are specialized to detect
aggression in dominant fish. However, treating dominant fish a fish’s own EOD and are used in active electrolocation. Mor-
with an aromatase inhibitor decreased aggression, indicating myromasts contain two types of electroreceptor cells. A-type
that aggression in the nonbreeding season depends on a nongo- cells encode the local amplitude of the EOD by firing at
nadal, estrogenic pathway. higher rates in response to stronger electric fields, but are
relatively insensitive to EOD phase or waveform; B-type cells
also encode EOD amplitude, but in addition are sensitive to
2.05.5 EOD Reception through the Electrosensory very small changes in EOD phase. Thus, they precisely
System encode EOD timing and waveform (Kawasaki, 1985). There
are also differences across mormyrid species in the relative
2.05.5.1 Electroreceptor Types, Function, and Tuning
sensitivities of A and B cells, which suggests mechanisms of
Weakly electric fish have two classes of electroreceptors: ampul- electroreceptive coding may vary across species (Bell, 1990).
lary receptors and tuberous receptors (Table 1). These receptors A second type of electroreceptor organ, the knollenorgan, is
are distributed over the fish’s body, although they are often used exclusively for social communication and detects the
most dense in the skin of the head and in some species along EOD pulses of other fish. Whenever a fish fires its own
the dorsal margin of the fish (Szabó, 1974). In the South electric organ, a corollary discharge from the electromotor
American ghost knifefishes (Apteronotidae), a specialized system strongly inhibits pathways from knollenorgans
structure that can be extended dorsally above the tail (dorsal through the nucleus of the ELL. This inhibition of responses
filament) also contains a high concentration of tuberous recep- to the fish’s own EOD allows the knollenorgan inputs to
tors (Franchina and Hopkins, 1996). Electroreceptors are respond specifically to the EODs of other fish (Bell, 1986;
evolutionarily derived from the neuromasts of the lateral line Bennett and Steinbach, 1969). The only mormyroid fish that
system and are innervated by the lateral line nerves. produces wave-type discharges, Gymnarchus niloticus, has two
90 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

Table 1 Types, functions, and sex differences in electroreceptors

Sex differences and hormonal


Taxon and EOD type Receptor type Stimulus encoded Function regulation

All electroreceptive fish Ampullary receptors Weak low-frequency/DC Passive electrolocationa Female stingrays: more ampullary
electric fieldsa Chirp duration in species in afferentsc
which chirps have DC Male stingrays: ampullary
componentsb receptors tuned to lower
frequencies in breeding seasond
Androgens lower frequency tuning
of ampullary receptorsd
Mormyrid pulse Knollenorgans Timing and waveform Communication (detecting Male knollenorgans tuned to lower
(zero-crossings) of and discriminating other frequency (male-like) EOD
other fishes’ EODse EODs)e spectraf
Androgens indirectly masculinize
tuningf
Mormyromasts Active electrolocation No known sex differences
A cells Local EOD amplitudeg
B cells Local EOD amplitudeg
Phase/waveform distortion
caused by object
capacitanceg
Gymnotiform pulse Tuberous receptors:
Burst duration coders EOD amplitudeh Active electrolocationh No known sex differences
Communicationi
Pulse markers EOD phase/timingh Active electrolocationh No known sex differences
(capacitance detection)
Communicationg–i (EOD
timing and waveform)
Gymnotiform wave Tuberous receptors:
P (probability)-type EOD amplitudej Active electrolocationj Receptors tuned to fishes’ own
Communication (beat/ EOD frequency (i.e., tuning
chirp detection)k sexual dimorphism matches)
Jamming avoidancel EOD sexual dimorphism
Androgens directly masculinize
tuning
T (timing)-type EOD phase/timingj Active electrolocationj Receptors tuned to fishes’ own
Communication (beat EOD frequency
phase shifts during Androgens directly masculinize
chirps)m tuning
Jamming avoidancel
a
Kalmijn (1974).
b
Metzner and Viete (1996), Naruse and Kawasaki (1998), and Stockl et al. (2014).
c
Kempster et al. (2013).
d
Sisneros and Tricas (2000).
e
Hopkins and Bass (1981).
f
Bass (1986) and Bass and Hopkins (1984).
g
Bell (1990) and Kawasaki (1985).
h
Bastian (1976).
i
Shumway and Zelick (1988).
j
Scheich et al. (1973).
k
Benda et al. (2006) and Nelson et al. (1997).
l
Heiligenberg (1986).
m
Stockl et al. (2014). EOD, electric organ discharge.

tuberous receptor types, gymnarchomast types I and II that are electrolocation and EODs of other fishes for electrocommuni-
somewhat morphologically and perhaps functionally similar cation. Pulse-type gymnotiforms have two types of tuberous
to the mormyromasts and knollenorgans of mormyrids receptor organs whose responses are analogous to those of
(Szabó, 1974; Zakon, 1986). The physiology and function of the A and B cells of the mormyrid mormyromasts. Pulse marker
gymnarchomasts, however, has not been well studied and electroreceptors fire at most a single spike to each EOD, but the
will not be further considered here. spike is phase-locked to the EOD and can thus serve to encode
Unlike mormyrids, gymnotiforms do not have tuberous EOD timing and waveform (Bastian, 1976). Burst duration
receptors dedicated only to detecting communication signals. coding electroreceptors are more sensitive than pulse markers
Instead, the tuberous receptors of gymnotiforms must perform and fire multiple spikes per EOD. Their firing rate encodes
the double duty of both detecting the fish’s own EOD for EOD amplitude. In both pulse-type gymnotiforms and
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 91

mormyrids, tuning of electroreceptors may vary across the pop- waveform. These species also lack the ELa circuitry to compare
ulation of electroreceptors on individual fish, but at least knollenorgan spike timing across the body and encode EOD
a subset are narrowly tuned to the power spectrum of the fish’s waveform differences (Carlson et al., 2011).
EOD (Bass and Hopkins, 1984; Bastian, 1976; Watson and Like the mormyrid knollenorgans, the pulse marker electro-
Bastian, 1979). receptors of pulse-type gymnotiforms have the capacity to
Wave-type gymnotiforms have two types of tuberous electro- generate a two-spike code to encode the timing of EOD zero-
receptor organs used for electrolocation communication crossings, and giant cells in the midbrain torus, like those in
(Table 1; Scheich et al., 1973). P-type electroreceptors, like the the ELa of mormyrids, can compare timing of zero-crossings
burst duration coders in pulse gymnotiforms and the A cells across electroreceptors in different locations on the skin.
in mormyrid mormyromasts, encode EOD amplitude with However, gymnotiforms may also compare the firing rates of
a rate code. P-type electroreceptor afferents fire once or not at narrowband and broadband burst duration encoding electrore-
all to each EOD, and their firing probability depends on EOD ceptors to encode the spectral content of the EOD and allow
amplitude. T-type electroreceptors are one to two orders of fish to discriminate between male EODs that have more power
magnitude more sensitive than P-type electroreceptors and in lower frequencies and female EODs that have more power in
typically fire once for every EOD cycle. T-type afferent firing to higher frequencies (Shumway and Zelick, 1988).
each EOD is phase-locked, and the timing of T-type afferent
action potentials thus precisely encodes EOD timing/phase. 2.05.5.2.2 Wave Fish
Wave-type gymnotiforms detect and differentially respond to
EOD frequency, and thus may be able to use EOD frequency
2.05.5.2 Electroreceptor Coding of EOD Waveforms
to obtain information about the species, sex, and/or social
2.05.5.2.1 Pulse Fish rank of other fish (Fugere and Krahe, 2010; Kolodziejski
The waveform of pulse EODs varies substantially across species et al., 2007; Triefenbach and Zakon, 2003). EOD waveform
and between sexes, and many pulse-type mormyrids and gym- also varies across and sometimes within species in wave-type
notiforms behaviorally discriminate among signals based on gymnotiforms, but the evidence that they use EOD waveform
pulse waveform (Hopkins, 1988; Hopkins and Bass, 1981; as a communication signal is mixed. For example, Eigenmannia
Shumway and Zelick, 1988). Variation in EOD pulse waveform can detect sex differences in EOD waveform, and both sexes
could theoretically be encoded by the electrosensory system in preferentially approach electrodes playing EODs with a female
one of two ways: (1) electroreceptors could encode ‘landmarks’ waveform (Kramer and Otto, 1988). In contrast, despite the
in the EOD pulse waveform by precisely encoding the timing of fact that EOD waveform is more variable across species of ghost
positive and negative zero-crossings of the EOD (i.e., when the knifefishes (Apteronotidae) than in any other clade of wave-
voltage of the EOD goes from negative to positive) and/or type gymnotiforms (Turner et al., 2007), a recent study did
(2) because the power spectrum of the EOD changes as its not find EOD waveform discrimination in A. leptorhynchus.
waveform changes, electroreceptors could encode EOD wave- Males did not approach or chirp more toward playbacks of
form by using electroreceptor populations with different EODs with conspecific versus heterospecific waveforms, which
frequency tuning to encode the spectral content of the EOD. suggests that at least in these contexts, EOD waveform is not
Pulse-type mormyrids use the first of these mechanisms a behaviorally relevant signal feature in this species (Fugere
(Hopkins and Bass, 1981). In pulse-type mormyrids that and Krahe, 2010). It remains to be determined whether EOD
have sexually dimorphic EOD waveforms and that discriminate waveform is discriminated by A. leptorhynchus in other contexts
EOD waveform, knollenorgan electroreceptors fire a single (e.g., courtship) or by other apteronotid species that have more
action potential precisely on positive zero-crossings of the complex EOD waveforms.
EOD. This allows them to encode the EOD pulse waveform Coding EOD waveform and/or frequency in wave-type fish
of other fish. Knollenorgans on one side of the body fire during is more complicated than in pulse-type fish because wave-type
positive to negative transitions of the EOD, and ones on the electric fish continuously produce a high-duty cycle EOD (i.e.,
other side fire during positive to negative transitions. This is the time between EODs is about equal to the duration of each
due to the geometry of current flow: current emanating from EOD). The tuberous receptors of wave-type fish thus simulta-
a neighboring fish enters one side of the body and exits the neously sense both the fish’s own EOD and that of neighboring
opposite side, depolarizing knollenorgans on the side it enters fish. This makes the EOD an unusual communication signal
and hyperpolarizing those on the side it exits. Neurons in the because the receiver cannot perceive the signal directly. Instead,
anterior exterolateral nucleus (ELa) in the mormyrid midbrain wave-type electric fish detect the presence of other EODs based
compare the timing of the EOD zero-crossings between knolle- on the interactions of the fish’s own EOD with those of nearby
norgans on each side of the body. These neurons thus encode conspecifics. Specifically, when two fish are near each other,
EOD waveform and help identify the species and sex of the their combined EODs produce a signal that beats (oscillates
sender’s EOD (Amagai, 1998; Xu-Friedman and Hopkins, in amplitude and comes in and out of phase) at a frequency
1999). Some mormyrid species do not have sexually dimorphic equal to the difference between the two fishes’ EOD frequencies
EOD waveforms, and their knollenorgans do not encode EOD (Heiligenberg, 1986). P-type tuberous electroreceptors (P-
waveform with a two-spike code (Baker et al., 2015). Instead, units) in the fishes’ skin encode the amplitude modulation of
the knollenorgans in these species spontaneously oscillate these beats, and T-type receptors in different parts of the
and produce synchronized, time-locked spikes in response to fishes’ skin encode the phase modulation. Because EOD
an EOD. This response optimizes the detection of other frequency is sex-specific in many species, the sex of a nearby
EODs, but does not allow discrimination based on EOD conspecific can be determined from the beat frequency. Fish
92 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

of the same sex have EODs of similar frequencies that produce of the ELL in the hindbrain (Carr and Maler, 1986). These
slow beats when combined, whereas fish of the opposite sex three ELL subregions process electrosensory input in parallel
have more distant EOD frequencies that produce higher beat for both electrolocation and communication and are
frequencies when their EODs interact. functionally specialized. Pyramidal cells in the centromedial
Much less is known about whether and how EOD waveform segment of the ELL have small spatial receptive fields,
is encoded by electroreceptors in wave-type fish. Information respond best to low-frequency beats, and are necessary for
about EOD waveform is reflected in the structure of the beat the jamming avoidance response. Pyramidal cells in the
produced by interacting EODs; fish with more complex EOD lateral segment of the ELL have large receptive fields, respond
waveforms produce complex EOD beats when they interact best to somewhat higher frequency beats, and are necessary
(Petzold and Smith, 2014). The complexity of the beat (and for fish to respond to conspecifics with chirping (Metzner,
thus EOD waveform) theoretically should be reflected in the 1999; Metzner and Juranek, 1997).
firing patterns of P-units, but because most studies of In A. leptorhynchus, the distinct sensory coding mechanisms
electroreceptors in wave-type electric fish have used for low-frequency (agonistic) chirps and high-frequency (court-
sinusoidal stimuli, the relationship between EOD waveform/ ship) chirps that are found in the periphery extends into the
beat complexity and electroreceptor coding has not been ELL. The ELL has two types of pyramidal cells that receive input
empirically examined. from P-units. E-cells receive direct input from P-type afferents
and are excited by increases in EOD amplitude. I-cells receive
indirect input from P-type afferents via an inhibitory
2.05.5.3 Sensory Coding of Chirps
interneuron and fire more in response to decreases in EOD
2.05.5.3.1 Electroreceptors amplitude. E-cells respond to low-frequency chirps on low-
The beat produced by the interaction of the EODs of two frequency beats (i.e., small chirps produced when same-sex
nearby wave-type electric fish is a rhythmic amplitude modula- individuals interact) by producing stereotyped bursts that
tion whose frequency is determined by the difference between facilitate the detection of these chirps, but do not allow
the frequencies of the two EODs. When a fish chirps, its EOD discrimination of subtle variation in the parameters of these
frequency increases abruptly, and in some cases the amplitude chirps (Marsat and Maler, 2012). In contrast, I-cells respond
of its EOD decreases. These transient frequency and amplitude to high-frequency chirps on high-frequency beats (i.e., big
changes disrupt the regular beat of two interacting EODs, and chirps produced during opposite-sex encounters) with
fish detect the chirps as the disruption of the beat. a heterogeneous, graded firing pattern that enables
Both P-type and T-type tuberous receptors and in some populations of them to encode the fine structure of the chirp
species even ampullary receptors encode chirps. P-type and thus enables fish to discriminate among different high-
receptors encode the amplitude disruption in the EOD beat frequency chirps. Thus, central processing mechanisms may
by either increasing or decreasing the synchrony of their be adapted with signal structure in this species to allow
firing (Benda et al., 2006; Stockl et al., 2014). T-type females to extract information about the quality of male
electroreceptors encode the disruption of the phase courtship chirps.
modulation in the beat by changing their interspike intervals. Less is known about how chirps are coded in the T-type
In some electric fish species, the EOD is interrupted during tuberous or ampullary pathways in the ELL or at higher levels
a chirp, which unmasks a head-negative DC component of in the electrosensory system. ELL neurons project to the
the EOD. This DC signal stimulates ampullary receptors and dorsal torus semicircularis (TSd) in the midbrain. The TSd
allows them to encode chirp duration (Metzner and Viete, contains populations of neurons that respond selectively to
1996; Naruse and Kawasaki, 1998; Stockl et al., 2014). chirps with a sparse code and that project to midbrain and
Chirp encoding by the electrosensory system has been most forebrain circuits mediating social behavior and diencephalic
extensively studied in brown ghost knifefish (A. leptorhynchus). circuits that mediate chirping (Metzner and Heiligenberg,
P-type electroreceptors in this species use distinct mechanisms 1991; Vonderschen and Chacron, 2011). However, how the
to encode different types of chirps. Small chirps, which are sparse code in the TSd arises and whether it allows merely
often emitted during same-sex interactions, are most the detection of chirps or fine discrimination of chirp
detectable by P-units (i.e., produce the most dramatic change properties are not yet clear.
in firing synchrony) when they occur on low-beat frequencies
created by the interaction of same-sex EODs. In contrast, big
2.05.5.4 Electroreception: Sex Differences and
chirps, which are produced during courtship, cause changes
Neuroendocrine Regulation
in the synchrony of P-unit firing when they occur on
a background of the high beat frequencies produced when Much less is known about sex differences and hormonal regula-
male and female EODs interact (Benda et al., 2006). These tion of the neural mechanisms by which EOD and chirps are
findings suggest a fascinating coevolution of the sensory perceived than is known about the mechanisms controlling sex
physiology of signal detection with signal structure and the differences in the production of electrocommunication signals.
social context (i.e., courtship vs aggression) in which the Most of what is known focuses on sex differences and hormonal
signals are used. regulation of tuning in electroreceptors. Some sex differences in
ampullary receptors have been found in elasmobranchs. Female
2.05.5.3.2 Central Nervous System stingrays have more ampullary afferents than males, which
Each P-type tuberous electroreceptor afferent projects to might make it easier for them to distinguish between approach-
pyramidal cells in three somatotopically mapped subregions ing conspecifics versus predators (Kempster et al., 2013).
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 93

The ampullary receptors of male stingrays are tuned to lower hormone treatment is a fertile one. Similarly, no studies have
frequencies in the breeding season than in the nonbreeding examined the possibility of sex differences or effects of gonadal
season, and it is hypothesized that this tuning facilitates locating hormones on the coding of EODs and chirps in the CNS.
mates buried in the substrate. The seasonal change in ampullary However, recent studies have found that neuromodulators
tuning in males is mediated by androgens; treatment with DHT may alter the excitability of pyramidal cells to influence sensory
lowers the best frequency of ampullary receptors (Sisneros and processing of EODs and chirps in the ELL. In particular, sero-
Tricas, 2000). tonin, acting on 5HT2-like receptors, enhances the excitability
In gymnotiform fish with wave-type discharges, tuning of of ELL pyramidal cells and selectively increases their responses
tuberous receptors is also sexually dimorphic and hormonally to stimuli created during same-sex encounters (low-frequency
regulated. Sex differences in EOD frequency are mediated EOD beats and small chirps), but not to signals produced
largely by activational effects of androgens and estrogens. during opposite-sex encounters (Deemyad et al., 2013; Larson
Because tuberous electroreceptors are precisely tuned in wave- et al., 2014). Combined with the ability of serotonin (via 5HT2
type fish, electroreceptors must shift their tuning when like receptors) to inhibit chirping (Maler and Ellis, 1987; Smith
hormones change EOD frequency to remain tuned to a fish’s and Combs, 2008), this finding led to the ‘shut up and listen’
own EOD. Like EOD frequency, the tuning of tuberous electro- hypothesis that serotonergic activation during aggressive
receptors is sexually dimorphic in wave-type gymnotiforms, encounters may modulate signaling by reducing the produc-
and fish treated with androgens show parallel shifts of EOD tion of chirps and enhancing detection and responsiveness to
frequency and electroreceptor tuning (Meyer et al., 1987; Meyer chirps (Deemyad et al., 2013).
and Zakon, 1982; Zakon and Meyer, 1983).
A key question is whether the hormones act directly or indi-
rectly on electroreceptors to change their tuning. That is, 2.05.6 Conclusions
hormones might act directly on the PN to induce a change in
EOD frequency and this change in the frequency at which the What general lessons can be reached from studying hormones
electroreceptors are driven might somehow reset electrorecep- and behavior in electric fish? First, although electrocommuni-
tor tuning to match. Alternatively, androgens might act inde- cation signals may be unique to only two orders of fish, they
pendently on electromotor circuits and on electroreceptors to are utilized in much the same ways as signals in other modal-
coordinate EOD frequency and receptor tuning. In wave-type ities and are similarly influenced by natural and sexual selec-
gymnotiforms, hormones appear to act independently on tion. As with other sexually dimorphic communication
motor circuits and electroreceptors. Androgens still shift elec- signals, the EOD conveys information on the species, age,
troreceptor tuning in Sternopygus whose EOD has been abol- sex, and individual identity of the signaler and can also trans-
ished by transecting the spinal cord or lesioning the PN mit information about breeding status, dominance/subordi-
(Ferrari and Zakon, 1989; Keller et al., 1986). Furthermore, nance, and motivation. As with many other sexually
androgen receptors have been localized immunohistochemi- dimorphic communication signals, EODs are often regulated
cally in electroreceptors, so they likely have the capacity to by gonadal steroid hormones through actions on the
respond directly to androgens (Gustavson et al., 1994). Thus, morphology and physiology of the neurons that control these
the actions of androgens on the electroreceptors, the pacemaker signals.
neurons, and the electric organ are coordinated to ensure Other similarities between systems are apparent in the
matched tuning, but this is accomplished via independent development and interaction of natural and sexual selection
hormone actions in each cell type. on the behavioral phenotypes. The EOD waveform of pulse
In mormyrid knollenorgans, electroreceptor tuning is also fish (and EOD frequency in many wave fish) is similar in juve-
sexually dimorphic. Pulse-type EODs have a broad power spec- nile and female gymnotiforms, reminiscent of the plumage
trum, and electroreceptors are broadly tuned. However, male similarities of juvenile and female birds. For many sexually
EODs have more power in lower frequencies and their knolle- dimorphic signals, particularly in species in which males
norgans are more sensitive to lower frequencies than those of compete for reproductive access to females, male signals are
females (Hopkins and Bass, 1981). This is interesting because more conspicuous and/or ornate than those of females. These
it implies that males are more sensitive to the EODs of other signals may carry costs, including increased risk of predation
males than they are to those of females. As in wave-type gym- and/or energetic costs (Stoddard, 1999; Salazar and Stoddard,
notiforms, the sex difference in receptor tuning in mormyrids is 2008). Similarly, in electric fish there is evidence that the elec-
regulated by androgens, which shift knollenorgan tuning trical signals produced by males are more conspicuous both to
curves toward lower frequencies. Unlike in gymnotiform conspecific females and to potential predators.
tuberous receptors, however, the tuning shift in knollenorgans Another striking feature of the electrocommunication
depends on the presence of the fish’s EOD. Spinal transection system in electric fish is the wealth of species diversity in the
of Brienomyrus blocked the effects of androgens on knollenor- biology of these fishes. This diversity includes variation in the
gan tuning (Bass, 1986; Bass and Hopkins, 1984). Thus, in magnitude and direction of sex differences in body size, EOD
mormyrids, sex differences in tuning likely result through indi- properties, and other electrical behaviors (i.e., chirping); diver-
rect actions of hormones. sity in habitat preferences, sociality, and associated diversity in
Unfortunately, no more recent studies have examined the physiology and patterning of the EOD; and variation in court-
hormonal regulation of sex differences in electroreceptor ship and parental behavior. This diversity is coupled with
tuning. In particular, the question of the biophysical mecha- numerous striking examples of convergence in the behavior
nisms by which receptor tuning is derived and shifted by and physiology between the two different orders of fish that
94 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

independently evolved electrocommunication. These features Bastian, J., 1976. Frequency-response characteristics of electroreceptors in weakly
lend themselves well to comparative studies of the hormonal electric fish (Gymnotoidei) with a pulse discharge. J. Comp. Physiol. 112,
165–180.
control of reproductive and communication behavior. For
Bastian, J., 1977. Variations in frequency-response of electroreceptors dependent on
example, evolutionary gains, losses, and even reversals in the receptor location in weakly electric fish (Gymnotoidei) with a pulse discharge.
direction of sex differences in EODs and chirps in apteronotid J. Comp. Physiol. 121, 53–64.
fishes make these signals an excellent model system for Batista, G., Zubizarreta, L., Perrone, R., Silva, A.C., 2012. Non-sex-biased dominance
studying how evolutionary lability of neuroendocrine mecha- in a sexually monomorphic electric fish: fight structure and submissive electric
signalling. Ethology 118, 398–410.
nisms that regulate sex differences can lead to species diversity Bell, C.C., 1986. Electroreception in mormyrid fish: central physiology. In: Bullock, T.H.,
in sexually dimorphic behavior. Heiligenberg, W. (Eds.), Electroreception. Wiley, New York, pp. 423–452.
A final highlight of the electrocommunication system is that Bell, C.C., 1990. Mormyromast electroreceptor organs and their afferent-fibers in
it has numerous features that allow us to study reproductive mormyrid fish 3. Physiological differences between two morphological types of
fibers. J. Neurophysiol. 63, 319–332.
communication behavior at multiple levels of analysis. These
Benda, J., Longtin, A., Maler, L., 2006. A synchronization-desynchronization code for
features include a signal that can be quantitatively analyzed, natural communication signals. Neuron 52, 347–358.
manipulated, and played back and a neural circuit that controls Bennett, M., 1961. Modes of operation of electric organs. Ann. N.Y. Acad. Sci. 94,
the EOD behavior that is well characterized, simple, and acces- 458–509.
sible enough to allow cellular analyses of the mechanisms Bennett, M.V.L., 1971. Electroreception. In: Hoar, W.S., Randall, D.J. (Eds.), Fish
Physiology, vol. V. Academic Press, New York, pp. 493–574.
underlying species, sex, and individual differences in behavior. Bennett, M., Steinbach, A.B., 1969. Influence of electric organ control system on
The recent addition of genomic and bioinformatic tools in this electrosensory afferent pathways in mormyrids. In: Llinas, R. (Ed.), Neurobiology of
system (Gallant et al., 2014; Salisbury et al., 2015) will facili- Cerebellare Evolution and Development. American Medical Association, Chicago,
tate future studies investigating both how hormones regulate pp. 207–214.
Black-Cleworth, P., 1970. The role of electrical discharges in the non-reproductive
gene expression to influence sex differences and how
social behaviour of Gymnotus carapo (Gymnotidae, Pisces). Anim. Behav. Monogr.
hormone–gene networks evolve to yield species diversity in 3, 1–IN1.
sexually dimorphic behavior. Bodznick, D., Montgomery, J.C., 2005. The physiology of low-frequency electro-
sensory systems. In: Bullock, T.H., et al. (Eds.), Electroreception. Springer, New
York, pp. 132–153.
Bodznick, D., Northcutt, R.G., 1981. Electroreception in lampreys - evidence that the
References earliest vertebrates were electroreceptive. Science 212, 465–467.
Boudinot, M., 1970. The effect of decreasing and increasing temperature on the
Allee, S.J., Markham, M.R., Stoddard, P.K., 2009. Androgens enhance plasticity of an frequency of the electric organ discharge in Eigenmannia sp. Comp. Biochem.
electric communication signal in female knifefish Brachyhypopomus pinnicaudatus. Physiol. 37, 601–603.
Horm. Behav. 56, 264–273. Bratton, B., Kramer, B., 1989. Patterns of the electric organ discharge during
Amagai, S., 1998. Time coding in the midbrain of mormyrid electric fish. II. Stimulus courtship and spawning in the mormyrid fish, Pollimyrus isidori. Behav. Ecol.
selectivity in the nucleus exterolateralis pars posterior. J. Comp. Physiol. A 182, Sociobiol. 24, 349–368.
131–143. Bass, A.H., Denizot, J.P., Marchaterre, M.A., 1986. Ultrastructural features and
Ardanaz, J., Silva, A., Macadar, O., 2001. Temperature sensitivity of the electric organ hormone-dependent sex differences of mormyrid electric organs. J. Comp. Neurol.
discharge waveform in Gymnotus carapo. J. Comp. Physiol. A 187, 853–864. 254, 511–528.
Arnegard, M.E., McIntyre, P.B., Harmon, L.J., Zelditch, M.L., Crampton, W.G.R., Bass, A.H., 1986b. A hormone-sensitive communication system in an electric fish.
Davis, J.K., Sullivan, J.P., Lavoué, S., Hopkins, C.D., 2010. Sexual signal evolution J. Neurobiol. 17, 131–156.
outpaces ecological divergence during electric fish species radiation. Am. Nat. Bass, A.H., Hopkins, C.D., 1985. Hormonal control of sex difference in the electric
176, 335–356. organ discharge (EoD) of mormyrid fishes. J. Comp. Physiol. A 156, 587–604.
Alves-Gomes, J., Hopkins, C., 1997. Molecular insights into the phylogeny of mor- Carlson, B.A., Hopkins, C.D., Thomas, P., 2000. Androgen correlates of socially
myriform fishes and the evolution of their electric organs. Brain Behav. Evol. 49, induced changes in the electric organ discharge waveform of a mormyrid fish.
324–351. Horm. Behav. 38, 177–186.
Alves-Gomes, J.A., Ortõ, G., Haygood, M., Heiligenberg, W., Meyer, A., 1995. Carlson, B.A., Hasan, S.M., Hollmann, M., Miller, D.B., Harmon, L.J., Arnegard, M.E.,
Phylogenetic analysis of the South American electric fishes (order gymnotiformes) 2011. Brain evolution triggers increased diversification of electric fishes. Science
and the evolution of their electrogenic system: a synthesis based on morphology. 332, 583–586.
Mol. Biol. Evol. 12, 298–318. Carr, C.E., Maler, L., 1986. Electroreception in gymnotiform fish: central anatomy and
Alves-Gomes, J., 1999. Systematic biology of gymnotiform and mormyriform electric physiology. In: Bullock, T.H., Heiligenberg, W. (Eds.), Electroreception. Wiley, New
fishes: phylogenetic relationships, molecular clocks and rates of evolution in the York, pp. 319–373.
mitochondrial rRNA genes. J. Exp. Biol. 202, 1167–1183. Cox Fernandes, C.C., Smith, G.T., Podos, J., Nogueira, A., Inoue, L., Akama, A.,
Arnegard, M.E., Bogdanowicz, S.M., Hopkins, C.D., 2005. Multiple cases of striking Ho, W.W., Alves-Gomes, J., 2010. Hormonal and behavioral correlates of
genetic similarity between alternate electric fish signal morphs in sympatry. morphological variation in an Amazonian electric fish (Sternarchogiton nattereri:
Evolution 59, 324–343. apteronotidae). Horm. Behav. 58, 660–668.
Assuncao, M., Schwassman, H., 1995. Reproduction and larval development of Crampton, W.G.R., 1998. Effects of anoxia on the distribution, respiratory strategies
Electrophorus electricus on Marajo Island (Para, Brazil). Ichthyol. Explor. Fresh- and electric signal diversity of gymnotiform fishes. J. Fish Biol. 53, 307–330.
waters 6, 175–184. Crampton, W.G.R., Albert, J.S., 2006. Evolution of electric signal diversity in gym-
Bennett, M., 1971. Electric organs. Fish. Physiol. 5, 347–491. notiform fishes. In: Ladich, F., Collin, S.P., Moller, P., Kapoor, B.G. (Eds.),
Baker, C.A., Huck, K.R., Carlson, B.A., 2015. Peripheral sensory coding through Communication in Fishes, vol. 2. Science Publishers, Inc, Enfield, NH,
oscillatory synchrony in weakly electric fish. Elife 4. pp. 647–731.
Bass, A.H., 1986. A hormone-sensitive communication system in an electric fish. Crampton, W.G.R., Rodriguez-Cattneo, A., Lovejoy, N.R., Caputi, A.A., 2013. Proxi-
J. Neurobiol. 17, 131–156. mate and ultimate causes of signal diversity in the electric fish Gymnotus. J. Exp.
Bass, A.H., Hopkins, C.D., 1983. Hormonal control of sexual differentiation: changes in Biol. 216, 2523–2541.
electric organ discharge waveform. Science 220, 971–974. Crawford, J.D., 1991. Sex recognition by electric cues in a sound-producing mormyrid
Bass, A.H., Hopkins, C.D., 1984. Shifts in frequency tuning of electroreceptors in fish, Pollimyrus isidori. Brain Behav. Evol. 38, 20–28.
androgen-treated mormyrid fish. J. Comp. Physiol. 155, 713–724. Crawford, J.D., 1992. Individual and sex specificity in the electric organ discharges of
Bastian, J., Schniederjan, S., Nguyenkim, J., 2001. Arginine vasotocin modulates breeding mormyrid fish (Pollimyrus isidori). J. Exp. Biol. 164, 79–102.
a sexually dimorphic communication behavior in the weakly electric fish Apter- Crawford, J.D., 1997. Hearing and acoustic communication in the mormyrid electric
onotus leptorhynchus. J. Exp. Biol. 204, 1909–1923. fishes. Mar. Freshw. Behav. Physiol. 29, 1–21.
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 95

Crawford, J.D., Hagedorn, M., Hopkins, C.D., 1986. Acoustic communication in an Dunlap, K.D., Tran, A., Ragazzi, M.A., Krahe, R., Salazar, V.L., 2016. Predators inhibit
electric fish, Pollimyrus isidori (Mormyridae). J. Comp. Physiol. A 159, 297–310. brain cell proliferation in natural populations of electric fish, Brachyhypopomus
Cuddy, M., Aubin-Horth, N., Krahe, R., 2012. Electrocommunication behaviour and occidentalis. Proc. R. Soc. B 283, 20152113.
non invasively-measured androgen changes following induced seasonal breeding in Ellis, D., Szabo, T., 1980. Identification of different cell types in the command
the weakly electric fish, Apteronotus leptorhynchus. Horm. Behav. 61, 4–11. (pacemaker) nucleus of several gymnotiform species by retrograde transport of
Crampton, W.G.R., Hopkins, C.D., 2005. Nesting and paternal care in the weakly horseradish peroxidase. Neuroscience 5, 1917–1929.
electric fish Gymnotus (Gymnotiformes: Gymnotidae) with descriptions of larval and Enger, P.S., Szabo, T., 1968. Effect of temperature on the discharge rates of the
adult electric organ discharges of two species. Copeia 2005, 48–60. electric organ of some gymnotids. Comp. Biochem. Physiol. 27, 625–627.
Curtis, C.C., Stoddard, P.K., 2003. Mate preference in female electric fish, Brachy- Engler, G., Zupanc, G.K., 2001. Differential production of chirping behavior evoked by
hypopomus pinnicaudatus. Anim. Behav. 66, 329–336. electrical stimulation of the weakly electric fish, Apteronotus leptorhynchus.
Carlson, B.A., Hopkins, C.D., 2004. Stereotyped temporal patterns in electrical J. Comp. Physiol. A 187, 747–756.
communication. Anim. Behav. 68, 867–878. Falconi, A., Borde, M., Hernández-Cruz, A., Morales, F., 1995. Mauthner cell-initiated
Deemyad, T., Metzen, M.G., Pan, Y.Z., Chacron, M.J., 2013. Serotonin selectively abrupt increase of the electric organ discharge in the weakly electric fish Gymnotus
enhances perception and sensory neural responses to stimuli generated by same- carapo. J. Comp. Physiol. A 176, 679–689.
sex conspecifics. Proc. Natl. Acad. Sci. U.S.A. 110, 19609–19614. Fernandes, C.C., Smith, G.T., Podos, J., Nogueira, A., 2010. Hormonal and behavioral
Deng, T.S., Tseng, T.C., 2000. Evidence of circadian rhythm of electric discharge in correlates of morphological variation in an Amazonian electric fish (Sternarchogiton
Eigenmannia virescens system. Chronobiol. Int. 17, 43–48. nattereri: Apteronotidae). Horm. Behav. 58, 660–668.
Dulka, J., Maler, L., 1994. Testosterone modulates female chirping behavior in the Ferrari, M.B., Zakon, H.H., 1989. The medullary pacemaker nucleus is unnecessary for
weakly electric fish, Apteronotus leptorhynchus. J. Comp. Physiol. A 174, electroreceptor tuning plasticity in Sternopygus. J. Neurosci. 9, 1354–1361.
331–343. Ferrari, M.B., McAnelly, L.M., Zakon, H.H., 1995. Individual variation in and
Dulka, J.G., Maler, L., Ellis, W., 1995. Androgen-induced changes in electro- androgen-modulation of the sodium current in electric organ. J. Neurosci. 15,
communicatory behavior are correlated with changes in substance P-like 4023–4032.
immunoreactivity in the brain of the electric fish Apteronotus leptorhynchus. Ferrari, M.B., Zakon, H.H., 1993. Conductances contributing to the action potential of
J. Neurosci. 15, 1879–1890. Sternopygus electrocytes. J. Comp. Physiol. A 173, 281–292.
Dunlap, K.D., 2002. Hormonal and body size correlates of electrocommunication Feulner, P.G.D., Kirschbaum, F., Schugardt, C., Ketmaier, V., Tiedemann, R., 2006.
behavior during dyadic interactions in a weakly electric fish, Apteronotus lepto- Electrophysiological and molecular genetic evidence for sympatrically occurring
rhynchus. Horm. Behav. 41, 187–194. cryptic species in African weakly electric fishes (Teleostei: Mormyridae : Campy-
Dunlap, K.D., McAnelly, L.M., Zakon, H.H., 1997. Estrogen modifies an electro- lomormyrus). Mol. Phylogenet. Evol. 39, 198–208.
communication signal by altering the electrocyte sodium current in an electric fish, Feulner, P.G.D., Kirschbaum, F., Mamonekene, V., Ketmaier, V., Tiedemann, R., 2007.
Sternopygus. J. Neurosci. 17, 2869–2875. Adaptive radiation in African weakly electric fish (Teleostei: Mormyridae: Campy-
Dunlap, K.D., Thomas, P., Zakon, H.H., 1998. Diversity of sexual dimorphism in lomormyrus) A combined molecular and morphological approach. J. Evol. Biol. 20,
electrocommunication signals and its androgen regulation in a genus of electric 403–414.
fish, Apteronotus. J. Comp. Physiol. A 183, 77–86. Few, W.P., Zakon, H.H., 2001. Androgens alter electric organ discharge pulse duration
Dunlap, K.D., Zakon, H.H., 1998. Behavioral actions of androgens and androgen despite stability in electric organ discharge frequency. Horm. Behav. 40, 434–442.
receptor expression in the electrocommunication system of an electric fish, Few, W.P., Zakon, H.H., 2007. Sex differences in and hormonal regulation of Kv1
Eigenmannia virescens. Horm. Behav. 34, 30–38. potassium channel gene expression in the electric organ: molecular control of
Dunlap, K.D., Smith, G.T., Yekta, A., 2000. Temperature dependence of electro- a social signal. Dev. Neurobiol. 67, 535–549.
communication signals and their underlying neural rhythms in the weakly electric Franchina, C.R., Hopkins, C.D., 1996. The dorsal filament of the weakly electric
fish, Apteronotus leptorhynchus. Brain Behav. Evol. 55, 152–162. Apteronotidae (Gymnotiformes; Teleostei) is specialized for electroreception. Brain
Dunlap, K.D., Oliveri, L.M., 2002. Retreat site selection and social organization in Behav. Evol. 47, 165–178.
captive electric fish, Apteronotus leptorhynchus. J. Comp. Physiol. A 188, Franchina, C.R., Stoddard, P., 1998. Plasticity of the electric organ discharge wave-
469–477. form of the electric fish Brachyhypopomus pinnicaudatus: I. Quantification of day-
Dunlap, K.D., Pelczar, P.L., Knapp, R., 2002. Social interactions and cortisol treatment night changes. J. Comp. Physiol. A 183, 759–768.
increase the production of aggressive electrocommunication signals in male Franchina, C.R., Salazar, V., Volmar, C.H., Stoddard, P., 2001. Plasticity of the electric
electric fish, Apteronotus leptorhynchus. Horm. Behav. 42, 97–108. organ discharge waveform of male Brachyhypopomus pinnicaudatus. II. Social
Dunlap, K.D., Larkins-Ford, J., 2003. Production of aggressive electrocommunication effects. J. Comp. Physiol. A 187, 45–52.
signals to progressively realistic social stimuli in male Apteronotus leptorhynchus. Freedman, E., Olyarchuk, J., Marcheterre, M., Bass, A.H., 1989. A temporal analysis
Ethology 109, 243–257. of testosterone-induced changes in electric organs and electric organ discharges of
Dunlap, K.D., Castellano, J.F., Prendaj, E., 2006. Social interaction and cortisol mormyrid fishes. J. Neurobiol. 20, 619–634.
treatment increase cell addition and radial glia fiber density in the diencephalic Fugere, V., Krahe, R., 2010. Electric signals and species recognition in the wave-type
periventricular zone of adult electric fish, Apteronotus leptorhynchus. Horm. Behav. gymnotiform fish Apteronotus leptorhynchus. J. Exp. Biol. 213, 225–236.
50, 10–17. Gallant, J.R., Traeger, L.L., Volkening, J.D., Moffett, H., Chen, P.H., Novina, C.D.,
Dunlap, K.D., McCarthy, E.A., Jashari, D., 2008. Electrocommunication signals alone Phillips, G.N., Anand, R., Wells, G.B., Pinch, M., Guth, R., Unguez, G.A.,
are sufficient to increase neurogenesis in the brain of adult electric fish, Apter- Albert, J.S., Zakon, H.H., Samanta, M.P., Sussman, M.R., 2014. Genomic basis
onotus leptorhynchus. Dev. Neurobiol. 68, 1420–1428. for the convergent evolution of electric organs. Science 344, 1522–1525.
Dunlap, K.D., DiBenedictis, B.T., Banever, S.R., 2010. Chirping response of weakly Gavassa, S., Silva, A.C., Stoddard, P.K., 2011. Tight hormonal phenotypic integration
electric knifefish (Apteronotus leptorhynchus) to low-frequency electric signals and ensures honesty of the electric signal of male and female Brachyhypopomus
to heterospecific electric fish. J. Exp. Biol. 213, 2234–2242. gauderio. Horm. Behav. 60, 420–426.
Dunlap, K.D., Jashari, D., Pappas, K.M., 2011a. Glucocorticoid receptor blockade Gavassa, S., Stoddard, P.K., 2012. Food restriction promotes signaling effort in
inhibits brain cell addition and aggressive signaling in electric fish, Apteronotus response to social challenge in a short-lived electric fish. Horm. Behav. 62,
leptorhynchus. Horm. Behav. 60, 275–283. 381–388.
Dunlap, K.D., Silva, A.C., Chung, M., 2011b. Environmental complexity, seasonality Goldina, A., Gavassa, S., Stoddard, P.K., 2011. Testosterone and 11 ketotestosterone
and brain cell proliferation in a weakly electric fish, Brachyhypopomus gauderio. have different regulatory effects on electric communication signals of male Bra-
J. Exp. Biol. 214, 794–805. chyhypopomus gauderio. Horm. Behav. 60, 139–147.
Dunlap, K.D., Chung, M., 2012. Social novelty enhances brain cell proliferation, cell Gustavson, S., Zakon, H., Prins, G., 1994. Androgen receptors in the brain, electro-
survival and chirp production in an electric fish, Apteronotus leptorhynchus. Dev. receptors, and electric organ of a wave-type electric fish. Abstr. Soc. Neurosci.
Neurobiol. 73, 324–332. 19, 371.
Dunlap, K.D., Chung, M., Castellano, J.F., 2013. Influence of long-term social inter- Hagedorn, M., 1988. Ecology and behavior of a pulse type electric fish, Hypopomus
action on chirping behavior, steroid levels and neurogenesis in weakly electric fish. occidentalis (Gymnotiformes, Hypopomidae), in a freshwater stream in Panama.
J. Exp. Biol. 216, 2434–2441. Copeia 2, 324–335.
Dunlap, K.D., Ragazzi, M.A., 2015. Thermal acclimation and thyroxine treatment Hagedorn, M., 1992. Social influences on the courtship and breeding of a gymnotiform
modify the electric organ discharge frequency in an electric fish, Apteronotus electric fish. In: Proceedings, IMS/CM2 International Australian Lungfish Breeding
leptorhynchus. Physiol. Behav. 151, 64–71. Workshop. Cleveland Metroparks Zoo.
96 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

Hagedorn, M., Carr, C., 1985. Single electrocytes produce a sexually dimorphic signal Keller, C.H., Zakon, H.H., Sanchez, D.Y., 1986. Evidence for a direct effect of
in South American electric fish, Hypopomus occidentalis (Gymnotiformes, Hypo- androgens upon electroreceptor tuning. J. Comp. Physiol. A 158, 301–310.
pomidae). J. Comp. Physiol. A 156, 511–523. Kempster, R.M., Garza-Gisholt, E., Egeberg, C.A., Hart, N.S., O’Shea, O.R.,
Hagedorn, M., Heiligenberg, W., 1985. Court and spark: electric signals in the Collin, S.P., 2013. Sexual Dimorphism of the electrosensory system: a quantitative
courtship and mating of gymnotoid fish. Anim. Behav. 33, 254–265. analysis of nerve axons in the dorsal anterior lateral line nerve of the blue-spotted
Hagedorn, M., Zelick, R., 1989. Male dominance is expressed in the electric organ fantail stingray (Taeniura lymma). Brain Behav. Evol. 81, 226–235.
discharge characteristics of a weakly electric fish. Anim. Behav. 38, 520–525. Kirschbaum, F., 1977. Electric-organ ontogeny: distinct larval organ precedes the adult
Hanika, S., Kramer, B., 1999. Electric organ discharges of mormyrid fish as a possible organ in weakly electric fish. Naturwissenschaften 64, 387.
cue for predatory catfish. Naturwissenschaften 86, 286–288. Kirschbaum, F., Westby, G., 1975. Development of the electric discharge in mormyrid
Hanika, S., Kramer, B., 2005. Intra-male variability of its communication signal in the and gymnotid fish (Marcusenius sp. and Eigenmannia virescens). Experientia 31,
weakly electric fish, (South African form), and possible functions. Behaviour 142, 1290–1293.
145–166. Kirschbaum, F., 1975. Environmental factors control the periodical reproduction of
Harvey-Girard, E., Tweedle, J., Ironstone, J., Cuddy, M., Ellis, W., Maler, L., 2010. tropical electric fish. Experientia 31, 1159–1160.
Long-term recognition memory of individual conspecifics is associated with Kirschbaum, F., 1995. Reproduction and development in mormyriform and gymnoti-
telencephalic expression of Egr-1 in the electric fish Apteronotus leptorhynchus. form fishes. In: Moller, P. (Ed.), Electric Fishes: History and Behavior. Chapman and
J. Comp. Neurol. 518, 2666–2692. Hall, London, pp. 267–301.
Henderson, P.A., Crampton, W.G.R., 1997. A comparison of fish diversity and Kolodziejski, J.A., Nelson, B.S., Smith, G.T., 2005. Sex and species differences in
abundance between nutrient-rich and nutrient-poor lakes in the Upper Amazon. neuromodulatory input to a premotor nucleus: a comparative study of substance P
J. Trop. Ecol. 13, 175–198. and communication behavior in weakly electric fish. J. Neurobiol. 62, 299–315.
Henninger, J., 2015. Social Interactions in Natural Populations of Weakly Electric Fish. Kolodziejski, J.A., Sanford, S.E., Smith, G.T., 2007. Stimulus frequency differentially
Doctoral Dissertation. Mathematisch-Naturwissenschaftlichen Fakultat, Universitat affects chirping in two species of weakly electric fish: implications for the evolution
Tubingen. of signal structure and function. J. Exp. Biol. 210, 2501–2509.
Heiligenberg, W., 1986. Jamming avoidance responses: model systems for neuro- Kramer, B., 1978. Spontaneous discharge rhythms and social signalling in the weakly
ethology. In: Bullock, T.H., Heiligenberg, W. (Eds.), Electroreception. Wiley, New York, electric fish Pollimyrus isidori (Cuvier et Valenciennes) (Mormyridae, Teleostei).
pp. 613–649. Behav. Ecol. Sociobiol. 4, 61–74.
Ho, W.W., Cox-Fernandes, C., Alves-Gomes, J.A., Smith, T.G., 2007. Electro- Kramer, B., 1979. Electric and motor responses of the weakly electric fish, Gnatho-
communication in the weakly electric fish ‘‘Apteronotus’’ bonapartii is sexually nemus petersii (Mormyridae), to playback of social signals. Behav. Ecol. Sociobiol.
monomorphic. Int. Congr. Neuroethol. Vancouver B. C. 8, 89. 6, 67–79.
Ho, W.W., Fernandes, C.C., Alves-Gomes, J.A., Smith, G.T., 2010. Sex differences in Kramer, B., Kirschbaum, F., Markl, H., 1981. Species specificity of electric organ
the electrocommunication signals of the electric fish Apteronotus bonapartii. discharges in a sympatric group of gymnotoid fish from Manaus (Amazonas). In:
Ethology 116, 1050–1064. T, S., Czéh, G. (Eds.), Sensory Physiology of Aquatic Lower Vertebrates. Akadémiai
Ho, W.W., Rack, J.M., Smith, G.T., 2013. Divergence in androgen sensitivity Kiadó, Budapest, pp. 195–219.
contributes to population differences in sexual dimorphism of electro- Kramer, B., Otto, B., 1988. Female discharges are more electrifying – spontaneous
communication behavior. Horm. Behav. 63, 49–53. preference in the electric fish, Eigenmannia (gymnotiformes, teleostei). Behav.
Hopkins, C.D., 1972. Sex differences in electric signaling in an electric fish. Science Ecol. Sociobiol. 23, 55–60.
176, 1035–1037. Kramer, B., Otto, B., 1991. Waveform discrimination in the electric fish Eigenmannia:
Hopkins, C.D., 1974a. Electric communication in the reproductive behavior of Ster- sensitivity for the phase differences between the spectral components of a stimulus
nopygus macrurus (Gymnotoidei). Z. Tierpsychol. 35, 518–535. wave. J. Exp. Biol. 159, 1–22.
Hopkins, C.D., 1974b. Electric communication: functions in the social behavior of Landsman, R.E., Moller, P., 1988. Testosterone changes the electric organ discharge
Eigenmannia virescens. Behavior 50, 270–305. and external morphology of the mormyrid fish, Gnathonemus petersii (Mormyr-
Hopkins, C.D., 1976. Stimulus filtering and electroreception - tuberous electro- iformes). Experientia 44, 900–903.
receptors in 3 species of gymnotoid Ffsh. J. Comp. Physiol. 111, 171–207. Larson, E.A., Metzen, M.G., Chacron, M.J., 2014. Serotonin modulates electrosensory
Hopkins, C.D., 1981. On the diversity of electric signals in a community of mormyrid processing and behavior via 5-Ht2-Like receptors. Neuroscience 271, 108–118.
electric fish in West Africa. Am. Zool. 21, 211–222. Lavoue, S., Sullivan, J.P., 2004. Simultaneous analysis of five molecular markers
Hopkins, C.D., 1988. Neuroethology of electric communication. Annu. Rev. Neurosci. provides a well-supported phylogenetic hypothesis for the living bony-tongue fishes
11, 497–535. (Osteoglossomorpha: teleostei). Mol. Phylogenet. Evol. 33, 171–185.
Hopkins, C.D., 1999. Design features for electric communication. J. Exp. Biol. 202, Lewis, J.E., Gilmour, K.M., Moorhead, M.J., Perry, S.F., Markham, M.R., 2014. Action
1217–1228. potential energetics at the organismal level reveal a trade-off in efficiency at high
Hopkins, C.D., Heiligenberg, W.H., 1978. Evolutionary designs for electric signals and firing rates. J. Neurosci. 34, 197–201.
electroreceptors in gymnotoid fishes of Surinam. Behav. Ecol. Sociobiol. 3, Liu, H., Wu, M., Zakon, H.H., 2007. Individual variation and hormonal modulation of
113–134. a sodium channel b subunit in the electric organ correlate with variation in a social
Hopkins, C.D., Bass, A.H., 1981. Temporal coding of species recognition signals in an signal. Dev. Neurobiol. 67, 1289–1304.
electric fish. Science 212, 85–87. Liu, H., Wu, M., Zakon, H.H., 2008. A novel Naþ channel splice form contributes to the
Hopkins, C.D., Comfort, N., Bastian, J., Bass, A.H., July 1, 1990. Functional analysis regulation of an androgen-dependent social signal. J. Neurosci. 28, 9173–9182.
of sexual dimorphism in an electric fish, Hypopomus pinnicaudatus, order Gym- Machnik, P., Kramer, B., 2008. A male’s playback signal turns female Marcusenius
notiformes. Brain Behav. Evol. 35 (6), 350–367. pongolensis receivers on or off depending on his behavioral state. Commun. Integr.
Hupe, G.J., Lewis, J.E., 2008. Electrocommunication signals in free swimming brown Biol. 1, 128–131.
ghost knifefish, Apteronotus leptorhynchus. J. Exp. Biol. 211, 1657–1667. Maler, L., Ellis, W.G., 1987. Inter-male aggressive signals in weakly electric fish are
Hupe, G.J., Lewis, J.E., Benda, J., 2008. The effect of difference frequency on modulated by monoamines. Behav. Brain Res. 25, 75–81.
electrocommunication: chirp production and encoding in a species of weakly Markham, M.R., Stoddard, P., 2005. Adrenocorticotropic hormone enhances the
electric fish, Apteronotus leptorhynchus. J. Physiol. Paris 102, 164–172. masculinity of an electric communication signal by modulating the waveform and
Jalabert, C., Quintana, L., Pessina, P., Silva, A.C., 2015. Extra-gonadal steroids modulate timing of action potentials within individual cells. J. Neurosci. 25, 8746–8754.
non-breeding territorial aggression in weakly electric fish. Horm. Behav. 72, 60–67. Markham, M.R., Stoddard, P.K., 2013. Cellular mechanisms of developmental and sex
Julian, D., Crampton, W.G.R., Wohlgemuth, A., Albert, J., 2003. Oxygen consumption differences in the rapid hormonal modulation of a social communication signal.
in weakly electric Neotropical fishes. Oecologia 137, 502–511. Horm. Behav. 63, 586–597.
Kalmijn, A.J., 1974. The detection of electric fields from inanimate and animate Markham, M.R., Kaczmarek, L.K., Zakon, H.H., 2013. A sodium-activated potassium
sources other than electric organs. In: Fessard, A. (Ed.), Handbook of Sensory channel supports high-frequency firing and reduces energetic costs during rapid
Physiology. Springer-Verlag, Heidelberg, pp. 147–200. modulations of action potential amplitude. J. Neurophysiol. 109, 1713–1723.
Kawasaki, M., 1985. Physiology of tuberous electrosensory systems. In: Bullock, T.H., Markham, M.R., McAnelly, L.M., Stoddard, P.K., Zakon, H.H., 2009. Circadian and
et al. (Eds.), Electroreception. Springer, New York, pp. 154–194. social cues regulate ion channel trafficking. PLoS Biol. 7, e1000203.
Kawasaki, M., Heiligenberg, W., 1989. Distinct mechanisms of modulation in Marsat, G., Maler, L., 2012. Cellular and circuit properties supporting different sensory
a neuronal oscillator generate different socialsignals in the electric fish Hypo- coding strategies in electric fish and other systems. Curr. Opin. Neurobiol. 22,
pomus. J. Comp. Physiol. A 165, 731–741. 686–692.
Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology 97

McAnelly, L.M., Zakon, H.H., 1996. Protein kinase A activation increases sodium Petzold, J.M., Smith, G.T., 2014. Harmonic content of waveform is nonlinearly related
current magnitude in the electric organ of Sternopygus. J. Neurosci. 16, to harmonic content of envelope during weakly electric fish social interactions.
4383–4388. Integr. Comp. Biol. 54, e330.
McAnelly, L.M., Zakon, H.H., 2000. Coregulation of voltage-dependent kinetics of Naþ Petzold, J.M., Smith, G.T., 2016. Androgens regulate sex differences in signaling but
and Kþ currents in electric organ. J. Neurosci. 20, 3408–3414. are not associated with male variation in morphology in the weakly electric fish
McAnelly, L.M., Silva, A.C., Zakon, H.H., 2003. Cyclic AMP modulates electrical Parapteronotus hasemani. Horm. Behav. 78, 67–71.
signaling in a weakly electric fish. J. Comp. Physiol. A 189, 273–282. Pouso, P., Quintana, L., Bolatto, C., Silva, A.C., 2010. Brain androgen receptor
McAnelly, L.M., Zakon, H., 2007. Androgen modulates the kinetics of the delayed expression correlates with seasonal changes in the behavior of a weakly electric
rectifying Kþ current in the electric organ of a weakly electric fish. Dev. Neurobiol. fish, Brachyhypopomus gauderio. Horm. Behav. 58, 729–736.
67, 1589–1597. Quintana, L., Silva, A.C., Berois, N., Macadar, O., 2004. Temperature induces gonadal
McGregor, P.K., Westby, G.W.M., 1992. Discrimination of individually characteristic maturation and affects electrophysiological sexual maturity indicators in Brachy-
electric organ discharges by a weakly electric fish. Anim. Behav. 43, 977–986. hypopomus pinnicaudatus from a temperate climate. J. Exp. Biol. 207,
Metzner, W., 1999. Neural circuitry for communication and jamming avoidance in 1843–1853.
gymnotiform electric fish. J. Exp. Biol. 202, 1365–1375. Quintana, L., Pouso, P., Fabbiani, G., Macadar, O., 2010. A central pacemaker that
Metzner, W., Heiligenberg, W., 1991. The coding of signals in the electric underlies the production of seasonal and sexually dimorphic social signals:
communication of the gymnotiform fish Eigenmannia - from electroreceptors to anatomical and electrophysiological aspects. J. Comp. Physiol. A 197, 75–88.
neurons in the torus semicircularis of the midbrain. J. Comp. Physiol. A 169, Reardon, E.E., Parisi, A., Krahe, R., Chapman, L.J., 2011. Energetic constraints on
135–150. electric signalling in wave-type weakly electric fishes. J. Exp. Biol. 214, 4141–4150.
Metzner, W., Juranek, J., 1997. A sensory brain map for each behavior? Proc. Natl. Rodríguez-Cattáneo, A., Aguilera, P., Cilleruelo, E., Crampton, W.G.R., Caputi, A.A.,
Acad. Sci. U.S.A. 94, 14798–14803. 2013. Electric organ discharge diversity in the genus Gymnotus: anatomo-
Metzner, W., Viete, S., 1996. The neuronal basis of communication and orientation in functional groups and electrogenic mechanisms. J. Exp. Biol. 216, 1501–1515.
the weakly electric fish, Eigenmannia. 2. Electrolocation and avoidance of jamming Rodriguez-Cattaneo, A., Pereira, A.C., Aguilera, P.A., Crampton, W.G.R., Caputi, A.A.,
by neighboring conspecifics. Naturwissenschaften 83, 71–77. 2008. Species-specific diversity of a fixed motor pattern: the electric organ
Meyer, J.H., Leong, M., Keller, C., 1987. Hormone-induced and maturational changes discharge of Gymnotus. PLoS One 3, e2038.
in electric organ discharges and electroreceptor tuning in the weakly electric fish Salazar, V.L., Stoddard, P.K., 2008. Sex differences in energetic costs explain sexual
Apteronotus. J. Comp. Physiol. A 160, 385–394. dimorphism in the circadian rhythm modulation of the electrocommunication signal
Meyer, J.H., Zakon, H.H., 1982. Androgens alter the tuning of electroreceptors. of the gymnotiform fish Brachyhypopomus pinnicaudatus. J. Exp. Biol. 211,
Science 217, 635–637. 1012–1020.
Meyer, J.H., 1983. Steroid influences upon the discharge frequencies of a weakly Salgado, J., Zupanc, G.K.H., 2011. Echo response to chirping in the weakly electric
electric fish. J. Comp. Physiol. A 153, 29–37. brown ghost knifefish (Apteronotus leptorhynchus): role of frequency and amplitude
Migliaro, A., Silva, A.C., 2016a. Melatonin regulates daily variations in electric behavior modulations. Can. J. Zool. 89, 498–508.
arousal in two species of weakly electric fish with different social structure. Brain Salisbury, J.P., Sirbulescu, R.F., Moran, B.M., Auclair, J.R., Zupanc, G.K.H.,
Behav. Evol. http://dx.doi.org/10.1159/000445494. Agar, J.N., 2015. The central nervous system transcriptome of the weakly electric
Migliaro, A., Silva, A.C., 2016b. Free running in the wild: circadian rhythmicity in brown ghost knifefish (Apteronotus leptorhynchus): de novo assembly, annotation,
electric behavior in two species of South American weakly electric fish. In: XII and proteomics validation. BMC Genomics 16, 1.
International Congress of Neuroethology. Schaefer, J.E., Zakon, H.H., 1996. Opposing actions of androgen and estrogen on
Mills, A., Zakon, H.H., 1987. Coordination of EOD frequency and pulse duration in in vitro firing frequency of neuronal oscillators in the electromotor system.
a weakly electric wave fish: the influence of androgens. J. Comp. Physiol. A 161, J. Neurosci. 16, 2860–2868.
417–430. Scheich, H., Bullock, T.H., Hamstra, R.H., 1973. Coding properties of 2 classes of
Mills, A., Zakon, H.H., 1991. Chronic androgen treatment increases action potential afferent nerve fibers - high-frequency electroreceptors in electric fish, Eigen-
duration in the electric organ of Sternopygus. J. Neurosci. 11, 2349–2361. mannia. J. Neurophysiol. 36, 39–60.
Mills, A., Zakon, H.H., Marchaterre, M., Bass, A.H., 1992. Electric organ morphology Shumway, C.A., Zelick, R.D., 1988. Sex recognition and neuronal coding of electric
of Sternopygus macrurus, a wave-type, weakly electric fish with a sexually organ discharge waveform in the pulse-type weakly electric fish, Hypopomus
dimorphic EOD. J. Neurobiol. 23, 920–932. occidentalis. J. Comp. Physiol. A 163, 465–478.
Naruse, M., Kawasaki, M., 1998. Possible involvement of the ampullary electro- Silva, A.C., Quintana, L., Galeano, M., Errandonea, P., Macadar, O., 1999. Water
receptor system in detection of frequency-modulated electrocommunication signals temperature sensitivity of EOD waveform in Brachyhypopomus pinnicaudatus.
in Eigenmannia. J. Comp. Physiol. A 183, 543–552. J. Comp. Physiol. A 185, 187–197.
Nelson, M.E., Xu, Z., Payne, J.R., 1997. Characterization and modeling of P-type Silva, A.C., Quintana, L., Ardanaz, J.L., Macadar, O., 2002. Environmental and
electrosensory afferent responses to amplitude modulations in a wave-type hormonal influences upon EOD waveform in gymnotiform pulse fish. J. Physiol.
electric fish. J. Comp. Physiol. A 181, 532–544. Paris 96, 473–484.
Oestreich, J., Zakon, H.H., 2005. Species-specific differences in sensorimotor Silva, A.C., Quintana, L., Galeano, M., Errandonea, P., 2003. Biogeography and
adaptation are correlated with differences in social structure. J. Comp. Physiol. A breeding in gymnotiformes from Uruguay. Environ. Biol. Fishes 66, 329–338.
191, 845–856. Silva, A.C., Perrone, R., Macadar, O., 2007. Environmental, seasonal, and social
Pappas, G., Waxman, S., Bennett, M., 1975. Morphology of spinal electromotor modulations of basal activity in a weakly electric fish. Physiol. Behav. 90,
neurons and presynaptic coupling in the gymnotid Sternarchus albifrons. 525–536.
J. Neurocytol. 4, 469–478. Silva, A.C., Quintana, L., Perrone, R., Sierra, F., 2008. Sexual and seasonal plasticity
Pedraja, F.P., Aguilera, P., Caputi, A.A., Budelli, R., 2014. Electric imaging through in the emission of social electric signals. Behavioral approach and neural bases.
evolution, a modeling study of commonalities and differences. PLoS Comput. Biol. J. Physiol. Paris 102, 272–278.
10, e1003722. Silva, A.C., Perrone, R., Zubizarreta, L., Batista, G., Stoddard, P.K., 2013. Neuro-
Pereira, A.C., Rodriguez-Cattaneo, A., Castello, M., Caputi, A.A., 2007. Post-natal modulation of the agonistic behavior in two species of weakly electric fish that
development of the electromotor system in a pulse gymnotid electric fish. display different types of aggression. J. Exp. Biol. 216, 2412–2420.
J. Exp. Biol. 210, 800–814. Sinnett, P.M., Markham, M.R., 2015. Food deprivation reduces and leptin increases
Perrone, R., 2012. La vasotocina modula el comportamiento social de dos especies de the amplitude of an active sensory and communication signal in a weakly electric
peces eléctricos con diferente socialidad. PEDECIBA, Universidad de la República, fish. Horm. Behav. 71, 31–40.
Montevideo. Sisneros, J.A., Tricas, T.C., 2000. Androgen-induced changes in the response
Perrone, R., Macadar, O., Silva, A.C., 2009. Social electric signals in freely moving dynamics of ampullary electrosensory primary afferent neurons. J. Neurosci. 20,
dyads of Brachyhypopomus pinnicaudatus. J. Comp. Physiol. A 195, 501–514. 8586–8595.
Perrone, R., Batista, G., Lorenzo, D., Macadar, O., Silva, A.C., 2010. Vasotocin actions Smith, G.T., 2006. Pharmacological characterization of ionic currents that regulate
on electric behavior: interspecific, seasonal, and social context-dependent differ- high-frequency spontaneous activity of electromotor neurons in the weakly electric
ences. Front. Behav. Neurosci. 4, 52. fish, Apteronotus leptorhynchus. J. Neurobiol. 66, 1–18.
Perrone, R., Migliaro, A., Comas, V., Quintana, L., Borde, M., Silva, A.C., 2014. Smith, G.T., 2013. Evolution and hormonal regulation of sex differences in the elec-
Local vasotocin modulation of the pacemaker nucleus resembles distinct electric trocommunication behavior of ghost knifefishes (Apteronotidae). J. Exp. Biol. 216,
behaviors in two species of weakly electric fish. J. Physiol. Paris 108, 203–212. 2421–2433.
98 Weakly Electric Fish: Behavior, Neurobiology, and Neuroendocrinology

Smith, G.T., Zakon, H.H., 2000. Pharmacological characterization of ionic currents Waxman, S., Pappas, G., Bennett, M., 1972. Morphological correlates of functional
that regulate the pacemaker rhythm in a weakly electric fish. J. Neurobiol. differentiation of nodes of Ranvier along single fibers in the neurogenic electric
42, 270. organ of the knife fish Sternarchus. J. Cell Biol. 53, 210–224.
Smith, G.T., Combs, N., 2008. Serotonergic activation of 5HT1A and 5HT2 receptors Weld, M.M., Maler, L., 1992. Substance P-like immunoreactivity in the brain of the
modulates sexually dimorphic communication signals in the weakly electric fish gymnotiform fish Apteronotus leptorhynchus: presence of sex differences.
Apteronotus leptorhynchus. Horm. Behav. 54, 69–82. J. Chem. Neuroanat. 5, 107–129.
Spiro, J.E., 1997. Differential activation of glutamate receptor subtypes on a single Weld, M.M., Kar, S., Maler, L., Quirion, R., 1994. The distribution of tachykinin binding
class of cells enables a neural oscillator to produce distinct behaviors. J. Neuro- sites in the brain of an electric fish (Apteronotus leptorhynchus). J. Chem. Neu-
physiol. 78, 835–847. roanat. 7, 123–139.
Stockl, A., Sinz, F., Benda, J., Grewe, J., 2014. Encoding of social signals in all three Westby, G., 1988. The ecology, discharge diversity and predatory behaviour of
electrosensory pathways of Eigenmannia virescens. J. Neurophysiol. 112, gymnotiforme electric fish in the coastal streams of French Guiana. Behav. Ecol.
2076–2091. Sociobiol. 22, 341–354.
Stoddard, P.K., Markham, M.R., Salazar, V.L., Allee, S., 2007. Circadian rhythms in Wong, R.Y., Hopkins, C.D., 2007. Electrical and behavioral courtship displays in the
electric waveform structure and rate in the electric fish Brachyhypopomus pin- mormyrid fish Brienomyrus brachyistius. J. Exp. Biol. 210, 2244–2252.
nicaudatus. Physiol. Behav. 90, 11–20. Xu-Friedman, M.A., Hopkins, C.D., 1999. Central mechanisms of temporal analysis in
Szabó, T., 1974. Anatomy of the specialized lateral line organs of electroreception. In: the knollenorgan pathway of mormyrid electric fish. J. Exp. Biol. 202, 1311–1318.
Fessard, A. (Ed.), Handbook of Sensory Physiology. Springer-Verlag, Heidelberg, Zakon, H.H., 1986. The electroreceptive periphery. In: Bullock, T.H., Heiligenberg, W.
pp. 14–58. (Eds.), Electroreception. Wiley, New York, pp. 103–156.
Stoddard, P., 1999. Predation enhances complexity in the evolution of electric fish Zakon, H.H., Dunlap, K.D., 1999. Sex steroids and communication signals in electric
signals. Nature 400, 254–256. fish: a tale of two species. Brain Behav. Evol. 54, 61–69.
Smith, G.T., 1999. Ionic currents that contribute to a sexually dimorphic communi- Zakon, H.H., 1999. Sex steroids and weakly electric fish: a model system for acti-
cation signal in weakly electric fish. J. Comp. Physiol. A 185, 379–387. vational mechanisms of hormone action. In: Matsumoto, A. (Ed.), Sexual Differ-
Tagliacollo, V.A., Bernt, M.J., Craig, J.M., Oliveira, C., Albert, J.S., 2016. Model-based entiation of the Brain. CRC Press, Boca Raton, FL, pp. 95–112.
total evidence phylogeny of neotropical electric knifefishes (teleostei, Gymnoti- Zakon, H.H., Meyer, J.H., 1983. Plasticity of electroreceptor tuning in the weakly
formes). Mol. Phylogenet. Evol. 95, 20–33. electric fish, Sternopygus dariensis. J. Comp. Physiol. 153, 477–487.
Tallarovic, S.K., Zakon, H.H., 2002. Electrocommunication signals in female brown Zakon, H.H., Thomas, P., Yan, H.Y., 1991. Electric organ discharge frequency and
ghost electric knifefish, Apteronotus leptorhynchus. J. Comp. Physiol. A 188, plasma sex steroid levels during gonadal recrudescence in a natural population of the
649–657. weakly electric fish Sternopygus macrurus. J. Comp. Physiol. A 169, 493–499.
Tan, E.W., Nizar, J.M., Carrera-G, E., Fortune, E.S., 2005. Electrosensory interference Zakon, H.H., Yan, H.Y., Thomas, P., 1990. Human chorionic gonadotropin-induced
in naturally occurring aggregates of a species of weakly electric fish, Eigenmannia shifts in the electrosensory system of the weakly electric fish, Sternopygus.
virescens. Behav. Brain Res. 164, 83–92. J. Neurobiol. 21, 826–833.
Telgkamp, P., Combs, N., Smith, G.T., 2007. Serotonin in a diencephalic nucleus Zakon, H.H., Oestreich, J., Tallarovic, S., Triefenbach, F., 2002. EOD modulations of
controlling communication in an electric fish: sexual dimorphism and relationship to brown ghost electric fish: JARs, chirps, rises, and dips. J. Physiol. Paris 96,
indicators of dominance. Dev. Neurobiol. 67, 339–354. 451–458.
Triefenbach, F.A., Zakon, H.H., 2003. Effects of sex, sensitivity and status on cue Zhou, M., Smith, G.T., 2006. Structure and sexual dimorphism of the electro-
recognition in the weakly electric fish Apteronotus leptorhynchus. Anim. Behav. 65, communication signals of the weakly electric fish, Adontosternarchus devenanzii.
19–28. J. Exp. Biol. 209, 4809–4818.
Triefenbach, F.A., Zakon, H.H., 2008. Changes in signalling during agonistic inter- Zubizarreta, L., Stoddard, P.K., Silva, A.C., 2015. Aggression levels affect social
actions between male weakly electric fish, Apteronotus leptorhynchus. Anim. interaction in the non-breeding territorial aggression of the weakly electric fish,
Behav. 75, 1263–1275. Gymnotus omarorum. Ethology 121, 8–16.
Turner, C.R., Derylon, M., de Santana, C.D., Alves-Gomes, J.A., Smith, G.T., Zucker, M.S., 1997. Hormonal Basis of Sexual Dimorphism in and Steroid Sensitivity of
2007. Phylogenetic comparative analysis of electric communication signals in the Electric Organ Discharge Frequency of the Gymnotiform Fish, Apteronotus
ghost knifefishes (Gymnotiformes: apteronotidae). J. Exp. Biol. 210, leptorhynchus (Ph.D. Dissertation). University of Texas at Austin.
4104–4122. Zupanc, G.K., Horschke, I., 1995. Proliferation zones in the brain of adult gymnotiform
Vonderschen, K., Chacron, M.J., 2011. Sparse and dense coding of natural stimuli by fish: a quantitative mapping study. J. Comp. Neurol. 353, 213–233.
distinct midbrain neuron subpopulations in weakly electric fish. J. Neurophysiol. Zupanc, G.K., Maler, L., 1993. Evoked chirping in the weakly electric fish Apteronotus
106, 3102–3118. leptorhynchus: a quantitative biophysical analysis. Can J. Zool. 71, 2301–2310.
Watson, D., Bastian, J., 1979. Frequency-response characteristics of electro- Zupanc, G.K.H., Maler, L., 1997. Neuronal control of behavioral plasticity: the pre-
receptors in the weakly electric fish, Gymnotus carapo. J. Comp. Physiol. 134, pacemaker nucleus of weakly electric gymnotiform fish. J. Comp. Physiol. A 180,
191–202. 99–111.
NON-MAMMALIAN
VERTEBRATES: AMPHIBIANS
This page intentionally left blank
2.06 Neuroendocrine Control of Social Behavior in Frogs
Walter Wilczynski, Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
Ó 2017 Elsevier Inc. All rights reserved.

2.06.1 Introduction 101


2.06.2 Seasonal Patterns of Hormones and Behavior 102
2.06.2.1 Seasonal Changes in Gonadal Steroids 102
2.06.2.2 Seasonal Changes in Social Behavior Match Changes in Gonadal Steroids 103
2.06.2.3 Seasonal Variation in Other Hormones: Adrenal Steroids 105
2.06.2.4 Seasonal Variation in Other Hormones: Melatonin 105
2.06.3 Steroid Hormone Receptors in the Brain 106
2.06.4 Hormone Effects on Hearing 108
2.06.4.1 Seasonal Changes in Hearing Track Seasonal Changes in Sex Steroids and Behavior 109
2.06.4.2 Experimental Manipulation of Circulating Gonadal Steroids Modifies the Auditory System 109
2.06.4.3 Implications for Social Behavior 110
2.06.5 Hearing Effects on Hormones 110
2.06.5.1 Anatomical Links between the Auditory Midbrain and Hypothalamic Areas Define an Audioendocrine
Circuit 110
2.06.5.2 Hearing Calls Elevates Levels of Circulating Steroid Hormones 111
2.06.5.3 Short-Term Exposure to Aggressive or Advertisement Calls Has Different Effects 111
2.06.5.4 Implications for Social Behavior 113
2.06.6 Summary and Conclusions 113
References 114

2.06.1 Introduction times of the year and become reproductively quiescent during
others. Frogs need water, or at least high local humidity, for
Anuran amphibians (frogs and toads) occupy a greater diversity successful egg deposition and larval development, and reproduc-
of habitats and express a wider variety of reproductive social tion is restricted to periods providing that. What defines the
behaviors than is generally appreciated outside of the field of season can differ. In temperate zones, the usual circannual
herpetology (Feder and Bruggren, 1992; Duellman and Trueb, patterns of temperature and day length define seasons. In other
1994; Wells, 2007). Anuran species range from the arctic to areas, seasonally occurring environmental cues dictate reproduc-
the tropical, and from dry desert regions to constantly wet habi- tion. Tropical species breed during defined rainy seasons, for
tats; some species, such as the Pipidae, are even fully aquatic. example. The season can be very long, as in many North Amer-
Anurans also vary in the degree to which they express different ican species such as green treefrogs (Hyla cinerea) that are repro-
kinds of social behavior. Most work on anuran social behavior ductively active, weather permitting, from late spring to early fall,
has focused on the male and female adult behaviors that lead or very short, sometimes being restricted to a matter of days in
to mate choice and ultimately reproduction. These behavioral very early spring, as in wood frogs (Rana sylvatica) which emerge
patterns are more consistent across species than are other types for a brief, frenetic breeding period in ephemeral ponds. In
of social behavior. Patterns of aggression for example, to date extreme cases, breeding ‘seasons’ in challenging habits such as
only described among males, vary in degree across species, and the southwest American desert occupied by spade foot toads
parental care can range from none (the most common condi- in the family Scaphiopodidae consist of unpredictable rain
tion), to depositing eggs in foam nests, to guarding or feeding showers that bring out immediate, but very brief, bursts of repro-
eggs and tadpoles by either males, females, or both. The variety ductive activity that can be over in a day. Regardless of the length
of ecological and behavioral profiles, viewed within the context or trigger, something must gate reproduction and coordinate the
of several largely conserved elements of social behavior that physiological, neural, and behavioral components of it. Gonadal
define this taxonomic group, provides ample opportunities for steroid hormones play this role throughout vertebrates. At least
comparative and evolutionary studies in brain–hormone– for temperate zone species with defined annual breeding
behavior relationships that have not been fully exploited. seasons, seasonal variation in gonadal steroids coincides with
The understanding of anuran social behavior, and the seasonal variation in reproductive behavior and physiology.
brain–hormone interactions behind it, is based on relatively Acoustic communication is the foundation for anuran social
few temperate and neotropical species that have long served behavior. Except for the small number of species in the most
as models for neuroethology, acoustic communication, and primitive frog families (the genera Ascaphus and Leiopelma) all
evolutionary animal behavior. With this caveat, there are anurans studied to date base their social interaction on vocali-
several features of anuran social behavior that define this group zations. Most species have a lek-like mating system (Höglund
of vertebrates. and Alatalo, 1995) in which males aggregate on communal
Reproduction is seasonal. Like most vertebrates, frogs engage in display sites (choruses) and produce advertisement, or mating,
reproduction and its attendant social behavior during specific calls to attract females (Wells, 1977, 2007). Females use the

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00026-2 101


102 Neuroendocrine Control of Social Behavior in Frogs

calls to find males and to compare them in order to make along with them. In amphibians, these seasonal changes corre-
a choice among them to initiate mating. Once a female has late most obviously with seasonal changes in the gonadal
picked a male, they enter into amplexus, in which the male steroid hormones, androgens and estrogens. Both the calling
clasps her from the dorsal side, and eggs and sperm are and the responses to calls are the most obvious seasonal social
co-released for external fertilization. Amplexus can last for behaviors in male and female frogs. As such, one would expect
hours in many species. This social system leads to intense selec- them to be gated by seasonal cycles of gonadal steroid
tion pressure on males to call vigorously throughout the hormones, which in fact they are. Kelley (in Kelley, Chapter
breeding season with clearly defined species-specific call 2.08, Hormones and Vocal Systems: Insights from Xenopus)
features attractive to females. Females in turn are under the discusses this for calling circuitry (see also Coddington,
pressure of their developing egg masses to find a mate within Chapter 2.07, The Vertebrate Brain Stem as the Primary Site
the cacophony of vocally competing males before their eggs of Behavior Command: Insights from an Amphibian, for
must be released fertilized or not. a similar analysis focusing on newts), and as such this chapter
In addition to advertisement calls, other vocalizations may will focus more on the auditory system providing the sensory
be produced. In some species males produce an aggressive call side of this communication. Hormones such as adrenal
that may be a modification of the advertisement call or a sepa- steroids and melatonin change seasonally as well. Their contri-
rate vocalization. In some species females produce recognition, butions to the control of social behavior remain less well
courtship, or release calls, the last signaling nonreceptivity. In understood but an important avenue for further study.
a few species, females have been found to produce a vocaliza-
tion that seems to function as some sort of advertisement call
2.06.2.1 Seasonal Changes in Gonadal Steroids
(Emerson and Boyd, 1999), although this is relatively rare (at
least among commonly observed species). As expected for vertebrates with sexually dimorphic, seasonally
Acoustic communication is a sexually differentiated behavior. gated, reproductive social behavior, seasonal variation in circu-
In the vast majority of anuran species, advertisement calling lating gonadal steroid levels is present in anurans (Licht et al.,
is a male trait, and vocal behavior overall is strongly male 1983; Pierantoni et al., 1984; Iela et al., 1986; Rastogi et al.,
biased. The sex difference can be so great that females do 1986; Itoh et al., 1990; Herman, 1992; Narayan et al., 2010;
not vocalize at all, a feature of several commonly studied Scaia et al., 2013). Studies that describe this pattern, coming
North American and neotropical species. In other commonly mainly from work in northern temperate species with pro-
studied species such as Xenopus sp. both sexes produce longed breeding seasons, indicate both a basic seasonal pattern
communication sounds, but the behavior and the features of endocrine changes plus variation within a season pegged to
of the underlying neural circuits and biomechanical mecha- reproductive cycles or environmental and social context. As in
nisms of the larynx clearly show a male-biased sex difference the yearly cycle seen in many seasonally breeding vertebrates,
(reviewed in Kelley, Chapter 2.08, Hormones and Vocal gonadal steroid levels are higher during the breeding season,
Systems: Insights from Xenopus). The great sex difference in when reproductive and aggressive social behavior is also
calling in many species provides a substrate for investigating high. In several species investigated, anuran males have been
sex differences in the neural and biomechanical mechanisms found to have the highest gonadal steroid (testosterone) levels
of this behavior. How these vary across species in which at the beginning of the breeding season; in others, androgens or
females do and do not call has not been studied in a systematic estrogens either remain constant or increase throughout the
way. breeding season. In either case, a rapid decline in gonadal
Although the sexes diverge in their capacity to call, both steroids marks the end of the breeding season, and the end of
males and females hear and respond to the advertisement calls reproductive social behavior. What the patterns look like in
that males produce. These responses, however, differ between frogs with extremely short breeding seasons, or with unpredict-
the sexes. Males respond by calling back to neighbors’ calls or able behavior based on local climate conditions, is less well
with aggressive responses that may lead to attacking nearby understood, but Harvey et al. (1997) described a similar
calling neighbors. Females respond by orienting and displaying pattern in the spadefoot toad, Scaphiopus couchii, an explosive
phonotaxis behavior (that is, moving toward a sound) to desert breeder that emerges and breeds quickly when rain
conspecific calls (which males may also do depending on the occurs. There, as in animals with a prolonged breeding season,
situation Bernal et al., 2007). It is clear that females evaluate androgens in males, and estrogens and progesterone in
the features of male calls, discriminate among them, and females, were highest when individuals had emerged to breed.
base their mate choice on their assessment (Ryan, 1985). In In addition to the seasonal hormone changes that gate the
general, males are much less discriminating in terms of the breeding season, individual variation in androgens and estro-
advertisement calls they will respond to, as predicted by theo- gens occurs within the breeding season. Some of this is driven
retical considerations of sex differences in the fitness costs of by individual reproductive cycles. For example, in species
recognition errors (Trivers, 1972). with long breeding seasons, females may cycle through
several clutches of eggs with estrogens and/or progesterone
levels cycling accordingly, peaking when eggs are ready to
2.06.2 Seasonal Patterns of Hormones and be released, then dropping quickly once mating is over
Behavior (Iela et al., 1986; Harvey et al., 1997; Medina et al., 2004;
Lynch and Wilczynski, 2005; Gramapurohit and Radder,
The hormonal state of a seasonal organism varies greatly over 2013; Gordon and Hellman, 2015). Interestingly, in one
a year as physiological demands change and behaviors shift species males were found to have a similar testosterone
Neuroendocrine Control of Social Behavior in Frogs 103

pattern: highest immediately before and during mating, then Wetzel and Kelley, 1983; Solís and Penna, 1997; Burmeister
dropping quickly once mating was over (Gramapurohit and and Wilczynski, 2001) and eliminated when males are cas-
Radder, 2013). More unpredictable context-dependent endo- trated (Schmidt, 1966; Palka and Gorbman, 1973; Kelley and
crine changes may also occur related to any number of envi- Pfaff, 1976; Burmeister and Wilczynski, 2001; Wilczynski
ronmental cues. The immediate social environment is one et al., 2005; Leary, 2009; Zornik and Kelley, 2011). Male calling
such cue. The effect of social interactions on endocrine state is not always expressed in lock-step with varying androgen
will be discussed in Section 2.06.5. levels, however, especially when naturally behaving animals
are studied. Solís and Penna (1997) found that androgen
concentrations and vocalization rates were correlated in a field
2.06.2.2 Seasonal Changes in Social Behavior Match
population of Batrachyla taeniata. Studies in other species have
Changes in Gonadal Steroids
produced conflicting results. In some cases, androgens are
Male and female reproductive social behavior waxes and wanes lower in calling anurans relative to noncallers (Mendonça
with the seasonal changes in gonadal steroids, providing strong et al., 1985), whereas in other species, androgens are higher
correlative evidence that these hormones are important modu- in calling males (Townsend and Moger, 1987; Marler and
lators of male calling and female phonotaxis. On average, Ryan, 1996). One of the reasons for the variety of findings is
seasonal changes in male calling track seasonal changes in that natural calling behavior is influenced by many factors.
androgen levels (Polzonetti-Magni et al., 1984; Varriale et al., Burmeister and Wilczynski (2001) provided some insight
1986; Itoh and Ishii, 1990; Gobbetti et al., 1991; Harvey into this variability in a study that distinguished spontaneous
et al., 1997). Male calling is often most vigorous at the point calling from evoked calling, that is, calling when males were
of the breeding season when testosterone is highest. In green isolated from other callers versus when males heard the calls
treefrogs, this is at the beginning of the season. One laboratory of other males. There, as expected, castration eliminated all
study (O’Bryant and Wilczynski, 2010) using this species held calling. In intact and castrated frogs implanted with testos-
environmental conditions constant throughout the summer terone capsules, testosterone levels correlated with sponta-
and found that both testosterone and spontaneous calling neous calling, but not evoked calling in the same animals.
declined together from June through September (Figure 1). When hearing other males’ calls, males with sufficient testos-
Experimental manipulation of gonadal steroid levels also terone vigorously responded by calling at high rates. This
clearly shows the role these hormones play in modulating pattern of antiphonal calling is a standard feature of frog social
frog social behavior. In males, vocal behavior is androgen behavior (Wells, 1977; Gerhardt and Huber, 2002). Similarly,
dependent (Wada et al., 1976; Wada and Gorbman, 1977; although spontaneous calling declined along with testosterone
in laboratory-held green treefrogs, evoked calling rates (calling
when chorus sounds were present) did not change over the
breeding season (O’Bryant and Wilczynski, 2010), suggesting
again that social cues can override whatever motivational effect
androgens may have. In a separate study (Burmeister et al.,
2001), a male’s latency to resume calling after capture was
also correlated with circulating androgen level. In total, this
work suggests that testosterone has a permissive effect and
increases the motivation to call, but once social cues are added,
male calling is at least, if not more, driven by the calls of
competitor males.
Several aspects of female reproductive behavior also vary
with fluctuating gonadal steroid levels particularly as females
with prolonged breeding seasons pass through different phases
of their ovulatory cycle (Lea et al., 2000; Bosch and Boyero,
2004; Lynch et al., 2006). Female sexual behavior most often
includes phonotaxis, or the orientation to and movement
toward conspecific calling males (Ryan, 1985; Gerhardt and
Huber, 2002), but, depending on the species, can also include
producing vocalizations (Tobias et al., 1988; Roy et al., 1995;
Emerson and Boyd, 1999), or exhibiting behaviors that reject
males, such as release calls and leg extensions (Diakow
and Nemiroff, 1981; Kelley, 1982; Boyd, 1992). Because
phonotaxis is such a common and robust indicator of female
receptivity, it is most often used in studies of female social
Figure 1 Mean (þSE) plasma testosterone (a) and log spontaneous
behavior. As for male calling, female phonotaxis toward
calls per hour (b) in May, July, and September in green treefrogs (Hyla
cinerea) held over their normal summer breeding season in constant conspecific calls is expressed during the breeding season
summer environmental conditions. Data from O’Bryant, E.L., Wilczynski, when estrogens and progesterone are high. Lynch and
W., 2010. Changes in plasma testosterone levels and brain AVT cell Wilczynski (2005) tracked the gonadal steroid levels over
number during the breeding season in the green treefrog. Brain Behav. the natural ovulatory cycle of field-caught túngara frogs
Evol. 75 (4), 271–281. (Physalaemus (¼Engystomops) pustulosus). Females collected at
104 Neuroendocrine Control of Social Behavior in Frogs

a breeding site prior to mating had low levels of estrogens and how receptive or permissive females become, they always elect
progesterone. Females that had entered into amplexus (the to go to an attractive call over an unattractive call if they have
condition in which a male tightly grasps a female to align their the choice. Chakraborty and Burmeister (2009) found that
bodies for sperm and egg release), indicating they were ready to estrogens alone could account for these behavioral changes.
release eggs, had peak estrogens and progesterone levels. As female frogs reach the point when their eggs are mature
Immediately after egg release, estrogens and progesterone enough to be released, they are under strong pressure to find
rapidly fell to their lowest levels during the breeding season. a suitable male. At some point, the egg mass becomes so large
Female phonotaxis behavior can assess the behavioral variation that it will be released regardless of whether mating is
that occurs with these natural cycles or after experimental underway to fertilize it. If this happens, females have wasted
manipulation of hormone levels and has been used in natu- the considerable resources that went into growing the egg
rally cycling and in hormonally treated túngara frog females mass, and wasted what may be one of their few, and perhaps
(Figure 2; Lynch and Wilczynski, 2005; Lynch et al., 2006; only, opportunities to procreate. As Lynch et al. (2006) argued,
Chakraborty and Burmeister, 2009). Receptivity, as defined estrogen-driven receptivity drives the female to search for and
by an approach to a recording of a male advertisement call, accept males. Estrogen-driven decreases in choosiness change
increases and decreases with the rise and fall of gonadal the search and decision process so that she will accept a wider
hormones, as happens in other cycling female vertebrates. At variety of males as the point of egg release becomes eminent. At
the same time, phonotaxis toward relatively unattractive male no time, however, does the female lose her ability to pick the
calls (‘permissiveness’) also increases when estrogens are more ‘attractive’ male among the choices she has.
high. However, females never lose the ability to discriminate Other studies have implicated progesterone (Gordon and
between attractive multisyllable calls and unattractive single Gerhardt, 2009), luteinizing hormone itself (Kelley, 1982),
syllable or artificially manipulated calls made to sound like and prostaglandin (Diakow and Nemiroff, 1981; Schmidt,
hybrid signals regardless of estrogens levels and reproductive 1985; Weintraub et al., 1985) in some aspects of female frog
state. That is, when estrogens and progesterone levels are reproductive social behavior. It is also possible that in those
high, which coincides with the point at which eggs are ready frog species where females produce advertisement vocaliza-
to be released, females are highly motivated to select a calling tions, testosterone might regulate this behavior as it does in
male and are more accepting of calls that, under lower males (Emerson and Boyd, 1999). There is, however, very little
hormone levels, they would reject. However, regardless of direct evidence that this is the case.

Receptivity
As estrogens increase:

Noise Normal
call
Response to a
normal male call
Permissiveness

Hybrid call Noise

Response to a
normally ignored call
Discrimination

One Two
syllable syllable
call call
Go to the more
attractive call

Figure 2 Diagram of phonotaxis experiments used in Lynch et al. (2005, 2006) to show changes in female túngara frog (Physalaemus pustulosus)
behavior as plasma levels of estrogens change. Females are placed in the center of a large experimental chamber and stimuli are broadcast from
speakers on either side. Receptivity (top panel) can be measured by the proportion of females moving toward a male call. Permissiveness (middle
panel) is measured by the proportion of females moving toward a normally unattractive call, such as an artificial call constructed as an acoustic
hybrid of a P. pustulosus and a Physalaemus enesfae call. Discrimination (bottom panel) is measured by the proportion of females moving toward
a two-syllable versus one-syllable call; receptive túngara females will display phonotaxis behavior to either when presented alone, but normally find
multisyllable calls more attractive. As estrogens increase receptivity increases (females are more likely to respond to any call), and permissiveness
increases (females are more likely to respond to a hybrid call), but females maintain the ability to discriminate between attractive two-syllable calls
and less attractive one-syllable calls by preferentially going toward the two-syllable call.
Neuroendocrine Control of Social Behavior in Frogs 105

2.06.2.3 Seasonal Variation in Other Hormones: Adrenal It may be, of course, that corticosterone levels can reach so
Steroids high a level that reproductive behavior ceases. However, an
alternative view of corticosteroid action may be that it too
Adrenal steroids, specifically corticosteroids like corticosterone,
has a seasonal pattern that like gonadal steroids is, on average,
are most often identified as ‘stress’ hormones rather than
higher in the breeding season than out of it. Rather than func-
seasonally varying endocrine products influencing social
tioning as an antagonist to calling or other social behaviors, it
behavior. But in fact, corticosteroids are linked to social
facilitates reproductive social behavior by supporting the
behavior and are found to be elevated during breeding periods
demand for increased energy mobilization during this impor-
in many amphibians (Licht et al., 1983; Mendonça et al., 1985;
tant period of an animal’s life (Eikenaar et al., 2012). Leary
Orchinik et al., 1988; Harvey et al., 1997; Romero, 2002;
et al. (2008) and de Assis et al. (2012) have in fact demon-
Moore and Jessop, 2003; Eikenaar et al., 2012). Frog calling
strated in different frog species that corticosterone levels corre-
is an extremely energetically demanding behavior (Taigen
late with vocal effort as expected if adrenal steroids are
and Wells, 1985; Marler and Ryan, 1996; Wells, 2001). As
mobilized to support this energetically intensive behavior. Of
such, the energetic stress resulting from intense vocal displays
course, the seasonal difference in adrenal steroid levels may
would be expected to trigger elevated corticosterone levels. In
not be environmentally controlled as for gonadal steroids,
fact, calling males do have high levels of corticosterone (Leary
but may be secondary to the energetic effort attendant to calling
et al., 2006, 2008; de Assis et al., 2012). What is the implication
and other reproductive efforts.
of high corticosteroids in frogs?
The Energetics-Hormone-Vocalization model remains an
Experimental manipulations of corticosterone in many
important insight, as something must trigger switches between
different vertebrates have resulted in a decrease in reproduction
behavioral states. If not adrenal steroids, are there other signals
and the social behavior supporting it. These studies have also
of energetic or related physiological condition that could
found that experimental elevation of corticosteroids yields
mediate this switch? As yet, these are unknown. Understanding
a suppression of testosterone levels in males. This observation
the interaction of metabolic regulation and social behavior is
gave rise to the ‘Energetics-Hormone-Vocalization’ hypothesis
important, and given the strong energetic demands of at least
(Emerson, 2001; Emerson and Hess, 2001) that corticosterone
male social behavior in frogs, these vertebrates could be useful
levels mediate a switch between calling and noncalling in male
models for investigating that process.
frogs. The idea is that calling effort results in energetic stress,
The work on corticosteroids and behavior in frogs has
which is signaled by raising corticosterone levels. As corticoste-
largely focused on males and with them, on the interaction
rone levels rise, androgen levels fall. At some point, androgen
of these adrenal steroids with androgens. The energetic stress
levels fall below the threshold for calling, and males switch
profile of females is unknown, and interactions between corti-
from reproductive behavior to foraging behavior. Once energy
costeroids and gonadal steroids in females is unclear except for
stores are rebuilt, corticosterone levels fall, testosterone levels
one study in green treefrog females (Davis and Leary, 2015)
rise, and males switch back from foraging to reproduction.
that found no effect of experimentally elevated corticosterone
This reasonable model is based on numerous laboratory
on female estrogens, progesterone, testosterone, or dihydrotes-
studies in many vertebrates showing that treatment with exog-
tosterone. That study did find, however, that high levels of
enous corticosteroids or infliction of stress of some kind
corticosterone reduced the strength of their preference among
reduces testosterone levels. In some seasonally breeding
male calls (Figure 3), but did not seem to change their motiva-
amphibians, chronic or acute corticosterone administration
tion to mate in phonotaxis tests. Davis and Leary speculated
inhibits courtship (Moore and Miller, 1984; Marler and Ryan,
that the corticosterone-mediated decrease in choosiness may
1996; Burmeister et al., 2001). Furthermore, smaller males in
be a mechanism for reducing risky extended searches for males
poorer condition that adopt noncalling satellite behavior in
under stressful conditions. In fact, factors such as high preda-
frog choruses have elevated corticosterone levels, as well as
tion risk have been argued to increase the cost of mate sampling
lower testosterone levels, compared to robustly calling males
and alter search strategies (Crowley et al., 1991; Rand et al.,
(Leary and Harris, 2013). Beyond this, however, the
1991).
Energetics-Hormone-Vocalization hypothesis has received little
support from studies that measure natural corticosterone–
2.06.2.4 Seasonal Variation in Other Hormones: Melatonin
androgen relationships. During the time of reproductive social
behavior, corticosterone and testosterone are most often seen Melatonin is a pineal hormone released in circadian and
to be elevated together and either positively correlated (Licht seasonal patterns in many vertebrates. In amphibians (and
et al., 1983; de Assis et al., 2012) or unrelated (Leary et al., some birds), the retina produces melatonin as well in particu-
2006, 2008, 2015) in naturally behaving male frogs. Moreover, larly high levels and may contribute significantly to circulating
exogenous arginine vasotocin treatment in male green treefrogs levels of this hormone (Acuña-Castroviejo et al., 2014).
stimulates calling (Burmeister et al., 2001), likely through its Amphibian melatonin cycles are the same as in other vertebrates,
effect on central nervous system areas (see also Coddington, with high levels at night and higher levels in winter, when the
Chapter 2.07, The Vertebrate Brain Stem as the Primary Site dark phase is longer, than in summer (Delgado and
of Behavior Command: Insights from an Amphibian), but it Vivien-Roels, 1989; d’Istria et al., 1994). In ectotherms tempera-
also increases levels of circulating corticosterone, likely through ture is another potent regulator of seasonal melatonin patterns
AVT/AVP’s well-known peripheral effect as a stimulator of (Rawding and Hutchison, 1992; Lutterschmidt and Mason,
adrenal steroid release. The increasing corticosterone does not 2009). Melatonin has been found to regulate reproduction in
negatively affect the calling induced by AVT. many animals (Revel et al., 2009; Barrett and Bolborea, 2012),
106 Neuroendocrine Control of Social Behavior in Frogs

Probability of choosing call broadcast at high


1

call rate (2.5 call/s) 0.8

0.6
* *

0.4

0.2

0
Control Saline 4 µg CORT 8 µg CORT 16 µg CORT
(n=9) (n=7) (n=8) (n=11) (n=10)
Figure 4 Number of melatonin type 1 receptor immunoreactive cells
Figure 3 The probability of female green treefrogs (Hyla cinerea) in (MT1-IR) in the nucleus accumbens of male and female green treefrogs
phonotaxis tests moving toward a call at a high call rate (2.5 calls/s) (Hyla cinerea) in summer and winter. Overall numbers are significantly
versus a low call rate (1.2 calls/s) following no treatment, treatment higher in the summer, when a significant sex difference is also
with saline vehicle, or different doses of corticosterone (CORT). Dotted apparent. From Howard, C.M., Lutterschmidt, D.I., 2015. The effects of
line indicates equal choice for each stimulus. At the highest corticoste- melatonin on brain arginine vasotocin: relationship with sex and
rone levels, female choice approaches random and is significantly seasonal differences in melatonin receptor type 1 in green treefrogs
different from the other treatments. Asterisks indicate significant differ- (Hyla cinerea). J. Neuroendocrinol. 27 (8), 670–679.
ence from control (no treatment) condition. From Davis, A.G., Leary,
C.J., 2015. Elevated stress hormone diminishes the strength of female
preferences for acoustic signals in the green treefrog. Horm. Behav. 69 brain area (Howard and Lutterschmidt, 2015). The different
(0), 119–122. sex and season effects of melatonin appear to be due to differ-
ences in melatonin receptor profiles. Amphibians, like
mammals, have three types of brain melatonin receptors
including courtship behavior, gonadal physiology, and sex (Wiechmann and Wirsig-Wiechmann, 1993; Li et al., 2013),
steroid hormone concentrations in some ectothermic vertebrates one of which, MT1(or Mel1a), has a role in regulating seasonal
(de Vlaming et al., 1974; Krohmer and Lutterschmidt, 2011). reproduction in other vertebrates (Dubocovich, 1988; Pre-
Experimental manipulation of melatonin levels in frogs, so far ndergast, 2010; Luridiana et al., 2015). MT1 immunoreactive
only in males, has failed to find any effect of melatonin on circu- cells are more numerous in the nucleus accumbens in males
lating testosterone (Pierantoni et al., 1986; Lutterschmidt and than in females and are less numerous there and in all brain
Wilczynski, 2012). This suggests that the seasonal decline in regions in winter males than in summer males (Figure 4).
testosterone that occurs as frogs move out of the breeding season The sex and season differences in the effects of melatonin on
is not due to the increasing level or nightly duration of mela- the brain AVT system thus appears to be due to sex and season
tonin that occurs in the winter, nonbreeding season. differences in the levels of MT1 receptors (Howard and Lut-
No work has related melatonin with social behavior in terschmidt, 2015).
frogs. However, melatonin does influence the amount of the It is important to remember that females also have seasonal
neuromodulatory peptide arginine vasotocin (AVT) in brain changes in reproductive behavior. AVT in general does not
areas related to social behavior. AVT and its mammalian seem to have as important a role in regulating female frog
homolog arginine vasopressin have been implicated in many social behavior, although AVT treatment does inhibit female
types of male social behavior, particularly social signaling. release calls (Diakow and Nemiroff, 1981; Boyd, 1992) and
The important role of AVT in amphibian courtship behavior may increase female phonotaxis in some species (Schmidt,
will be discussed at length in the chapter by Coddington in 1984; Boyd, 1994). No other neural peptide has been found
this volume (Coddington, Chapter 2.07, The Vertebrate Brain to influence female frog social behavior.
Stem as the Primary Site of Behavior Command: Insights from
an Amphibian) and is reviewed in Emerson and Boyd (1999)
and in Wilczynski et al. (2005). Briefly, as in other vertebrates, 2.06.3 Steroid Hormone Receptors in the Brain
AVT cell number is sexually dimorphic and higher in males.
Treatments that elevate AVT stimulate advertisement calling Across vertebrates, receptors for gonadal steroids are wide-
in males and result in advertisement calls that females find spread across the brain. Androgen and estrogen receptors are
more attractive. Implantation of melatonin capsules in particularly noticeable within the forebrain areas connected
breeding-condition green treefrogs in the summer results in into brain networks for social decision making (Goodson,
a decrease in the number of AVT immunoreactivity in two 2005; Goodson and Thompson, 2010) and reward (Berridge
brain areas, the nucleus accumbens and the suprachiasmatic and Kringelbach, 2013). The pattern of these and other neuro-
nucleus (Lutterschmidt and Wilczynski, 2012). The effect chemical markers are conserved across vertebrates (O’Connell
was restricted to males; brain AVT seemed relatively insensitive and Hofmann, 2012). Androgen and estrogen receptors are
to melatonin changes in females. Furthermore, the same treat- seen here in frogs, as well as in motor and sensory areas,
ment in winter males in nonbreeding condition also results in and in the thalamus and midbrain, suggesting that as in other
no significant changes in AVT immunoreactive cells in any vertebrates, gonadal steroids have widespread modulatory
Neuroendocrine Control of Social Behavior in Frogs 107

effects on multiple aspects of social behavior. The distribution The torus is composed of several subdivisions, the laminar,
of sex steroid receptors indicates that gonadal hormones influ- principal, and magnocellular nuclei being the most prominent.
ence both the vocal and the auditory components of the frog Toral nuclei are rich in receptors for a variety of hormones
social system. The torus semicircularis, a large midbrain area and neuromodulators (Endepols et al., 2000; Wilczynski and
homologous to the inferior colliculus (Wilczynski and Ende- Endepols, 2007) including receptors for gonadal steroids
pols, 2007) where auditory sensory and motor systems (Figure 5). The laminar nucleus, which provides a significant
come together, is rich in androgen, estrogen, and progesterone portion of the torus’ ascending and descending outputs, has
receptors (Figure 5). the highest concentration of estrogen and androgen receptors
The anatomical organization of the frog auditory system is within the torus (Kelley et al., 1975; Morrell et al., 1975;
reviewed in detail in Wilczynski and Capranica (1984) and Kelley, 1980; Chakraborty and Burmeister, 2010). Androgen,
in Wilczynski and Endepols (2007). Like the mammalian ERalpha, and ERbeta receptors are found there, as well as in
inferior colliculus, the amphibian torus is the site where the principal nucleus (Chakraborty and Burmeister, 2010).
ascending auditory input from lower brainstem auditory areas Progesterone receptors are also expressed in the laminar
converges and where auditory information is then relayed to nucleus, as well as in the magnocellular nucleus (O’Connell
a variety of thalamic nuclei as well as sent via downstream et al., 2011). Although there is no sex difference in the locations
connections to the brainstem reticular formation and auditory of toral steroid receptors, the expression levels do differ
areas. Neuronal responses with complex spectral and temporal (Chakraborty and Burmeister, 2010). Females express more
properties emerge in the torus (reviewed in Rose and Gooler, ERbeta in the laminar nucleus and more ERalpha in the prin-
2007). These feature-detecting properties of toral neurons are cipal nucleus; males express more androgen receptor in the
linked to the detection of the species’ advertisement calls, principal nucleus.
accentuating a bias toward responding to call-related acoustic Brain areas caudal to the midbrain show little evidence
signals seen throughout the frog auditory system. Significant of sex steroid binding in frogs. Androgen receptors are
sex differences in auditory responses also appear in the present in the pretrigeminal nucleus (also called the DTAM),
midbrain (Hoke et al., 2008, 2010; Wilczynski and Ryan, a key part of the frog vocal control system, as well as on the
2010). The response of the torus to acoustic stimuli as a whole motor neurons in the XI-X complex innervating the larynx
closely mirrors behavioral responses to conspecific signals (Emerson and Boyd, 1999; see also Kelley, Chapter 2.08,
(Hoke et al., 2008; Mangiamele and Burmeister, 2011). Hormones and Vocal Systems: Insights from Xenopus).
With its strategic anatomical position and neurophysiological Neither androgen nor estrogen receptors have been
bias toward responding to call features, the torus acts as a key described in the amphibian brainstem auditory areas,
sensory motor interface, and more generally a control point although progesterone receptor protein, as identified by
regulating connections between auditory input, and motor, immunocytochemistry, seems to be widespread throughout
endocrine, and social responses (Wilczynski and Endepols, brainstem reticular areas. Forebrain areas, on the other hand,
2007; Wilczynski and Ryan, 2010). are rich in all gonadal steroid receptors (Kelley et al., 1975;

Figure 5 Locations of androgen (AR), estrogen (ER) (reported in Chakraborty and Burmeister, 2010 based on in situ hybridization), and proges-
terone receptors (PR) (reported in O’Connell et al., 2011 based on immunocytochemistry) in the torus semicircularis (the auditory midbrain) and
surrounding areas in túngara frogs (Physalaemus pustulosus). Abbreviations: TE, optic tectum; Teg, tegmental areas; Tl, laminar nucleus of the torus
semicircularis; Tm, magnocellular nucleus of the torus semicircularis; Tp, principal nucleus of the torus semicircularis.
108 Neuroendocrine Control of Social Behavior in Frogs

(a) MP
(AR,ER)

(ER) Auditory
St TS brainstem
A
C (AR,ER)
(AR, P
ER) VM
Sep SI

(AR,
POA/Hy
ER)

Endocrine
control

(b)
MP

St Auditory
(AR, A (ER) TS brainstem
ER) C (AR,ER)
P
(AR)
Sep VM SI

POA/Hy

Motor
systems

Figure 6 Diagram of the audioendocrine (a) and audiomotor (b) systems of the frog brain defined Wilczynski and Endepols (2007) based on
ascending connections from the torus semicircularis. Presence of moderate to high levels of androgen (AR) and estrogen (ER) receptor expression
within the systems are indicated. Abbreviations: A, anterior thalamic nucleus; C, central thalamic nucleus; Hy, hypothalamus; MP, medial pallium;
P, posterior thalamic nucleus; POA, preoptic area; Sep, septal complex; Si, secondary isthmal nucleus; St, striatopallidal complex; TS, torus semi-
circularis; VM, ventromedial thalamic nucleus.

Morrell et al., 1975; Chakraborty and Burmeister, 2010; particular association with vocal or auditory centers is
O’Connell et al., 2011). This includes the targets of ascending apparent, including no strong presence in the toral nuclei
auditory connections, and the motor and basal forebrain with high gonadal steroid receptor expression. The nature
areas implicated in the control of vocalization (Emerson and distribution of glucocorticoid receptors and other
and Boyd, 1999). Wilczynski and Endepols (2007) components of the brain’s stress signaling system in
conceptualized ascending circuits emerging from the torus as amphibians deserves further attention, especially given
comprising audiomotor, audioendocrine, and audiolimbic new work showing significant effects of adrenal steroids
components based on their forebrain targets (Figure 6). Each on acoustic communication in frogs (Leary et al., 2006;
includes several nuclei with estrogen, androgen, and Davis and Leary, 2015) and the substantial previous work
progesterone receptors, including thalamic nuclei, areas of on their strong influence on olfactory-driven courtship
the basal forebrain implicated in male and female social behavior in salamanders (reviewed in Coddington, Chapter
behavior such as the preoptic area and ventral hypothalamus, 2.07, The Vertebrate Brain Stem as the Primary Site of
and limbic centers such as the medial septum and the medial Behavior Command: Insights from an Amphibian).
pallium (the amphibian homolog of the mammalian
hippocampal complex) (Chakraborty and Burmeister, 2010;
O’Connell et al., 2011). 2.06.4 Hormone Effects on Hearing
Glucocorticoids also influence social behavior in frogs,
and presumably work at least in part through neural systems The auditory system detects and analyzes the acoustic social
to do so. They interact with brain arginine vasotocin systems cues guiding frog social interactions and transmits this infor-
to modulate courtship behavior in newts, which is described mation to motor, endocrine, and limbic areas responsible for
in detail by Coddington (Coddington, Chapter 2.07, The orchestrating the behavioral and physiological components
Vertebrate Brain Stem as the Primary Site of Behavior of frog social behavior (Figure 6). The sensory systems respon-
Command: Insights from an Amphibian), although whether sible for inputting information about social signals are often
they do the same in frogs is unknown. Yao et al. (2008) thought of as the stable component of social neural networks.
found glucocorticoid receptor protein widely distributed In fact, growing evidence shows that sensory processing is
throughout the brain in Xenopus with particularly high levels remarkably plastic. Several lines of evidence support the idea
in forebrain limbic and hypothalamic areas. Receptors were that hormone-dependent changes in hearing are present in
also found throughout the midbrain and hindbrain. No amphibians and that this results in changes in the detection
Neuroendocrine Control of Social Behavior in Frogs 109

and response to the male advertisement call. This may be 2.06.4.2 Experimental Manipulation of Circulating Gonadal
a major component of the hormonal regulation of anuran Steroids Modifies the Auditory System
reproductive social behavior.
Several studies have directly manipulated androgens and estro-
gens to show the effect these hormones have on auditory pro-
2.06.4.1 Seasonal Changes in Hearing Track Seasonal
cessing. Nearly all have focused on the midbrain, where
Changes in Sex Steroids and Behavior
estradiol injections were reported to increase the amplitude
Social behavior waxes and wanes seasonally in frogs along of toral-evoked potentials to tones (Yovanof and Feng,
with their levels of circulating sex steroid hormones. In 1983). Testosterone’s effects have been more inconsistent.
nonbreeding seasons, frogs are less responsive to their species’ Penna et al. (1992) found no testosterone effects on midbrain
vocal communication signals. Changes in the auditory system thresholds, which may not be surprising given that most
may contribute to this. By several measures, midbrain studies of seasonal variation in midbrain auditory responses
responses to auditory stimulation are more robust in the find little or no change in thresholds. Testosterone appears to
summer breeding season than when frogs are in a nonbreeding induce more complicated sex- and stimulus-specific changes
condition. Goense and Feng (2005) found that more toral than simple threshold changes in toral auditory responses.
auditory cells could be isolated in Lithobates (¼Rana) pipiens Testosterone treatment yielded no statistically significant
when recording in the summer, latencies were shorter, and changes in midbrain thresholds in male green treefrogs
phase locking to amplitude modulated signals stronger. Earlier (Miranda and Wilczynski, 2009b). In females, however, testos-
work found greater suprathreshold multiunit responses in the terone increased thresholds to tones with frequencies within
torus in fire-bellied toads (Bombina bombina; Walkowiak, the spectral bands of the male advertisement call, but not to
1980) and gray treefrogs (Hyla chrysoscelis; Hillary, 1984) those outside those bands, and also increased thresholds to
during the breeding season. Seasonal threshold changes at the advertisement call itself. These results bear an interesting
the single neuron level may (Hillary, 1984) or may not relationship to a natural sex difference in green treefrog sensi-
(Penna et al., 1992; Goense and Feng, 2005) be present. tivity to the male call. In the hormone state appropriate for
However, the overall picture of the anuran midbrain is that breeding condition animals, female toral multiunit responses
stronger auditory responses are present in the summer, to the male call have lower thresholds than do the responses
when hormone levels are high, than in the winter, when levels of males to the same stimulus (Miranda and Wilczynski,
are low. 2009b). After testosterone treatment, female thresholds
In females, estrogen levels drop precipitously immedi- increase to match those of males, eliminating the sex difference
ately after eggs are laid, and along with this, females become in sensitivity.
unreceptive to male advertisement calls (Lynch et al., 2006). Quantification of immediate early gene (IEG) mRNA
Toral auditory responses change cyclically in a way that expression also shows the stimulatory effect of gonadal
matches the change in receptivity and which is also consis- hormones on toral auditory activity. Treating túngara frog
tent with the seasonal changes. In postmated female green females with gonadotropin (Lynch and Wilczynski, 2008),
treefrogs, toral response strength to band-limited noise which elevates estrogen levels, or with estrogen itself (Chakra-
stimuli is significantly less than in premated females borty and Burmeister, 2015) results in an increased midbrain
(Miranda and Wilczynski, 2009a). The effect is restricted to response to conspecific advertisement calls (Figure 7). Both
lower frequencies in the frog’s hearing range, which are ulti- endocrine treatments cause an increase in IEG expression
mately derived from the ear’s amphibian papilla, one of two even without accompanying auditory stimulation. There is
main auditory end organs in the amphibian ear (for reviews thus an additive effect of hormones and stimulus presentation,
of the amphibian ear, see Zakon and Wilczynski, 1988; resulting in a higher level of midbrain activity to calls when
Simmons et al., 2007). Responses to the higher frequencies estrogens are high (in females; a matching study of males
derived from stimulation of the frog’s second auditory end and testosterone has not been done). It is not clear what might
organ, the basilar papilla, were stable throughout the repro- be the electrophysiological correlate of the estrogen-induced
ductive cycle. Similarly, Goense and Feng (2005) found that elevation of IEG expression in the absence of an auditory stim-
seasonal changes were apparent in lower frequencies, ulus. Hormone-dependent changes in spontaneous activity, as
whereas responses to the higher basilar papilla frequencies measured by single- or multiunit spike counts, have not been
were unchanged across seasons. consistently reported. It may be, however, that gonadal
The midbrain may not be the first auditory processing stage hormones induce subthreshold depolarizations or represent
at which seasonal and reproductive-cycle changes in hearing other, nonelectrophysiological, changes in midbrain cells that
occur. Zhang et al. (2012) found that peripheral auditory adds to the response strength of midbrain auditory neurons
thresholds were lower during the breeding season than the to calls under the endocrine conditions found in reproductively
nonbreeding season in the Emei Music frog (Babina daunchina). active frogs.
This is the only frog in which a seasonal change in the periph- Chakraborty and Burmeister (2015) is the only study that
eral auditory system has been documented. However, seasonal extended the analysis of hormone effects on hearing beyond
or hormone-dependent changes to the ear’s sensitivity have the midbrain. They found that the facilitating, additive effects
been described in several vertebrates (Sisneros and Bass, of estradiol on the responses to conspecific calls extended to
2003; Sisneros et al., 2004; Caras, 2013; Gall et al., 2013). many areas of the limbic forebrain that are sensitive to auditory
Further studies on the effects of steroid hormones on the ear stimulation. In most cases, as in the torus, both conspecific calls
itself are clearly warranted. and estradiol alone resulted in an elevation of IEG expression,
110 Neuroendocrine Control of Social Behavior in Frogs

(a) (b)

(c) (d)

Figure 7 Photomicrographs of immediate early gene egr-1 mRNA expression in the laminar nucleus of the torus semicircularis in female túngara
frogs (Physalaemus pustulosus). (a) Egr-1 expression in saline-treated frogs with no acoustic stimuli; (b) Egr-1 expression in saline-treated frogs
hearing chorus sounds; (c) Egr-1 expression in saline-treated frogs treated with human chorionic gonadotropin (HCG) with no acoustic stimuli; (d)
Egr-1 expression in HCG-treated frogs hearing chorus sounds. From Lynch, K.S., Wilczynski, W., 2008. Reproductive hormones modify reception of
species-typical communication signals in a female anuran. Brain Behav. Evol. 71 (2), 143–150.

and together the effects were additive. The one exception was triggers an endocrine response of some kind. In anurans,
the preoptic area. There, a statistical call by hormone interac- acoustic signals in the form of male advertisement calls are
tion was apparent, suggesting that preoptic auditory responses the most common and most salient sensory cues associated
are dependent on estradiol level, not just additive to it. with social behavior. Hormonal condition influences how
Gonadal steroids (or at least estrogens in females, which is strongly the nervous system responds to those cues. Hearing
the only condition studied to date) appear to influence the those cues, in turn, influences the endocrine state of both males
strength of auditory responses beyond the midbrain, to the and females.
forebrain areas to which it feeds information.
2.06.5.1 Anatomical Links between the Auditory Midbrain
2.06.4.3 Implications for Social Behavior and Hypothalamic Areas Define an Audioendocrine Circuit
Social behavior in frogs, or in any animal, consists of detecting Neuroanatomical pathways emerging from the torus semicircu-
social signals and responding with appropriate behaviors. laris provide the substrate by which auditory stimulation by
Between signal detection and response integrative processes calls can modulate the endocrine system. Ascending toral effer-
link sensory, motor, and physiological systems and modify ents, mainly from the laminar nucleus, reach multiple thalamic
a variety of motivational, affective, and cognitive responses. nuclei (Wilczynski et al., 1993; Wilczynski and Endepols,
Hormones can and do influence the process at any stage. The 2007) that together relay auditory information to the basal
input, or sensory, side of the system is modified on a seasonal ganglia, limbic pallial and subpallial areas, and directly to
and cyclic basis by gonadal hormones. The auditory system hypothalamic and preoptic areas (Figure 6). Wilczynski and
changes make the advertisement call more detectable, more Endepols (2007) termed the toral-thalamic-hypothalamic
stimulatory, and thus more salient when gonadal steroids are pathway the audioendocrine system. Three distinct circuits
elevated. These changes should enhance the probability of comprise this system. Auditory inputs to the ventral hypothal-
both males and females to respond to the call in their own amus arise from the central and anterior thalamic nuclei (Neary
sex-specific way. and Wilczynski, 1986; Hall and Feng, 1987; Neary, 1988;
Allison and Wilczynski, 1991). Connections to the anterior
preoptic area arise mainly from the anterior thalamic nucleus,
2.06.5 Hearing Effects on Hormones supplemented by a small projection from the central thalamic
nucleus. Auditory input to both the ventral hypothalamus and
A traditional approach in behavioral endocrinology is to inves- anterior preoptic area also comes from the secondary isthmal
tigate how hormones modulate behavior. But behavior also nucleus (Neary and Wilczynski, 1986; Neary, 1988; Allison
modulates hormonal state. Engaging in any social behavior and Wilczynski, 1991), an area of the isthmal tegmentum
Neuroendocrine Control of Social Behavior in Frogs 111

that receives a heavy toral input (Neary, 1988). The secondary (Hoke et al., 2007), and in both preoptic and hypothalamic
isthmal area is similar in location to the mammalian parabra- areas gonadal hormones add to or interact with the acoustically
chial region, which is recognized as relaying sensory informa- driven IEG expression (Chakraborty and Burmeister, 2015).
tion to the hypothalamus. It is studied in mammals mainly The preoptic area and ventral hypothalamus region respon-
in regards to the transmission of behaviorally relevant chemo- sive to acoustic call stimuli are part of the neuroendocrine
sensory or pain signals. Connections from thalamic and isth- system controlling gonadotropin release. The extent of the
mal sensory areas are not unique to frogs, but the anatomical connections and the widespread acoustic sensitivity
connections relaying auditory information are exceptionally across hypothalamic nuclei suggest that acoustic social signals,
robust in these vertebrates. and perhaps other auditory stimuli, may have a broader impact
The acoustic sensitivity of preoptic and hypothalamic on endocrine and physiological regulation. This, however, has
areas have been confirmed by electrophysiology (Urano and not been investigated.
Gorbman, 1981; Wilczynski and Allison, 1989; Allison, 1992)
and by studies using immediate early gene expression as a mar-
2.06.5.2 Hearing Calls Elevates Levels of Circulating Steroid
ker of neural activity (Hoke et al., 2005, 2007; Chakraborty and
Hormones
Burmeister, 2015). Response properties of preoptic and hypo-
thalamic neurons are characterized by long latencies and firing Observations across multiple vertebrate species have docu-
rates that declines gradually after the termination of the stimulus mented that engaging in social interactions stimulates the
(Figure 8). In both areas, most acoustically sensitive neurons are reproductive system. In frogs, male Rana esculenta maintained
insensitive to tones or noise bursts, but fire strongly when stim- mature testes longer if they were exposed to conspecific calls
ulated with a conspecific call. IEG studies show additional compared to control males exposed to nonsocial sound stimuli
complexity in the responses in these and other hypothalamic or no sound (Brzoska and Obert, 1980), and in female midwife
and limbic centers. Responses in hypothalamic areas are often toads, Alytes muletensis, hearing conspecific calls facilitated
a combination of auditory- and movement-related activation oocyte retention (Lea et al., 2000). There is substantial direct
evidence that hearing conspecific advertisement calls results
in an elevation of gonadal steroids in both males and females.
Hearing conspecific calls assembled into a simulated chorus
over 10 nights increases circulating androgen levels in male
green treefrogs (Burmeister and Wilczynski, 2000) (Figure 9).
Hearing random tones matched in time and amplitude have
no such effect, and in fact result is no difference from remaining
in silence for the duration of the experiment. The same occurs
in southern leopard frogs, Rana sphenocephala (Chu and
Wilczynski, 2001). Interestingly, the effect appears to be a direct
result of hearing, rather than calling or engaging in any other
behavior. Both testosterone and dihydrotestosterone increase
in treefrogs after hearing calls regardless of whether or not the
listener calls back, and males hearing tones have no elevation
in androgens whether or not they themselves call. Gonadal
steroids are also elevated in female túngara frogs exposed to
conspecific male advertisement calls with the same 10-day
exposure (Lynch and Wilczynski, 2006) (Figure 9). In those
females, estrogens, but not testosterone, increased significantly.
In male treefrogs, the elevation in testosterone coincides with
an increase in the number of GnRH-immunoreactive cells in
the preoptic area (Burmeister and Wilczynski, 2005). The
strong connections from auditory nuclei to the preoptic area
and other hypothalamic areas would provide the substrate by
which the constant stimulation by conspecific social signals
can stimulate production of GnRH and ultimately the
increased release of androgens and estrogens downstream of
GnRH release. A similar coordinated elevation of both GnRH
and estrogens, mediated by social interactions, occurs in
Figure 8 Poststimulus–time histograms showing single unit newts (Propper and Moore, 1991).
responses of a preoptic area (a) and a ventral hypothalamic (b) neuron
in the green treefrog (Hyla cinerea). Repetitive white noise (wn) and
mating call (mc) presentations alternate with no stimulus period (ns). 2.06.5.3 Short-Term Exposure to Aggressive or
Responses to the conspecific call are marked by long latency, persis- Advertisement Calls Has Different Effects
tent responses; spontaneous activity can remain elevated after the call
stimulus ends. From Allison, J., 1992. Acoustic modulation of neural Social stimulation of reproductive physiology and endocrine
activity in the preoptic area and ventral hypothalamus of the green tree- state occurs under conditions that mimic the state of frogs in
frog (Hyla cinerea). J. Comp. Physiol. A 171 (3), 387–395. a breeding chorus, where males and females hear a changing
112 Neuroendocrine Control of Social Behavior in Frogs

Figure 9 Increases in plasma testosterone (a) in green treefrog (Hyla cinerea) males and estradiol (b) in túngara frog (Physalaemus pustulosus)
females after 10 consecutive nights of hearing chorus sounds. Similar exposure to tones (and silence in males) has no effect. Male data from
Burmeister, S., Wilczynski, W., 2000. Social signals influence hormones independently of calling behavior in the treefrog (Hyla cinerea). Horm. Behav.
38 (4), 201–209; female data from Lynch, K.S., Wilczynski, W., 2006. Social regulation of plasma estradiol concentration in a female anuran. Horm.
Behav. 50 (1), 101–106.

pattern of conspecific advertisement calls for many hours each testosterone levels than winners. It is difficult to know whether
night for many nights. But within a chorus, behaving individuals losing triggered the fall in testosterone or whether the low
can engage in rapid, short-range interactions as well. Male frogs testosterone was a preexisting condition causing the losing
in many species, for example, occupy and defend call sites from
which they respond with matching advertisement calls to males (a) 30
calling close-by and will escalate their vocalization and occasion-
a
ally engage in physical fights when calling intruders encroach on 25
Corticosterone (ng ml–1)

their site. Fewer researchers have explored the hormonal conse-


quences of these rapid, short-range interactions, but those that 20
have find a decrease, rather than an increase, in androgens.
Experimental simulation of a territorial intrusion in the neotrop- 15
ical treefrog, Hypsiboas faber, by presenting the male with taped
advertisement calls for 10 min caused a rapid decrease in testos- 10 a
terone in the threatened male (de Assis et al., 2012). The same
effect occurred in male green treefrogs after exposure to conspe- 5
cific advertisement calls in a similar simulated vocal intruder
paradigm (Leary, 2014). There are clearly complicated and 0
Pre-broadcast Post-broadcast
context-dependent patterns of behaviorally induced hormone (b)
changes. 22.5
a b
Behavioral triggers of adrenal steroids were unknown until 20
recently, until Leary (2014) explored the effects of aggressive
17.5 b
Testosterone (ng ml–1)

calls in male green treefrogs. Green treefrogs produce a separate


15 a
aggressive call when engaged in escalating, short-range interac-
tions with other calling males. Hearing aggressive calls triggered 12.5
an increase in corticosteroid levels within 45 min of the stim- 10
ulus (Figure 10). The corticosterone increase was accompanied
7.5
by a rapid reduction in androgen levels. Hearing advertisement
calls, on the other hand, did not stimulate adrenal steroid 5
changes in this species or in male H. faber (de Assis et al., 2.5
2012). It is not known if changes of this type occur in females.
0
The consequence of natural aggressive interactions between Pre-broadcast Post-broadcast
males may also lead to a rapid shift in steroid hormone levels.
In many vertebrates, there is a rapid elevation of testosterone Figure 10 Plasma corticosterone (a) and testosterone (b) levels
when a male wins an aggressive interaction; losers of these chal- before and after a 45 min presentation of nearby aggressive calls
(black), advertisement calls (gray), or silent control (white) in male
lenges often see a slower decrease in testosterone. Calling male
green treefrogs (Hyla cinerea). Testosterone decreases after hearing
frogs will act aggressively toward nearby males intruding on
either the advertisement or aggressive call, whereas corticosterone
their calling site; losers of these fights stop calling and adopt levels increase after hearing the aggressive calls; a and b above bars
a silent satellite strategy (reviewed in Gerhardt and Huber, indicates significant differences in pre- and post-treatment hormone
2002; Wells, 2007). Leary and Harris (2013) reported that levels within treatment groups. From Leary, C.J., 2014. Close-range
males that lost short-range aggressive vocal interactions with vocal signals elicit a stress response in male green treefrogs: resolution
other males (that is, fell silent or moved away) had lower of an androgen-based conflict. Anim. Behav. 96, 39–48.
Neuroendocrine Control of Social Behavior in Frogs 113

condition; but in either case, the relationship between steroid levels may play some part in these shifts (Leary et al.,
hormone level and social status is very interesting. 2006; Leary and Harris, 2013).

2.06.5.4 Implications for Social Behavior


2.06.6 Summary and Conclusions
Frog social behavior is dependent on adequate levels of
gonadal steroid hormones. Frog social behavior also requires The effect of hormones on frog social behavior aligns with
males and females to listen to a variety of conspecific advertise- effects seen in seasonal vertebrates generally. Amphibian social
ment calls in varying patterns over a long period of time. This behavior offers a way to examine this in the context of verte-
experience elevates sex steroid hormones, resulting in a positive brates heavily dependent on acoustic communication, and
feedback system that can help maintain high hormone levels, with a strong sex difference in the way males and females
behavior, and reproductive capacity throughout a breeding produce and respond to those signals. With that in mind,
season and slow the inevitable decline in endocrine state that work on the behavioral neuroendocrinology of anuran social
leads to the termination of the breeding season (O’Bryant behavior can be considered in light of the three aspects of their
and Wilczynski, 2010). social behavior noted in Section 2.06.1.
For males, the interaction between androgens and calling Reproduction is seasonal, and under that pattern, gonadal
helps them coordinate their social breeding behavior. Frog steroids vary seasonally and with them so does reproductive
choruses are structured as lek-like communal displays in which social behavior in both males and females. Both correlative
males advertise and compete for females (for a review of leks, data from naturally behaving frogs and experimental work
see Höglund and Alatalo, 1995). As argued by Wilczynski and manipulating hormones provide compelling evidence that
Burmeister (2016), reproductive success in a lek depends on androgens in males and estrogens in females drive the expres-
a combination of cooperation and competition. A successful sion of male calling and female responses to the call, respec-
lek depends on multiple males displaying at the same time tively. More recent work being done on adrenal steroids lays
and place, as females are attracted to sites with large numbers the foundation for interesting research to untangle the role
of displaying males. Within the lek, however, males are only these hormones play in naturally occurring social behavior.
successful if they outcompete other males, that is, display This includes the question of whether baseline corticosterone
more vigorously or more often, than neighboring males. A posi- levels change seasonally in response to external seasonal cues
tive feedback system between hormones and behavior maintains in the same way gonadal steroids are regulated or whether the
calling behavior in the presence of other calling males. It also elevations seen when animals are in their social reproductive
stimulates, or at least helps maintain, reproductive physiology state are a reaction to the specific social, energetic, or other
when competition, in the form of other calling males, is intense. conditions encountered by the individual at that time. The
For females, the implications are more complicated. way in which melatonin influences social behavior is also ripe
Behavioral-endocrine interactions help coordinate the level for further studies, as are the potential effects of other mono-
of social behavior with reproductive physiology and egg amine and peptide hormones. Of particular interest is the ques-
maturation and help increase the drive to receptivity when tion of what endocrine or neuromodulatory agents regulate
vigorously calling males are present. On the other hand, shifts in behavioral state from reproduction to foraging or other
female reproductive stimulation by male calls can be viewed behavioral modes, an important topic in the field of environ-
as a way for males to manipulate female physiology, and mental physiology (Lutterschmidt and Maine, 2014), but
with it their behavior, to make them more receptive and which is unexplored in amphibians except for work testing
driven to accept a male. As reviewed in Section 2.06.2.1, as the Energetics-Hormone-Vocalization hypothesis.
female estrogens rise, receptivity increases, but choosiness Acoustic communication is the foundation for anuran social
decreases, meaning that the strength of female preference behavior, and we can see from the work on frogs that endocrine
declines (Lynch et al., 2006; Chakraborty and Burmeister, modulation influences both the sensory and the motor compo-
2009). A male under intense male–male competition may nents of this communication system to change the way frogs
benefit from driving a female toward amplexus both behav- hear and respond to conspecific vocal signals when they are
iorally and physiologically. engaged in seasonal reproductive social behavior. The social
The short-term endocrine effects of hearing advertisement behavior and communication system of frogs offers an opportu-
and aggressive calls are quite different from the effects of nity to explore this phenomenon further, and in particular the
chronic exposure to a chorus. The elevation of corticosterone mechanisms behind the changes in both components of a social
and the decrease in androgens may be part of a behavioral communication, and how the input and output changes interact
system in which male agonistic interactions lead to behav- through the integrative networks of the brain. The work on audi-
ioral state differences within the chorus, with males adopting tory changes highlights a growing appreciation that social behav-
different strategies. Frogs with extended breeding seasons ior’s input side is as plastic as its output side and that steroid
and large choruses often have males with different behav- hormones have a particularly strong influence on auditory func-
ioral strategies (Wells, 2007). Some call persistently from tions from the ear to the forebrain (Bass et al., 2016).
defended sites, some adopt satellite behavior, in which Acoustic communication is a sexually differentiated behavior in
they stay near calling males, remain silent, and attempt to frogs, from the large sex difference in behavior to more subtle
intercept females moving toward the calling male, and differences in the response properties of the ear and higher
some move away from callers to find another call site from areas of the auditory system. With this comes the opportunity
which to broadcast their vocal signals. Behaviorally adjusted to examine the morphological, physiological, neural, and
114 Neuroendocrine Control of Social Behavior in Frogs

behavioral sex differences in specific components of a social Davis, A.G., Leary, C.J., 2015. Elevated stress hormone diminishes the strength of
system in a truly integrative way. Work on the vocal production female preferences for acoustic signals in the green treefrog. Horm. Behav. 69
(0), 119–122.
component of frog social behavior is an exceptional example of
Delgado, M.J., Vivien-Roels, B., 1989. Effect of environmental temperature and
this (Kelley, Chapter 2.08, Hormones and Vocal Systems: photoperiod on the melatonin levels in the pineal, lateral eye, and plasma of the
Insights from Xenopus) and a model for pursuing research on frog, Rana perezi: importance of ocular melatonin. Gen. Comp. Endocrinol. 75
the sensory side of this social system. (1), 46–53.
Work on the behavioral endocrinology and neuroethology of Diakow, C., Nemiroff, A., 1981. Vasotocin, prostaglandin, and female reproductive
behavior in the frog, Rana pipiens. Horm. Behav. 15 (1), 86–93.
amphibian behavior has made significant contributions to Dubocovich, M.L., 1988. Pharmacology and function of melatonin receptors. FASEB
understanding general principles of vertebrate social behavior J. 2 (12), 2765–2773.
and acoustic communication. Amphibians continue to be valu- Duellman, W.E., Trueb, L., 1994. Biology of Amphibians. Johns Hopkins Press,
able model organisms for understanding how the sensory, Baltimore, MD.
Eikenaar, C., Husak, J., Escallón, C., Moore, I.T., 2012. Variation in testosterone and
integrative, and motor components of social behavior interact
corticosterone in amphibians and reptiles: relationships with latitude, elevation,
and are coordinated by a variety of hormones. and breeding season length. Am. Nat. 180 (5), 642–654.
Emerson, S.B., 2001. Male advertisement calls: behavioral variation and physiological
processes. In: Ryan, M.J. (Ed.), Anuran Communication. Smithsonian Institution
References Press, Washington, DC, pp. 36–44.
Emerson, S.B., Boyd, S.K., 1999. Mating vocalizations of female frogs: control and
Acuña-Castroviejo, D., Escames, G., Venegas, C., Díaz-Casado, M., Lima-Cabello, E., evolutionary mechanisms. Brain Behav. Evol. 53 (4), 187–197.
López, L., Rosales-Corral, S., Tan, D.-X., Reiter, R., 2014. Extrapineal melatonin: Emerson, S.B., Hess, D.L., 2001. Glucocorticoids, androgens, testis mass, and the
sources, regulation, and potential functions. Cell. Mol. Life Sci. 71 (16), 2997–3025. energetics of vocalization in breeding male frogs. Horm. Behav. 39 (1), 59–69.
Allison, J., 1992. Acoustic modulation of neural activity in the preoptic area and ventral Endepols, H., Walkowiak, W., Luksch, H., 2000. Chemoarchitecture of the anuran
hypothalamus of the green treefrog (Hyla cinerea). J. Comp. Physiol. A 171 (3), auditory midbrain. Brain Res. Rev. 33 (2–3), 179–198.
387–395. Feder, M.E., Bruggren, W.W. (Eds.), 1992. Environmental Physiology of the
Allison, J.D., Wilczynski, W., 1991. Thalamic and midbrain auditory projections to the Amphibians. University of Chicago Press, Chicago.
preoptic area and ventral hypothalamus in the green treefrog (Hyla cinerea). Brain Gall, M.D., Salameh, T.S., Lucas, J.R., 2013. Songbird frequency selectivity and
Behav. Evol. 38 (6), 322–331. temporal resolution vary with sex and season. Proc. R. Soc. Lond. B Biol. Sci. 280
de Assis, V.R., Navas, C.A., Mendonça, M.T., Gomes, F.R., 2012. Vocal and territorial (1751), 20122296.
behavior in the Smith frog (Hypsiboas faber ): relationships with plasma levels of Gerhardt, H.C., Huber, F., 2002. Acoustic Communication in Insects and Anurans.
corticosterone and testosterone. Comp. Biochem. Physiol. Part A Mol. Integr. University of Chicago Press, Chicago.
Physiol. 163 (3–4), 265–271. Gobbetti, A., Zerani, M., Bolelli, G.F., Botte, V., 1991. Seasonal changes in plasma
Barrett, P., Bolborea, M., 2012. Molecular pathways involved in seasonal body weight prostaglandin F2a and sex hormones in the male water frog, Rana esculenta. Gen.
and reproductive responses governed by melatonin. J. Pineal Res. 52 (4), 376–388. Comp. Endocrinol. 82 (3), 331–336.
Bass, A.H., Sisneros, J.A., Fay, R.R., Popper, A.N. (Eds.), 2016. Hearing and Goense, J.B.M., Feng, A.S., 2005. Seasonal changes in frequency tuning and
Hormones: Springer Handbook of Auditory Research. Springer, New York. temporal processing in single neurons in the frog auditory midbrain. J. Neurobiol.
Bernal, X.E., Stanley Rand, A., Ryan, M.J., 2007. Sex differences in response to 65, 22–36.
nonconspecific advertisement calls: receiver permissiveness in male and female Goodson, J.L., 2005. The vertebrate social behavior network: evolutionary themes and
túngara frogs. Anim. Behav. 73 (6), 955–964. variations. Horm. Behav. 48 (1), 11–22.
Berridge, K.C., Kringelbach, M.L., 2013. Neuroscience of affect: brain mechanisms of Goodson, J.L., Thompson, R.R., 2010. Nonapeptide mechanisms of social cognition,
pleasure and displeasure. Curr. Opin. Neurobiol. 23 (3), 294–303. behavior and species-specific social systems. Curr. Opin. Neurobiol. 20 (6),
Bosch, J., Boyero, L., 2004. Reproductive stage and phonotactic preferences of 784–794.
female midwife toads (Alytes cisternasii ). Behav. Ecol. Sociobiol. 55 (3), 251–256. Gordon, N.M., Gerhardt, H.C., 2009. Hormonal modulation of phonotaxis and
Boyd, S.K., 1992. Sexual differences in hormonal control of release calls in bullfrogs. advertisement-call preferences in the gray treefrog (Hyla versicolor ). Horm. Behav.
Horm. Behav. 26 (4), 522–535. 55 (1), 121–127.
Boyd, S.K., 1994. Arginine vasotocin facilitation of advertisement calling and call Gordon, N.M., Hellman, M., 2015. Dispersal distance, gonadal steroid levels and body
phonotaxis in bullfrogs. Horm. Behav. 28 (3), 232–240. condition in gray treefrogs (Hyla versicolor): seasonal and breeding night variation
Brzoska, J., Obert, H.-J., 1980. Acoustic signals influencing the hormone production in females. J. Herpetol. 49, 655–661.
of the testes in the grass frog. J. Comp. Physiol. A 140 (1), 25–29. Gramapurohit, N.P., Radder, R.S., 2013. Changes in plasma sex steroids in relation to
Burmeister, S., Somes, C., Wilczynski, W., 2001. Behavioral and hormonal effects of reproductive behavior of the toad, Bufo melanostictus (Schn.). Copeia 2013 (2),
exogenous vasotocin and corticosterone in the green treefrog. Gen. Comp. 332–337.
Endocrinol. 122 (2), 189–197. Hall, J.C., Feng, A.S., 1987. Evidence for parallel processing in the frog’s auditory
Burmeister, S., Wilczynski, W., 2000. Social signals influence hormones independently of thalamus. J. Comp. Neurol. 258, 407–419.
calling behavior in the treefrog (Hyla cinerea). Horm. Behav. 38 (4), 201–209. Harvey, L.A., Propper, C.R., Woodley, S.K., Moore, M.C., 1997. Reproductive endo-
Burmeister, S.S., Wilczynski, W., 2001. Social context influences androgenic effects crinology of the explosively breeding desert spadefoot toad, Scaphiopus couchii.
on calling in the green treefrog (Hyla cinerea). Horm. Behav. 40 (4), 550–558. Gen. Comp. Endocrinol. 105 (1), 102–113.
Burmeister, S.S., Wilczynski, W., 2005. Social signals regulate gonadotropin-releasing Herman, C.A., 1992. Endocrinology. In: Feder, M.E., Bruggren, W.W. (Eds.), Envi-
hormone neurons in the green treefrog. Brain Behav. Evol. 65 (1), 26–32. ronmental Physiology of the Amphibians. University of Chicago Press, Chicago,
Caras, M.L., 2013. Estrogenic modulation of auditory processing: a vertebrate pp. 40–54.
comparison. Front. Neuroendocrinol. 34 (4), 285–299. Hillary, C.M., 1984. Seasonality of two midbrain auditory responses in the treefrog,
Chakraborty, M., Burmeister, S.S., 2009. Estradiol induces sexual behavior in female Hyla chrysoscelis. Copeia 4, 844–852.
túngara frogs. Horm. Behav. 55 (1), 106–112. Höglund, J., Alatalo, R.V., 1995. Leks. Princeton University Press, Princeton NJ.
Chakraborty, M., Burmeister, S.S., 2010. Sexually dimorphic androgen and estrogen Hoke, K.L., Ryan, M.J., Wilczynski, W., 2005. Social cues shift functional
receptor mRNA expression in the brain of túngara frogs. Horm. Behav. 58 (4), connectivity in the hypothalamus. Proc. Natl. Acad. Sci. U.S.A. 102 (30),
619–627. 10712–10717.
Chakraborty, M., Burmeister, S.S., 2015. Effects of estradiol on neural responses to Hoke, K.L., Ryan, M.J., Wilczynski, W., 2007. Integration of sensory and motor pro-
social signals in female túngara frogs. J. Exp. Biol. 218, 3671–3677. cessing underlying social behaviour in túngara frogs. Proc. R. Soc. B Biol. Sci. 274
Chu, J., Wilczynski, W., 2001. Social influences on androgen levels in the southern (1610), 641–649.
leopard frog, Rana sphenocephala. Gen. Comp. Endocrinol. 121 (1), 66–73. Hoke, K.L., Ryan, M.J., Wilczynski, W., 2008. Candidate neural locus for sex differ-
Crowley, P.H., Travers, S.E., Linton, M.C., Cohn, S.L., Sih, A., Sargent, R.C., 1991. ences in reproductive decisions. Biol. Lett. 4 (5), 518–521.
Mate density, predation risk, and the seasonal sequence of mate choices: Hoke, K.L., Ryan, M.J., Wilczynski, W., 2010. Sexually dimorphic sensory gating drives
a dynamic game. Am. Nat. 137 (4), 567–596. behavioral differences in túngara frogs. J. Exp. Biol. 213 (Pt 20), 3463–3472.
Neuroendocrine Control of Social Behavior in Frogs 115

Howard, C.M., Lutterschmidt, D.I., 2015. The effects of melatonin on brain arginine Lynch, K.S., Wilczynski, W., 2005. Gonadal steroids vary with reproductive stage in
vasotocin: relationship with sex and seasonal differences in melatonin receptor type a tropically breeding female anuran. Gen. Comp. Endocrinol. 143 (1), 51–56.
1 in green treefrogs (Hyla cinerea). J. Neuroendocrinol. 27 (8), 670–679. Lynch, K.S., Wilczynski, W., 2006. Social regulation of plasma estradiol concentration
Iela, L., Rastogi, R.K., Delrio, G., Bagnara, J.T., 1986. Reproduction in the Mexican in a female anuran. Horm. Behav. 50 (1), 101–106.
leaf frog, Pachymedusa dacnicolor: III. The female. Gen. Comp. Endocrinol. 63 Lynch, K.S., Wilczynski, W., 2008. Reproductive hormones modify reception of
(3), 381–392. species-typical communication signals in a female anuran. Brain Behav. Evol. 71
d’Istria, M., Monteleone, P., Serino, I., Chieffi, G., 1994. Seasonal variations in the (2), 143–150.
daily rhythm of melatonin and NAT activity in the Harderian gland, retina, pineal Mangiamele, L.A., Burmeister, S.S., 2011. Auditory selectivity for acoustic features
gland, and serum of the green frog, Rana esculenta. Gen. Comp. Endocrinol. 96 that confer species recognition in the túngara frog. J. Exp. Biol. 214 (17),
(1), 6–11. 2911–2918.
Itoh, M., Inoue, M., Ishii, S., 1990. Annual cycle of pituitary and plasma gonadotropins Marler, C.A., Ryan, M.J., 1996. Energetic constraints and steroid hormone correlates
and plasma sex steroids in a wild population of the toad, Bufo japonicus. Gen. of male calling behaviour in the túngara frog. J. Zool. (Lond.) 240, 379–409.
Comp. Endocrinol. 78 (2), 242–253. Medina, M.F., Ramos, I., Crespo, C.A., González-Calvar, S., Fernández, S.N., 2004.
Itoh, M., Ishii, S., 1990. Changes in plasma levels of gonadotropins and sex steroids in Changes in serum sex steroid levels throughout the reproductive cycle of Bufo
the toad, Bufo japonicus, in association with behavior during the breeding season. arenarum females. Gen. Comp. Endocrinol. 136 (2), 143–151.
Gen. Comp. Endocrinol. 80 (3), 451–464. Mendonça, M.T., Licht, P., Ryan, M.J., Barnes, R., 1985. Changes in hormone levels
Kelley, D.B., 1980. Auditory and vocal nuclei in the frog brain concentrate sex in relation to breeding behavior in male bullfrogs (Rana catesbeiana) at the indi-
hormones. Science 207 (4430), 553–555. vidual and population levels. Gen. Comp. Endocrinol. 58 (2), 270–279.
Kelley, D.B., 1982. Female sex behaviors in the South African clawed frog, Xenopus Miranda, J.A., Wilczynski, W., 2009a. Female reproductive state influences the
laevis: gonadotropin-releasing, gonadotropic, and steroid hormones. Horm. Behav. auditory midbrain response. J. Comp. Physiol. A 195 (4), 341–349.
16 (2), 158–174. Miranda, J.A., Wilczynski, W., 2009b. Sex differences and androgen influences on
Kelley, D.B., Morrell, J.I., Pfaff, D.W., 1975. Autoradiographic localization of midbrain auditory thresholds in the green treefrog, Hyla cinerea. Hearing Res. 252
hormone-concentrating cells in the brain of an amphibian, Xenopus laevis. I. (1–2), 79–88.
Testosterone. J. Comp. Neurol. 164 (1), 47–61. Moore, F.L., Miller, L.J., 1984. Stress-induced inhibition of sexual behavior: cortico-
Kelley, D.B., Pfaff, D.W., 1976. Hormone effects on male sex behavior in adult South sterone inhibits courtship behaviors of a male amphibian (Taricha granulosa).
African clawed frogs, Xenopus laevis. Horm. Behav. 7 (2), 159–182. Horm. Behav. 18 (4), 400–410.
Krohmer, R.W., Lutterschmidt, D.I., 2011. Environmental and neuroendocrine control Moore, I.T., Jessop, T.S., 2003. Stress, reproduction, and adrenocortical modulation in
of reproduction in snakes. In: Lamieson, B.G.M. (Ed.), Reproductive Biology and amphibians and reptiles. Horm. Behav. 43 (1), 39–47.
Phylogeny, vol. 9. CRC Press, Boca Raton, FL, pp. 289–346. Morrell, J.I., Kelley, D.B., Pfaff, D.W., 1975. Autoradiographic localization of
Lea, J., Halliday, T., Dyson, M., 2000. Reproductive stage and history affect the hormone-concentrating cells in the brain of an amphibian, Xenopus laevis. II.
phonotactic preferences of female midwife toads, Alytes muletensis. Anim. Behav. Estradiol. J. Comp. Neurol. 164 (1), 63–77.
60 (4), 423–427. Narayan, E.J., Molinia, F.C., Christi, K.S., Morley, C.G., Cockrem, J.F., 2010. Annual
Leary, C.J., 2009. Hormones and acoustic communication in anuran amphibians. cycles of urinary reproductive steroid concentrations in wild and captive endan-
Integr. Comp. Biol. 49 (4), 452–470. gered Fijian ground frogs (Platymantis vitiana). Gen. Comp. Endocrinol. 166 (1),
Leary, C.J., 2014. Close-range vocal signals elicit a stress response in male green 172–179.
treefrogs: resolution of an androgen-based conflict. Anim. Behav. 96, 39–48. Neary, T.J., 1988. Forebrain auditory pathways in ranid frogs. In: Fritzsch, B.,
Leary, C.J., Garcia, A.M., Knapp, R., 2006. Elevated corticosterone levels elicit Ryan, M.J., Wilczynski, W., Heatherington, T., Walkowiak, W. (Eds.), The Evolution
non-calling mating tactics in male toads independently of changes in circulating of the Amphibian Auditory System. John Wiley & Sons, New York, pp. 233–252.
androgens. Horm. Behav. 49 (4), 425–432. Neary, T.J., Wilczynski, W., 1986. Auditory pathways to the hypothalamus in ranid
Leary, C.J., Garcia, A.M., Knapp, R., Hawkins, D.L., 2008. Relationships among frogs. Neurosci. Lett. 71 (2), 142–146.
steroid hormone levels, vocal effort and body condition in an explosive-breeding O’Bryant, E.L., Wilczynski, W., 2010. Changes in plasma testosterone levels and brain
toad. Anim. Behav. 76 (1), 175–185. AVT cell number during the breeding season in the green treefrog. Brain Behav.
Leary, C.J., Harris, S., 2013. Steroid hormone levels in calling males and males Evol. 75 (4), 271–281.
practicing alternative non-calling mating tactics in the green treefrog, Hyla cinerea. O’Connell, L.A., Hofmann, H.A., 2012. Evolution of a vertebrate social decision-making
Horm. Behav. 63 (1), 20–24. network. Science 336, 1154–1157.
Leary, C.J., Lippincott, J., Harris, S., Hawkins, D.L., 2015. A test of the O’Connell, L.A., Ding, J.H., Ryan, M.J., Hofmann, H.A., 2011. Neural distribution of the
energetics-hormone vocalization model in the green treefrog. Gen. Comp. Endo- nuclear progesterone receptor in the túngara frog, Physalaemus pustulosus.
crinol. 213 (0), 32–39. J. Chem. Neuroanat. 41 (3), 137–147.
Li, D., Smith, D., Hardeland, R., Yang, M., Xu, H., Zhang, L., Yin, H., Zhu, Q., 2013. Orchinik, M., Licht, P., Crews, D., 1988. Plasma steroid concentrations change in
Melatonin receptor genes in vertebrates. Int. J. Mol. Sci. 14 (6), 11208. response to sexual behavior in Bufo marinus. Horm. Behav. 22 (3), 338–350.
Licht, P., McCreery, B.R., Barnes, R., Pang, R., 1983. Seasonal and stress related Palka, Y.S., Gorbman, A., 1973. Pituitary and testicular influenced sexual behavior in
changes in plasma gonadotropins, sex steroids, and corticosterone in the bullfrog, male frogs, Rana pipiens. Gen. Comp. Endocrinol. 21 (1), 148–151.
Rana catesbeiana. Gen. Comp. Endocrinol. 50 (1), 124–145. Penna, M., Capranica, R., Somers, J., 1992. Hormone-induced vocal behavior and
Luridiana, S., Mura, M.C., Daga, C., Diaz, M.L., Bini, P.P., Cosso, G., Carcangiu, V., midbrain auditory sensitivity in the green treefrog, Hyla cinerea. J. Comp. Physiol. A
2015. The relationship between melatonin receptor 1A gene (MTNR1A) poly- 170 (1), 73–82.
morphism and reproductive performance in Sarda breed sheep. Livest. Sci. 171, Pierantoni, R., Iela, L., Delrio, G., Rastogi, R.K., 1984. Seasonal plasma sex steroid
78–83. levels in the female Rana esculenta. Gen. Comp. Endocrinol. 53 (1), 126–134.
Lutterschmidt, D.I., Maine, A.R., 2014. Sex or candy? Neuroendocrine regu- Pierantoni, R., Varriale, B., Minucci, S., Di Matteo, L., Fasano, S., D’Antonio, M.,
lation of the seasonal transition from courtship to feeding behavior in male Chieffi, G., 1986. Regulation of androgen production by frog (Rana esculenta)
red-sided garter snakes (Thamnophis sirtalis parietalis). Horm. Behav. 66 testis: an in vitro study on the effects exerted by estradiol, 5a-dihy-
(1), 120–134. drotestosterone, testosterone, melatonin, and serotonin. Gen. Comp. Endocrinol.
Lutterschmidt, D.I., Mason, R.T., 2009. Endocrine mechanisms mediating 64 (3), 405–410.
temperature-induced reproductive behavior in red-sided garter snakes (Thamnophis Polzonetti-Magni, A., Botte, V., Bellini-Cardellini, L., Gobbetti, A., Crasto, A., 1984.
sirtalis parietalis). J. Exp. Biol. 212 (19), 3108–3118. Plasma sex hormones and post-reproductive period in the green frog, Rana
Lutterschmidt, D.I., Wilczynski, W., 2012. Sexually dimorphic effects of melatonin on esculenta complex. Gen. Comp. Endocrinol. 54 (3), 372–377.
brain arginine vasotocin immunoreactivity in green treefrogs (Hyla cinerea). Brain Prendergast, B.J., 2010. MT1 melatonin receptors mediate somatic, behavioral, and
Behav. Evol. 80 (3), 222–232. reproductive neuroendocrine responses to photoperiod and melatonin in Siberian
Lynch, K.S., Crews, D., Ryan, M.J., Wilczynski, W., 2006. Hormonal state influences hamsters (Phodopus sungorus). Endocrinology 151 (2), 714–721.
aspects of female mate choice in the túngara frog (Physalaemus pustulosus). Propper, C.R., Moore, F.L., 1991. Effects of courtship on brain gonadotropin
Horm. Behav. 49 (4), 450–457. hormone-releasing hormone and plasma steroid concentrations in a female
Lynch, K.S., Rand, A.S., Ryan, M.J., Wilczynski, W., 2005. The influence of repro- amphibian (Taricha granulosa). Gen. Comp. Endocrinol. 81 (2), 304–312.
ductive stage in producing within-individual plasticity in female mate choice. Anim. Rand, A.S., Bridarolli, M.E., Dries, L., Ryan, M.J., 1991. Light levels influence female
Behav. 69 (3), 689–699. choice in túngara frogs: predation risk assessment? Copeia 2, 447–450.
116 Neuroendocrine Control of Social Behavior in Frogs

Rastogi, R.K., Iela, L., Delrio, G., Bagnara, J.T., 1986. Reproduction in the Mexican leaf de Vlaming, V.L., Sage, M., Charlton, C.B., 1974. The effects of melatonin treatment
frog, Pachymedusa dacnicolor: II. The male. Gen. Comp. Endocrinol. 62 (1), 23–35. on gonadosomatic index in the teleost, Fundulus similis, and the tree frog, Hyla
Rawding, R.S., Hutchison, V.H., 1992. Influence of temperature and photoperiod on cinerea. Gen. Comp. Endocrinol. 22 (4), 433–438.
plasma melatonin in the mudpuppy, Necturus maculosus. Gen. Comp. Endocrinol. Wada, M., Gorbman, A., 1977. Relation of mode of administration of testosterone to
88 (3), 364–374. evocation of male sex behavior in frogs. Horm. Behav. 8 (3), 310–319.
Revel, F.G., Masson-Pévet, M., Pévet, P., Mikkelsen, J.D., Simonneaux, V., 2009. Wada, M., Wingfield, J.C., Gorbman, A., 1976. Correlation between blood level of
Melatonin controls seasonal breeding by a network of hypothalamic targets. androgens and sexual behavior in male leopard frogs, Rana pipiens. Gen. Comp.
Neuroendocrinology 90 (1), 1–14. Endocrinol. 29 (1), 72–77.
Romero, L.M., 2002. Seasonal changes in plasma glucocorticoid concentrations in Walkowiak, W., 1980. The coding of auditory signals in the torus semicircularis of the
free-living vertebrates. Gen. Comp. Endocrinol. 128 (1), 1–24. fire-bellied toad and the grass frog: responses to simple stimuli and to conspecific
Rose, G.J., Gooler, D.M., 2007. Function of the amphibian central auditory system. In: calls. J. Comp. Physiol. 138 (2), 131–148.
Narins, P.M., Feng, A.S., Fay, R.R., Popper, A.N. (Eds.), Springer Handbook of Weintraub, A.S., Kelley, D.B., Bockman, R.S., 1985. Prostaglandin E2 induces
Auditory Research: Hearing and Sound Communication in Amphibians. Springer, receptive behaviors in female Xenopus laevis. Horm. Behav. 19 (4), 386–399.
New York, pp. 250–290. Wells, K.D., 1977. The social behaviour of anuran amphibians. Anim. Behav. 25,
Roy, D.B., Borah, B., Sarma, A., 1995. Analysis and significance of female reciprocal 666–693.
call in frogs. Curr. Sci. 69, 265–270. Wells, K.D., 2001. The energetics of calling in frogs. In: Ryan, M.J. (Ed.), Anuran
Ryan, M.J., 1985. The Túngara Frog: A Study in Sexual Selection and Communication. Communication. Smithsonian Institution Press, Washington, DC, pp. 45–60.
University of Chicago Press, Chicago. Wells, K.D., 2007. The Ecology and Behavior of Amphibians. University of Chicago
Scaia, M.F., Regueira, E., Volonteri, M.C., Ceballos, N.R., 2013. Estradiol production Press, Chicago.
by the Bidder’s organ of the toad Rhinella arenarum (Amphibia, Anura). Seasonal Wetzel, D.M., Kelley, D.B., 1983. Androgen and gonadotropin effects on male mate calls
variations in plasma estradiol. J. Exp. Zool. Part A Ecol. Genet. Physiol. 319 (6), in South African clawed frogs, Xenopus laevis. Horm. Behav. 17 (4), 388–404.
355–364. Wiechmann, A.F., Wirsig-Wiechmann, C.R., 1993. Distribution of melatonin receptors
Schmidt, R.S., 1966. Hormonal mechanisms of frog mating calling. Copeia 1966, in the brain of the frog Rana pipiens as revealed by in vitro autoradiography.
637–644. Neuroscience 52 (2), 469–480.
Schmidt, R.S., 1984. Mating call phonotaxis in the female American toad: induction by Wilczynski, W., Allison, J.D., 1989. Acoustic modulation of neural activity in the
hormones. Gen. Comp. Endocrinol. 55, 150–156. hypothalamus of the leopard frog. Brain Behav. Evol. 33 (6), 317–324.
Schmidt, R.S., 1985. Prostagladin-induced mating call phonotaxis in female American Wilczynski, W., Allison, J.D., Marler, C.A., 1993. Sensory pathways linking social and
toad: facilitation by progesterone and arginine vasotocin. J. Comp. Physiol. A 156, environmental cues to endocrine control regions of amphibian forebrains. Brain
823–829. Behav. Evol. 42 (4–5), 252–264.
Simmons, D.D., Meenderink, S.W.F., Vassilakis, P.N., 2007. Anatomy, physiology, and Wilczynski, W., Burmeister, S., 2016. Effects of steroid hormones on hearing and
function of the auditory end-organs in the frog inner ear. In: Narins, P.M., communication in frogs. In: Bass, A.H., Sisneros, J.A., Fay, R.R., Popper, A.N.
Feng, A.S., Fay, R.R., Popper, A.N. (Eds.), Hearing and Sound Communication in (Eds.), Hearing and Hormones: Springer Handbook of Auditory Research. Springer,
Amphibians. Springer, New York, pp. 184–220. New York, vol. 57, pp. 53–75.
Sisneros, J.A., Bass, A.H., 2003. Seasonal plasticity of peripheral auditory frequency Wilczynski, W., Capranica, R.R., 1984. The auditory system of anuran amphibians.
sensitivity. J. Neurosci. 23 (3), 1049–1058. Prog. Neurobiol. 22 (1), 1–38.
Sisneros, J.A., Forlano, P.M., Deitcher, D.L., Bass, A.H., 2004. Steroid-dependent Wilczynski, W., Endepols, H., 2007. Central auditory pathways in anuran amphibians:
auditory plasticity leads to adaptive coupling of sender and receiver. Science the anatomical basis of hearing and sound communication. In: Narins, P.M.,
305, 404–407. Feng, A.S., Fay, R.R., Popper, A.N. (Eds.), Hearing and Sound Communication in
Solís, R., Penna, M., 1997. Testosterone levels and evoked vocal responses in Amphibians: Springer Handbook of Auditory Research. Springer, New York,
a natural population of the frog Batrachyla taeniata. Horm. Behav. 31 (2), pp. 221–249.
101–109. Wilczynski, W., Lynch, K.S., O’Bryant, E.L., 2005. Current research in amphibians:
Taigen, T., Wells, K., 1985. Energetics of vocalization by an anuran amphibian (Hyla studies integrating endocrinology, behavior, and neurobiology. Horm. Behav. 48
versicolor ). J. Comp. Physiol. B 155 (2), 163–170. (4), 440–450.
Tobias, M.L., Viswanathan, S.S., Kelley, D.B., 1988. Rapping, a female receptive call, Wilczynski, W., Ryan, M.J., 2010. The behavioral neuroscience of anuran social signal
initiates male–female duets in the South African clawed frog. Proc. Natl. Acad. Sci. processing. Curr. Opin. Neurobiol. 20 (6), 754–763.
U.S.A. 95, 1870–1875. Yao, M., Hu, F., Denver, R.J., 2008. Distribution and corticosteroid regulation of
Townsend, D.S., Moger, W.H., 1987. Plasma androgen levels during male parental glucocorticoid receptor in the brain of Xenopus laevis. J. Comp. Neurol. 508 (6),
care in a tropical frog (Eleutherodactylus). Horm. Behav. 21 (1), 93–99. 967–982.
Trivers, R., 1972. Parental investment and sexual selection. In: Campbell, B. (Ed.), Yovanof, S., Feng, A.S., 1983. Effects of estradiol on auditory evoked responses from
Sexual Selection and Descent of Man. Aldine, Chicago. the frog’s auditory midbrain. Neurosci. Lett. 36, 291–297.
Urano, A., Gorbman, A., 1981. Effects of pituitary hormonal treatment on respon- Zakon, H., Wilczynski, W., 1988. The physiology of the anuran eighth nerve. In:
siveness of anterior preoptic neurons in male leopard frogs, Rana pipiens. J. Comp. Fritzsch, B., Walkowiak, W., Ryan, M.J., Wilczynski, W., Heatherington, T. (Eds.),
Physiol. 141 (2), 163–171. The Evolution of the Amphibian Auditory System. Wiley, New York, pp. 125–155.
Varriale, B., Pierantoni, R., Di Matteo, L., Minucci, S., Fasano, S., D’Antonio, M., Zhang, D., Cui, J., Tang, Y., 2012. Plasticity of peripheral auditory frequency sensitivity
Chieffi, G., 1986. Plasma and testicular estradiol and plasma androgen profile in in Emei music frog. PLoS One 7 (9), e45792.
the male frog Rana esculenta during the annual cycle. Gen. Comp. Endocrinol. 64 Zornik, E., Kelley, D.B., 2011. A neuroendocrine basis for the hierarchical control of
(3), 401–404. frog courtship vocalizations. Front. Neuroendocrinol. 32 (3), 353–366.
2.07 The Vertebrate Brain Stem as the Primary Site of Behavior Command:
Insights from an Amphibian
Emma Coddington, Willamette University, Salem, OR, USA
Ó 2017 Elsevier Inc. All rights reserved.

2.07.1 Scope of This Chapter 117


2.07.2 Broadening the Theoretical Framework Underlying Our Understanding of Sex Behaviors 117
2.07.2.1 A Broader Framework for the Evolutionary Significance of Sex Behaviors 117
2.07.2.2 A Modular Approach to Understanding Sex Behaviors 118
2.07.3 Current Understanding of Motor Control of Vertebrate Behaviors and Taricha Clasping 119
2.07.3.1 Taricha Clasp Behavior 119
2.07.3.2 Motor Control of Vertebrate Behaviors Including Clasp Behavior 119
2.07.3.3 Salamanders Provide an Ideal Model to Examine Brainstem Command of Behavior 122
2.07.4 Neuromodulation of Clasping 123
2.07.4.1 VT Enhances Social Behaviors, Including Clasping 123
2.07.4.2 Acute Stress Rapidly Suppresses Sex Behavior Through CORT Actions 124
2.07.4.3 How Does CORT Rapidly Modify Behaviors? 124
2.07.4.3.1 Context-Dependent Interactions of CORT and VT 125
2.07.4.3.2 CORT Modifies VT V1a Receptor Availability in mRF Neurons 125
2.07.4.4 Identity of Membrane CORT Receptors 126
2.07.5 Outlook 126
Acknowledgments 126
References 127

2.07.1 Scope of This Chapter 2.07.2 Broadening the Theoretical Framework


Underlying Our Understanding of Sex Behaviors
This chapter begins by asking us to broaden the scope of how
2.07.2.1 A Broader Framework for the Evolutionary
we define and discuss sex behaviors in order to identify previ-
Significance of Sex Behaviors
ously unconsidered questions, allowing us to grow our under-
standing of the evolutionary and proximal mechanisms that Sex behaviors are often referred to synonymously with repro-
mediate sex behaviors. The current understanding of motor ductive behaviors, motivated toward gamete exchange. If we
control of sex behaviors in vertebrates is summarized and dis- define behavior as the musculoskeletal movements that animals
cussed in the context of neural control of clasping in the newt, make, then sex/reproductive behaviors are the suite of coordi-
Taricha granulosa, a urodele amphibian. This chapter is centered nated musculoskeletal movements made toward locating,
on the key role of the rostromedial medullary reticular forma- attracting, competing, choosing, and persuading a conspecific(s)
tion (mRF) in the vertebrate brainstem and its principal output to join them in sexual interactions. Classically, sex behaviors
cells, the reticulospinal neurons. The concept of the brainstem have been defined and limited to the act of gamete exchange,
as an essential region in motor control of behavior is extended with a large amount of effort expended toward understanding
by current data that show how neuromodulation and penile erection and ejaculation, resource competition, and
metamodulation provide for situationally relevant expression mate selection, in concert with understanding female receptivity
of clasping. There are at least two key hormones, corticosterone to male attentions. The significance of these studies requires us
(CORT) and vasotocin (VT), that act on mRF neurons to to agree that the only and primary drive motivating sex behav-
modulate context-dependent expression of sex behavior in iors is to exchange gametes. We could, however, consider that
newts (clasping). Studies in Taricha examining the nature of the suite of behaviors involved in sexual interactions may also
the interaction between these two hormones expose two serve an arguably even more fundamental animal drive, the
main findings: (1) endocannabinoid signaling is critical for drive for affiliation and social connection through touch.
CORT effects on sensorimotor processing leading to From this perspective, our working definition of sex behavior
clasping and (2) CORT suppresses V1a receptor availability can encompass same-sex, heterosexual, and affiliative behaviors
in a novel mechanism of metamodulation. Given the conser- that can be, but not necessarily always, used toward sexual inter-
vation of motor circuits and neuromodulatory controls of actions (Zuk, 2006). This directly applies to how we are begin-
behavior, we can derive and apply fundamental principles ning to view Taricha clasping behavior, and how we could view
learned from the study of Taricha clasping to a broader under- sex behaviors across all vertebrates.
standing of how basic behaviors of vertebrates are controlled I am compelled to adopt this broadened definition of sex
by brainstem neural circuits and modulated to allow for behaviors because it allows us to ask previously unconsidered
context-appropriate expression. questions and therefore grow our understanding of the

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00025-0 117


118 The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian

evolutionary and proximal mechanisms mediating behaviors behaviors, the smallest functional unit of behaviors akin to
in general. If we view sex behaviors on a continuum from genes, being organized in a predictable order toward an
completely solitary to paired sexual interactions, then several easily identifiable goal. Behavioral biologists construct etho-
inferences can be made. The suite of sexual behaviors grams to capture the predictable and precise sequence of
possible, and used in one species, will vary according to behavioral modules composing a behavioral suite, such as
evolutionary constraints – defining the neural, hormonal, sex or feeding behavior. Some modules are common across
and musculoskeletal architecture and therefore specific behav- animals within entire taxa, some are species-specific, some
iors that any one individual has to work with. The notion of can be skipped or repeated an individual’s ethogram, and
social selection has been offered to better explain the evolu- some can occur across behavioral suites of the same species
tion of diverse sexual strategies and behaviors observed in across sexes. Applying the comparative approach to this
the animal kingdom (Roughgarden et al., 2006; Milam et view of behaviors would allow us to examine a module
al., 2011; Roughgarden, 2012). The manifestation of specific common across species, across sexes within a species, or
behaviors at a given time point in any one individual will within a sex across different ontogenetic stages or situational
vary according to ontogenetic, social, environmental, and contexts, revealing fundamental principles of neural function
immediate physiological influences that are coded by a flex- and hormone action. This modular approach to under-
ible combination of neural and endocrine systems. standing behaviors has been employed in robotics as
How we define the suite of sexual behaviors involved, and a bottom-up basis of designing behaviorally adaptive robots
the selective forces acting on them, will shift how we view the (Lenser et al., 2001) and has been suggested as a useful
behaviors and underlying neural control and will therefore framework to understand the neurobiological basis of sex
shift how we ask questions and reveal any emergent under- differences in behavior (Yang and Shah, 2014). If behavioral
standing of fundamental principles. The species-specific and biologists view discrete behaviors as modules, then we can
sex-specific manifestation of sex behaviors is typically viewed begin to realize that courtship behavior is distinct from affili-
as different behaviors, yet there are modules of behaviors and ative or feeding behaviors, not due to the specific modules
neural circuits that are common across species and sexes per se, but due to the order of modules employed with the
(Dominici et al., 2011). For example, female and male newts addition or elimination of specific modules. Examining the
both employ clasping behavior, where the hind- and fore- neural and endocrine regulation of Taricha clasping behavior
limbs are bilaterally and rhythmically engaged to grasp as a behavioral module offers the opportunity to apply this
something between their limbs. Females use this behavior modified comparative method to reveal previously unappre-
to lay eggs on the underside of sticks and reeds in ponds; ciated proximal mechanisms.
males use this same behavior to grasp and persuade the Understanding the fundamental principles of how the
female to ultimately exchange gametes, but males and neural and endocrine systems work in concert to facilitate
females also use clasping behavior with conspecifics of any reflexive or goal-directed behaviors is not trivial. The nervous
sex when exuberantly responding to food placement in their and endocrine systems are both dynamic; they influence
tanks. each other in highly flexible ways and function across very
The modular nature of the motor control circuits in combi- different spatial and temporal scales. Historically, researchers
nation with the ability of neuromodulators to shift the have approached questions in this field by focusing on either
command allows for the exhibition of apparently ‘different’ the neural or the endocrine control of behaviors. Few models
behaviors. If we begin to realize the common elements bring the two fields together. The most comprehensively
composing these behaviors we can begin to examine the under- examined models exploit the relative simplicity of inverte-
lying fundamental principles of neural control and hormone brates such as the crayfish stomatogastric system controlling
action. The behavior itself becomes goal-directed, sex- and food digestion behaviors, nudibranch escape response, and
species-specific, when engaged within a specific window of leech locomotion. Favored vertebrate models include bird-
time – during specific life stages, seasons, and circadian time song with an emphasis on cortical circuits, rodent lordosis
frames. behavior with an emphasis on the hypothalamus, and
teleost and Xenopus vocalization circuits that include the
brainstem.
2.07.2.2 A Modular Approach to Understanding Sex
We are examining questions about the role of the brain-
Behaviors
stem in a urodele amphibian, rough-skinned newts (T. gran-
In order to derive fundamental principles of neuronal and ulosa). Our overarching working hypothesis asserts that (1)
hormonal regulation of behaviors, it is also useful to redefine clasping behavior can be considered a modified version of
how we (de)construct, and therefore measure, behaviors in locomotion; (2) a given hormone will modulate behavior
general and sex behaviors specifically. Interestingly, a recent in a species-, sex-, and context-specific manner, enabling
survey revealed that behavioral biologists do not yet agree a different order and frequency of modules employed to
on the definition of behavior (Levitis et al., 2009). We manifest a specific goal-directed behavioral suite; (3) a given
suggest that this is largely because behaviors are often hormone will activate different behavior suites in the same
defined based on their functional endpoint: sexual interac- organism because it uses a cascade of different cellular
tions that result in gamete exchange, territorial defense, mechanisms acting on different timescales from milliseconds
foraging, parental or affiliative behaviors. Yet these behaviors to seasons; and (4) metamodulation of hormone actions
listed above are more accurately termed ‘behavioral suites’ is essential for the expression of context-appropriate
and consist of a concert of many discrete modules of behaviors.
The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian 119

2.07.3 Current Understanding of Motor Control behaviors (Beach, 1967; Kelley and Pfaff, 1976; Moralí et al.,
of Vertebrate Behaviors and Taricha Clasping 2003; Crews and Moore, 2005; Adkins-Regan, 1990, 2009;
Carter, 1998; Pfaus et al., 2001; Ball and Balthazart, 2004; Moore
2.07.3.1 Taricha Clasp Behavior
and Rose, 2002; Coddington and Moore, 2003; Hull and Dom-
Taricha is a urodele amphibian that exhibits a robust sex inguez, 2007; Lutterschmidt and Maine, 2014), we can under-
behavior, amplectic clasping, which is modulated by experi- stand that sexual interactions are composed of a suite of
ence and exposure to hormones (Moore and Rose, 2002; behaviors that are initiated in a seasonally appropriate manner,
Moore et al., 2005). The clasp itself is one element of a complex modulated to be situationally appropriate, and maintained by
sequence of behavioral modules underlying a number of interconnected microcircuits of neurons in the brainstem and
behaviors in Taricha including reproductive behaviors. Court- spinal cord. Seasonally, circadian, and physiologically appro-
ship clasping by males entails a sequence of events that are priate motivation (goal-directed information) is provided by
guided by various sensory modalities (vision, olfaction, inputs from basal ganglia, striatum, cerebellum, hypothalamus,
somatic sensation) (Propper, 1991). The suite of courtship thalamus, and cortex to the mRF of the brainstem (Grillner,
behaviors include a coordinated sequence of behavioral 2006, 2011). The reticulospinal neurons in the mRF provide
modules in which the male pursues and clasps the female the primary output commands to initiate and maintain specific
and then rubs the female’s snout with his chin. The clasp is behaviors in all vertebrates (Grillner, 2011; Humphries et al.,
stereotyped: an unstressed mature male grasps the back of 2007). In salamanders specifically, descending reticulospinal
a sexually attractive ovulating female and synchronously neurons influence the onset, offset, and duration of the clasp
tightens his hind legs around the female’s dorsally presented motor reflex (Naujoks Manteuffel and Manteuffel, 1988; Rose
torso (Propper, 1991; Moore and Rose, 2002). This response et al., 1995). The mRF region is subject to multisensory and
is robustly triggered and maintained by tactile somatosensory hormonal neuromodulation and metamodulation (Bowsher
pressure applied to the cloaca, located between the hind limbs. et al., 1968; Rose et al., 1993, 1995; Martin et al., 2010, 2011).
Neuromodulation effectively broadens the operational range
of neuronal circuits allowing for flexible modification of basic
The clasp as a modified version of locomotion
The clasp behavior can be considered a fundamental behavioral
behaviors (Katz, 1999; Mesce, 2002; Marder and Prinz, 2003;
module that engages the same limbs as other forms of locomotion, Marder et al., 2014), while metamodulation is the second-
recruiting the same muscles, motoneurons, spinal central pattern order modulation of neuromodulator actions (Katz, 1999;
generators (CPGs), and rostromedial medullary reticular formation Mesce, 2002). Collectively, these provide modulatory input
(mRF), but in a behavior-specific order and rhythmicity. Viewing
that allows for context-appropriate behaviors.
the clasp from this perspective informs how we consider all vertebrate
behaviors that involve limb movements and torso postures. The basic The brainstem and spinal cord circuitry in communication
neural circuitry driving the behavior involves the same brainstem and with the musculoskeletal system are necessary and sufficient
spinal cord regions and the same neuromuscular units. The mRF is to produce situationally specific behaviors in all vertebrates
one of the primary sites where vertebrates manifest their urgent (Figure 1). Original studies in cats and dogs by Sherrington
behavior decisions to fuck/fight/flight/or freeze, and the decision is
made based on modulation and metamodulation of the local
(1910) and nonhuman primates by Lawrence and Kuypers
circuitry. The reticulospinal neurons provide the primary output (1968a,b) revealed the essential and sufficient role of the
from the mRF, dictating the order of CPG recruitment and rate of medulla oblongata in the motor control of the trunk and prox-
rhythmicity. The most rostral reticular region, the mesencephalic imal and distal limb segments. Decerebrate cats and dogs
locomotor region (MLR), provides drive to the mRF, because the
maintain the ability to walk, run, jump, and climb. However,
MLR receives and integrates most of the inputs coming from cortical
and subcortical brain regions. These supra-brainstem regions provide the speed of movements was far from normal. These coordi-
important goal-directed salience and greater flexibility to the basic nated behaviors are entirely achievable by an intact brainstem
suite of behaviors encoded by the brainstem and spinal cord. and spinal cord, with no input from rostral parts of the central
nervous system (CNS). When decerebration leaves the hypo-
thalamus intact with brainstem, these decerebrate animals
can also homeostatically regulate functions such as body
2.07.3.2 Motor Control of Vertebrate Behaviors Including
temperature and hunger (Lawrence and Kuypers, 1968b).
Clasp Behavior
Since these early studies, however, most investigations inter-
All behaviors, including clasping, are enacted by the coordinated ested in understanding motor control of behavior shifted their
engagement of muscles on each limb, torso, head, neck, and tail focus to supra-brainstem structures underpinned by the arguably
required to enact the behavior. Drawing on the collective knowl- false assumption that ‘higher vertebrates’ are dominated by
edge of locomotor circuitry from studies of lamprey (Grillner, ‘higher brain function.’ This has led to tunnel vision and the
2006; Grillner et al., 2008; Kozlov et al., 2014), amphibians brainstem being hidden in a collective blind spot. Only recently
(Rose, 1990a,b; Rose et al., 1995; Chevallier et al., 2008; Ryczko has the focus broadened again – heavily influenced and inspired
et al., 2010; Ryczko and Dubuc, 2013; Sánchez-Camacho et al., by the discoveries in lamprey of a brainstem command circuit
2001; Ten Donkelaar, 1990), fish (Goodson and Bass, 2002; for locomotor behaviors (Grillner et al., 2008; Grillner, 2011)
Bass et al., 2008; Korn and Faber, 1996, 2005; Fetcho, 1992; and in teleosts, a brainstem Mauthner neuron command for
Finger, 2009; Eaton et al., 2001), and mammal locomotion the C-start escape response (Korn and Faber, 1996; 2005;
(Grillner, 2011; Humphries et al., 2007; Jordan et al., 2008; Eaton et al., 2001). For example, an intriguing hypothesis has
Kimura et al., 2013; McCrea and Rybak, 2008; Tresch, 2010; been proposed that reticulospinal neurons within the nucleus
Whelan, 1996; Siegel, 1979) in combination with our current reticularis gigantocellularis (NGC, also referred to as the mRF)
understanding of behavioral neuroendocrine control of sex might be evolutionary analogs of Mauthner neurons, and the
120 The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian

Figure 1 Representation of the vertebrate nervous system involved in sensorimotor selection, generation, and neuromodulation of behaviors.
Green boxes indicate regions that are essential for the generation of basic motor programs and a broad selection of postures and limb move-
ments, such as walking, clasping/hugging, or swimming. These brain regions also provide information regarding the onset and offset of the
behavior, maintenance, and intensity. mRF, rostromedial medullary reticular formation located in the rostral aspect of the medulla oblongata;
MLR, mesencephalic locomotor region located at the boundary between the brainstem and midbrain; and CPG, central pattern generator, a distrib-
uted network of neurons that are flexibly connected and together coordinate a set of muscles toward an endpoint, such as one limb movement.
Gray symbols indicate sensory input for initiation and feedback for ongoing modulation of behavior; from top down: olfaction, vision, hearing and
lateral line, taste, touch, and proprioception. Tan boxes indicate rostral brain regions that provide modulatory neuronal input to the brainstem
motor circuits. The removal of inhibitory input from the basal ganglia allows motor programs to proceed. There is anatomical evidence that the
cortex and vestibular systems also have some direct connections to the spinal cord as well. Orange ovals indicate brain regions known to provide
neuroendocrine modulatory input to the mRF, providing broader context to the motor programs including intrinsic and extrinsic information such
as physiological state and seasonal information. The final motor output to muscles is provided by the spinal motoneurons located within spinal
CPGs innervating muscle fibers.

NGC/mRF contributes to decision-making by functioning as behaviors. Situationally relevant information to the mRF is
a primary site of general arousal (Machne et al., 1955; Pfaff received from three key neuromodulatory inputs: (1) multi-
et al., 2012). This hypothesis is supported by the findings that modal sensory information and feedback (Pfaff et al.,
the brainstem and neurons within the mRF respond to and 2012; Rose et al., 1995; Martin et al., 2010; Azim et al.,
provide emotional context (Coimbra et al., 2006; Vertes and 2014); (2) general physiology and seasonal information
Miller, 1976). from the medial hypothalamus, periaqueductal gray (PAG),
The mRF is highly conserved across taxa (Grillner, 2011; and lateral hypothalamus (Jordan, 1998; Jordan et al.,
Jordan et al., 2008; Matsuyama et al., 2004a) and can be 2008); and (3) acute hormone changes in response to the
viewed as the major sensorimotor integration region in all immediate situation (Coddington et al., 2007; Rose et al.,
vertebrates mediating socially and situationally appropriate 1995, 1998; Rose and Moore, 2002). Sensory inputs arriving
The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian 121

at the mRF originate from a broad range of sources Reticulospinal neurons are responsible for modifying the
(Figure 1) and provide necessary and sufficient sensory strength of connections between CPGs and the timing of ipsi-
input to affect immediate context-appropriate behavioral lateral- and contralateral-CPG communication (Grillner,
commands. These include tactile/somatosensory (Rose 2006). Therefore, the mRF provides key information to the
et al., 1995; Rose and Sutin, 1973; Saadé et al., 1983), olfac- CPGs that results in specific behavior of limbs and posture.
tory (Bowsher et al., 1968; Thompson et al., 2008), visual For example, a walk gait would involve asymmetric limb
(Martin et al., 2010), auditory (Bowsher et al., 1968; Martin flexion–extension patterns in a distinctly different order
et al., 2010), lateral line (Bodznick and Northcutt, 1980), compared with a gallop (Grillner, 2006). In Taricha, reticulo-
vestibular (Ladpli and Brodal, 1968; Peterson and Abzug, spinal neurons respond to multimodal sensory and hormone
1975), thermal (Farham and Douglas, 1985), nociceptive information and are modulated by hormone input to produce
(Fort et al., 1994; Willis, 1985), visceral (Brown et al., the key output that commands walking, swimming, or clasping
2002; Curtis et al., 2002; Kaddumi and Hubscher, 2006), (Moore and Rose, 2002; Rose et al., 1993, 1995). Clasping
baroreceptive (Guilbaud et al., 1973; Haxhiu and Loewy, behavior involves strong, rhythmic, bilateral flexion of fore-
1996), and chemosensory stimuli (Andreatta-Van Leyen and hind limbs and therefore requires recruitment of the
et al., 1990; Guyenet et al., 2005; Silver and Finger, 2009). same limb CPGs that control each limb in a pattern and order
This sensory information is integrated at the mRF with distinctly different from that used for walking or swimming. To
socially relevant information (Shelley et al., 2006), energy understand the relationship between reticulospinal neurons,
balance (Grill and Hayes, 2012), and the need for general CPGs, and the subsequent behavior, we suggest that clasping
vigilance and anxiety as conveyed by corticotropin-releasing is a modified form of locomotion that employs reticulospinal
hormone (Lowry et al., 1996; Hubbard et al., 2010). Collec- neurons and limb and postural CPGs in a species-specific
tively, the neuromodulatory input provides the situational manner to produce symmetrical bilateral flexion of paired
information required to the mRF. limbs.
The integrated context-appropriate motor commands are There are inputs to the mRF from more rostral brainstem
sent to the spinal cord by the projection neurons of the regions that provide additional nuance and salience to any
mRF, the reticulospinal neurons. Also highly conserved across motor program. The mRF output is directly influenced by
taxa (Murakami et al., 2004; Fetcho, 1992), reticulospinal the mesencephalic locomotor region (MLR) (Figure 1).
neurons are sometimes referred to as second-order premotor This highly conserved structure located at the brainstem–
neurons. However, this terminology belies the enormously midbrain boundary contributes to locomotion in all verte-
integrative functions they provide. These neurons are related brates investigated to date (Bachmann et al., 2013; Cabel-
to Mauthner neurons, the enormous neurons located in the guen et al., 2003; Garcia-Rill et al., 1987; Garcia-Rill and
mRF that are responsible for the startle response in fish, Skinner, 1987; Shik et al., 1967; Sirota et al., 2000). Early
tadpoles, and salamanders (Eaton et al., 2001; Korn and studies performed in cats revealed that unilateral or bilateral
Faber, 2005; Oda, 2015; Sillar, 2009; Zottoli and Faber, stimulation of neurons in MLR elicit switching between
2000). Smaller and more numerous than Mauthner neurons, walking, trotting, and galloping in high-decerebrate cats
reticulospinal neurons typically range from 25 to 50 mm in (Shik et al., 1967). The MLR mediates its effects through
length and project in two directions: rostrally to supra- monosynaptic projections to the mRF where a majority of
brainstem sites and caudally to the spinal cord (Newman the mRF neurons are activated in synchrony with MLR output
et al., 1983; Roberts and Alford, 1986; Sakai et al., 2009). (67%) and behavioral pattern (Orlovskiĭ, 1970a,b). This
Their descending output to the spinal cord provides the key highly reliable circuit is conserved from teleosts to primates
information to select behaviors by modulating the order and (Grillner, 2006; Ryczko et al., 2016). Voluntary selection
frequency of motor rhythm produced by the spinal central (goal-directed selection) of motor output is provided by
pattern generators (CPGs) (Grillner, 2006; Grillner and Wal- output from the basal ganglia to the MLR and is also
len, 1985; Grillner et al., 2008; Matsuyama et al., 2004b; conserved across all vertebrates (Cabelguen et al., 2010; Grill-
McCrea and Rybak, 2008). ner, 2006, 2011; Ryczko et al., 2016).
CPGs are evolutionarily conserved across the animal Studies of decerebrate mammals have demonstrated that
kingdom; a fundamental functional unit for the motor control supra-brainstem regions are not necessary for the full
of invertebrates and vertebrates (Marder and Bucher, 2001). suite of limb and torso motor commands; nevertheless,
They are a functionally connected and spatially distributed supra-brainstem input is important for the greatly nuanced
group of neurons that coordinate a subset of movements. In versions of the basic motor programs provided by the
vertebrates, CPGs in the spinal cord coordinate limb move- brainstem þ spinal cord. More recent studies in rodents
ments, while those in the hindbrain coordinate movements confirm that spinal interneurons receive very weak input
by the torso, head, and neck (Grillner, 2006). The coordinated from corticospinal neurons, but are powerfully innervated
rhythmic activity of neurons in CPGs underlies the rhythmic on both the cell bodies and dendrites by reticulospinal
patterns of behaviors. A key feature of CPG function is that neurons (Mitchell et al., 2016). This is not to say that supra-
they are essentially modular and can function in isolation brainstem inputs are unimportant, but that their roles are
and be flexibly connected with each other to varying degrees key for appropriate expression of skilled goal-directed behav-
of strength. This flexibility allows for variability in the order iors. Brainstem motor networks receive direct descending
of activation (left, right), regulates the intensity of the move- dopaminergic inputs that potentiate the locomotor output
ment, and therefore the behavior is enacted (Buchanan, (Ryczko et al., 2016), and recurrent information from spinal
2011; Grillner, 2006). cord interneurons to mRF and cortex, in parallel with spinal
122 The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian

cord to cerebellum, play a crucial role in skilled hand þ arm and MLR in selecting and mediating locomotion and clasping
reaching (Azim et al., 2014). behaviors for several reasons. Primarily, the control of motor
output is highly conserved; therefore general principles about
neural control of tetrapod behaviors can be derived from inves-
2.07.3.3 Salamanders Provide an Ideal Model to Examine
tigations in salamanders (Grillner, 2006; Ryczko et al., 2016;
Brainstem Command of Behavior
Sánchez-Camacho et al., 2001). Secondly, there are some
Taricha granulosa are ideal model organisms for neuroethology taxon-specific anatomical arrangements that render the sala-
studies because (1) the males have robust, ethologically rele- mander an ideal model to examine mRF: the cells of the mRF
vant behaviors that are measurably modulated by hormones are presented on the floor of the fourth ventricle unobscured
(Moore et al., 2005; Moore and Zoeller, 1985; Rose and Moore, by the cerebellum and available for in vivo single-unit and
2002); (2) the neural system regulating those behaviors is iden- whole-cell electrophysiological examination, and there is
tified and amenable to interrogation because the neurons a wealth of knowledge in this species regarding hormonal
controlling the clasp behavior are large (20–100 mm) and modulation of clasping and locomotion to rely on. To date,
highly accessible for functional imaging and electrophysiology what is known about the mRF and its functional role in sala-
(Rose et al., 1993); (3) the molecular and pharmacological mander clasp and locomotor behavior modules comes primarily
identity of many receptors and signaling molecules has been from in vivo single-unit electrophysiology studies (Coddington
identified, including the cannabinoid type 1 receptor (CB1) et al., 2007; Hubbard et al., 2010; Lewis et al., 2005; Lewis
(Hollis et al., 2006; Soderstrom, 2009; Soderstrom et al., and Rose, 2003; Lowry et al., 1996; Rose, 2000; Rose et al.,
2000) and V1a receptors (Searcy et al., 2011); and (4) these 1995, 1998, 1993; Ryczko et al., 2016; Smetana et al., 2010).
animals are abundant in the Pacific Northwest of the The mRF in the hindbrain initiates and maintains clasping
USA and offer a rare opportunity to observe organisms’ (Moore and Rose, 2002; Rose et al., 1993, 1995: Figure 2).
behavior in nature. In vivo single-unit recordings from freely behaving animals
Salamanders, in general, provide an ideal model to examine have revealed that the neurons in the mRF respond to clasp-
brainstem command of behavior and therefore the role of mRF inducing somatosensory stimulation to the cloaca, and the

Figure 2 Neuroethology model of Taricha clasping behavior. (a) Schematic of neural control of clasping. (b) Transverse section of a male Taricha
brainstem at the level of the mRF region, located in the rostromedial aspect of the medulla oblongata. Abbreviations indicate specific regions – mRF,
rostromedial medullary reticular formation; ra, raphe nucleus; rp, parvocellular reticular formation; VIIIvn, eighth cranial nerve. Box indicates location
of image in c. Scale bar ¼ 100 mm. (c) Location of Taricha CB1 receptors in the hindbrain mRF (in situ hybridization), scale bar ¼ 20 mm. (d) Repre-
sentative traces of single-unit recordings from mRF neurons of awake and behaving Taricha illustrating how CORT diminishes spontaneous and
sensory stimulated responses. Gray bars below spike traces indicate the timing (onset to offset) of pressure applied to the cloaca designed to simu-
late cloacal stimulation, a necessary and sufficient somatosensory stimulus to elicit clasp behavior. The lower trace indicates time in 1 s divisions.
Reprinted with permission from Coddington, E., Lewis, C., Rose, J.D., Moore, F.L., 2007. Endocannabinoids mediate the effects of acute stress and
corticosterone on sex behavior. Endocrinology 148, 493–500.
The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian 123

reticulospinal neurons provide the output that is coincident broadly distributed within the CNS. Both VT and VP affect
with motor control of the clasp. By focusing on the a variety of social behaviors in vertebrates (Goodson and
somatosensory-evoked clasp, the neurobehavioral model Bass, 2001): mammals (Young et al., 2001), avians (Panzica
system is refined to a single, readily controlled sensory stimulus et al., 2001), teleosts (Bass and Grober, 2001), and amphibians
and simple, readily elicited, and quantifiable neural and behav- (Deviche and Moore, 1987; Emerson and Boyd, 1999; Iwata
ioral responses. et al., 2000; Moore et al., 1992).
In Taricha, VT acts as a neuromodulator to regulate male
clasping behavior. The action of VT at the mRF is proposed
2.07.4 Neuromodulation of Clasping to occur by influencing sensorimotor integration of socially
relevant sensory information by mRF neurons. This principle
The manifestation of courtship behaviors by males and females is based on multiple lines of evidence. Administration of VT
is seasonally and ontogenetically constrained and modulated increases the incidence of male clasping behaviors and VT
by hormones. The specific roles of a variety of hormones and antagonists suppress these behaviors (Moore and Miller,
transmitters in activating courtship behaviors have been well 1984; Moore and Zoeller, 1979). Reticulospinal and interneu-
characterized in Taricha: sex-specific gonadal steroid actions, rons within the mRF respond robustly to intracranial adminis-
rapid actions of CORT and its membrane receptor, sex- tration of VT with an increase in spontaneous activity and
specific and seasonal variation in VT, and dopamine have sensory responsiveness to clasp-triggering cloacal stimulation
been reviewed in earlier publications (Moore et al., 2005; (Moore and Rose, 2002; Rose et al., 1995). Interestingly, VT’s
Moore and Rose, 2002). influence is not limited to sexually specific stimuli. The behav-
The incidence and maintenance of clasping is enhanced by ioral responses of males to visual and olfactory stimuli origi-
VT (Moore and Miller, 1984; Rose et al., 1995) and suppressed nating from food or sexual partners are markedly enhanced
by exposure to acute stress, an effect mediated by CORT by VT administration (Thompson and Moore, 2000). Further-
(Coddington et al., 2007; Coddington and Moore, 2003; more, the current notion is that social experience results in
Moore and Zoeller, 1985; Rose and Moore, 1999). Our more elevated VT at the mRF. Female Taricha sex pheromones
recent work has established that suppression of clasping by promote courtship clasping in males (Thompson and Moore,
acute stress and CORT will not occur if CB1 receptors are 2000) and do so by amplifying the sensory responsiveness of
blocked (Coddington et al., 2007). These receptors are located mRF neurons to clasp-triggering somatosensory stimulation
in the mRF (Hollis et al., 2006), and their pharmacology has of the cloaca (Thompson et al., 2008). This neuromodulation
been well characterized in Taricha (Soderstrom et al., 2000). of olfactory signals is dependent on endogenously released
Importantly, VT and CORT both provide information about VT, because V1a antagonist blocks pheromone-induced behav-
situational context to the brain, and their effects are themselves ioral amplification (Thompson et al., 2008).
context-dependent (Coddington and Moore, 2003): if an While V1a receptors for VT are broadly distributed
animal is exposed to CORT or acute stress first, VT does not throughout the CNS (Evans et al., 2000), single-unit
enhance clasping. electrophysiology predicted that a primary site of action is the
Several lines of evidence have revealed that VT and CORT mRF circuitry (Rose et al., 1995). Neuroanatomical location of
interact in a time-dependent manner to provide different these receptors was, however, unknown until recently,
context-appropriate behavioral outcomes, and at the core of because there were no acceptable antibodies available for
this switch is a third signaling molecule, eCB (Coddington immunohistochemical identification of V1a receptor proteins.
et al., 2007; Coddington and Moore, 2003). Endocannabinoids In lieu of this, a functional imaging method was developed
are best known for their unique role in suppressing presynaptic using VT conjugated to a fluorophore, VT–Oregon Green
neurotransmitter release to postsynaptic neurons (Heifets and (VT-OG) (Lewis et al., 2004). This method is predicated on
Castillo, 2009; Kreitzer and Regehr, 2001; Wilson and Nicoll, the understanding that when VT binds its V1a receptor, they
2001). They are also well known to play a critical role in medi- behave like all G-protein-coupled receptors analyzed thus far,
ating vertebrate responses to stress and anxiety (Coddington by clustering with other V1a–VT complexes, and undergoing
et al., 2007; Hill, 2012; McEwen and Gianaros, 2011; Riebe receptor-mediated endocytosis (Fukunaga et al., 2006). Proof
and Wotjak, 2011; Taber and Hurley, 2009). By focusing on of concept experiments verified that the VT-OG molecule
the peptide VT and the steroid hormone, this section examines retains physiological function identical to unlabeled VT, and
the interactions between these two hormones, which promote its action is blocked by V1a antagonists (Lewis et al., 2004).
switching between very different behavioral outcomes: VT The advantage of this method is that we can use functional
promotes prosocial behaviors, including promoting courtship imaging to assess the location and availability of functional
behaviors, while CORT promotes stress-avoidance and stress- V1a receptors by tracking the VT-OG molecule as it is
coping behaviors in all vertebrates, including blocking court- packaged into vesicles visible by confocal microscopy (Lewis
ship behaviors. et al., 2005). VT-OG is endocytosed into 70% of mRF
reticulospinal neurons and 80% mRF interneurons,
supporting the notion that a large population of mRF
2.07.4.1 VT Enhances Social Behaviors, Including Clasping
neurons can respond to this peptide (Davis et al., 2015; Lewis
VT is a nonmammalian homologue of vasopressin (VP); VT/VP et al., 2005). Collectively, evidence from behavioral,
are nonapeptide hormones that have conserved functions electrophysiological, and functional imaging support the
influencing peripheral physiology via release from the notion that VT is elevated in response to social
neurohypophysis and behaviors via release from neurons experience and modulates sensorimotor integration in mRF
124 The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian

neurons to modify the expression of clasp behavior in a context- Parsimony would predict that there is a single mechanism
appropriate manner. that mediates the rapid response to CORT. For example,
some researchers have argued that intracellular GRs and MRss
alone explain the rapid response to CORT (Joëls and Van
2.07.4.2 Acute Stress Rapidly Suppresses Sex Behavior
Riel, 2004), while others argue that a novel membrane receptor
Through CORT Actions
is responsible (Moore, 2003; Orchinik et al., 1991; Tasker et al.,
Activation of the hypothalamo–pituitary–adrenal (HPA) axis 2006). I propose that parsimony, in this case, is limiting our
by acute stress leads to a rapid release of CORT into circulation. full understanding of rapid responses to stress. Instead, CORT
A majority of research has focused on the negative effects of must engage a cascade of effects across different timescales,
CORT in the induction of chronic disease states that develop from milliseconds to days and seasons, and multiple receptors
after prolonged stress (Hunter et al., 2015; McEwen and and mechanisms must be employed for a context-appropriate
Gianaros, 2011). These effects are mediated by the intracellular response to stress and for recovery from stress to occur. Our
glucocorticoid receptor (GR) and mineralocorticoid receptor research suggests that CORT mediates behavioral responses to
(MR), which act as ligand-activated transcription factors to acute stress through one established mechanism and one novel
influence genomic responses. However, high concentrations of mechanism acting on two different ‘rapid’ timescales: (1)
CORT, which are released into general circulation after an acute CORT modifies synaptic communication by upregulating
threat, affect behavior on a much faster timescale than transcrip- endogenous cannabinoid (eCB) signaling and (2) CORT
tional control: a few seconds to 5 min to initiate behavior modifies subsequent modulatory influence by interfering
changes and 20–60 min to peak, before declining equally with other receptors’ availability.
precipitously (Rankin et al., 2012). These acute effects of eCBs act as major neuromodulatory signaling molecules
CORT are crucial to an animal’s ability to respond appropriately that regulate many different physiological and behavioral
to a broad range of perceived threats. Rapid adaptive behavioral functions and have been identified as a key mediator of rapid
responses are therefore thought to be mediated by nongenomic CORT effects in a variety of brain functions necessary for appro-
mechanisms mediated through membrane receptors. priate decision-making during stress. They are critical for extinc-
A fundamental premise of my framework is that a primary tion of fear memories (Marsicano and Lafenetre, 2009;
response to a perceived threat is behavioral and that neural Marsicano et al., 2002), relief from anxiety (Riebe and Wotjak,
correlates of CORT actions on behavior must be present. 2011), providing negative feedback to the HPA axis (Di et al.,
Many different behavior suites are modified by acutely 2003, 2005), habituation of the HPA axis to repeated stress
elevated CORT, including reproductive behaviors in (Hill, 2012; Hill et al., 2010a), and modifying behavioral
mammals (Etgen and Barfield, 1986), teleosts (Remage- responses to stress (Coddington et al., 2007). Studies on the
Healey and Bass, 2006), and amphibians (Moore and Zoeller, mechanism of CORT–eCB interaction have focused on the rat
1985); locomotor activity of mammals (Meijer et al., 1998) hypothalamus, where CORT is the trigger for typical eCB
and birds (Breuner et al., 1998; Breuner and Wingfield, signaling (Evanson et al., 2010; Hill et al., 2010b; Hill and
2000); and aggression in mammals (Bertsch et al., 2011). At Tasker, 2012); where CORT initiates the synthesis and release
a neural systems level, these changes in behavior are of eCBs from postsynaptic neurons, and eCBs then travel retro-
hypothesized to be mediated by nongenomic actions of gradely to suppress neurotransmitter release by presynaptic
CORT on the processing of external sensory information neurons (Crosby and Bains, 2012; Crosby et al., 2011; Di
(environment) and/or motor output, leading to context- et al., 2003, 2005; Hill et al., 2010b). This retrograde eCB
appropriate behaviors (Coddington and Moore, 2003). Simi- signaling mechanism is a remarkably elegant way in which
larly, CORT rapidly induces changes in the animal’s attention postsynaptic neurons can switch off one presynaptic pathway
from ongoing activity to the perceived threat (Putman et al., of information and therefore tune into the remaining salient
2007). These behavioral and perceptive changes predict that inputs.
CORT directly impacts neuron function, although this predic- The CORT-initiated eCB signaling observed in rodents also
tion has rarely been directly tested. occurs in Taricha and is one of the primary mechanisms
employed by CORT to exert rapid effects in sensorimotor
processing necessary for context-appropriate clasping.
2.07.4.3 How Does CORT Rapidly Modify Behaviors?
Administration of a CB1 receptor antagonist blocks the
The challenge to understanding rapid actions of CORT is due to typically suppressive actions of CORT on male clasping
behavioral and cellular responses being context- or state- behavior. CB1 receptor antagonist also blocks CORT’s
dependent, and we have yet to consider the possibility of inhibitory effect on mRF neurons; in fact CORT post-CB1
response and recovery involving a cascade of mechanisms blockade resulted in 25% of mRF neurons actually increasing
that can occur within the ‘rapid’ millisecond-to-minute time in spontaneous activity and sensory responsiveness to clasp-
frame. Early work in Taricha revealed that CORT rapidly generating tactile stimuli (Coddington et al., 2007). A
suppresses male clasping behavior in a dose-dependent reasonable conclusion from these studies in Taricha is that
manner (Orchinik et al., 1991, 1994). The cellular mecha- CORT acts in the mRF in a manner similar to that observed
nism(s) by which CORT might rapidly influence neuronal pro- in rodent hypothalamus – binding a receptor on presynaptic
cessing and immediate behavioral responses are too rapid to be membranes, initiating synthesis and secretion of an
genomic and occur in the absence of gene transcription endocannabinoid that travels retrogradely to inhibit
(Evanson et al., 2010; Haller et al., 1998; Mikics et al., 2004; neurotransmitter release from presynaptic neurons. The
Orchinik et al., 1991). added information that we gain from the Taricha model is
The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian 125

that we have learned that eCB signaling provides a key administration of VT in lieu of courtship experience with
molecular switch allowing CORT to have variable effects on a female mirrored the effect of prosocial behavior, preventing
behavioral outcomes on the same sensorimotor circuitry. the typically suppressive effects of CORT on clasping (Codding-
Additional support for this mechanism comes from char- ton and Moore, 2003).
acterization of Taricha-specific cannabinoid receptors. Taricha A parsimonious explanation for this time-dependent inter-
CB1 receptors were cloned, sequenced, and pharmacologically action relates to the time constraints involved in eCB synthesis,
identified (Soderstrom et al., 2000). They are densely release, and subsequent modification of presynaptic release of
expressed in Taricha brain tissue and have a high sequence neurotransmitters. Evidence definitely supports the notion that
homology with mammalian CB1 receptors. Importantly, Tar- this mechanism is in play in sensorimotor processing involved
icha CB1 receptors functionally bind agonists and antagonists in the decision process leading to clasping or escape. However,
in a pharmacological signature identical to mammalian CB1 it is helpful to adopt a broader view and consider whether there
receptors (Soderstrom et al., 2000). A plausible hypothesis are other mechanisms involved in the escape – clasp, CORT–VT
was that CORT, an equally small lipophilic molecule, might interplay that might also occur in the ‘rapid’ nongenomic time
co-opt CB1 receptors to effect rapid neuronal changes. frame.
However, this hypothesis was ruled out by pharmacological
studies (Soderstrom et al., 2000). In situ hybridization reveals 2.07.4.3.2 CORT Modifies VT V1a Receptor Availability in mRF
that the density and distribution of CB1 receptors is consistent Neurons
with other vertebrates, and fundamental to our working Another plausible mechanism that would explain the time
model they are densely expressed in the mRF (Hollis et al., span of the interactions observed between CORT and VT
2006). Therefore, in support of our working model, func- includes CORT decreasing the availability of V1a receptors
tional CB1 receptors are found in the hindbrain region, which and therefore inhibiting subsequent VT-mediated effects. We
is known to mediate clasping, and respond to CORT tested this hypothesis with the use of VT conjugated to
administration. a fluorescent label that is endocytosed (internalized in
neurons) through a receptor-mediated process when the
2.07.4.3.1 Context-Dependent Interactions of CORT and VT ligand binds to V1a receptors (Fukunaga et al., 2006; Wieffer
The suppressive effect of CORT on clasping behavior is only et al., 2009). VT endocytosis was blocked by mid- and high
apparent when administered to solitary males; if males doses (akin to levels during an acute stress) of CORT in the
have already recently engaged in courtship behavior, then clasp-controlling region of the hindbrain, mRF (Figure 3;
neither acute stress nor exogenous CORT suppresses clasping Davis et al., 2015). The percent of VT-containing vesicles
(Coddington and Moore, 2003). A parallel study where court- significantly declined with increasing doses of CORT, which
ship experience was replaced with administration of VT prior to indicates that there were fewer receptor-mediated endocytosis
acute stress or CORT led to a similar outcome: VT completely events, and the intensity per vesicle also significantly
blocked the typically suppressive effects of CORT on clasping decreased, indicating that there were fewer receptor–VT pairs
(Coddington et al., 2007). per vesicle (Figure 3). These data collectively suggest that
The context-dependent effect of acute stress on clasping CORT is affecting the availability of V1a receptors. The
behavior is time sensitive and involves two primary hormones specific subcellular mechanism remains unclear but could be
(VT and CORT) that seem to convey opposing information; VT explained by (1) a decrease in the number of V1a receptors
says go, and CORT says no. In vivo single-unit electrophysiolog- expressed at the membrane, (2) a decrease in the affinity of
ical recordings of mRF neurons in male Taricha show that the extant V1a receptors, and/or (3) a suppression of the
typical suppressive effects of CORT are not seen if VT is admin- machinery by which receptor-mediated endocytosis occurs.
istered between 10 and 60 min prior to CORT administration The behavioral significance of these data was strengthened
(Rose et al., 1995). The converse is also true: the typically by two further findings: (1) the influence of CORT on VT endo-
enhancing effects of VT are not apparent if CORT is adminis- cytosis in the mRF is only apparent during the breeding season,
tered between 2 and 20 min prior to administration of VT i.e., when VT is modulating the expression of clasping behavior,
(Rose et al., 1995). Based on these early electrophysiological and (2) the CORT-induced suppression of VT endocytosis
findings, we supposed that effects observed at the single-cell observed in the mRF was not observed in the inferior
level would be reproduced at the behavioral levels. In other reticular formation (iRF) (Davis et al., 2015) – a region
words, if the animals experienced prosocial behaviors prior to thought to be homologous to one of the premotor regions
exposure to a threat, then the driving force on the animal that regulate vocalization in all vertebrates (Bass et al., 2008).
would be to continue employing prosocial behaviors. In contrast to the mRF breeding-season data where CORT
Conversely, if the animals were solitary and then exposed to significantly reduced key measures of endocytosis (number of
an acute threat, then they would fall back onto the three ‘f’s’: vesicles containing VT, the total intensity of VT/mm3, and
fight, flight, or freeze. Behavioral studies confirmed this percent area covered by vesicles), there was no significant
hypothesis. Male Taricha allowed to engage in courtship with effect of CORT on these three measures in the iRF during the
a female for 1 h (courtship typically extends for 3–20 h in breeding season. But there was a significant increase in
lab), readily reengaged in courtship after separation and expo- amount of VT found per endocytosed vesicle in the iRF after
sure to 2 min restraint stress or administration of CORT (Cod- pretreatment with high-stress CORT concentrations – polar
dington and Moore, 2003). This is remarkable given that 95– opposite effects in the two functionally different reticular
100% of solitary males exposed to 2 min restraint stress or regions. Collectively these functional imaging data suggest
CORT administration will not engage in courtship. In addition, that CORT metamodulates VT effects by modifying V1a
126 The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian

Figure 3 Corticosterone (CORT) reduces the availability of V1a receptors as indicated by a decrease in receptor-mediated endocytosis of VT conju-
gated to Oregon green (VT-OG) into hindbrain mRF cells. (a and b) are representative confocal images captured at 63x. Orange arrows indicate DAPI-
labeled nuclei. Fuchsia arrows indicate putative vesicles that have endocytosed VT-OG with V1a receptors. (c) CORT significantly decreased the
average number of endocytosed vesicles containing VT-OG compared with brains pretreated with vehicle control solution. (d) CORT significantly
decreased the intensity of VT-OG per vesicle. Collectively, these data reveal that CORT reduced the availability of functional V1a receptors and affected
the endocytic machinery so that fewer pairs of V1a receptor þ VT-OG were internalized per vesicle. Quantification of images was achieved using
Axiovision software to analyze maximum projection images of confocal slices (N ¼ 4 newts per group). Scale bars ¼ 20 mm. Graphs show data as
mean  SE, and asterisks above bars indicate significant differences between groups (p < 0.01). Reproduced based on Davis, A., Abraham, E.,
Mcevoy, E., Sonnenfeld, S., Lewis, C., Hubbard, C.S., Dolence, E.K., Rose, J.D., Coddington, E., 2015. Corticosterone suppresses vasotocin-
enhanced clasping behavior in male rough-skinned newts by novel mechanisms interfering with V1a receptor availability and receptor-mediated
endocytosis. Horm. Behav. 69, 39–49.

receptor availability. This effect is consistent with the natural was able to block the kappa-mediated suppression of
and context-dependent expression of clasping behavior, as clasping (Lombana et al., 2015). Therefore, while the in vitro
CORT only modified VT endocytosis in the mRF and not in data provide evidence that the mCR might be a kappa-opiate-
the iRF, and in a season-dependent manner. We suggest that like receptor, our in vivo behavioral data suggest that kappa-
metamodulation of other hormone receptors’ availability is mediated suppression of clasping occurs via a different subset
an unappreciated mechanism of hormone action common of membrane receptors, and endocannabinoids do not
among vertebrates (Von Zastrow and Williams, 2012) and is mediate the kappa-induced suppression of clasping. The
a major player in the neural processing leading to context- molecular identity of mCR remains elusive to this date.
appropriate behaviors.

2.07.5 Outlook
2.07.4.4 Identity of Membrane CORT Receptors
The role of the brainstem is pivotal for the manifestation of
CORT influences clasping by binding a G-protein-coupled
context-appropriate behaviors, yet we know very little about
receptor located at the cell surface (Haller et al., 2008; Orchinik
this highly conserved brain region in any vertebrate and
et al., 1994). There is conflicting evidence concerning the
even less in tetrapod vertebrates. Harnessing what is known
nature of this receptor: it could be a new membrane receptor
about the brainstem, sensorimotor control of vertebrate loco-
specific to CORT (Orchinik et al., 1991) or a kappa-opioid-
motion, and hormone effects on sex behaviors, we can better
like receptor (Bradford et al., 2005; Evans et al., 2000; Moore,
understand the evolutionary and proximal mechanisms that
2003). Early behavioral studies revealed that kappa agonists
drive sex behaviors specifically and behaviors involving
suppress clasping behavior in a dose-dependent manner
limb and torso in general. Using whole-cell electrophysiology
(Deviche and Moore, 1987), in a manner identical to CORT.
in concert with functional imaging, we can reveal previously
Additionally, in vitro assays showed that membrane CORT
unappreciated mechanisms that neural circuits and hormones
receptors (mCRs) recognize kappa-opioid receptor agonists
are using to modify behaviors in the rapid nongenomic time
and antagonists (Bradford et al., 2005; Evans et al., 2000).
frame. There is much to be learned through the lens of Taricha
Combined, these studies suggested that CORT and kappa
clasping behavior.
agonists might suppress clasping behavior via the same
pathway: an mCR-initiated upregulation of eCB signaling.
However, further behavioral pharmacological examination Acknowledgments
revealed that kappa agonists suppress clasping via a pathway
independent of eCB signaling, which is independent of the Gratitude to Dan Yaeger and Jim Rose for conversations about the
pathway activated by CORT. Pretreatment with a CB1 vertebrate brainstem. And thanks to Barbara Stebbins-Boaz and Jennifer
antagonist failed to block the kappa-mediated suppression of Butler for their editorial support. This work is funded by NSF (IOS-
clasping, whereas a typical kappa-opiate antagonist, nor-BNI, 1351129).
The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian 127

References Crosby, K.M., Inoue, W., Pittman, Q.J., Bains, J.S., 2011. Endocannabinoids gate
state-dependent plasticity of synaptic inhibition in feeding circuits. Neuron 71,
Adkins-Regan, E., 1990. Is the snark still a boojum? The comparative approach to 529–541.
reproductive behavior. Neurosci. Biobehav. Rev. 14, 243–252. Curtis, K.S., Krause, E.G., Contreras, R.J., 2002. Fos expression in non-
Adkins-Regan, E., 2009. Neuroendocrinology of social behavior. ILAR J. Natl. Res. catecholaminergic neurons in medullary and pontine nuclei after volume deple-
Council Inst. Lab. Anim. Resour. 50, 5–14. tion induced by polyethylene glycol. Brain Res. 948, 149–154.
Andreatta-Van Leyen, S., Averill, D.B., Guertzenstein, P.G., 1990. Cardiorespiratory Davis, A., Abraham, E., Mcevoy, E., Sonnenfeld, S., Lewis, C., Hubbard, C.S.,
effects induced by acetazolamide on the ventromedullary surface of the cat. Dolence, E.K., Rose, J.D., Coddington, E., 2015. Corticosterone suppresses
J. Physiol. 421, 171–184. vasotocin-enhanced clasping behavior in male rough-skinned newts by novel
Azim, E., Jiang, J., Alstermark, B., Jessell, T.M., 2014. Skilled reaching relies on mechanisms interfering with V1a receptor availability and receptor-mediated
a V2a propriospinal internal copy circuit. Nature 508, 357–363. endocytosis. Horm. Behav. 69, 39–49.
Bachmann, L.C., Matis, A., Lindau, N.T., Felder, P., Gullo, M., Schwab, M.E., Deviche, P., Moore, F.L., 1987. Opioid k-receptor agonists suppress sexual behaviors
2013. Deep brain stimulation of the midbrain locomotor region improves in male rough-skinned newts (Taricha granulosa). Horm. Behav. 21, 371–383.
paretic hindlimb function after spinal cord injury in rats. Sci. Transl. Med. 5 Di, S., Boudaba, C., Popescu, I.R., Weng, F.J., Harris, C., Marcheselli, V.L., Bazan, N.G.,
(208), ra146–208ra146. Tasker, J.G., 2005. Activity-dependent release and actions of endocannabinoids in
Ball, G.F., Balthazart, J., 2004. Hormonal regulation of brain circuits mediating male the rat hypothalamic supraoptic nucleus. J. Physiol. 569, 751–760.
sexual behavior in birds. Physio. Behav. 83, 329–346. Di, S., Malcher-Lopes, R., Halmos, K.C., Tasker, J.G., 2003. Nongenomic glucocor-
Bass, A.H., Gilland, E.H., Baker, R., 2008. Evolutionary origins for social vocalization in ticoid inhibition via endocannabinoid release in the hypothalamus: a fast feedback
a vertebrate hindbrain-spinal compartment. Science 321, 417–421. mechanism. J. Neurosci. 23, 4850–4857.
Bass, A.H., Grober, M.S., 2001. Social and neural modulation of sexual plasticity in Dominici, N., Ivanenko, Y.P., Cappellini, G., D’avella, A., Mondì, V., Cicchese, M.,
teleost fish. Brain Behav. Evol. 57, 293–300. Fabiano, A., Silei, T., Di Paolo, A., Giannini, C., Poppele, R.E., Lacquaniti, F., 2011.
Beach, F.A., 1967. Cerebral and hormonal control of reflexive mechanisms involved in Locomotor primitives in newborn babies and their development. Science 334,
copulatory behavior. Physiol. Rev. 47, 289–316. 997–999.
Bertsch, K., Böhnke, R., Kruk, M.R., Richter, S., Naumann, E., 2011. Exogenous Eaton, R.C., Lee, R.K., Foreman, M.B., 2001. The Mauthner cell and other identified
cortisol facilitates responses to social threat under high provocation. Horm. Behav. neurons of the brainstem escape network of fish. Prog. Neurobiol. 63, 467–485.
59, 428–434. Emerson, S.B., Boyd, S.K., 1999. Mating vocalizations of female frogs: control and
Bodznick, D., Northcutt, R.G., 1980. Segregation of electro- and mechanoreceptive evolutionary mechanisms. Brain Behav. Evol. 53, 187–197.
inputs to the elasmobranch medulla. Brain Res. 195, 313–321. Etgen, A.M., Barfield, R.J., 1986. Antagonism of female sexual behavior with intra-
Bowsher, D., Mallart, A., Petit, D., Albe-Fessard, D., 1968. A bulbar relay to the centre cerebral implants of antiprogestin RU 38486: correlation with binding to neural
median. J. Neurophysiol. 31, 288–300. progestin receptors. Endocrinology 119, 1610–1617.
Bradford, C.S., Walthers, E.A., Searcy, B.T., Moore, F.L., 2005. Cloning, heterologous Evans, S.J., Searcy, B.T., Moore, F.L., 2000. A subset of kappa opioid ligands bind to
expression and pharmacological characterization of a kappa opioid receptor from the membrane glucocorticoid receptor in an amphibian brain. Endocrinology 141,
the brain of the rough-skinned newt, Taricha granulosa. J. Mol. Endocrinol. 34, 2294–2300.
809–823. Evanson, N.K., Tasker, J.G., Hill, M.N., Hillard, C.J., Herman, J.P., 2010. Fast
Breuner, C.W., Greenberg, A.L., Wingfield, J.C., 1998. Noninvasive corticosterone feedback inhibition of the HPA axis by glucocorticoids is mediated by endo-
treatment rapidly increases activity in Gambel’s white-crowned sparrows (Zono- cannabinoid signaling. Endocrinology 151, 4811–4819.
trichia leucophrys gambelii). Gen. Comp. Endocrinol. 111, 386–394. Farham, C.J., Douglas, R.J., 1985. The response of neurons of the medial ponto-
Breuner, C.W., Wingfield, J.C., 2000. Rapid behavioral response to corticosterone medullary reticular formation of rats to peripheral thermal stimuli. Brain Res. 336,
varies with photoperiod and dose. Horm. Behav. 37, 23–30. 107–115.
Brown, R.E., Sergeeva, O.A., Eriksson, K.S., Haas, H.L., 2002. Convergent excitation Fetcho, J.R., 1992. The spinal motor system in early vertebrates and some of its
of dorsal raphe serotonin neurons by multiple arousal systems (orexin/hypocretin, evolutionary changes. Brain Behav. Evol. 40, 82–97.
histamine and noradrenaline). J. Neurosci. 22, 8850–8859. Finger, T.E., 2009. Evolution of gustatory reflex systems in the brainstems of fishes.
Buchanan, J.T., 2011. Flexibility in the patterning and control of axial locomotor Integr. Zool. Electron. 4, 53–63.
networks in lamprey. Integr. Comp. Biol. 51, 869–878. Fort, P., Luppi, P.H., Jouvet, M., 1994. Afferents to the nucleus reticularis parvi-
Cabelguen, J.-M., Bourcier-Lucas, C., Dubuc, R., 2003. Bimodal locomotion elicited by cellularis of the cat medulla oblongata: a tract-tracing study with cholera toxin B
electrical stimulation of the midbrain in the salamander Notophthalmus vir- subunit. J. Comp. Neurol. 342, 603–618.
idescens. J. Neurosci. 23, 2434–2439. Fukunaga, S., Setoguchi, S., Hirasawa, A., Tsujimoto, G., 2006. Monitoring ligand-
Cabelguen, J.-M., Ijspeert, A., Lamarque, S., Ryczko, D., 2010. Axial dynamics during mediated internalization of G protein-coupled receptor as a novel pharmacolog-
locomotion in vertebrates lesson from the salamander. Prog. Brain Res. 187, ical approach. Life Sci. 80, 17–23.
149–162. Garcia-Rill, E., Houser, C.R., Skinner, R.D., Smith, W., Woodward, D.J., 1987.
Carter, C.S., 1998. Neuroendocrine perspectives on social attachment and love. Locomotion-inducing sites in the vicinity of the pedunculopontine nucleus. Brain
Psychoneuroendocrinology 23, 779–818. Res. Bull. 18, 731–738.
Chevallier, S., Jan Ijspeert, A., Ryczko, D., Nagy, F., Cabelguen, J.-M., 2008. Orga- Garcia-Rill, E., Skinner, R.D., 1987. The mesencephalic locomotor region. I. Activation
nisation of the spinal central pattern generators for locomotion in the salamander: of a medullary projection site. Brain Res. 411, 1–12.
biology and modelling. Brain Res. Rev. 57, 147–161. Goodson, J.L., Bass, A.H., 2001. Social behavior functions and related anatomical
Coddington, E., Lewis, C., Rose, J.D., Moore, F.L., 2007. Endocannabinoids mediate characteristics of vasotocin/vasopressin systems in vertebrates. Brain Res. Brain
the effects of acute stress and corticosterone on sex behavior. Endocrinology 148, Res. Rev. 35, 246–265.
493–500. Goodson, J.L., Bass, A.H., 2002. Vocal-acoustic circuitry and descending vocal
Coddington, E., Moore, F.L., 2003. Neuroendocrinology of context-dependent stress pathways in teleost fish: convergence with terrestrial vertebrates reveals conserved
responses: vasotocin alters the effect of corticosterone on amphibian behaviors. traits. J. Comp. Neurol. 448, 298–322.
Horm. Behav. 43, 222–228. Grill, H.J., Hayes, M.R., 2012. Hindbrain neurons as an essential hub in the neuro-
Coimbra, N.C., De Oliveira, R., Freitas, R.L., Ribeiro, S.J., Borelli, K.G., Pacagnella, R.C., anatomically distributed control of energy balance. Cell Metab. 16, 296–309.
Moreira, J.E., Da Silva, L.A., Melo, L.L., Lunardi, L.O., Brandão, M.L., 2006. Grillner, S., 2006. Biological pattern generation: the cellular and computational logic of
Neuroanatomical approaches of the tectum-reticular pathways and immunohisto- networks in motion. Neuron 52, 751–766.
chemical evidence for serotonin-positive perikarya on neuronal substrates of the Grillner, S., 2011. Human locomotor circuits conform. Science 334, 912–913.
superior colliculus and periaqueductal gray matter involved in the elaboration of the Grillner, S., Wallen, P., 1985. Central pattern generators for locomotion, with special
defensive behavior and fear-induced analgesia. Exp. Neurol. 197, 93–112. reference to vertebrates. Annu. Rev. Neurosci. 8, 233–261.
Crews, D., Moore, M.C., 2005. Historical contributions of research on reptiles to Grillner, S., Wallén, P., Saitoh, K., Kozlov, A., Robertson, B., 2008. Neural bases of
behavioral neuroendocrinology. Horm. Behav. 48, 384–394. goal-directed locomotion in vertebratesdan overview. Brain Res. Rev. 57, 2–12.
Crosby, K.M., Bains, J.S., 2012. The intricate link between glucocorticoids and Guilbaud, G., Besson, J.M., Oliveras, J.L., Wyon-Maillard, M.C., 1973. Modifications of
endocannabinoids at stress-relevant synapses in the hypothalamus. Neuroscience the firing rate of bulbar reticular units (nucleus gigantocellularis) after intra-arterial
204, 31–37. injection of bradykinin into the limbs. Brain Res. 63, 131–140.
128 The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian

Guyenet, P.G., Mulkey, D.K., Stornetta, R.L., Bayliss, D.A., 2005. Regulation of ventral Lenser, S., Bruce, J., Veloso, M., 2001. A Modular Hierarchical Behavior-based
surface chemoreceptors by the central respiratory pattern generator. J. Neurosci. Architecture. Springer Berlin Heidelberg, Berlin, Heidelberg.
25, 8938–8947. Levitis, D.A., Lidicker Jr., W.Z., Freund, G., 2009. Behavioural biologists do not agree
Haller, J., Halasz, J., Makara, G.B., Kruk, M.R., 1998. Acute effects of glucocorticoids: on what constitutes behaviour. Anim. Behav. 78, 103–110.
behavioral and pharmacological perspectives. Neurosci. Biobehav. Rev. 23, Lewis, C.M., Dolence, E.K., Hubbard, C.S., Rose, J.D., 2005. Identification of
337–344. roughskin newt medullary vasotocin target neurons with a fluorescent vasotocin
Haller, J., Mikics, É., Makara, G.B., 2008. The effects of non-genomic glucocorticoid conjugate. J. Comp. Neurol. 491, 381–389.
mechanisms on bodily functions and the central neural system. A critical evaluation Lewis, C.M., Dolence, E.K., Zhang, Z., Rose, J.D., 2004. Fluorescent vasotocin
of findings. Front. Neuroendocrinol. 29, 273–291. conjugate for identification of the target cells for brain actions of vasotocin. Bio-
Haxhiu, M.A., Loewy, A.D., 1996. Central connections of the motor and sensory vagal conjugate Chem. 15, 909–914.
systems innervating the trachea. J. Aut. Nerv. Syst. 57, 49–56. Lewis, C.M., Rose, J.D., 2003. Rapid corticosterone-induced impairment of amplectic
Heifets, B.D., Castillo, P.E., 2009. Endocannabinoid signaling and long-term synaptic clasping occurs in the spinal cord of roughskin newts (Taricha granulosa). Horm.
plasticity. Annu. Rev. Physiol. 71, 283–306. Behav. 43, 93–98.
Hill, M.N., 2012. Introduction to the special issue on stress, emotional behavior, and Lombana, K., Middleton, N., Coddington, E., 2015. Suppression of sex behavior by
the endocannabinoid system: a decade of research. Neuroscience 204, 1–4. kappa opiates and stress steroids occurs via independent neuroendocrine path-
Hill, M.N., Karatsoreos, I.N., Hillard, C.J., Mcewen, B.S., 2010a. Rapid elevations in ways. Gen. Comp. Endocrinol. 210, 81–86.
limbic endocannabinoid content by glucocorticoid hormones in vivo. Psychoneur- Lowry, C.A., Rose, J.D., Moore, F.L., 1996. Corticotropin-releasing factor enhances
oendocrinology 35, 1333–1338. locomotion and medullary neuronal firing in an amphibian. Horm. Behav. 30,
Hill, M.N., Patel, S., Campolongo, P., Tasker, J.G., Wotjak, C.T., Bains, J.S., 2010b. 50–59.
Functional interactions between stress and the endocannabinoid system: from Lutterschmidt, D.I., Maine, A.R., 2014. Sex or candy? Neuroendocrine regulation of the
synaptic signaling to behavioral output. J. Neurosci. 30, 14980–14986. seasonal transition from courtship to feeding behavior in male red-sided garter
Hill, M.N., Tasker, J.G., 2012. Endocannabinoid signaling, glucocorticoid-mediated snakes (Thamnophis sirtalis parietalis). Horm. Behav. 66, 120–134.
negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis. Machne, X., Calma, I., Magoun, H.W., 1955. Unit activity of central cephalic brain stem
Neuroscience 204, 5–16. in EEG arousal. J. Neurophysiol. 18, 547–558.
Hollis, D.M., Coddington, E.J., Moore, F.L., 2006. Neuroanatomical distribution of Marder, E., Bucher, D., 2001. Central pattern generators and the control of rhythmic
cannabinoid receptor gene expression in the brain of the rough-skinned newt, movements. Curr. Biol. 11, R986–R996.
Taricha granulosa. Brain Behav. Evol. 67, 135–149. Marder, E., O’leary, T., Shruti, S., 2014. Neuromodulation of circuits with variable
Hubbard, C.S., Dolence, E.K., Rose, J.D., 2010. Brainstem reticulospinal neurons are parameters: single neurons and small circuits reveal principles of state-dependent
targets for corticotropin-releasing factor-Induced locomotion in roughskin newts. and robust neuromodulation. Annu. Rev. Neurosci. 37, 329–346.
Horm. Behav. 57, 237–246. Marder, E., Prinz, A.A., 2003. Current compensation in neuronal homeostasis. Neuron
Hull, E.M., Dominguez, J.M., 2007. Sexual behavior in male rodents. Horm. Behav. 52, 37, 2–4.
45–55. Marsicano, G., Lafenetre, P., 2009. Roles of the endocannabinoid system in learning
Humphries, M.D., Gurney, K., Prescott, T.J., 2007. Is there a brainstem substrate for and memory. Curr. Top. Behav. Neurosci. 1, 201–230.
action selection? Philos. Trans. R. Soc. B Biol. Sci. 362, 1627–1639. Marsicano, G., Wotjak, C.T., Azad, S.C., Bisogno, T., Rammes, G., Cascio, M.G.,
Hunter, R.G., Gagnidze, K., Mcewen, B.S., Pfaff, D.W., 2015. Stress and the dynamic Hermann, H., Tang, J., Hofmann, C., Zieglgansberger, W., Di Marzo, V., Lutz, B.,
genome: steroids, epigenetics, and the transposome. Proc. Natl. Acad. Sci. U.S.A. 2002. The endogenous cannabinoid system controls extinction of aversive
112, 6828–6833. memories. Nature 418, 530–534.
Iwata, T., Toyoda, F., Yamamoto, K., Kikuyama, S., 2000. Hormonal control of urodele Martin, E.M., Devidze, N., Shelley, D.N., Westberg, L., Fontaine, C., Pfaff, D.W.,
reproductive behavior. Comp. Biochem. Physiol. Part B 126, 221–229. 2011. Molecular and neuroanatomical characterization of single neurons in the
Joëls, M., Van Riel, E., 2004. Mineralocorticoid and glucocorticoid receptor-mediated mouse medullary gigantocellular reticular nucleus. J. Comp. Neurol. 519,
effects on serotonergic transmission in health and disease. Ann. N.Y. Acad. Sci. 2574–2593.
1032, 301–303. Martin, E.M., Pavlides, C., Pfaff, D., 2010. Multimodal sensory responses of nucleus
Jordan, L.M., 1998. Initiation of locomotion in mammals. Ann. N.Y. Acad. Sci. 860, reticularis gigantocellularis and the responses’ relation to cortical and motor
83–93. activation. J. Neurophysiol. 103, 2326–2338.
Jordan, L.M., Liu, J., Hedlund, P.B., Akay, T., Pearson, K.G., 2008. Descending Matsuyama, K., Mori, F., Nakajima, K., Drew, T., Aoki, M., Mori, S., 2004a. Locomotor
command systems for the initiation of locomotion in mammals. Brain Res. Rev. 57, role of the corticoreticular-reticulospinal-spinal interneuronal system. Prog. Brain
183–191. Res. 143, 239–249.
Kaddumi, E.G., Hubscher, C.H., 2006. Convergence of multiple pelvic organ inputs in Matsuyama, K., Nakajima, K., Mori, F., Aoki, M., Mori, S., 2004b. Lumbar commis-
the rat rostral medulla. J. Physiol. 572, 393–405. sural interneurons with reticulospinal inputs in the cat: morphology and discharge
Katz, P.S., 1999. Beyond Neurotransmission: Neuromodulation and its Importance for patterns during fictive locomotion. J. Comp. Neurol. 474, 546–561.
Information Processing. Oxford University Press, New York. McCrea, D.A., Rybak, I.A., 2008. Organization of mammalian locomotor rhythm and
Kelley, D.B., Pfaff, D.W., 1976. Hormone effects on male sex behavior in adult South pattern generation. Brain Res. Rev. 57, 134–146.
African clawed frogs, Xenopus laevis. Horm. Behav. 7, 159–182. McEwen, B.S., Gianaros, P.J., 2011. Stress- and allostasis-induced brain plasticity.
Kimura, Y., Satou, C., Fujioka, S., Shoji, W., Umeda, K., Ishizuka, T., Yawo, H., Annu. Rev. Med. 62, 431–445.
Higashijima, S.I., 2013. Hindbrain V2a neurons in the excitation of spinal locomotor Meijer, O.C., Kortekaas, R., Oitzl, M.S., De Kloet, E.R., 1998. Acute rise in cortico-
circuits during zebrafish swimming. Curr. Biol. 23, 843–849. sterone facilitates 5-HT(1A) receptor-mediated behavioural responses. Eur. J.
Korn, H., Faber, D.S., 1996. Escape behavior - brainstem and spinal cord circuitry and Pharmacol. 351, 7–14.
function. Curr. Opin. Neurobiol. 6, 826–832. Mesce, K.A., 2002. Metamodulation of the biogenic amines: second-order modulation
Korn, H., Faber, D.S., 2005. The Mauthner cell half a century later: a neurobiological by steroid hormones and amine cocktails. Brain Behav. Evol. 60, 339–349.
model for decision-making? Neuron 47, 13–28. Mikics, É., Kruk, M.R., Haller, J., 2004. Genomic and non-genomic effects of
Kozlov, A.K., Kardamakis, A.A., Kotaleski, J.H., Grillner, S., 2014. Gating of steering glucocorticoids on aggressive behavior in male rats. Psychoneuroendocrinology 29,
signals through phasic modulation of reticulospinal neurons during locomotion. 618–635.
Proc. Natl. Acad. Sci. U.S.A. 111, 3591–3596. Milam, E.L., Millstein, R.L., Potochnik, A., Roughgarden, J.E., 2011. Sex and sensi-
Kreitzer, A.C., Regehr, W.G., 2001. Cerebellar depolarization-induced suppression of bility: the role of social selection. Metascience 20 (2), 253–277.
inhibition is mediated by endogenous cannabinoids. J. Neurosci. 21, RC174. Mitchell, E.J., Mccallum, S., Dewar, D., Maxwell, D.J., 2016. Corticospinal and
Ladpli, R., Brodal, A., 1968. Experimental studies of commissural and reticular reticulospinal contacts on cervical commissural and long descending propriospinal
formation projections from the vestibular nuclei in the cat. Brain Res. 8, 65–96. neurons in the adult rat spinal cord; evidence for powerful reticulospinal connec-
Lawrence, D.G., Kuypers, H.G., 1968a. The functional organization of the motor tions. PLoS One 11, e0152094.
system in the monkey. I. The effects of bilateral pyramidal lesions. Brain 91, Moore, F.L., 2003. Evidence that a Membrane Corticosteroid Receptor Is an Opioid-
1–14. like Receptor. The Identities of Membrane Steroid Receptors Boston. Springer
Lawrence, D.G., Kuypers, H.G., 1968b. The functional organization of the motor US, MA.
system in the monkey. II. The effects of lesions of the descending brain-stem Moore, F.L., Boyd, S.K., Kelley, D.B., 2005. Historical perspective: hormonal regulation
pathways. Brain 91, 15–36. of behaviors in amphibians. Horm. Behav. 48, 373–383.
The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian 129

Moore, F.L., Miller, L.J., 1984. Stress-induced inhibition of sexual behavior: cortico- Rose, J.D., Marrs, G.S., Moore, F.L., 1998. Rapid, corticosterone-induced disruption
sterone inhibits courtship behaviors of a male amphibian (Taricha granulosa). of medullary sensorimotor integration related to suppression of amplectic
Horm. Behav. 18, 400–410. clasping in behaving roughskin newts (Taricha granulosa). Horm. Behav. 34,
Moore, F.L., Rose, J.D., 2002. Sensorimotor processing model: how vasotocin and 268–282.
corticosterone interact and control reproductive behaviors in an amphibian. In: Rose, J.D., Moore, F.L., 1999. A neurobehavioral model for rapid actions of cortico-
Pfaff, D.W. (Ed.), Hormones, first ed. sterone on sensorimotor integration. Steroids 64, 92–99.
Moore, F.L., Wood, R.E., Boyd, S.K., 1992. Sex steroids and vasotocin interact in Rose, J.D., Moore, F.L., 2002. Behavioral neuroendocrinology of vasotocin and
a female amphibian (Taricha granulosa) to elicit female-like egg-laying behavior or vasopressin and the sensorimotor processing hypothesis. Front. Neuroendocrinol.
male-like courtship. Horm. Behav. 26, 156–166. 23, 317–341.
Moore, F.L., Zoeller, R.T., 1979. Endocrine control of amphibian sexual behavior: Rose, J.D., Moore, F.L., Orchinik, M., 1993. Rapid neurophysiological effects of
evidence for a neurohormone-androgen interaction. Horm. Behav. 13, 207–213. corticosterone on medullary neurons: relationship to stress-induced suppression of
Moore, F.L., Zoeller, R.T., 1985. Stress-induced inhibition of reproduction: evidence of courtship clasping in an amphibian. Neuroendocrinology 57, 815–824.
suppressed secretion of LH-RH in an amphibian. Gen. Comp. Endocrinol. 60, Rose, J.D., Sutin, J., 1973. Responses of single units in the medulla to genital
252–258. stimulation in estrous and anestrous cats. Brain Res. 50, 87–99.
Moralí, G., Asunción Pía Soto, M., Luis Contreras, J., Arteaga, M., González- Roughgarden, J., 2012. The social selection alternative to sexual selection. Philos.
Vidal, M.D., Beyer, C., 2003. Detailed analysis of the male copulatory motor Trans. R. Soc. Lond. Ser. B Biol. Sci. 367, 2294–2303.
pattern in mammals: hormonal bases. Scand. J. Psychol. 44, 279–288. Roughgarden, J., Oishi, M., Akçay, E., 2006. Reproductive social behavior: cooperative
Murakami, Y., Pasqualetti, M., Takio, Y., Hirano, S., Rijli, F.M., Kuratani, S., 2004. games to replace sexual selection. Science 311, 965–969.
Segmental development of reticulospinal and branchiomotor neurons in lamprey: Ryczko, D., Auclair, F., Cabelguen, J.-M., Dubuc, R., 2016. The mesencephalic
insights into the evolution of the vertebrate hindbrain. Development 131, locomotor region sends a bilateral glutamatergic drive to hindbrain reticulospinal
983–995. neurons in a tetrapod. J. Comp. Neurol. 524, 1361–1383.
Naujoks Manteuffel, C., Manteuffel, G., 1988. Origins of descending projections to the Ryczko, D., Dubuc, R., 2013. The multifunctional mesencephalic locomotor region.
medulla oblongata and rostral medulla spinalis in the urodele Salamandra sala- Curr. Pharm. Des. 19, 4448–4470.
mandra (amphibia). J. Comp. Neurol. 273, 187–206. Ryczko, D., Dubuc, R., Cabelguen, J.-M., 2010. Rhythmogenesis in axial locomotor
Newman, D.B., Cruce, W.L., Bruce, L.L., 1983. The sources of supraspinal afferents to networks: an interspecies comparison. Prog. Brain Res. 187, 189–211.
the spinal cord in a variety of limbed reptiles. I. Reticulospinal systems. J. Comp. Saadé, N.E., Salibi, N.A., Banna, N.R., Towe, A.L., Jabbur, S.J., 1983. Spinal input
Neurol. 215, 17–32. pathways affecting the medullary gigantocellular reticular nucleus. Exp. Neurol. 80,
Oda, Y., 2015. Escape behaviors and its underlying neuronal circuits. Brain Nerve 582–600.
Shinkei kenkyu no shinpo 67, 1173–1183. Sakai, S.T., Davidson, A.G., Buford, J.A., 2009. Reticulospinal neurons in the pon-
Orchinik, M., Moore, F.L., Rose, J.D., 1994. Mechanistic and functional studies of tomedullary reticular formation of the monkey (Macaca fascicularis). Neuroscience
rapid corticosteroid actions. Ann. N.Y. Acad. Sci. 746, 101–112. 163, 1158–1170.
Orchinik, M., Murray, T.F., Moore, F.L., 1991. A corticosteroid receptor in neuronal Sánchez-Camacho, C., Marín, O., Ten Donkelaar, H.J., González, A., 2001.
membranes. Science 252, 1848–1851. Descending supraspinal pathways in amphibians. I. A dextran amine tracing study
Orlovskiĭ, G.N., 1970a. Relations between reticulo-spinal neurons and locomotor of their cells of origin. J. Comp. Neurol. 434, 186–208.
regions of the brain stem. Biofizika 15, 171–178. Searcy, B.T., Bradford, C.S., Thompson, R.R., Filtz, T.M., Moore, F.L., 2011. Identi-
Orlovskiĭ, G.N., 1970b. Work of reticulo-spinal neurons during locomotion. Biofizika 15, fication and characterization of mesotocin and V1a-like vasotocin receptors in
728–737. a urodele amphibian, Taricha granulosa. Gen. Comp. Endocrinol. 170, 131–143.
Panzica, G.C., Aste, N., Castagna, C., Viglietti-Panzica, C., Balthazart, J., 2001. Shelley, D.N., Choleris, E., Kavaliers, M., Pfaff, D.W., 2006. Mechanisms underlying
Steroid-induced plasticity in the sexually dimorphic vasotocinergic innervation of sexual and affiliative behaviors of mice: relation to generalized CNS arousal. Soc.
the avian brain: behavioral implications. Brain Res. Brain Res. Rev. 37, Cogn. Affect. Neurosci. 1, 260–270.
178–200. Sherrington, C.S., 1910. Flexion-reflex of the limb, crossed extension-reflex, and reflex
Peterson, B.W., Abzug, C., 1975. Properties of projections from vestibular nuclei to stepping and standing. J. Physiol. 40, 28–121.
medial reticular formation in the cat. J. Neurophysiol. 38, 1421–1435. Shik, M.L., Severin, F.V., Orlovskiĭ, G.N., 1967. Structures of the brain stem
Pfaff, D.W., Martin, E.M., Faber, D., 2012. Origins of arousal: roles for medullary responsible for evoked locomotion. Fiziologicheskii zhurnal SSSR Im. I. M.
reticular neurons. Trends Neurosci. 35, 468–476. Sechenova 53, 1125–1132.
Pfaus, J.G., Kippin, T.E., Centeno, S., 2001. Conditioning and sexual behavior: Siegel, J.M., 1979. Behavioral functions of the reticular formation. Brain Res. 180, 69–105.
a review. Horm. Behav. 40, 291–321. Sillar, K.T., 2009. Mauthner cells. Curr. Biol. 19, R353–R355.
Propper, C.R., 1991. Courtship in the rough-skinned newt Taricha granulosa. Anim. Silver, W.L., Finger, T.E., 2009. The anatomical and electrophysiological basis of
Behav. 41, 547–554. peripheral nasal trigeminal chemoreception. Ann. N.Y. Acad. Sci. 1170,
Putman, P., Hermans, E.J., Koppeschaar, H., Van Schijndel, A., Van Honk, J., 2007. 202–205.
A single administration of cortisol acutely reduces preconscious attention for fear in Sirota, M.G., Di Prisco, G.V., Dubuc, R., 2000. Stimulation of the mesencephalic
anxious young men. Psychoneuroendocrinology 32, 793–802. locomotor region elicits controlled swimming in semi-intact lampreys. Eur. J.
Rankin, J., Walker, J.J., Windle, R., Lightman, S.L., Terry, J.R., 2012. Characterizing Neurosci. 12, 4081–4092.
dynamic interactions between ultradian glucocorticoid rhythmicity and acute stress Smetana, R., Juvin, L., Dubuc, R., Alford, S., 2010. A parallel cholinergic brainstem
using the phase response curve. PLoS One 7, e30978. pathway for enhancing locomotor drive. Nat. Publ. Group 13, 731–738.
Remage-Healey, L., ̄ Bass, A.H., 2006. A rapid neuromodulatory role for steroid Soderstrom, K., 2009. Lessons from nonmammalian species. Curr. Top. Behav.
hormones in the control of reproductive behavior. Brain Res. 1126, 27–35. Neurosci. 1, 173–198.
Riebe, C.J., Wotjak, C.T., 2011. Endocannabinoids and stress. Stress 14, Soderstrom, K., Leid, M., Moore, F.L., Murray, T.F., 2000. Behavioral, pharmaco-
384–397. logical, and molecular characterization of an amphibian cannabinoid receptor.
Roberts, A., Alford, S.T., 1986. Descending projections and excitation during fictive J. Neurochem. 75, 413–423.
swimming in Xenopus embryos: neuroanatomy and lesion experiments. J. Comp. Taber, K.H., Hurley, R.A., 2009. Endocannabinoids: stress, anxiety, and fear.
Neurol. 250, 253–261. J. Neuropsychiatry Clin. Neurosci. 21, iv-109–113.
Rose, J.D., 1990a. Brainstem influences on sexual behavior. In: Klemm, W., Vertes, R. Tasker, J.G., Di, S., Malcher-Lopes, R., 2006. Minireview: rapid glucocorticoid
(Eds.), Brainstem Mechanisms of Behavior. JW Wiley and Sons, New York. signaling via membrane-associated receptors. Endocrinology 147, 5549–5556.
Rose, J.D., 1990b. Forebrain influences on brainstem and spinal mechanisms of Ten Donkelaar, H.J., 1990. Brainstem mechanisms of behavior: comparative aspects.
copulatory behavior: a current perspective on Frank Beach’s contribution. Neurosci. In: Klemm, W., Vertes, R. (Eds.), Brainstem Mechanisms of Behavior.
Biobehav. Rev. 14, 207–215. Thompson, R.R., Dickinson, P.S., Rose, J.D., Dakin, K.A., Civiello, G.M., Segerdahl, A.,
Rose, J.D., 2000. Corticosteroid actions from neuronal membrane to behavior: Bartlett, R., 2008. Pheromones enhance somatosensory processing in newt brains
neurophysiological mechanisms underlying rapid behavioral effects of corticoste- through a vasotocin-dependent mechanism. Proc. R. Soc. B Biol. Sci. 275,
rone. Biochem. Cell Biol. 78, 307–315. 1685–1693.
Rose, J.D., Kinnaird, J.R., Moore, F.L., 1995. Neurophysiological effects of vasotocin Thompson, R.R., Moore, F.L., 2000. Vasotocin stimulates̆ appetitive responses to the
and corticosterone on medullary neurons: implications for hormonal control of visual and pheromonal stimuli used by male roughskin newts during courtship.
amphibian courtship behavior. Neuroendocrinology 62, 406–417. Horm. Behav. 38, 75–85.
130 The Vertebrate Brain Stem as the Primary Site of Behavior Command: Insights from an Amphibian

Tresch, M.C., 2010. A neural system for boosting locomotion. Nat. Publ. Group 13, Wilson, R.I., Nicoll, R.A., 2001. Endogenous cannabinoids mediate retrograde sig-
659–660. nalling at hippocampal synapses. Nature 410, 588–592.
Vertes, R.P., Miller, N.E., 1976. Brain stem neurons that fire selectively to a condi- Yang, C.F., Shah, N.M., 2014. Representing sex in the brain, one module at a time.
tioned stimulus for shock. Brain Res. 103, 229–242. Neuron 82, 261–278.
Von Zastrow, M., Williams, J.T., 2012. Modulating neuromodulation by receptor Young, L.J., Lim, M.M., Gingrich, B., Insel, T.R., 2001. Cellular mechanisms of social
membrane traffic in the endocytic pathway. Neuron 76, 22–32. attachment. Horm. Behav. 40, 133–138.
Whelan, P.J., 1996. Control of locomotion in the decerebrate cat. Prog. Neurobiol. 49, Zottoli, S.J., Faber, D.S., 2000. Review: the Mauthner cell: what has it taught us?
481–515. Neurosci. 6 (1), 26–38.
Wieffer, M., Maritzen, T., Haucke, V., 2009. Snapshot: endocytic trafficking. Cell 137, Zuk, M., 2006. Family values in black and white. Nature 439, 917.
382.e1–e3.
Willis, W.D., 1985. Nociceptive pathways: anatomy and physiology of nociceptive
ascending pathways. Philos. Trans. R. Soc. B Biol. Sci. 308, 253–270.
2.08 Hormones and Vocal Systems: Insights from Xenopus
Erik Zornik, Reed College, Portland, OR, USA
Darcy B Kelley, Columbia University, New York, NY, USA
Ó 2017 Elsevier Inc. All rights reserved.
This chapter is a revision of the previous edition chapter by E.J. Yang, D.B. Kelley, volume 1, pp. 693–706, Ó 2009, Elsevier Inc.

2.08.1 Introduction 131


2.08.2 Mating and Vocal Behaviors 132
2.08.2.1 Vocalizations Depend on Social Context 132
2.08.2.1.1 Male Advertisement Calls 132
2.08.2.1.2 Female Unreceptive Calls 132
2.08.2.1.3 Female Rapping and Male Answer Call 132
2.08.2.1.4 Amplectant Call 132
2.08.2.1.5 Male–Male Interaction Calls 132
2.08.2.2 Clasping, Reproductive Status, and Social Hierarchies 134
2.08.2.2.1 Female Receptivity 134
2.08.2.2.2 Male–Female Clasping 134
2.08.2.2.3 Male–Male Clasping 134
2.08.2.2.4 Vocal Dominance 135
2.08.2.3 Hormone-Dependence of Vocal Behaviors 135
2.08.2.3.1 Advertisement Calls 135
2.08.2.3.2 Female Calling 135
2.08.2.3.3 Male Interaction Calls 137
2.08.2.3.4 Insights from Endocrine Disrupting Compounds 137
2.08.3 Sex Differences and Hormone Targets in the Vocal System 137
2.08.3.1 Sex Differences and Hormone Targets in the Larynx 137
2.08.3.2 Sex Differences and Hormone Targets in the Vocal Circuit 137
2.08.3.2.1 Motor Circuit 137
2.08.3.2.2 Auditory Pathways 138
2.08.4 Generating Sexually Differentiated Vocalizations 138
2.08.4.1 Generating Fast and Slow Calls 138
2.08.4.1.1 Larynx 138
2.08.4.1.2 Central Pattern Generator 139
2.08.4.2 Generating Complex and Simple Vocal Patterns 140
2.08.4.2.1 Biphasic versus Monophasic Calling 140
2.08.4.2.2 Intensity Modulation of Vocal Trills 140
2.08.5 Processing Vocal Signals 141
2.08.5.1 Sexually Differentiated Auditory Processing 141
2.08.5.2 Generating Socially Appropriate Vocal Responses 142
2.08.6 Evolution of Vocal Effectors in Xenopus 142
2.08.7 Conclusion 142
References 143

2.08.1 Introduction mechanisms at each level must be complemented with studies


that integrate across mechanism and level of organization.
The stunning diversity of patterned movements that make up In this chapter, we review research on the sexually differen-
behavioral repertoires are governed by neural circuits at tiated vocal behaviors of South African clawed frogs, Xenopus
multiple levels and supported by specialized features of muscle laevis. The Xenopus vocal system is unusual because Xenopus is
effectors. In vertebrates, behaviors that are specific to each sex entirely aquatic throughout its life cycle; vocalization is
are also regulated, during development and in adulthood, by produced under water and is not powered by air movement
the endocrine signals that control reproduction. The complex (breathing). The essential involvement of breathing circuits in
nature of hormone action, with multiple targets and tempo- terrestrial vertebrates greatly complicates the study of hind-
rally distinct but overlapping sensitive periods, creates a major brain control of vocalization (McLean et al., 2013; Tupal
challenge in deciphering the causal relevance of hormone et al., 2014). Vocalization without breathing in Xenopus
signaling at each level of control. To overcome this hurdle, provides many experimental advantages that include two
investigations of the physiological, cellular, and molecular powerful reduced experimental preparations: the isolated

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00023-7 131


132 Hormones and Vocal Systems: Insights from Xenopus

(ex vivo) larynx and the isolated brain. ‘Fictive’ vocal behaviors by a single male in field recordings (Tobias et al., 2010). The
can be evoked in either preparation independently of the other male advertisement call also attracts gravid females (Picker,
(Tobias and Kelley, 1987; Rhodes et al., 2007). The ex vivo 1983) and evokes a specific call, rapping (Figure 1(b); Tobias
larynx preparation has shed light on how the periphery et al., 1998b).
constrains sound production, as well as on how hormones
support the production of sex-specific behaviors (Zornik and 2.08.2.1.2 Female Unreceptive Calls
Kelley, 2011). The ex vivo brain preparation has provided During the breeding season, most female X. laevis are sexually
important insights into the essential components of the vocal unreceptive at any given time. When clasped by a male, nongra-
central pattern generator (CPG) and the neural circuits that vid females produce a slow series of sound pulses at a rate of 4–
generate different vocal rhythms (Rhodes et al., 2007; Yu and 8 Hz called ticking (Figure 1(c)). Unlike male advertisement
Yamaguchi, 2009, 2010; Yamaguchi et al., 2010; Zornik and calling, the sound pulses in ticking are spectrally broad, with
Yamaguchi, 2012). In vivo studies of intact animals comple- a single DF around 1.2 kHz (Figure 2). The temporal pattern
ment findings ex vivo. Lesion studies, combined with the is simple, consisting of an ongoing, monotonous trill, and
ex vivo brain preparation, have provided insight into forebrain pulses do not vary systematically in intensity as the call prog-
mechanisms that govern context-dependent social signaling resses (Tobias et al., 1998b). Nongravid females also extend
(Hall et al., 2013). Finally, recent advances in understanding their hindlimbs when clasped (Kelley, 1982). When males
auditory processing will facilitate future integrative syntheses are exposed to playbacks of female unreceptive calls during
of sensory, sensorimotor, and motor levels of control (Elliott advertisement calling, they exhibit temporary vocal suppres-
et al., 2011; Hall et al., 2016). sion (Tobias et al., 2004; Elliott and Kelley, 2007).

2.08.2.1.3 Female Rapping and Male Answer Call


2.08.2 Mating and Vocal Behaviors
Female X. laevis also advertise for mates using the rapping call.
Rapping can be evoked in gravid females by advertisement
In X. laevis, the most extensively studied Xenopus species,
calls, and pairs of sexually receptive males and females produce
mating occurs over a prolonged period (6 months), and
rapping/advertisement calling duets (Tobias et al., 1998b).
only a small percentage of females are sexually receptive at
Rapping trill rates average 12 Hz. Playbacks of rapping evoke
any given time (Tobias et al., 1998b). Because these fully
advertisement and answer calling. The answer call, a modified
aquatic frogs mate under water at night and typically live in
advertisement call, contains elongated fast trills, shortened
turbid bodies of water with many conspecifics, vocal signals
slow trills, and enhanced intensity modulation (Figure 1(b)).
drive courtship as they are well suited to locating a suitable
mate. Each call is made up of brief (5–10 ms; Vignal and Kel-
ley, 2007) sound pulses produced with different temporal 2.08.2.1.4 Amplectant Call
patterns. Eight unique call types have been described (Figure 1), Upon clasping a female or another male (amplexus), males
and each can be distinguished through a combination of the generate a low-intensity amplectant call consisting of two to
pulse rate, duration, and period (repetition rate) of each trill, three sound pulses at 150 ms intervals (Figure 1(d); Picker,
as well as the pulse intensity (loudness) and frequency (pitch). 1980; Zornik and Kelley, 2008). These sounds coincide with
male forelimb contraction and ventrally directed head move-
ments, potential signals for synchronizing egg and sperm release.
2.08.2.1 Vocalizations Depend on Social Context
2.08.2.1.1 Male Advertisement Calls 2.08.2.1.5 Male–Male Interaction Calls
The male advertisement call (Figure 1(a)) is the most spectrally All of the six call types given by males can be recorded from
and temporally complex in the X. laevis repertoire. It can be male/male pairs; three (chirping, growling, and male ticking)
divided broadly into two phases: the fast trill and the slow trill. are directed exclusively toward males (Tobias et al., 2004).
Fast and slow trills alternate during calling bouts. Across the Chirping, a brief (5 pulses) 60–80 Hz burst repeated
fast phase (60 Hz; 200–300 ms duration), sound pulses approximately four times per second (Figure 1(e)), can be
increase in amplitude. For the slow phase, sound pulses are recorded as one male approaches another and is reliably
produced at 30 Hz; several ‘loud slow’ pulses often precede produced by the clasping male (Tobias et al., 2004) as well
the longer-duration (700–800 ms) slow trills. Fast trill sound as in response to playbacks of synthetic male-like calls (Vignal
pulses contain two dominant frequencies (DF1: 1.9 kHz; and Kelley, 2007). Chirping could be a male–male aggressive
DF2: 2.2 kHz), while slow trill pulses contain a third, lower signal, though as noted above, advertisement calls suffice for
DF 1.2 kHz (Figure 2). vocal suppression.
During the peak reproductive season (August in Cape Town, In male/male pairs, the clasped male produces growling,
South Africa), the male-specific advertisement call can be a rapid, variable-length trill (Figure 1(e); 40–80 Hz Tobias
recorded using an underwater microphone for prolonged et al., 2004). Unlike the sound pulses of other male calls,
periods (>45 min; Tobias et al., 2004). In the lab, males also growling sound pulses have a lower DF around 1 kHz (Tobias
call for prolonged periods when alone. When paired with et al., 2004).
a conspecific, males switch from advertisement to answer Male ticking is a rare call, and it is not clear whether it is eli-
calling (Figure 1(b)). In the laboratory, broadcasts of advertise- cited by amplexus, and if so, whether the clasper or claspee
ment calls suppress calling by other males (Tobias et al., 2004) produces the call (Tobias et al., 2004). The sound pulse rate
accounting for vocal dominance and the prevalence of calling in male ticking is 4 Hz, as in female ticking (Figure 1(c)),
Hormones and Vocal Systems: Insights from Xenopus 133

Male calls Female calls

Advertisement calling
(a)

Fast trill Slow trill


500 ms

(b)
Answer calling Rapping

Fast trill Slow trill 500 ms

(c) Ticking

500 ms

Amplectant calling
(d)

500 ms

Chirping

(e)

250 ms
Growling

250 ms

Figure 1 The social context-dependent Xenopus laevis vocal repertoire. (a) Male X. laevis produce advertisement calls in isolation, as well as in the
presence of conspecific females and males. Advertisement calls are temporally complex. Sound pulses occur in alternating trills: fast trill pulses are
produced at a rate of 60 Hz, while slow trill pulse rates average 30 Hz. Sound pulse intensity changes within each call. Calls produced by a single
animal can continue uninterrupted for up to 45 min. (b) When sexually receptive females are about to oviposit, they often generate a call, rapping,
consisting of an 12 Hz pulse rate that attracts males. When males hear rapping, they approach the female (or loudspeaker playing an artificial
call) and generate a modified version of the advertisement call, answer call. Answer calling consists of elongated fast trills and truncated slow trills.
(c) When unreceptive females are clasped by a male, they extend their hindlimbs and generate a slower, 4 Hz, call: ticking. Ticking suppresses male
calling. Ticking has also been recorded from male/male pairs (Tobias et al., 2004). While the sound pulse rate (4 Hz) does not differ from female
ticking, sound pulses frequencies (1.7–2.3 kHz) in male ticking are higher than in female ticking (1.2 kHz). (d) When females are reproductively
active, they respond to male clasps with flexion of the hindlimbs, which promotes fertilization during oviposition. During this prolonged amplexus,
males generate a low-intensity, low pulse rate call: amplectant call. (e) Males produce all call types in the presence of other males. Two call types
most commonly associated with male–male interactions are chirping and growling, which are believed to represent agonistic signals. Chirping
appears to be largely associated with dominant males, while growling is produced by clasped males. Pulse intervals for both calls are fast
(40–80 Hz), with distinctive temporal patterns; chirps are brief and repeated multiple times per second, each growling trill tend to last several
hundred milliseconds. Reproduced from Zornik, E., Kelley, D.B., 2011. A neuroendocrine basis for the hierarchical control of frog courtship vocaliza-
tions. Front. Neuroendocrinol. 32, 353–366.

but spectral frequencies of male pulses are higher (1.8 kHz) recording that includes advertisement calling and growling
than in females (1.2 kHz). indicates a male/male pair in which one is clasping the other
The distinctive features of different calls given by the sexes (and thus sexually active). A recording that includes rapping
allow the social context in which a recording was obtained to and answer calling indicates interactions between a sexually
be determined from the calls alone. For example, a laboratory active female/male pair. Support for the reliability of these
134 Hormones and Vocal Systems: Insights from Xenopus

4000 Hz

3000 Hz

2000 Hz

1000 Hz

Fast trill Slow trill

Figure 2 Spectrograms of sexually differentiated Xenopus laevis calls. Left, male advertisement calls are spectrally complex. Fast trill sound pulses
contain two dominant frequencies (DFs) at 1.9 and 2.2 kHz. Slow trill sound pulses contain a lower frequency component 1.2 kHz. Right,
female ticking sound pulses have less finely tuned frequency ranges, with a single DF at 1.2 kHz. Scale bars: 250 ms.

inferences comes from the ability to reproduce the outcomes of metabolized to the androgen androstenedione (Lutz et al.,
social interactions in isolated frogs using sound broadcasts. 2001). Injection of GnRH into ovariectomized females treated
Rapping broadcasts, for example, stimulate male answer calling with progesterone and estradiol increased the likelihood of
(Vignal and Kelley, 2007) while intense advertisement calling clasping by males and led to prolonged amplexus, an effect
produces prolonged vocal suppression in males (Tobias et al., not present after hCG administration. Thus GnRH might be
2010). The broadcast paradigm has been useful in functional acting directly on brain targets rather the pituitary to elicit
dissection of forebrain influences on vocal responses, described receptive behaviors.
below (Hall et al., 2013). Prostaglandin E2 (PGE2) injection has also been shown to
increase female receptivity (Weintraub et al., 1985). PGE2
injections led to dramatic and rapid (30 s–3 min) reduction
2.08.2.2 Clasping, Reproductive Status, and Social of unreceptive leg extensions. Because these effects occur in
Hierarchies both intact and ovariectomized females, it is likely that
a PGE2 acts directly on neuronal targets to mediate these
2.08.2.2.1 Female Receptivity
behavioral changes. Similar rapid actions of prostaglandins
Nearly a century of experimental work has probed the role of
have been described in fish (Stacey and Goetz, 1982; Juntti
steroids in female X. laevis reproduction. Early studies found
et al., 2016).
that anterior pituitary extracts could induce ovulation in
X. laevis females, while hypophysectomy lead to dramatic
ovarian retrogression (Hogben et al., 1931). These results 2.08.2.2.2 Male–Female Clasping
set the stage for the widespread use of Xenopus as a test for Clasping is a critical component of Xenopus mating behavior; in
pregnancy; injection of urine containing human chorionic the laboratory, amplexus can be maintained for several days
gonadotropin (hCG, a hormone produced by the placenta) (Kelley, 1982). Male clasping is androgen-dependent. Castra-
leads to egg ripening and oviposition in Xenopus (Elkan, tion abolishes clasping, while T or DHT implants reinstate
1938). Although little is known about the relation between the behavior (Kelley and Pfaff, 1976). Although intact females
hormonal fluctuations in adult female X. laevis to their repro- do not clasp males, gonadectomized females treated with
ductive cycles in nature, one study found a strong positive either T or DHT do clasp males, and clasp durations do not
correlation between hypothalamic GnRH (also known as differ from hormone-replaced males. The finding that DHT
LHRH) and breeding season (King and Millar, 1979). Thus, can induce clasping in both sexes suggests androgens are suffi-
it is likely that seasonal elevation of GnRH leads to gonado- cient to induce clasping; E on the other hand does not induce
tropin (LH)-induced oocyte maturation and oviposition, clasping.
two events that can be recapitulated by exposure to hCG.
Although it was initially assumed that hCG injections 2.08.2.2.3 Male–Male Clasping
promote oocyte maturation via progesterone signaling, recent Group-housed male X. laevis are often observed clasping each
experiments indicate that plasma progesterone is an order of other, sometimes for prolonged periods. A recent study
magnitude lower than androgens and that androgens instead suggests that these male–male interactions do not necessarily
appear responsible for oocyte maturation (Lutz et al., 2001). reflect aggression or failure to discriminate sex, but may instead
Both hCG and GnRH induce a receptive posture – flexion represent an alternative mating strategy (Rhodes et al., 2014).
of the hindlimbs – in intact females clasped by males In triad experiments, in which two males were tested with
(Figure 1(d); Kelley, 1982). Ovariectomy eliminates this one female, most males preferentially clasped females;
behavior, which can be reinstated by injections of progesterone however, a subset of males preferentially directed clasping
and estradiol together, although each hormone by itself does toward other males. Males that are less likely to clasp a male
not induce the receptive response (Kelley, 1982). These results in male–male pairs are more likely to ‘win’ clasps of females
do not rule out a role for androgens in female responses to in subsequent male–male–female triads. Conversely, males
clasping, as progesterone has been shown to be rapidly that preferentially clasped the other male in the male–male
Hormones and Vocal Systems: Insights from Xenopus 135

pair rarely gained access to the female, but continued to clasp pairs. However, when one or both males in a pair is injected
the other male. Thus clasping in this study does not appear (100 IU, 24 h and 6–8 h before each trial), the amount of
to represent a means of demonstrating or establishing repro- calling is greatly increased, with all six call types represented.
ductive dominance. Together, these results support the hypoth- Although some of this effect is likely due to an increase in circu-
esis that some males use an alternative mating strategy in which lating androgens, hCG also appears to affect calling by directly
they gain physical proximity to gravid females via clasping an activating vocal nuclei in the brain. The original evidence for
amplexing male; such physical proximity could allow fertiliza- a central mechanism of hCG function came from experiments
tion of some eggs as they are laid by the female. in castrated, hormone-replaced frogs (Wetzel and Kelley,
1983). As described above, calling was rapidly eliminated
2.08.2.2.4 Vocal Dominance following castration, but could be reinstated by replacing T or
The finding that, in any given pond, only one or a few males DHT. Calling rates in hormone-replaced animals were signifi-
produce advertisement calls each night prompted laboratory cantly lower than intact frogs. However, when these animals
investigations into the mechanisms regulating calling. Tobias were injected with hCG, time spent calling increased as it did
et al. (2004) showed that pairing two males led to vocal during presurgery control recordings. Since the testes were no
silencing of one. Transitive, linear vocal dominance hierarchies longer present, an extragonadal target was implicated.
can be inferred from tests of multiple male pairs and remain Later experiments provided strong support for the hypoth-
stable for up to 2 weeks (Tobias et al., 2010). Physical contact esis that gonadotropins enhance advertisement calling via
is not required to establish vocal dominance, and the amount direct action in the brain (Yang et al., 2007). Intracerebroven-
of male clasping did not match vocal dominance patterns. Play- tricular (ICV) injection of hCG was effective at enhancing
back of male advertisement calls alone was sufficient to vocally calling at far lower doses than systemic injections. This effect
suppress all exposed males (Tobias et al., 2010). Vocal suppres- appears to be transduced via luteinizing hormone receptors,
sion was achieved whether the playback recordings were which are expressed in the central amygdala (CeA), a known
derived from a vocally dominant or subordinate animal; low- vocal control region (Figure 3; Brahic and Kelley, 2003; Hall
intensity playbacks of either call were less effective in vocal et al., 2013). Exposure of the isolated brain to hCG induced
suppression than high-intensity playbacks. Quantity or inten- upregulation of the immediate early gene egr-1 in the CeA,
sity of calling thus appears to be a determining factor in estab- even when synaptic transmission was blocked. Endogenous
lishing dominance. gonadotropin signaling must not be sufficient to induce
Whether the establishment or maintenance of vocal domi- calling, however, because hCG injections in castrated males
nance is hormone-dependent has yet to be investigated. do not enhance male calling (Zornik and Yamaguchi,
Because advertisement calling depends on circulating andro- 2011). Thus, androgens and gonadotropins are both required
gens (see below), differential levels of circulating androgens in the brain to allow normal production of male advertise-
could be correlated with vocal dominance, as observed in social ment calls.
dominance in other species such as cichlids (Maruska, 2014), As described above, linear vocal hierarchies between males
electric fish (Cuddy et al., 2012), and rodents (Shen et al., can be stable for up to 2 weeks, but subordinate males only
2015). Another possible contributor is corticosterone, as the suppress their calling when in the presence of a dominant
stress-related glucocorticoids are known to be correlated male. The mechanisms that underlie rapid suppression of
with social dominance in many species (Abbott et al., 2003; calling is unknown, but must involve processing of auditory
Goymann and Wingfield, 2004; Jeffrey et al., 2012; Ode information, given that playbacks alone are sufficient to
et al., 2015). Because of its prolonged nature, advertisement suppress calling. A likely candidate nucleus for regulating vocal
calling could be energetically costly, perhaps contributing to suppression is the CeA. Because this region receives auditory
the scarcity of calling in any given population during the information (Figure 3(a); Hall et al., 2013) and is a target of
breeding season. In a corticosterone-regulated pattern of vocal gonadotropin (Yang et al., 2007), one intriguing possibility is
dominance, males in an optimal physiological state (i.e., well- that vocal suppression may be a result of changes in endoge-
fed, metabolically stable, low stress) would be most likely nous gonadotropin signaling in the CeA.
to gain vocal dominance, leading to greater reproductive
success. 2.08.2.3.2 Female Calling
While females do not use vocalizations when paired with other
females (Tobias et al., 2004), reproductive state is reflected in
2.08.2.3 Hormone-Dependence of Vocal Behaviors
their vocalizations when paired with a male. Unreceptive
2.08.2.3.1 Advertisement Calls females produce the slow call, ticking, while receptive females
Male advertisement calling depends on endocrine state. Most produce the faster call, rapping (Figure 1; Tobias et al., 1998b).
castrated males show a complete loss of advertisement calling Although hormones that promote vitellogenin synthesis (e.g.,
after about 1 month (Wetzel and Kelley, 1983), although estradiol), oocyte maturation (e.g., androgens) and female
some animals do produce small amounts of abnormal calling receptivity (GnRH, progesterone, and estradiol) have been
as long as 1 year after castration (Zornik and Yamaguchi, described, we do not have a complete picture of how endoge-
2011). Normal calling in castrated males resumes following nous hormones modulate female vocal behaviors. The time
treatment with either T or DHT (Wetzel and Kelley, 1983). course of changes in female vocal behaviors induced by hCG
In the lab, male calling is greatly enhanced by injection of in the presence of an advertising, clasping male has been fol-
hCG (Wetzel and Kelley, 1983; Tobias et al., 2004; Yang lowed in intact females (Wu et al., 2001). All uninjected
et al., 2007). Very little calling is observed in uninjected male females ticked when clasped; 6 h after injection, all females
136 Hormones and Vocal Systems: Insights from Xenopus

(a)
TS
CT
DTAM
CeA n.IX-X

VN

LHR ER AR
(b)

CeA
A TS
S

n.IX-X
DTAM
VN

Figure 3 Auditory and vocal pathways in Xenopus laevis. (a) Diagram (top) and photograph (bottom) of a sagittal view of the X. laevis brain and
anterior spinal cord (anterior is left, dorsal is up). Key auditory, sensorimotor and vocal central pattern generator (CPG) nuclei are shown with their
known connections. The midbrain torus semicircularis (TS) processes auditory information and projects to the central thalamus (CT), which in turn
projects to the central amygdala (CeA). CeA projections to the hindbrain are thought to regulate social context-dependent activation of the vocal CPG.
CPG nuclei DTAM and n.IX-X are reciprocally connected. (b) Diagram (top) and photograph (bottom) of a dorsal view of the X. laevis brain and
anterior spinal cord (anterior is left). Color coded nuclei indicate adult patterns of hormone receptor expression (LHR, luteinizing hormone receptor;
ER, estrogen receptor; AR, androgen receptor). Laryngeal motor neuron axons exit the brain via the caudal root of cranial nerve IX-X, the vocal
nerve (VN).

were silent when clasped. Rapping in response to male calling local action triggers the final reduction divisions of meiosis
appeared in a smaller subset of females by 9 h postinjection responsible for oocyte formation (Lutz et al., 2001). While T
and was recorded out to 18 h postinjection. and DHT treatment masculinize female vocal behavior, the
What are the mechanisms underlying suppression of ticking primary circulating androgen in females, androstenedione,
and promotion of rapping? A potential endocrine suppressor might preferentially act on a subset of neural target cells that
of ticking is prostaglandin; PGE2 rapidly suppresses ticking promote rapping. An acute increase in circulating androgen,
behaviors when given exogenously but does not elicit rapping associated with imminent oviposition, might activate neural
(Weintraub et al., 1985). In cichlid fish, PGF2a activates female targets responsible for producing rapping. Another possibility
reproductive behaviors and appears to act directly on CNS is that rapping arises in response to gonadotropin signaling
targets via a gonadal hormone-induced PGF2 receptor (Juntti in the CeA. Recent experiments in the ex vivo brain
et al., 2016). A similar scenario might operate via PGE2 in preparation (described below) showed that CeA electrical
Xenopus. stimulation preferentially induce rappinglike vocal patterns,
What is the trigger for rapping? hCG-induced serum steroid suggesting that increased activity in the CeA may promote
hormones in females are dominated by androgens, and their receptive vocalizations in females (Ballagh, 2014).
Hormones and Vocal Systems: Insights from Xenopus 137

2.08.2.3.3 Male Interaction Calls effects of long-term (5–18 months) castration (Zornik and
As described above, Rhodes et al. (2014) observed that males Yamaguchi, 2011). These calls (referred to as ‘isolated slow
clasping other males (male preference) were less likely to clasp trill’ in Zornik and Yamaguchi) therefore may be generated by
females in a male–male–female triad, indicating that male– males with relatively low androgen signaling, but can be
male clasping may represent an alternative (i.e., ‘sneaker rapidly eliminated by exogenous androgens. The behavioral
male’) mating strategy rather than an agonistic interaction. relevance of ‘isolated slow trills’ is not understood.
Because they did not report vocal patterns in their study,
however, it is not clear whether vocal dominance is associated
with female-preference or male-preference clasping. 2.08.3 Sex Differences and Hormone Targets in the
In an earlier study, Tobias et al. (2004) showed that vocally Vocal System
dominant males produce many chirps while clasping, while
vocally subordinate males clasping another male produced The vocal behaviors of X. laevis are highly differentiated between
few chirps. These results support the idea that chirping is an the sexes. Nearly all neural and muscular components of the
aggressive vocal behavior that functions in regulating reproduc- vocal circuits are regulated by one or more steroid hormones.
tive dominance. Clasping by a dominant male may represent The vocal organ, which contains the single muscle group needed
aggression, while clasping by a subordinate male may represent to generate vocal sounds, differs markedly in males and females
a ‘sneaker’ strategy. Males in the Rhodes et al. (2014) study (Sassoon and Kelley, 1986). In the brain, many nuclei related to
were not injected with hCG, while both males in the Tobias auditory processing and vocal motor production were first iden-
et al. (2004, 2010) paired recordings were. The hCG treatments tified by their ability to concentrate steroid hormones (Kelley
may have led to aggressive clasping absent from the Rhodes et al., 1975; Morrell et al., 1975; Kelley, 1980) and have been
et al.’s study. shown to express mRNA for the androgen receptor (Pérez
These laboratory results leave open the question of how, et al., 1996). More recently, functional contributions of these
and under what circumstances, endogenous gonadotropins vocal system components and the mechanisms whereby steroid
act to promote aggressive clasping and chirping. Field record- hormones influence the vocal system have been investigated. In
ings in South Africa reveal that while advertisement calling is the following section we describe known components of the
produced at high levels throughout the breeding season, chirp- vocal system and briefly describe the hormones that regulate
ing peaks at the end of the season (Tobias et al., 2004). their development and function.
Growling, produced by males clasped by other males, drops
immediately prior to the surge in chirping and answer calling.
2.08.3.1 Sex Differences and Hormone Targets in the Larynx
This rapid change could reflect a population-wide surge in
endogenous gonadotropin in the late South African winter, as The adult X. laevis vocal organ is markedly sexually dimorphic
has been observed in females (King and Millar, 1979). Testing both in size and in tissue components and is functionally
this hypothesis will require replication of the vocal results specialized for producing sex-specific vocal behaviors
together with hormone measurements. (described in detail below). Male larynges contain a greater
number of muscle fibers than female larynges and express
2.08.2.3.4 Insights from Endocrine Disrupting Compounds a number of structural and functional adaptations that support
Most hormone manipulation studies in Xenopus have addressed the production of fast rhythmic vocalizations. Female laryngeal
long-term (i.e., weeks to months) effects. Recently, a series of features support the production of slower calls (Tobias and
studies investigating the effects of endocrine disrupting Kelley, 1987; Sassoon and Kelley, 1986; Sassoon et al., 1987;
compounds have begun to address short-term effects of endo- Marin et al., 1990; Tobias et al., 1995). Larynges of both sexes
crine signaling on vocal behaviors. In one study, exposure of express androgen and estrogen receptors (Segil et al., 1987;
hCG-injected males to the antiandrogen flutamide (10 6 M) Fischer et al., 1995; Wu et al., 2003). At metamorphosis,
significantly reduced the amount of advertisement calling male and female larynges are structurally and functionally
compared to hCG-injected controls 3 and 4 days after similar; sex-specific traits arise during juvenile development.
exposure, resulting in calling levels as low as non-hCG- All identified sexually distinct laryngeal traits are established
injected animals (Behrends et al., 2010). Similar reductions in and maintained by either androgens or estrogens. Although
calling were observed during exposure to 10 6 M of the some traits gradually lose hormone sensitivity, most sex-
antiandrogen vinclozolin; advertisement calling and chirping specific traits remain at least partially steroid sensitive in adult-
were reduced while ticking and growling were increased hood (Zornik and Kelley, 2011).
(Hoffmann and Kloas, 2010). In contrast, 4-day exposure to
low doses (30.5 ng l 1, 3.05 mg l 1, 30.5 mg l 1) of the xeno-
2.08.3.2 Sex Differences and Hormone Targets in the Vocal
androgen methyldihydrotestosterone (MDHT) induced
Circuit
significant increases in the percentage of advertisement calling
(Hoffmann and Kloas, 2012a). The increase in percentage of 2.08.3.2.1 Motor Circuit
advertisement calls produced by males was paralleled by Laryngeal muscle fibers are innervated by motor neurons located
a significant decrease in a call the authors referred to as in cranial nerve nucleus (n.) IX-X in the caudal hindbrain
rasping. The spectrogram of rasping reveals a call that appears (Kelley, 1980; Wetzel et al., 1985). Molecular markers
similar to an elongated (over 5 s) slow trill uninterrupted by (transcription factors and neurotransmitter enzymes) support
fast trills (Hoffmann and Kloas, 2012b). Interestingly, the homology of the Xenopus n.IX-X to nucleus ambiguus of
a similar pattern of calling was observed in a study of the mammals (Albersheim-Carter et al., 2016). As in the sexually
138 Hormones and Vocal Systems: Insights from Xenopus

dimorphic larynx with its greater number of muscle fibers in 1. Rate: Except for ticking and amplectant calls, male vocali-
males, the number of male vocal motor neurons in n.IX-X is zations consist of rapid sound pulses, between 30 and
greater than in females (Kay et al., 1999). Unlike the larynx, 80 Hz, whereas the fastest female call does not exceed
however, differences in laryngeal motor neuron number 12 Hz.
develop before metamorphosis (Kelley and Dennison, 1990), 2. Complexity: Male advertisement and answer calls are
arising through an androgen-dependent reduction in cell death temporally complex: trills with different durations and
in males compared to females, a process that can be partially pulse repetition rate alternate. Both female calls are simpler
rescued in females by androgen exposure (Kay et al., 1999). In trains of sound pulses.
adulthood, motor neurons remain sensitive to androgens, and 3. Spectral features: Male advertisement and answer calls are
many, but not all, of the laryngeal motor neurons express spectrally complex: each sound pulse includes two well-
androgen receptors (Perez et al., 1996). Following axotomy in defined frequency peaks. Sound pulses in both female
adult males, the total number of n.IX-X motor neurons is calls contain a single, less sharply defined peak.
greatly reduced, and cell loss can be partly prevented by DHT
In this section, we discuss each class of sexually differenti-
treatment in both males and females (Pérez and Kelley, 1996).
ated vocal features, focusing on the operation of multiple levels
Androgens upregulate expression of the calcium-binding
of mechanistic control and the role of hormonal regulation.
protein calbindin in axotomized female vocal motor neurons,
suggesting a possible mechanism for its neuroprotective effects
(Perez and Kelley, 1996). 2.08.4.1 Generating Fast and Slow Calls
Vocal rhythms are generated by a hindbrain CPG (Rhodes
et al., 2007). The two primary vocal CPG nuclei are n.IX-X in The most dramatic vocal difference between the sexes is pulse
the caudal medulla, which contains both motor neurons and rate. The biphasic male advertisement call consists of two
interneurons, and a nucleus, DTAM (used as a proper noun), rate-specific phases, fast trill and slow trill. The typical fast trill
located in the anterior brain stem (Figure 3(a)). These two pulse rate is 60 Hz (interpulse interval: 16 ms), while the
CPG nuclei are reciprocally connected via n.IX-X projection slow trill pulse rate is 30 Hz (interval, 30 ms). In contrast,
neurons (Wetzel et al., 1985; Zornik and Kelley, 2007), and rates of sound pulses in the female receptive call, rapping, range
communication between them is critical for generating vocal from 8 to 12 Hz (interval, 80–125 ms), much slower than
rhythms (Rhodes et al., 2007; Zornik et al., 2010). In male advertisement calls. The female unreceptive call, ticking,
addition to motor neurons, some interneurons in n.IX-X also is even slower, with pulse rates at 4 Hz. How are these sex
express androgen receptors, as do neurons in DTAM (Pérez differences in sound pulse rhythms (biphasic vs monophasic)
et al., 1996). Thus the two primary CPG nuclei are both and rates (fast vs slow) produced?
under direct control of androgens (Figure 3(b)). Two reduced preparations – the isolated larynx and brain –
have greatly advanced our understanding of the mechanisms
2.08.3.2.2 Auditory Pathways underlying sex-specific vocal behaviors. We review our current
Androgen receptors are expressed in the auditory ganglion, understanding of how sex-specific vocalizations are generated
indicating that peripheral coding of auditory information is and perceived and discuss experiments aimed at determining
regulated by gonadal steroids (Pérez et al., 1996). An important the precise role of hormones in shaping vocal output.
auditory processing nucleus, the laminar nucleus of the torus
semicircularis (the homologue of the mammalian inferior 2.08.4.1.1 Larynx
colliculus) located in the midbrain, concentrates three classes Xenopus vocalizations are produced under water by the contrac-
of steroid hormones: androgens, estrogens, and progesterone tion of a single muscle group, the laryngeal dilators. Each
(Figure 3(b); Morrell et al., 1975; Kelley, 1980). The basic sound pulse in a call is generated by a synchronous bilateral
sensory–motor integration pathways have been identified contraction of these muscles (Tobias and Kelley, 1987; Yager,
anatomically. Auditory information appears to primarily travel 1992). Investigations of laryngeal mechanisms of sound
from the torus to central thalamus (CT) and then on the central production have been greatly facilitated by the fact that the iso-
amygdala (CeA), which completes the vocal pathway circuit by lated larynx can generate sounds in vitro when both laryngeal
projecting back to the CPG via DTAM (Figure 3(b); Hall et al., nerves are stimulated synchronously (Tobias and Kelley,
2013). Neurons in the CeA express estrogen receptors and 1987). This experimental preparation – the ‘vox in vitro’ – has
luteinizing hormone receptors (Figure 3(b); Morrell et al., allowed the identification of numerous sex-specific traits of
1975; Yang et al., 2007). Thus, nearly every component of male and female vocal muscle.
the anatomically identified auditory circuit is sensitive to Sound pulses are generated when laryngeal cartilages, the
reproductive hormones, indicating that sensory processing of arytenoid discs, are rapidly pulled apart by a tendon into which
vocal signals and sensorimotor integration are likely to be as the laryngeal muscle inserts; before the next pulse can be gener-
sexually distinct as the behaviors themselves. ated, muscles must relax completely to allow laryngeal carti-
lages to regain contact (Yager, 1992). Speed of contraction
and relaxation are major functional differences between male
2.08.4 Generating Sexually Differentiated and female larynges. Male laryngeal muscle fibers are fast
Vocalizations twitch, and the muscles can fully contract and relax when stim-
ulated at fast trill rates, 60 Hz (Tobias and Kelley, 1987). In
Vocal behaviors of males and females differ markedly in at least contrast, female laryngeal muscle cannot generate sound pulses
three ways. faster than 30 Hz (associated with the upper limit for pulse
Hormones and Vocal Systems: Insights from Xenopus 139

intervals in rapping; Tobias and Kelley, 1987; Tobias et al., spontaneous vocal-like nerve activity patterns can also be
1998b). Above these sex-typical rate ceilings, the fused tension recorded from the isolated brain (e.g., fictive amplectant
that can be recorded from the tendon prevents the arytenoid calling; Zornik and Kelley, 2008). Though 5-HT initiates fictive
discs from regaining contact; subsequent sound pulses cannot calling in both sexes, the patterns are sex-specific as they are
be generated in response to nerve activity. in vivo; 5-HT induces fictive advertisement calls in male brains
The sexually differentiated expression pattern of a larynx- and fictive ticking in female brains.
specific myosin heavy chain is a major factor underlying sex- The isolated brain preparation facilitates localization of the
specific contraction rates. Adult male larynges contain a single vocal pattern generator to major brain regions. Transections of
fiber type – medium-sized, fast-twitch muscle fibers – whereas the isolated brain at the border between the midbrain and
female larynges contain a mixture of fast-twitch and slow- hindbrain do not eliminate 5-HT-induced fictive calls, indi-
twitch fibers (Sassoon and Kelley, 1986). The sex difference cating that the vocal CPG is located in the hindbrain (Yu and
in fiber types reflects expression of a laryngeal-specific myosin Yamaguchi, 2010). The vocal CPG includes two reciprocally
heavy chain (LM) in all male muscle fibers, but only in a subset connected nuclei: n.IX-X in the caudal hindbrain and
of female muscle fibers (Catz et al., 1992). The development of a premotor nucleus, DTAM, in the rostral hindbrain (Brahic
ubiquitous LM expression in males requires androgen, and and Kelley, 2003; Rhodes et al., 2007). In Xenopus, both vocal
DHT-exposure in juvenile females upregulates LM expression. CPG nuclei were first identified by their ability to concentrate
In addition, androgen treatment in females leads to androgen (Kelley, 1980), and both are sexually dimorphic
a dramatic increase in the number of histologically identified (Wetzel et al., 1985; Kay et al., 1999; Brahic and Kelley,
fast-twitch fiber types (Sassoon et al., 1987) and also leads to 2003). Homologues of these two nuclei are also believed to
rapid increase in contraction and relaxation rate of the participate in generating vocal rhythms in a distantly related
muscle (Potter et al., 2005). Although long-term castration of frog species, Rana pipiens (Schmidt, 1992), and expiration in
adult males does not affect the histological profiles of male lamprey (‘paratrigeminal respiratory group’ nucleus; Cinelli
muscle fibers (Sassoon et al., 1987), there is a dramatic et al., 2013; Leininger and Kelley, 2015) and mammals
slowing of muscle contraction and an accompanying increase (parabrachial complex; Dick et al., 1994; Song and Poon,
in fused tension during nerve stimulation trains at fast trill 2009). It is likely that vocal CPGs in many, if not all, frog
rates (Zornik and Yamaguchi, 2011). Whether this castration- species comprise these two nuclei and that both contribute to
induced slowing of the male larynx is due to effects on LM coordinating vocalization with breathing in mammals.
(as opposed to other components of the muscle’s contractile
machinery) is not known. In summary, larynges of males and 2.08.4.1.2.1 Motor Neurons
females support the sex-specific calls: male larynges are As described above, many, but not all, laryngeal motor neurons
specialized for generating fast calls, while female vocal organs express androgen receptors (Kelley, 1980; Pérez et al., 1996),
can generate only relatively slow calls. These laryngeal which suggests an ongoing role for androgens in maintaining
characteristics are androgen-dependent and many remain appropriate motor neuron function throughout adulthood.
hormone-dependent in adulthood. Motor neuron firing rates are significantly higher during fictive
vocalizations in males than in females, and membrane proper-
2.08.4.1.2 Central Pattern Generator ties and firing patterns of motor neurons express sexually
Laryngeal muscles provide constraints on the rates of sound distinct features for generating their sex-specific motor patterns.
pulse generation but do not shape most features of vocal The majority of male motor neurons spike with short latency
patterning; these are generated within the brain. Yamaguchi following current injection and are thus well suited to
and Kelley (2000) recorded from the laryngeal nerve of male the demands of rapid firing with high temporal precision
and female frogs during vocalization and found that each (Yamaguchi et al., 2003). In contrast, female motor neurons
sound pulse is preceded by a compound action potential have longer spike onset times (Yamaguchi et al., 2003), which
(CAP). For both sexes and all call types, synchronous firing may account for the less precise firing patterns, and resulting
of laryngeal motor neurons in the brain generates a CAP in longer nerve CAPs, than males during both in vivo and ex vivo
the nerve that, in turn, induces rapid contraction of the laryn- vocalizations (Yamaguchi and Kelley, 2000; Rhodes et al.,
geal dilators and a resulting sound pulse. Thus, the temporal 2007). Patch-clamp recordings in slice preparations also
features of each Xenopus call primarily reflect patterns of brain revealed that male motor neurons have higher capacitance
activity. than female motor neurons (Yamaguchi et al., 2003). Larger
Because the temporal patterns of nerve activity and sound motor neuron somata volumes in male motor neurons could
are matched, vocal ‘intention’ can be identified unambiguously contribute to these sex differences (Potter et al., 2005).
by measuring nerve activity. This feature of the Xenopus vocal The size difference is highly plastic, and female motor neurons
system has been put to effective use through the development increase to male size after only 1 week of testosterone
of a ‘singing brain in a dish’ preparation (Rhodes et al., 2007). treatment.
The CNS from forebrain to hindbrain is removed from the
frog, pinned to a coated dish, and maintained in an oxygenated 2.08.4.1.2.2 Premotor Neurons
artificial cerebrospinal fluid while recording from the laryngeal The major source of input to laryngeal motor nucleus is the
nerve, the most caudal nerve rootlet of N.IX-X (Simpson et al., premotor nucleus, DTAM, located in the anterior hindbrain
1986). Bath-application of serotonin (5-HT) reliably induces (Wetzel et al., 1985; Zornik and Kelley, 2007). Whole-cell
‘fictive vocalizations’ – nerve activity that matches the sound patch-clamp recordings in isolated male brains during
pulse patterns of singing frogs (Rhodes et al., 2007); some fictive advertisement calling have revealed a population of
140 Hormones and Vocal Systems: Insights from Xenopus

premotor neurons that are active only during fictive fast trills changes in these neurons that drive the ontogeny of fast trill
(Zornik and Yamaguchi, 2012). These fast trill neurons rhythms is a challenge for the future. A goal is to identify
produce long-lasting depolarizations that coincide with each both CPG components that remain androgen-sensitive, as
fast trill. During these depolarizations, fast trill neurons produce well as those that lose androgen-sensitivity and thus constrain
action potentials that precede each fast trill CAP recorded from masculinization in adulthood.
the nerve. Most fast trill neurons project directly to n.IX-X,
suggesting that these neurons provide premotor signals that
2.08.4.2 Generating Complex and Simple Vocal Patterns
drive motor output. Earlier physiological studies showed that
DTAM neurons provide monosynaptic glutamatergic excitatory 2.08.4.2.1 Biphasic versus Monophasic Calling
input to laryngeal motor neurons (Zornik and Kelley, 2008), Fast trill neurons in DTAM produce long-lasting depolariza-
supporting the notion that synchronous firing of fast trill tions that coincide with each fast trill (Zornik and Yamaguchi,
neurons is responsible for generating each fast trill CAP. 2012). These depolarizations require NMDA receptor
When depolarized by current steps, fast trill neurons signaling and are blocked by NMDA receptor antagonists. In
preferentially generate spikes around 60 Hz, the approximate the presence of TTX (to block spike-mediated synaptic trans-
rate of fast trills (Zornik and Yamaguchi, 2012). Intrinsic fast mission) and NMDA (to activate NMDA receptors), fast trill
trill neuron membrane properties may be tuned to fire at neurons generate membrane potential oscillations in which
60 Hz, and thus be critical for setting the fast trill rhythm. the duration and period of each depolarization is similar to
DTAM neurons express androgen receptors (Kelley, 1981; Pérez the duration and period of fast trills. The constellation of ion
et al., 1996), and it is likely that fast trill neurons in particular currents in DTAM must support membrane potential oscilla-
are directly regulated by androgens. When androgens were tions with advertisement call patterns: that is, about one call
ablated by castration, the rate of fictive fast trills was reduced per second, with each fast trill lasting about 300 ms. Slow trills
over a period of months, indicating a role for androgens in only occur while fast trill neurons are hyperpolarized. Thus fast
maintaining normal fast trill rhythms (Zornik and Yamaguchi, trill neurons may be largely responsible for controlling the
2011). We hypothesize that androgens act directly on fast temporal patterns of male advertisement calls.
trill neurons to ensure high levels of expression of androgen- Because fast trill neurons produce these membrane poten-
regulated proteins that permit firing at 60 Hz for long tial oscillations synchronously, population–level activity in
periods and that the loss of these properties in the absence of DTAM can be readily recorded as local field potential (LFP)
androgen shifts vocal rhythms. Exactly how androgens act to waves that correspond to fast trills. DTAM activity in females
regulate fast trill neurons and possible roles for an activity- does not reveal the LFP waves seen in males (Ballagh, 2014).
dependent mechanism remain to be explored. During androgen treatment, however, females gradually
The sensitivity of sexually differentiated behavioral systems develop biphasic calls. Masculinization of existing neurons in
to reproductive hormones depends on developmental stage: female DTAM probably involves the rearrangement of ion
early actions can produce permanent changes in the behavioral currents to promote long-lasting depolarizations that lead to
repertoire, while actions during adulthood can be transient membrane oscillations with an 1 Hz period.
(Phoenix et al., 1959). Sensitivity to androgens is often studied During 5-HT evoked fictive ticking in the ex vivo female
by manipulating hormone levels across development. For brain, CAP rates recorded from the laryngeal nerve occur
X. laevis, androgen promotion of clasping serves as an example at 4 Hz (Rhodes et al., 2007). In the initial stages of
of a transient action of a hormone: abolished in adult males by testosterone-induced masculinization, single sound pulses
gonadectomy, reinstated by androgen replacement, and transition to doublet, triplet, and quadruplet pulse bursts
induced, in a form not distinguishable from that of males, in (Hannigan and Kelley, 1986; Potter et al., 2005). The appear-
adult females by androgen treatment (Kelley, 1982). Similarly, ance of pulse bursts could be due to prolonged depolarization
male calling is abolished by castration and reinstated by in female premotor neurons.
androgen replacement (Wetzel and Kelley, 1983). However, Which neuronal properties are most strongly regulated by
masculinization of calling in females appears less malleable. androgens and which neuronal properties are most closely
Watson and Kelley (1992) provided testis implants or associated with overall circuit activity? Elongating bursts may
androgen pellets to juveniles at a number of postmetamorphic be correlated with increasing NMDA receptor current density.
stages. They noted a decline in the ability to masculinize call Alternatively, NMDA currents may change little during mascu-
temporal patterns with age in females but not in males. linization; instead, an upregulation of slower outward currents
Long-term testis implants and androgen treatment (10– could underlie changes in temporal patterns of vocal output.
15 months) in adult females, however, did induce some Combining androgen-induced masculinization treatments in
biphasic advertisement calls. Potter et al. (2005) found that adult females with physiological recordings in the isolated
within 8 weeks, androgen-treated adult females produced brain will permit the identification of steroid-induced neuronal
biphasic, advertisement call-like vocalizations. Their fast trills traits that contribute to sex-specific vocal characteristics.
approached, but did not quite equal, the 60 Hz rates typical
of males. These results confirm that the adult female CPG can 2.08.4.2.2 Intensity Modulation of Vocal Trills
be masculinized to generate male-like vocal rhythms, albeit 2.08.4.2.2.1 Synaptic Strength in the Larynx
incompletely. While the processes underlying this androgen- In addition to rhythm and rate, the intensity modulation of
induced masculinization of adult females is unclear, cells fated male and female calls also differs. In male advertisement and
to become fast trill neurons are likely participants. Identifying answer calls, sound pulses early in fast trills are of low
androgen-induced functional, structural, and/or synaptic intensity and become gradually louder throughout each trill;
Hormones and Vocal Systems: Insights from Xenopus 141

female ticking and rapping sound pulses, in contrast, do not 2.08.5 Processing Vocal Signals
vary systematically in intensity (Figure 1; Tobias et al.,
2.08.5.1 Sexually Differentiated Auditory Processing
1998b). The laryngeal neuromuscular synapse contributes to
intensity modulation. When the male laryngeal nerve is stimu- Auditory sensitivity also appears to differ in females and males.
lated at advertisement call rates ex vivo, amplitudes of the elec- While no sex differences in spectral preference were identified
tromyogram, tension, and sound amplitudes all increase in auditory nerve fibers using single-cell electrophysiological
progressively throughout the stimulus train (Tobias and Kelley, recordings (N.VIII; Elliott et al., 2007), auditory brain stem
1987). This increase in amplitudes reflects the use-dependent response (ABR) measurements have revealed sex-specific
facilitation of the weak male laryngeal synapse (Tobias et al., frequency preferences (Hall et al., 2016). Xenopus laevis male
1995; Ruel et al., 1998). In contrast, females have strong laryn- sound pulses contain two major DFs: 1.9 and 2.2 kHz
geal synapses, well suited to ensuring that slower rates of nerve (Figure 2). As would be predicted from the matched-filter
activity are faithfully converted to sound. The sex difference in hypothesis (Frishkopf et al., 1968; Capranica and Moffat,
synaptic strength arises presynaptically and is due to elevated 1983; Moreno-Gomez et al., 2013; Simmons, 2013), in
quantal content at the synaptic terminals of female motor females, ABR thresholds for combinations of these two
neurons (Tobias et al., 1995). Strong female laryngeal synapses frequencies are lower than either frequency alone; small shifts
arise during juvenile development and require prolonged in either abolish this enhanced sensitivity. This enhanced sensi-
elevated estrogen levels; ovariectomy leads to a decrease in tivity is absent in males. Instead, ABR sensitivity peaks at lower
synaptic strength (Tobias et al., 1998a). In contrast, androgen frequencies, just under 1.5 kHz.
treatment weakens female synapses (Tobias and Kelley, These results indicate that, at the level of the hindbrain, the
1995). In adult males, long-term castration appears to reduce female auditory system is more highly tuned to male call
male-typical EMG potentiation during nerve stimulation frequencies than is the male auditory system; this acoustic
trains, reflecting castration-dependent synaptic strengthening advantage may facilitate locating a mate. Males also communi-
(Zornik and Yamaguchi, 2011). cate vocally, but most of their vocal interactions are close
Laryngeal neuromuscular junctions are well suited to gener- range, often while clasping, and enhanced sensitivity may be
ating intensity-modulated male calls via weak facilitating less important. However, males do establish vocal dominance
synapses and slower female calls via strong synapses, both of hierarchies, which are sensitive to advertisement call intensity
which depend on appropriate androgen and estrogen levels (Tobias et al., 2010). Relative insensitivity to advertisement
during development and in adulthood. Thus, as is the case calls could release males from a vocally subordinate condition
for clasping behavior, synaptic strength displays endocrine- at shorter distances from a dominant male. Field recordings
regulated flexibility, even in adulthood. do indicate that very few frogs advertise on any given night
(Tobias et al., 2004); the number may depend on how well
sound travels in particular bodies of water.
2.08.4.2.2.2 Motor Neuron Recruitment An alternative possibility is that slow trills, which contain
Laryngeal nerve recordings in singing frogs revealed that CAP a lower frequency band below 1.5 kHz not present in fast trill
amplitudes during fast trills also progressively increase sound pulses (Figure 2; Yamaguchi et al., 2010), may be the
throughout each trill (Yamaguchi and Kelley, 2000), and this more salient feature for establishing dominance. This hypoth-
pattern is preserved in fictive vocalizations (Rhodes et al., esis has some support from experimental evidence: a subset
2007). The most likely mechanism is sequential recruitment of of males was preferentially vocally suppressed by artificial
motor neurons. The neural basis for this recruitment appears slow-trill-only stimuli, which were more effective than either
to have at least two components. First, the premotor fast trill fast-trill-only stimuli or natural advertisement call playbacks
neurons are gradually recruited across each trill. Early in a trill (Tobias et al., 2010). If true, these findings may indicate
only a few fast trill neurons spike; the number of spiking a sex-specific tuning to distinct features of the male advertise-
neurons then increases, and nearly all fast trill neurons spike ment call: females attending to fast trills, males attending to
at the end of a trill (Zornik and Yamaguchi, 2012). Second, slow trills. This arrangement is analogous to sex-specific prefer-
the DTAM-to-motor neuron synapses facilitate; when DTAM is ences in the tree frog, Eleutherodactylus coqui, in which the male
electrically stimulated in trains from 20 to 60 Hz, nerve CAPs auditory system preferentially responds to the first ‘co’ note of
progressively increase in amplitude, as is also seen during a male calls, while female auditory systems preferentially
in vivo calling (Zornik and Kelley, 2008). Thus, intensity respond to the second ‘qui’ component (Narins and Capranica,
modulation of fast trills is coded in three ways: premotor 1976).
neuron activation, premotor-to-motor synaptic facilitation, Male auditory sensitivity may also be tuned to female
and neuromuscular junction facilitation. We know that the frequencies, which have a DF 1.2 kHz (Vignal and Kelley,
weak, facilitating male neuromuscular synapse is regulated by 2007). Although the unreceptive call, ticking, is used at close
hormones. Because both DTAM and motor neurons express range, the female ‘advertisement call,’ rapping, is a high-inten-
androgen receptors, it is likely that some aspects of this sity signal that should carry longer distances under water,
trait are also androgen-dependent. Finally, because of the allowing males to find distant receptive mates (Tobias et al.,
redundant mechanisms underlying intensity modulation, this 1998b).
aspect of fast trills could be a behaviorally critical component Sex-specific auditory sensitivities are steroid-dependent.
of the communication signal, a hypothesis supported by the Ovariectomy masculinized female sensitivity to DF1–DF2
even more dramatic intensity modulation of the answer call dyads and to frequencies between 1 and 1.5 kHz, while
during male/female duets (Tobias et al., 1998b). androgen treatment with DHT prevented this masculinization
142 Hormones and Vocal Systems: Insights from Xenopus

(Hall et al., 2016). These results are consistent with the finding long-lasting (trill-type (as in X. laevis)), fast and brief (burst-
that androgens are the primary circulating steroids in female type), and slow and ongoing (click-type). Each type occurs in
X. laevis (Lutz et al., 2001). These results also suggest a possible multiple branches of the phylogeny, and parsimony suggests
functional role for the observation that androgen receptors are that the ancestral call type was burst-type. The click-type pattern
expressed in the auditory ganglion (Pérez et al., 1996), but one occurs in two species – Xenopus boumbaensis and Xenopus borealis
would then expect a similar change in males. It is possible that- – which are found on distantly related branches of the Xenopus
male and female auditory ganglia respond differently to phylogeny, indicating that their click-type calls evolved indepen-
androgen exposure, or that there is a dose–response difference dently. As in X. laevis, the X. boumbaensis larynx is strongly
between the sexes. Alternatively, sex-specific production of sexually dimorphic; male fibers are large and fast twitch, while
the DHT metabolite 5a-androstane-3b,17b-diol (3b-diol), female fibers are small and mixed twitch type. Because male
a potent agonist of ERb, could be a DHT-dependent source of X. boumbaensis laryngeal fibers are mostly fast twitch, they could
estrogen (Oliveira et al., 2007). Although ERb expression in support the production of fast trills as found in X. laevis.
the Xenopus auditory periphery has not been explored, if However, the CPG generates very short vocal bursts consisting
estrogen receptors are expressed in hair cells of the inner ear, of two CAPs in the nerve (Leininger and Kelley, 2013). These
as they are in vocal fish (Forlano et al., 2015), then selective doublets are translated into single sound pulses due to the laryn-
production of 3b-diol only in female hair cells could account geal synapse’s requirement for facilitation (see Section
for DHT-dependent feminization of the auditory periphery. 2.08.4.2.2.1). In X. borealis the larynges are less dramatically
sexually dimorphic. Male and female muscle fibers are both
a mixture of fast and slow twitch, though male fibers are larger
2.08.5.2 Generating Socially Appropriate Vocal Responses
and greater in number (Leininger et al., 2015). The X. borealis
To facilitate effective social interactions, vocal responses of vocal CPG produces only single CAPS. Because the male
a receiver must be appropriately based on auditory information X. borealis laryngeal synapse does not require facilitation, the
from the sender. Examples of socially appropriate vocal single CAPs in the nerve faithfully produce single sound pulses.
responses in X. laevis include the stimulation of male answer Thus, the vocal CPG rhythm and the laryngeal synapse
calling by female rapping, the transient suppression of male in X. borealis are similar to features associated with ticking in
calling by female ticking, and the prolonged suppression of female X. laevis. Since male laryngeal muscle fibers in the very
male calling by intense advertisement calling of another male distantly related species Xenopus tropicalis are entirely fast twitch
(Figure 1; Tobias et al., 1998b, 2004; Elliott and Kelley, (Baur et al., 2008), this feature is presumed to represent the
2007). Activation of CNS auditory regions must be sufficient ancestral state. The convergent evolution of click-type calls in
to promote appropriate vocal responses since broadcast calls X. borealis and X. boumbaensis are thus due to distinct mecha-
are effective in eliciting these responses. The CeA plays a critical nisms: in X. boumbaensis, the number of CAPs produced by the
role in matching auditory signals to call-type responses (Hall CPG is greatly reduced, while in X. borealis, the developmental
et al., 2013). Neurons in the CeA receive synaptic input from program for the CPG, the laryngeal synapse, and muscle fiber
the CT, which in turn receives inputs from the auditory types are all demasculinized (Leininger and Kelley, 2015).
midbrain (Figure 3). When the CeA is lesioned, males continue
to generate spontaneous advertisement calls, and thus are not
mute. Lesioned males also can still respond to intense adver- 2.08.7 Conclusion
tisement call broadcasts with prolonged vocal suppression,
and thus are not deaf. However, both ticking and rapping Despite the conceptual value of the organization-activation
also elicit prolonged vocal suppression, a response that intact dichotomy (Phoenix et al., 1959), this stark distinction is often
males do not normally make to either female call. In addition an oversimplification with many exceptions (Arnold and
to socially inappropriate vocal suppression to female calling, Breedlove, 1985; Tobet and Fox, 1992; Arnold, 2009; McCarthy
CeA-lesioned males also produce a lower proportion of and Nugent, 2013). Hormonal regulation occurs throughout
answer calls when paired with another male. These results indi- development and into adulthood. While some traits are irre-
cate that the CeA may be a positive regulator of vocal responses, versibly set during a narrow critical period, others retain full
as such responses are diminished after lesions. Because the hormone sensitivity throughout life, while still others retain
CeA is reciprocally connected with the CPG nucleus DTAM intermediate sensitivity. Xenopus vocalizations are complex,
(Figure 3), there is a direct anatomical pathway by which context-dependent and sexually differentiated, making them
CeA may activate socially appropriate vocal responses. excellent subjects for identifying the role of reproductive
hormones in regulating behaviors. The two ex vivo vocal prepa-
rations, the isolated larynx and brain, have permitted
2.08.6 Evolution of Vocal Effectors in Xenopus numerous advances in understanding the neural and muscular
mechanisms of vocal production. Each can produce fictive
The ex vivo brain and larynx preparations have been used to vocalizations independent of the other, a feature that has
probe the mechanisms underlying the evolutionary convergence enabled identification of separate hormonal effects on discrete
of a rare vocal pattern in Xenopus (Leininger and Kelley, 2013; vocal effectors that work together in the animal. For example,
Leininger et al., 2015). Each Xenopus species has a unique adver- although the larynges of many androgen-treated females are
tisement call that can be distinguished by call temporal patterns not capable of generating sound pulses at fast trill rates, the
and spectral properties of sound pulses (Tobias et al., 2011). isolated brain preparation permits an accurate assessment of
Three classes of call pattern can be identified: fast and the hormone sensitivity of the vocal CPG in adults. Similarly,
Hormones and Vocal Systems: Insights from Xenopus 143

castration leads to nearly complete elimination of in vivo male Goymann, W., Wingfield, J.C., 2004. Allostatic load, social status and stress
calling, yet fictive vocalizations can still be generated by the hormones: the costs of social status matter. Anim. Behav. 67, 591–602.
Hall, I.C., Ballagh, I.H., Kelley, D.B., 2013. The Xenopus amygdala mediates socially
isolated brain, allowing the identification of castration-
appropriate vocal communication signals. J. Neurosci. 33, 14534–14548.
induced functional changes in the CPG. Because the full Hall, I.C., Woolley, S.M., Kwong-Brown, U., Kelley, D.B., 2016. Sex differences and
acoustic communication pathway from ear to brain to vocal endocrine regulation of auditory-evoked, neural responses in African clawed frogs
organ has been outlined, with nearly every component known (Xenopus). J. Comp. Physiol. A Neuroethol. Sens. Neural Behav. Physiol. 202,
to be hormone-sensitive, the Xenopus vocal system provides an 17–34.
Hannigan, P., Kelley, D.B., 1986. Androgen-induced alterations in vocalizations of
experimentally powerful preparation for discovering novel female Xenopus laevis: modifiability and constraints. J. Comp. Physiol. A 158,
roles for hormone-directed behaviors and should offer new 517–527.
insights about fundamental mechanisms for sexually differen- Hoffmann, F., Kloas, W., 2010. An environmentally relevant endocrine-disrupting
tiated vertebrate behaviors. antiandrogen, vinclozolin, affects calling behavior of male Xenopus laevis. Horm.
Behav. 58, 653–659.
Hoffmann, F., Kloas, W., 2012a. Effects of environmentally relevant concentrations of
the xeno-androgen, methyldihydrotestosterone, on male and female mating
References behavior in Xenopus laevis. Chemosphere 87, 1246–1253.
Hoffmann, F., Kloas, W., 2012b. Estrogens can disrupt amphibian mating behavior.
Abbott, D.H., Keverne, E.B., Bercovitch, F.B., Shively, C.A., Mendoza, S.P., PLoS One 7, e32097.
Saltzman, W., Snowdon, C.T., Ziegler, T.E., Banjevic, M., Garland Jr., T., Hogben, L., Charles, E., Slome, D., 1931. Studies on the pituitary VIII. J. Exp. Biol. 8
Sapolsky, R.M., 2003. Are subordinates always stressed? A comparative analysis (4), 345–354.
of rank differences in cortisol levels among primates. Horm. Behav. 43, 67–82. Jeffrey, J.D., Esbaugh, A.J., Vijayan, M.M., Gilmour, K.M., 2012. Modulation of
Albersheim-Carter, J., Blubaum, A., Ballagh, I.H., Missaghi, K., Siuda, E.R., hypothalamic-pituitary-interrenal axis function by social status in rainbow trout.
McMurray, G., Bass, A.H., Dubuc, R., Kelley, D.B., Schmidt, M.F., Wilson, R.J., Gen. Comp. Endocrinol. 176, 201–210.
Gray, P.A., 2016. Testing the evolutionary conservation of vocal motoneurons in Juntti, S.A., Hilliard, A.T., Kent, K.R., Kumar, A., Nguyen, A., Jimenez, M.A.,
vertebrates. Respir. Physiol. Neurobiol. 224, 2–10. Loveland, J.L., Mourrain, P., Fernald, R.D., 2016. A neural basis for control of
Arnold, A.P., Breedlove, S.M., 1985. Organizational and activational effects of sex cichlid female reproductive behavior by prostaglandin F2alpha. Curr. Biol. 26,
steroids on brain and behavior: a reanalysis. Horm. Behav. 19, 469–498. 943–949.
Arnold, A.P., 2009. The organizational-activational hypothesis as the foundation for Kay, J.N., Hannigan, P., Kelley, D.B., 1999. Trophic effects of androgen: development
a unified theory of sexual differentiation of all mammalian tissues. Horm. Behav. and hormonal regulation of neuron number in a sexually dimorphic vocal motor
55, 570–578. nucleus. J. Neurobiol. 40, 375–385.
Ballagh, Irene H., 2014. Sex Differences in the Structure, Function and Regulation Kelley, D.B., Dennison, J., 1990. The vocal motor neurons of Xenopus laevis:
of Vocal Circuits in Xenopus (Ph.D. thesis). Columbia University. development of sex differences in axon number. J. Neurobiol. 21, 869–882.
Baur, L.A., Nasipak, B.T., Kelley, D.B., 2008. Sexually differentiated, androgen- Kelley, D.B., Pfaff, D.W., 1976. Hormone effects on male sex behavior in adult South
regulated, larynx-specific myosin heavy-chain isoforms in Xenopus tropicalis; African clawed frogs, Xenopus laevis. Horm. Behav. 7, 159–182.
comparison to Xenopus laevis. Dev. Genes Evol. 218, 371–379. Kelley, D.B., Morrell, J.I., Pfaff, D.W., 1975. Autoradiographic localization of hormone-
Behrends, T., Urbatzka, R., Krackow, S., Elepfandt, A., Kloas, W., 2010. Mate calling concentrating cells in the brain of an amphibian, Xenopus laevis. I. Testosterone.
behavior of male South African clawed frogs (Xenopus laevis) is suppressed by the J. Comp. Neurol. 164, 47–59.
antiandrogenic endocrine disrupting compound flutamide. Gen. Comp. Endocrinol. Kelley, D.B., 1980. Auditory and vocal nuclei in the frog brain concentrate sex
168, 269–274. hormones. Science 207, 553–555.
Brahic, C.J., Kelley, D.B., 2003. Vocal circuitry in Xenopus laevis: telencephalon to Kelley, D.B., 1981. Locations of androgen-concentrating cells in the brain of Xenopus
laryngeal motor neurons. J. Comp. Neurol. 464, 115–130. laevis: autoradiography with 3H-dihydrotestosterone. J. Comp. Neurol. 199,
Capranica, R.R., Moffat, A.J.M., 1983. Neurobehavioral correlates of sound commu- 221–231.
nication in anurans. In: Ewert, J.P., Capranica, R.R., Ingle, D. (Eds.), Advances in Kelley, D.B., 1982. Female sex behaviors in the South African clawed frog, Xenopus
Vertebrate Neuroethology. Plenum Press, New York, pp. 701–730. laevis: gonadotropin-releasing, gonadotropic, and steroid hormones. Horm. Behav.
Catz, D.S., Fischer, L.M., Moschella, M.C., Tobias, M.L., Kelley, D.B., 1992. Sexually 16, 158–174.
dimorphic expression of a laryngeal-specific, androgen-regulated myosin heavy King, J.A., Millar, R.P., 1979. Hypothalamic luteinizing hormone-releasing hormone
chain gene during Xenopus laevis development. Dev. Biol. 154, 366–376. content in relation to the seasonal reproductive cycle of Xenopus laevis. Gen.
Cinelli, E., Robertson, B., Mutolo, D., Grillner, S., Pantaleo, T., Bongianni, F., 2013. Comp. Endocrinol. 39, 309–312.
Neuronal mechanisms of respiratory pattern generation are evolutionary conserved. Leininger, E.C., Kelley, D.B., 2013. Distinct neural and neuromuscular strategies
J. Neurosci. 33, 9104–9112. underlie independent evolution of simplified advertisement calls. Proc. Biol. Sci.
Cuddy, M., Aubin-Horth, N., Krahe, R., 2012. Electrocommunication behaviour and 280, 20122639.
non invasively-measured androgen changes following induced seasonal breeding in Leininger, E.C., Kelley, D.B., 2015. Evolution of courtship songs in Xenopus :
the weakly electric fish, Apteronotus leptorhynchus. Horm. Behav. 61, 4–11. vocal pattern generation and sound production. Cytogenet Genome Res. 145,
Dick, T.E., Bellingham, M.C., Richter, D.W., 1994. Pontine respiratory neurons in 302–314.
anesthetized cats. Brain Res. 636, 259–269. Leininger, E.C., Kitayama, K., Kelley, D.B., 2015. Species-specific loss of sexual
Elkan, E.R., 1938. The Xenopus pregnancy test. Br. Med. J. 2, 1253–1256, 1274–2. dimorphism in vocal effectors accompanies vocal simplification in African clawed
Elliott, T.M., Kelley, D.B., 2007. Male discrimination of receptive and unreceptive frogs (Xenopus). J. Exp. Biol. 218, 849–857.
female calls by temporal features. J. Exp. Biol. 210, 2836–2842. Lutz, L.B., Cole, L.M., Gupta, M.K., Kwist, K.W., Auchus, R.J., Hammes, S.R.,
Elliott, T.M., Christensen-Dalsgaard, J., Kelley, D.B., 2007. Tone and call responses of 2001. Evidence that androgens are the primary steroids produced by
units in the auditory nerve and dorsal medullary nucleus of Xenopus laevis. Xenopus laevis ovaries and may signal through the classical androgen
J. Comp. Physiol. A Neuroethol. Sens. Neural Behav. Physiol. 193, 1243–1257. receptor to promote oocyte maturation. Proc. Natl. Acad. Sci. U.S.A. 98,
Elliott, T.M., Christensen-Dalsgaard, J., Kelley, D.B., 2011. Temporally selective pro- 13728–13733.
cessing of communication signals by auditory midbrain neurons. J. Neurophysiol. Marin, M.L., Tobias, M.L., Kelley, D.B., 1990. Hormone-sensitive stages in the sexual
105, 1620–1632. differentiation of laryngeal muscle fiber number in Xenopus laevis. Development
Fischer, L.M., Catz, D., Kelley, D.B., 1995. Androgen-directed development of the 110, 703–711.
Xenopus laevis larynx: control of androgen receptor expression and tissue differ- Maruska, K.P., 2014. Social regulation of reproduction in male cichlid fishes. Gen.
entiation. Dev. Biol. 170, 115–126. Comp. Endocrinol. 207, 2–12.
Forlano, P.M., Sisneros, J.A., Rohmann, K.N., Bass, A.H., 2015. Neuroendocrine McCarthy, M.M., Nugent, B.M., 2013. Epigenetic contributions to hormonally-mediated
control of seasonal plasticity in the auditory and vocal systems of fish. Front. sexual differentiation of the brain. J. Neuroendocrinol. 25, 1133–1140.
Neuroendocrinol. 37, 129–145. McLean, J., Bricault, S., Schmidt, M.F., 2013. Characterization of respiratory neurons
Frishkopf, L.S., Capranica, R.R., Goldstein Jr., M.H., 1968. Neural coding in the in the rostral ventrolateral medulla, an area critical for vocal production in song-
bullfrog’s auditory systemda teleological approach. Proc. IEEE 56, 969–980. birds. J. Neurophysiol. 109, 948–957.
144 Hormones and Vocal Systems: Insights from Xenopus

Moreno-Gomez, F.N., Sueur, J., Soto-Gamboa, M., Penna, M., 2013. Female frog Tobias, M.L., Kelley, D.B., Ellisman, M., 1995. A sex difference in synaptic efficacy at
auditory sensitivity, male calls, and background noise: potential influences on the the laryngeal neuromuscular junction of Xenopus laevis. J. Neurosci. 15,
evolution of a peculiar matched filter. Biol. J. Linn. Soc. 110, 814–827. 1660–1668.
Morrell, J.I., Kelley, D.B., Pfaff, D.W., 1975. Autoradiographic localization of hormone- Tobias, M.L., Tomasson, J., Kelley, D.B., 1998a. Attaining and maintaining strong
concentrating cells in the brain of an amphibian, Xenopus laevis. II. Estradiol. vocal synapses in female Xenopus laevis. J. Neurobiol. 37, 441–448.
J. Comp. Neurol. 164, 63–77. Tobias, M.L., Viswanathan, S.S., Kelley, D.B., 1998b. Rapping, a female receptive call,
Narins, P.M., Capranica, R.R., 1976. Sexual differences in the auditory system of the initiates male-female duets in the South African clawed frog. Proc. Natl. Acad. Sci.
tree frog Eleutherodactylus coqui. Science 192, 378–380. U.S.A. 95, 1870–1875.
Ode, M., Asaba, A., Miyazawa, E., Mogi, K., Kikusui, T., Izawa, E., 2015. Sex-reversed Tobias, M.L., Barnard, C., O’Hagan, R., Horng, S.H., Rand, M., Kelley, D.B., 2004.
correlation between stress levels and dominance rank in a captive non-breeder Vocal communication between male. Anim. Behav. 67, 353–365.
flock of crows. Horm. Behav. 73, 131–134. Tobias, M.L., Corke, A., Korsh, J., Yin, D., Kelley, D.B., 2010. Vocal competition in
Oliveira, A.G., Coelho, P.H., Guedes, F.D., Mahecha, G.A., Hess, R.A., male Xenopus laevis frogs. Behav. Ecol. Sociobiol. 64, 1791–1803.
Oliveira, C.A., 2007. 5alpha-Androstane-3beta,17beta-diol (3beta-diol), an Tobias, M.L., Evans, B.J., Kelley, D.B., 2011. Evolution of advertisement calls in
estrogenic metabolite of 5alpha-dihydrotestosterone, is a potent modulator of African clawed frogs. Behaviour 148, 519–549.
estrogen receptor ERbeta expression in the ventral prostrate of adult rats. Tupal, S., Rieger, M.A., Ling, G.Y., Park, T.J., Dougherty, J.D., Goodchild, A.K.,
Steroids 72, 914–922. Gray, P.A., 2014. Testing the role of preBötzinger Complex somatostatin neurons
Perez, J., Kelley, D.B., 1996. Trophic effects of androgen: receptor expression and the in respiratory and vocal behaviors. Eur. J. Neurosci. 40, 3067–3077.
survival of laryngeal motor neurons after axotomy. J. Neurosci. 16, 6625–6633. Vignal, C., Kelley, D., 2007. Significance of temporal and spectral acoustic cues for
Pérez, J., Cohen, M.A., Kelley, D.B., 1996. Androgen receptor mRNA expression in sexual recognition in Xenopus laevis. Proc. Biol. Sci. 274, 479–488.
Xenopus laevis CNS: sexual dimorphism and regulation in laryngeal motor nucleus. Watson, J.T., Kelley, D.B., 1992. Testicular masculinization of vocal behavior in
J. Neurobiol. 30, 556–568. juvenile female Xenopus laevis reveals sensitive periods for song duration, rate,
Phoenix, C.H., Goy, R.W., Gerall, A.A., Young, W.C., 1959. Organizing action of and frequency spectra. J. Comp. Physiol. A 171, 343–350.
prenatally administered testosterone propionate on the tissues mediating mating Weintraub, A.S., Kelley, D.B., Bockman, R.S., 1985. Prostaglandin E2 induces
behavior in the female guinea pig. Endocrinology 65, 369–382. receptive behaviors in female Xenopus laevis. Horm. Behav. 19, 386–399.
Picker, M., 1980. Xenopus laevis (Anura: Pipidae) mating systems-a preliminary Wetzel, D.M., Kelley, D.B., 1983. Androgen and gonadotropin effects on male mate
synthesis with some data on the female phonoresponse. South Afr. J. Zool. 15 (3), calls in South African clawed frogs, Xenopus laevis. Horm. Behav. 17, 388–404.
150–158. Wetzel, D.M., Haerter, U.L., Kelley, D.B., 1985. A proposed neural pathway for
Picker, M.D., 1983. Hormonal induction of the aquatic phonotactic response of vocalization in South African clawed frogs, Xenopus laevis. J. Comp. Physiol. A
Xenopus. Behavior 84, 74–90. 157, 749–761.
Potter, K.A., Bose, T., Yamaguchi, A., 2005. Androgen-induced vocal transformation in Wu, K.H., Tobias, M.L., Kelley, D.B., 2001. Estrogen and laryngeal synaptic strength in
adult female African clawed frogs. J. Neurophysiol. 94, 415–428. Xenopus laevis: opposite effects of acute and chronic exposure. Neuroendocri-
Rhodes, H.J., Yu, H.J., Yamaguchi, A., 2007. Xenopus vocalizations are controlled by nology 74, 22–32.
a sexually differentiated hindbrain central pattern generator. J. Neurosci. 27, Wu, K.H., Tobias, M.L., Thornton, J.W., Kelley, D.B., 2003. Estrogen receptors in
1485–1497. Xenopus: duplicate genes, splice variants, and tissue-specific expression. Gen.
Rhodes, H.J., Stevenson, R.J., Ego, C.L., 2014. Male-male clasping may be part of an Comp. Endocrinol. 133, 38–49.
alternative reproductive tactic in Xenopus laevis. PLoS One 9, e97761. Yager, D.D., 1992. A unique sound production mechanism in the pipid anuran Xen-
Ruel, T.D., Kelley, D.B., Tobias, M.L., 1998. Facilitation at the sexually differentiated opus borealis. Zool. J. Linn. Soc. Lond. 104, 351–375.
laryngeal synapse of Xenopus laevis. J. Comp. Physiol. A 182, 35–42. Yamaguchi, A., Kelley, D.B., 2000. Generating sexually differentiated vocal patterns:
Sassoon, D., Kelley, D.B., 1986. The sexually dimorphic larynx of Xenopus laevis: laryngeal nerve and EMG recordings from vocalizing male and female African
development and androgen regulation. Am. J. Anat. 177, 457–472. clawed frogs (Xenopus laevis). J. Neurosci. 20, 1559–1567.
Sassoon, D.A., Gray, G.E., Kelley, D.B., 1987. Androgen regulation of muscle fiber type Yamaguchi, A., Kaczmarek, L.K., Kelley, D.B., 2003. Functional specialization of male
in the sexually dimorphic larynx of Xenopus laevis. J. Neurosci. 7, 3198–3206. and female vocal motoneurons. J. Neurosci. 23, 11568–11576.
Schmidt, R.S., 1992. Neural correlates of frog calling: production by two semi- Yamaguchi, A., Munoz, M.M., Bose, T.O., Oberlander, J.G., Smith, S., 2010. Sexually
independent generators. Behav. Brain Res. 50, 17–30. distinct development of vocal pathways in Xenopus laevis. Dev. Neurobiol. 70,
Segil, N., Silverman, L., Kelley, D.B., 1987. Androgen-binding levels in a sexually 862–874.
dimorphic muscle of Xenopus laevis. Gen. Comp. Endocrinol. 66, 95–101. Yang, E.J., Nasipak, B.T., Kelley, D.B., 2007. Direct action of gonadotropin in brain
Shen, W., Zhang, X.Y., Liu, D.Z., Wang, D.H., 2015. Hormones orchestrated pre- and integrates behavioral and reproductive functions. Proc. Natl. Acad. Sci. U.S.A 104,
post-copulatory sexual traits in male Mongolian gerbils. Physiol. Behav. 143, 2477–2482.
90–96. Yu, H.J., Yamaguchi, A., 2009. 5-HT2C-like receptors in the brain of Xenopus laevis
Simmons, A.M., 2013. “To ear is human, to frogive is divine”: Bob Capranica’s legacy initiate sex-typical fictive vocalizations. J. Neurophysiol. 102, 752–765.
to auditory neuroethology. J. Comp. Physiol. A Neuroethol. Sens. Neural Behav. Yu, H.J., Yamaguchi, A., 2010. Endogenous serotonin acts on 5-HT2C-like receptors
Physiol. 199, 169–182. in key vocal areas of the brain stem to initiate vocalizations in Xenopus laevis.
Simpson, H.B., Tobias, M.L., Kelley, D.B., 1986. Origin and identification of fibers in J. Neurophysiol. 103, 648–658.
the cranial nerve IX-X complex of Xenopus laevis: Lucifer Yellow backfills in vitro. Zornik, E., Kelley, D.B., 2007. Breathing and calling: neuronal networks in the Xenopus
J. Comp. Neurol. 244, 430–444. laevis hindbrain. J. Comp. Neurol. 501, 303–315.
Song, G., Poon, C.S., 2009. Lateral parabrachial nucleus mediates shortening of Zornik, E., Kelley, D.B., 2008. Regulation of respiratory and vocal motor pools in the
expiration and increase of inspiratory drive during hypercapnia. Respir. Physiol. isolated brain of Xenopus laevis. J. Neurosci. 28, 612–621.
Neurobiol. 165, 9–12. Zornik, E., Kelley, D.B., 2011. A neuroendocrine basis for the hierarchical control of
Stacey, N.E., Goetz, F.W., 1982. Role of prostaglandins in fish reproduction. Can. J. frog courtship vocalizations. Front. Neuroendocrinol. 32, 353–366.
Fish. Aquat. Sci. 39, 92–98. Zornik, E., Yamaguchi, A., 2011. Vocal pathway degradation in gonadectomized
Tobet, S.A., Fox, T.O., 1992. Sex differences in neuronal morphology influenced Xenopus laevis adults. J. Neurophysiol. 105, 601–614.
hormonally throughout life. In: Gerall, A.A., Moltz, H., Ward, I.L. (Eds.), Sexual Zornik, E., Yamaguchi, A., 2012. Coding rate and duration of vocalizations of the frog,
Differentiation. Springer, USA, pp. 41–83. Xenopus laevis. J. Neurosci. 32, 12102–12114.
Tobias, M.L., Kelley, D.B., 1987. Vocalizations by a sexually dimorphic isolated larynx: Zornik, E., Katzen, A.W., Rhodes, H.J., Yamaguchi, A., 2010. NMDAR-dependent
peripheral constraints on behavioral expression. J. Neurosci. 7, 3191–3197. control of call duration in Xenopus laevis. J. Neurophysiol. 103, 3501–3515.
Tobias, M.L., Kelley, D.B., 1995. Sexual differentiation and hormonal regulation of
the laryngeal synapse in Xenopus laevis. J. Neurobiol. 28, 515–526.
2.09 Endocrinology of Complex Life Cycles: Amphibians
Robert J Denver, The University of Michigan, Ann Arbor, MI, USA
Ó 2017 Elsevier Inc. All rights reserved.

2.09.1 Most Amphibian Species Have Complex Life Cycles 146


2.09.2 Endocrinology of Metamorphosis 146
2.09.2.1 Overview 146
2.09.2.2 Thyroid Hormone 148
2.09.2.2.1 Role of Thyroid Hormone in Amphibian Development 148
2.09.2.2.2 Thyroid Gland Development and Hormone Production 148
2.09.2.2.3 Thyroid Hormone Metabolism 148
2.09.2.2.4 Thyroid Hormone Transport in Blood 151
2.09.2.2.5 Cellular Uptake of Thyroid Hormone 152
2.09.2.2.6 Mechanisms of Thyroid Hormone Action 152
2.09.2.3 Corticosteroids 154
2.09.2.3.1 Development of and Hormones Produced by Amphibian Interrenal Glands 154
2.09.2.3.2 Roles of Corticosteroids in Amphibian Growth and Development 154
2.09.2.4 Neuroendocrine Control of Metamorphosis 155
2.09.2.4.1 Developmental Expression and Regulation of TSH 156
2.09.2.4.2 Neurohormones Regulating TSH 156
2.09.2.4.3 Other Neurohormones That Regulate TSH Secretion 157
2.09.2.4.4 Negative Feedback Regulation of TSH 158
2.09.2.4.5 Developmental Expression and Regulation of Adrenocorticotropic Hormone 159
2.09.2.4.6 Prolactin and Growth Hormone 159
2.09.3 Role of the Neuroendocrine System in Mediating Environmental Influences on the Timing
of Metamorphosis 160
2.09.4 Future Directions 161
Acknowledgment 162
References 162

Glossary
Adrenocorticotropic hormone (ACTH) Small Premetamorphosis Stage of amphibian larval
polypeptide hormone derived from a larger precursor development when the animal grows but little or no
(proopiomelanocortin) produced by the anterior morphological change occurs; plasma thyroid hormone
pituitary gland that stimulates the adrenal cortex concentrations are low.
(interrenal glands in nonmammalian species) to produce Prolactin (PRL) Simple polypeptide hormone produced
corticosteroids (primarily glucocorticoids). by the anterior pituitary gland that stimulates lactation
Corticotropin-releasing hormone (CRH) Forty-one- in mammals and has antimetamorphic effects in
amino-acid polypeptide produced in the hypothalamus amphibians.
and extrahypothalamic sites that stimulates the release of Prometamorphosis Stage of amphibian larval
ACTH (all vertebrates studied) and TSH (nonmammalian development when metamorphosis begins. Hindlimb
vertebrates) by the anterior pituitary gland. CRH is growth and development is evident externally. The thyroid
a member of a family of peptides of similar size that gland becomes active and secretes thyroid hormone in
include the urocortins, urotensin I, and sauvagine. CRH- response to increasing plasma concentrations of pituitary
like peptides play central roles in developmental, thyroid stimulating hormone (TSH).
behavioral, and physiological responses to stressors. Thyroid stimulating hormone (TSH) Glycoprotein
Growth hormone (GH) Simple polypeptide hormone hormone comprised of two subunits produced by the
produced by the anterior pituitary gland that stimulates anterior pituitary gland that stimulates the production of
growth in all vertebrate species studied. thyroid hormone by the thyroid gland.
Metamorphic climax The final and most rapid phase of Thyrotropin-releasing hormone (TRH) Tripeptide
morphological change when thyroid activity is at its peak. produced in the hypothalamus and extrahypothalamic
Paedomorphosis Reproductive maturity is attained while sites that stimulates the release of TSH by the anterior
in a larval or branchiate form. pituitary gland.

Hormones, Brain, and Behavior, 3rd edition, Volume 2 http://dx.doi.org/10.1016/B978-0-12-803592-4.00024-9 145


146 Endocrinology of Complex Life Cycles: Amphibians

2.09.1 Most Amphibian Species Have Complex Life availability, pond drying or predation – reviewed by Denver,
Cycles 1997b). Thus, body size and stage of development may interact
in complex ways to determine the phenotypic response to
Amphibians exhibit considerable diversity in behavioral, phys- specific environmental variables.
iological, and life history strategies. They are geographically Amphibian larvae exhibit plasticity in the timing of
widespread, occupying a diverse range of habitats. The life metamorphosis and can capitalize on favorable conditions
history strategies of amphibian species include complex life for growth as long as such conditions last (up until a genetically
cycles (e.g., metamorphosis) and direct development (Callery determined upper limit to the length of the larval period)
et al., 2001). The majority of amphibian species exhibit (Newman, 1992; Rudolf and Rodel, 2007). Such plasticity
complex life cycles, and thus have two very different life stages may permit amphibian larvae to match their phenotype
that are affected differently by environmental factors. Most (morphology, physiology, metamorphic timing) to prevailing
anuran (frog) larvae are aquatic, and tadpoles are found in environmental conditions. Animals capable of phenotypic
a wide variety of habitats, ranging from water-filled crevices plasticity may have a higher probability of surviving in unpre-
in rocks, logs, or leaves to larger ponds or streams. Most then dictable habitats compared with those with a genetically fixed,
undergo morphological, biochemical, and physiological trans- or canalized phenotype (Stearns, 1989; Newman, 1992;
formation into adults, which are sensitive to different environ- Van Buskirk, 2002; Gomez-Mestre and Buchholz, 2006).
mental variables than larvae, due to this shift in habitat The upper and lower limits to the length of the larval period
(Duellman and Trueb, 1994). Some amphibians have lost are determined by genetic factors that are subject to natural
the larval form and develop directly into the adult morphology selection. The plasticity of larval period length within these
(direct development); others do not metamorphose but repro- limits is also subject to natural selection and is influenced at
duce in the aquatic habitat while retaining the larval both proximate and ultimate levels by the environment. While
morphology (paedomorphosis). metamorphic timing is determined by both genetic and envi-
Metamorphosis is a stage of many amphibian life cycles ronmental factors, its expression depends on the development
characterized by dramatic morphological transformation that and activity of endocrine glands and the actions of the
is accompanied by a transition in ecological niche and behav- hormones that these glands produce (see below).
ioral mode. Amphibians that undergo a metamorphosis Among the most extreme evolutionary modifications of the
exhibit strong variation, both between and within species, in ancestral, complex life history is paedomorphosis. Most
the duration of the larval period (Wilbur and Collins, 1973; amphibian larvae undergo a metamorphosis to an adult form
Werner, 1986). Larvae encounter diverse ecological conditions before becoming sexually mature. Some species of urodele
during development. Variation in abiotic factors (e.g., water amphibians (e.g., salamanders, newts) exhibit paedomor-
availability, temperature, photoperiod) as well as biotic factors phosis, where reproductive maturity is attained while in a larval
(e.g., intra- and interspecific competition, predation) can or branchiate form. Paedomorphosis refers to the retention of
interact in complex ways to influence larval growth and devel- juvenile characteristics in sexually mature adults (Gould,
opment (Semlitsch, 1987; Sredl and Collins, 1992; Rowe and 1977). Paedomorphosis can either be obligate or facultative
Dunson, 1995; Taylor and Scott, 1997; Alvarez and Nicieza, depending on the species. Obligate paedomorphs never
2002; Relyea, 2002; Downie et al., 2004; Relyea, 2007). The undergo metamorphosis and remain in an aquatic habitat their
timing of metamorphosis is a central amphibian life history entire lives (e.g., Necturus, Proteus, Amphiuma, Ambystoma mexi-
trait that likely reflects the quality and relative permanence of canum – axolotl). Facultatively paedomorphic species can
the larval habitat. Species that breed in predictable habitats either become paedomorphic and remain in the aquatic
(i.e., permanent or semipermanent lakes and ponds) tend to habitat, or metamorphose and move into the terrestrial
have longer larval periods. Species that breed in unpredictable environment where they become sexually mature (e.g., Ambys-
habitats (i.e., ephemeral pools) generally have much shorter toma tigrinum, Ambystoma talpoideum, Ambystoma gracile,
larval periods (Denver, 1997b). Notophthalmus viridescens) (Duellman and Trueb, 1994). The
Wilbur and Collins (1973) suggested that there is developmental ‘decision’ to become paedomorphic or to meta-
a threshold of minimum body size that must be reached before morphose in facultative species depends on the prevailing envi-
metamorphosis is possible and that larval growth rates deter- ronmental conditions rather than the animal’s genotype
mine the timing of metamorphosis after this minimum size (Harris, 1987; Semlitsch, 1987; Licht, 1992; Jackson and
has been attained. Werner (1986) added mortality risk in the Semlitsch, 1993; Denoel and Poncin, 2001) and may be
larval and adult habitat to the list of factors that ultimately controlled by the interplay of antagonistic hormonal pathways
influence metamorphosis. The effects of environmental factors (Wakahara, 1994, 1996; Rosenkilde and Ussing, 1996). The
may differ depending on the animal’s stage of development. reader is referred to a recent review by Johnson and Voss
These observations led Day and Rowe (2002) to incorporate (2013) for a detailed discussion of the hormonal basis for
developmental thresholds into the Wilbur–Collins model. paedomorphosis in salamanders.
Environmental factors that influence growth rate or mortality
risk therefore should alter the timing of metamorphosis, and
the effects of the environment may be influenced by the stage 2.09.2 Endocrinology of Metamorphosis
of development that has been achieved. For example, the
2.09.2.1 Overview
same factor may be inhibitory to growth if present early in
the larval phase or stimulatory to development if present Hormones orchestrate the diverse morphological and physio-
during metamorphosis (e.g., population density, food logical changes that occur during metamorphosis and also
Endocrinology of Complex Life Cycles: Amphibians 147

function as mediators of environmental effects on develop- thyroidectomy results in metamorphic stasis (Dodd and
ment. A striking characteristic of amphibian metamorphosis Dodd, 1976; Kikuyama et al., 1993). Neurohormones
is that a single signaling molecule produced by the thyroid produced in the hypothalamus control hormone biosynthesis
gland (thyroid hormone – TH) can orchestrate the entire suite and secretion by the pituitary gland, and the hypothalamus
of molecular, biochemical, and morphological changes. mediates the interaction between the external and internal
Gudernatsch (1912) first showed that the vertebrate thyroid environments, and the production of hormones that control
gland contained a factor that could induce precocious meta- metamorphosis.
morphosis if fed to tadpoles. This compound, later identified The work of William Etkin laid much of the foundation for
as 3,5,30 ,50 -tetraiodothyronine (thyroxine) (Kendall, 1915; our current understanding of the endocrine control of meta-
Harrington, 1926; Harrington and George, 1927) and referred morphosis. Etkin, (1968) proposed a model for the hormonal
to as thyroid hormone (TH), is now known to be the primary changes that occur during amphibian metamorphosis. He also
hormone controlling amphibian metamorphosis. Thyroid coined the terms in common use today among amphibian
hormone is required for amphibian metamorphosis (Brown endocrinologists for describing the stages of anuran develop-
and Cai, 2007); the hormone initiates gene expression ment: ‘premetamorphosis,’ when the larvae grow but little or
programs in diverse tissues that lead to cell proliferation, death, no morphological change occurs and plasma TH concentra-
differentiation, or migration (Brown and Cai, 2007). While tions are low; ‘prometamorphosis,’ when hindlimb growth
hormones produced by the anterior pituitary gland and the accelerates and plasma TH concentration rises; ‘metamorphic
interrenal glands (amphibian homologs of the mammalian climax,’ the final and most rapid phase of morphological
adrenal cortex; corticosteroids – CS) influence the rate of meta- change when thyroid activity is at its peak (Dodd and Dodd,
morphosis by controlling TH production and action on target 1976; White and Nicoll, 1981; Table 1).
tissues, exogenous TH alone can induce the entire suite of The objective of the following section is to describe the cast
tissue transformations. Furthermore, chemical or surgical of endocrine characters that interact to control metamorphosis.

Table 1 A comparison of three of the most widely cited staging tables for postembryonic, feeding stages of anuran larvae

NF a staging for Major, common diagnostic


Xenopus laevis features/morphological changes TK staging b Gosner staging c Terminology of Etkin d

1–45 Nonfeeding stages (comparable to 1–25


Shumway stagese 1–24)
46 I 26 Premetamorphosis
47–48 Feeding begins II 27
49–50 III 28
51 IV 29
V 30
52 Foot paddle stages
VI 31
53 VII 32
VIII 33
54 IX 34
X 35
55 Hindlimb stages XI 36 Prometamorphosis
XII 37
56 XIII 38
57–58 XIV–XVI 39–40
59 Tadpole reaches maximum length XVII 40
60 XVIII 41
XIX
61 XX
62 Rapid tail resorption begins, front XXI 42 Climax
limbs erupt*
63 XXII 43
64 XXIII 44
65 Stump of tail remains XXIV 45
66 Tail completely resorbed, juvenile frog XXV 46

Note: Table is derived from similar tables published by Nieuwkoop and Faber (1956), Dodd and Dodd (1976), and Kikuyama et al. (1993) with the
addition of Gosner staging. Note that the table is modified somewhat with respect to the table published by Kikuyama et al. (1993) with deference to
the comparison between Xenopus laevis and the staging of Rana pipiens (Taylor and Kollros, 1946) made by Nieuwkoop and Faber (1956).
Comparison of Taylor and Kollros (1946) with Gosner (1960) staging tables is based on that of Gosner (1960).
*In Xenopus species the front limbs do not erupt, but instead develop outside of the body cavity.
a
Stages of Nieuwkoop and Faber (1956).
b
Stages of Taylor and Kollros (1946).
c
Stages of Gosner (1960).
d
Terminology of Etkin (1968).
e
Stages of Shumway (1940).
148 Endocrinology of Complex Life Cycles: Amphibians

For each endocrine axis involved in metamorphosis I will first sensitive and quantitative methods for determining plasma
examine its developmental schedule. This should allow predic- TH concentrations. Early methods relied on the determination
tions of when the endocrine system is sufficiently developed to of protein-bound iodide to estimate plasma TH titers (Just,
allow the animal to become competent to respond to the 1972). Subsequently, sensitive and specific radioimmunoas-
external environmental. I will also examine the multiple levels says (RIAs) were developed that allowed determinations of
at which the activity and functioning of each endocrine axis can plasma thyroxine (T4; the primary product of the thyroid
be regulated. The goal, which is addressed in Section 2.09.3 on gland) and 3,5,30 -triiodothyronine (T3; derived from T4 by
integration, is to understand how the endocrine system deter- monodeiodination in target tissues) concentrations during
mines the timing of metamorphosis and mediates environ- metamorphosis. These studies confirmed earlier studies and
mental effects on amphibian development. the predictions of Etkin by demonstrating low-to-nondetect-
able plasma TH concentrations during premetamorphosis,
increasing concentrations during prometamorphosis, and
2.09.2.2 Thyroid Hormone
a dramatic peak at metamorphic climax (Figure 1(b); Leloup
2.09.2.2.1 Role of Thyroid Hormone in Amphibian and Buscaglia, 1977; Miyauchi et al., 1977; Regard et al., 1978;
Development Mondou and Kaltenbach, 1979; Suzuki and Suzuki, 1981;
Perhaps the most striking characteristic of amphibian meta- Weil, 1986; Niinuma et al., 1991a; Weber et al., 1994; Denver,
morphosis, from the perspective of hormonal control, is that 1998a; Krain and Denver, 2004).
a single signaling molecule, produced by a highly restricted Because of the difficulty of obtaining blood from small
group of cells (the thyroid epithelial cells), can orchestrate tadpoles for analysis by RIA, until recently only those species
the entire suite of molecular, biochemical, and morphological with tadpoles large enough to obtain a serum sample were
changes. Depending on the tissue, TH can induce cell prolifer- analyzed. Thus, most blood measurements have been done
ation, cell death, differentiation, or migration. Target cells for on ranid species (e.g., Rana catesbeiana; Rana clamitans;
TH are now known to activate both similar and different sets however, Leloup and Buscaglia (1977) and Tata et al. (1993)
of genes according to the concentration of this single signaling measured THs in plasma pools of Xenopus laevis; see also
molecule. Specific tissues exhibit different dose sensitivities to Buscaglia et al. (1985) for measures of plasma T3 and T4 in
TH, and the challenge for investigators studying the molecular other Xenopus spp.). In species with small tadpoles, develop-
basis of TH action during metamorphosis is to determine how mental changes in TH content of whole bodies and individual
and why individual tissues respond differently to the hormone tissues have been determined. These analyses have shown that
and exhibit differential dose responses (see Brown and Cai, changes in whole body TH content in the smaller species essen-
2007). Thyroid hormone regulation of metamorphosis has tially parallel changes observed in the plasma of tadpoles of the
been best studied in anuran amphibians, which is the primary larger species (Bufo japonicus (Niinuma et al., 1991a); Spea
focus of this chapter, but it is important to note that TH is also hammondii (Denver, 1993, 1998a); X. laevis (Krain and Denver,
the primary morphogen in metamorphosing urodele (sala- 2004); Bufo marinus (Weber et al., 1994)). The peak in whole-
mander) species (reviewed by Johnson and Voss, 2013). body T3 and T4 coincides with peak uptake of 131I in B. japonicus
(Niinuma et al., 1991a). Thus, it is likely that determination of
2.09.2.2.2 Thyroid Gland Development and Hormone whole-body hormone content provides a reasonable estimate
Production of physiological changes in TH production in species for which
The thyroid gland develops early in the amphibian embryo blood samples are unobtainable.
when the anlage consists of a thickening of the pharyngeal
epithelium; these cells are capable of synthesizing small iodo- 2.09.2.2.3 Thyroid Hormone Metabolism
proteins (reviewed by Dodd and Dodd, 1976; Regard et al., The major product of the amphibian thyroid gland is T4 with
1978). The gland matures functionally at the time of hatching minor amounts of T3 produced (Rosenkilde, 1978; Buscaglia
when it separates into two distinct lobes and is essentially et al., 1985). The result is that plasma T4 concentration tends
completely developed by late premetamorphosis/early prome- to be an order of magnitude greater than T3 (Regard et al.,
tamorphosis (Nieuwkoop and Faber, 1956; Saxen et al., 1978; Larras-Regard et al., 1981). The only case where this rela-
1957a,b; Kaye, 1959, 1961; Dodd and Dodd, 1976; Regard tionship may not hold is for X. laevis where the reported plasma
et al., 1978). Multiple measures of thyroid activity including concentrations of T3 and T4 are very similar, and the T3/T4 ratio
radioiodine uptake, gland ultrastructure, and plasma concen- may even exceed one at metamorphic climax (Leloup and
tration or tissue content of THs show that thyroid activity Buscaglia, 1977; Buscaglia, 1985). Measures of tissue content
increases markedly during prometamorphosis (Table 1; of T4 and T3 in various species show that the two hormones
Figure 1), peaks at metamorphic climax, and declines there- are present in roughly similar amounts (Niinuma et al.,
after to reach an ‘adult’ level of activity (Kaye, 1959, 1960; 1991a; Weber et al., 1994; Denver, 1997a, 1998a). Although
Dodd and Dodd, 1976; Regard et al., 1978; Kikuyama et al., a comprehensive analysis of both blood concentrations and
1993). Ultrastructural analyses show a dramatic increase in tissue contents of THs has not been done for any species, it is
thyroid follicular cell height during prometamorphosis with likely that the higher T3:T4 ratio in tissues compared with
a peak at metamorphic climax that corresponds to the peak T3:T4 ratio in blood reflects high tissue 50 -monodeiodinase
in plasma concentrations (and tissue content) of THs (Dodd activity (see below).
and Dodd, 1976; Regard et al., 1978). An important point of control of TH bioactivity is at the
When Etkin proposed his endocrine-based model for target tissues, where monodeiodinase enzymes convert T4 to
metamorphosis, investigators at the time did not have T3, or inactivate T4 and T3 (Figure 2). The monodeiodinases
Endocrinology of Complex Life Cycles: Amphibians 149

Figure 1 Changes in thyroid hormones and pituitary thyroid stimulating hormone (TSH) throughout metamorphosis in Xenopus laevis. (a) Tadpoles
at Nieuwkoop–Faber (NF) stages 52, 58, and 64 (photos by David Bay). (b) Changes in whole-body thyroxine (T4) and 3,5,30 -triiodothyronine (T3)
throughout metamorphosis. Asterisks denote stage at which there was a statistically significant increase in whole-body hormone content. (Modified
from Krain, L.P., Denver, R.J., 2004. Developmental expression and hormonal regulation of glucocorticoid and thyroid hormone receptors during
metamorphosis in Xenopus laevis. J. Endocrinol. 181 (1), 91–104.) (c) and (d) Changes in TSHb and the common glycoprotein hormone subunit
(a-GSU) mRNAs in the pituitary gland of X. laevis throughout metamorphosis as determined by semiquantitative RT-PCR. Different letters indicate
significant differences among means. (Modified from Manzon, R.G., Denver, R.J., 2004. Regulation of pituitary thyrotropin gene expression during
Xenopus metamorphosis: negative feedback is functional throughout metamorphosis. J. Endocrinol. 182 (2), 273–285.)

catalyze two basic reactions: a 50 monodeiodination (outer expressed in the thyroid gland and is strongly induced by TH
ring) that results in bioactivation and a 5 monodeiodination in the intestine during metamorphosis via a TH response
(inner ring) that results in bioinactivation of the substrate, T4 element (TRE) located in the promoter of the gene (Fujimoto
or T3. There are three types of vertebrate deiodinases (types I, et al., 2012).
II, and III) that differ in their substrate specificity, kinetics, Three deiodinase genes have been isolated in amphibian
and sensitivity to inhibitors. Type I catalyzes both 5 and 50 , species (Brown, 2005). The Dio2 (type II) and Dio3 (type III)
type II 50 , and type III 5 deiodination (St Germain et al., genes exhibit tissue-specific and developmental stage-specific
2009). Types II and III, but not type I, enzyme activities have expression patterns (Becker et al., 1997; Cai and Brown,
been detected in tadpole tissues, and although frogs have 2004; Brown, 2005; Duarte-Guterman et al., 2012). The expres-
a type I (Dio1) gene, little is known about its expression or sion patterns correlate with the asynchronous tissue
function (Becker et al., 1997; Dubois et al., 2006; Kuiper morphogenesis and the roles that the deiodinases play in
et al., 2006). An iodotyrosine deiodinase that is involved modulating intracellular T3 concentration during metamor-
with recycling of iodine from mono- and diiodotyrosine is phosis (Brown, 2005; St Germain et al., 2009). In many cells
150 Endocrinology of Complex Life Cycles: Amphibians

Figure 2 Central and peripheral organization of the thyroid and stress endocrine axes controlling amphibian metamorphosis. Shown is a schematic
representation of the hypothalamo–pituitary–thyroid (HPT) and hypothalamo–pituitary–interrenal (HPI; stress) axes in amphibian tadpoles, their regu-
lation by input from the external environment, transduction of this input by neural and neuroendocrine pathways, and synergistic interactions among
thyroid hormones and corticosteroids in target cells leading to the promotion of metamorphosis. The two endocrine axes are controlled centrally by
CRH which acts on the anterior pituitary gland (AP) to stimulate the release of thyrotropin (TSH) and corticotropin (ACTH). TSH acts on the thyroid
gland to stimulate release of thyroxine (T4) and 3,5,30 -triiodothyronine (T3). Thyroid hormones are transported in the blood bound by serum binding
proteins (transthyretin, thyroxine binding globulin, and albumin). ACTH acts on interrenal cortical cells in the interrenal glands to stimulate biosyn-
thesis and release of glucocorticoids which are transported in the blood bound to corticosteroid binding globulin. Cellular uptake of T3 and T4 is ach-
ieved by organic anion, monocarboxylate, and amino acid transporters; there is also evidence that thyroid hormones may enter cells bound to
transthyretin via a receptor-mediated process. Glucocorticoids enter cells by passive diffusion across the plasma membrane. Upon entering the cell,
thyroid hormone is bound by cytosolic binding proteins, some of which (the monodeiodinases) convert the hormone to either active (T3; deiodinases
types I and II) or inactive forms (reverse T3 (rT3), diiodothyronine (T2); deiodinase type III). Thyroid hormone receptors (TR) form heterodimers with
retinoid X receptors (RXR) and are bound to DNA in the unliganded form where they actively repress gene transcription. Upon thyroid hormone
Endocrinology of Complex Life Cycles: Amphibians 151

both enzymes may be expressed, and the relative expression gene based on its rapid induction in response to T3, and accu-
levels may establish a type of push–pull mechanism that regu- mulation of the mRNA in response to T3 treatment is resistant
lates intracellular T3 concentration (St Germain et al., 2009). to protein synthesis inhibition; i.e., it does not require a new
Alternatively, in some tissues the two genes show different protein to be made to activate the gene, only preexisting TRs
temporal dynamics, leading to hormone inactivation or activa- (Wang and Brown, 1993; St Germain et al., 1994; Becker
tion at different developmental stages. et al., 1995; Denver et al., 1997; Kawahara et al., 1999; Hogan
For example, Dio3 mRNA is expressed in several cell types in et al., 2007; Das et al., 2009; Duarte-Guterman et al., 2012).
tadpole tail, but not in tail muscle cells (Berry et al., 1998); both Thyroid hormone positively regulates 50 deiodinase
Dio3 mRNA and 5 deiodinase activity increase during late activity and Dio2 mRNA in tadpoles (Buscaglia, 1985; Brown,
prometamorphosis (Nieuwkoop-Faber (NF) stages 59–61; 2005; Hogan et al., 2007; Duarte-Guterman et al., 2012).
refer Table 1 for a definition of developmental stages) but However, unlike Dio3, which is an early T3 response gene,
then decline sharply at metamorphic climax (St Germain Dio2 exhibits delayed response kinetics; the gene was not iso-
et al., 1994; Brown et al., 1996). This pattern of Dio3 expression lated in screens for early response, direct TR target genes in Xen-
may protect the tadpole tail, an essential locomotory organ, opus tadpole tissues (Brown, 2005; Das et al., 2006; Buchholz
from premature resorption (Brown, 2005). By contrast, Dio2 et al., 2007; Bonett et al., 2010). The dependence of Dio2 expres-
expression, which occurs mainly in tail fibroblasts, is undetect- sion on TH may vary among tissues. Tissues in which cell
able before late prometamorphosis at which time expression proliferation occurs as an early response to TH constitutively
increases markedly and is maintained through the end of meta- express relatively high levels of Dio2 (e.g., neurogenic zones of
morphosis (Cai and Brown, 2004). This late expression of Dio2 the brain and spinal cord, limb buds); whereas, in tissues that
is hypothesized to generate bioactive T3 at an appropriate devel- transform later, Dio2 is upregulated by TH (Cai and Brown,
opmental stage to accelerate tail resorption. Neither Dio2 nor 2004). Treatment of early prometamorphic tadpoles with T4
Dio3 mRNAs are expressed in tail muscle cells (Berry et al., can induce cell proliferation in these tissues, which can be
1998; Bonett et al., 2010); nonmuscle tail cells may inactivate blocked by the deiodinase inhibitor iopanoic acid (Cai and
T4 during pre- and prometamorphosis to protect tail muscle Brown, 2004). Dio2 mRNA in brain and spinal cord declines
cells from apoptosis and subsequently generate high local at metamorphic climax, as does TH-dependent cell
concentrations of T3 to promote tail muscle cell apoptosis at proliferation (Cai and Brown, 2004; Denver et al., 2009).
metamorphic climax. While the decline in Dio2 mRNA may be permissive for the
Tissue transformations during metamorphosis are asynchro- reduction in TH-dependent cell proliferation in the brain, it
nous: some tissues respond early to low plasma concentrations does not alone explain why the brain becomes refractory to
of TH (e.g., hindlimb, brain), while other tissues respond later TH action since treatment with T3 could not increase cell
in development and require high TH concentration (e.g., intes- proliferation (which normally declines) as the animals
tine, tail – discussed above). The expression patterns of the approach metamorphic climax (Denver et al., 2009). The
monodiodinase genes may play a key role in establishing tissue decline in cell proliferation is likely due to processes, probably
competence to respond to the TH signal. For example, for tissues under TH control, that lead to a reduction in the stem cell/
that respond early in metamorphosis to TH such as the retina progenitor pool in the ventricular/subventricular zones of the
and hindlimb, Dio2 expression was high during early prometa- tadpole brain. Dio2 expression then appears in late-responding
morphosis but declined at metamorphic climax. The importance tissues such as the intestine, tail, and anterior pituitary and
of 50 deiodinase activity for hindlimb development is supported may be induced at this time by rising plasma titers of TH
by findings that T4 has no effect on the hindlimb in the presence (Huang et al., 2001; Cai and Brown, 2004; Manzon and
of the deiodinase inhibitor iopanoic acid (Brown, 2005). Dio2 Denver, 2004; discussed more below). Physiological roles for
mRNA expression showed a progressive decline in the brain tissue monodeiodinases in the timing of metamorphosis are
throughout metamorphosis, while brain Dio3 mRNA increased supported by experiments with iopanoic acid and transgenesis
during late prometamorphosis and metamorphic climax overexpression of Dio3 (Buscaglia, 1985; Galton, 1989; Becker
(Hogan et al., 2007). Thyroid hormone induces cell proliferation et al., 1997; Huang et al., 1999, 2001; Marsh-Armstrong et al.,
in the early prometamorphic tadpole brain, but cells of the 1999; Cai and Brown, 2004).
neurogenic zone become refractory to TH action on cell prolifer-
ation as metamorphic climax approaches, which may be 2.09.2.2.4 Thyroid Hormone Transport in Blood
explained by the temporal patterns of Dio2 and Dio3 expression Thyroxine synthesized by thyroid follicular cells diffuses into
and actions (Cai and Brown, 2004; Denver et al., 2009). the bloodstream where it is reversibly bound by plasma
Thyroid hormone regulates the expression of the Dio2 and proteins that transport the hormone from the site of produc-
Dio3 genes. The Dio3 gene appears to be a direct T3 response tion to its target tissues (Figure 2). Two plasma binding

=
binding to TR gene transcription is derepressed and activated. Upon entering the cell glucocorticoids bind to corticosteroid receptors (glucocorticoid
receptor (GR) or mineralocorticoid receptor (MR)) that are located in the cytosol bound to heat shock proteins (the ‘foldosome’). Hormone binding
causes a conformational change in the receptor, the release of heat shock proteins, and dimerization and translocation of receptors to the nucleus
where they bind DNA to activate or repress target genes. When cells are exposed to low concentrations of thyroid hormone plus glucocorticoids,
genes such as the TRs, deiodinase type 2, and the thyroid hormone-inducible transcription factor Klf9 are activated in a synergistic manner. This
leads to enhanced sensitivity of cells to the actions of thyroid hormone, which accelerates metamorphosis. þ, indicates an increase; , indicates
a decrease in the regulated variable.
152 Endocrinology of Complex Life Cycles: Amphibians

proteins that bind T4 and T3 with moderate to high affinities has been demonstrated in tadpole red blood cells (Galton
have been identified in vertebrates. Thyroxine-binding globulin et al., 1986; Yamauchi et al., 1989; Murata and Yamauchi,
(TBG) binds T4 with high affinity and low capacity but is found 2005). The genes that encode TH transport proteins could be
only in large, eutherian mammals (Power et al., 2000). Trans- important loci for the modulation of the timing of
thyretin (TTR; also known as prealbumin) is found in all verte- metamorphosis.
brates and it binds T4 with moderate affinity and intermediate There are three general classes of proteins that allow for
capacity. Both TBG and TTRs can also bind T3, although in active uptake of TH by cells: the organic anion transporters
most cases with 10 times lower affinity than T4 (Power et al., (OATC), monocarboxylate transporters (MCT), and the L
2000). However, the situation in amphibia is the reverse, where amino acid permeases (LAT) (Ritchie et al., 1999, 2003;
TTR binds T3 with greater affinity than T4 (Yamauchi et al., Friesema et al., 2005; Jansen et al., 2005; Visser et al., 2008).
1993, 1998, 1999, 2000). The two primary sites for TTR expres- Orthologs of OATC, MCT, and LAT genes have been isolated
sion in vertebrates are the liver and the choroid plexus from frogs, and patterns of expression throughout metamor-
(although it is expressed at other sites (Power et al., 2000)). phosis have been described (Shi and Brown, 1993; Liang
In amphibians TTR is expressed primarily in the liver (Power et al., 1997; Connors et al., 2010; Choi et al., 2015a). Only
et al., 2000). An essential function of TTR is its interaction the amino acid permeases have so far received attention with
with retinol-binding protein, which acts as a carrier for all- regard to their function during tadpole metamorphosis. The
trans-retinol in the blood. The functional significance of T3-inducible gene IU12 from X. laevis intestine (Shi and Brown,
this interaction is not known, but it is intriguing that T3 and 1993; Liang et al., 1997) encodes a subunit of a heterodimeric
9-cis-retinoic acid (which is a metabolite of all-trans-retinol) amino acid permease complex (System L (Torrents et al., 1998;
serve as ligands for the TR-retinoid X receptor (RXR) Ritchie et al., 2003)). This permease complex efficiently trans-
heterocomplex that regulates TH target genes. Serum albumin ports T3 and T4 when expressed in the Xenopus oocyte expres-
also binds T3 and T4 in many species with low affinity and sion system, but is inhibited by reverse T3 (Ritchie et al.,
high capacity, and Power et al. (2000) suggested that 1999). Overexpression of System L in Xenopus oocytes
albumin might be the principal T4-binding protein in increased cytoplasmic and nuclear delivery of THs from the
amphibia. external medium and enhanced transcriptional activation by
Circulating TTR protein is present in tadpoles during preme- TH receptors (TRs) (Ritchie et al., 2002). By contrast, blocking
tamorphosis and prometamorphosis when thyroid activity is endogenous System L activity in mammalian cells reduced
increasing, but declines at metamorphic climax (Yamauchi both TH uptake and TR function (Ritchie et al., 2003). The
et al., 1998, 2000; Prapunpoj et al., 2000). The free hormone fact that IU12 is a T3-inducible gene suggests that it could
hypothesis (Mendel, 1989; Ekins, 1990) leads to the prediction play a role in mediating T3 uptake by cells during tadpole meta-
that TTR during pre- and early prometamorphosis serves to morphosis (Liang et al., 1997). Choi et al. (2015a) recently
reduce the free fraction of TH in blood thus limiting bioavail- showed that forced expression of LAT1 in tadpole tail muscle
ability. Conversely, hormone-binding proteins can serve as cells accelerated T3-induced cell loss.
a reservoir for hormone in the blood; TTR could therefore Upon entering cells THs may bind to a series of intracel-
help to sustain increasing plasma TH concentrations prior to lular binding proteins, termed cytoplasmic TH binding
the acceleration of thyroid gland activity induced by rising proteins (CTHBPs) that represent several classes of multifunc-
plasma TSH titers. The TTR concentration in the blood declines tional proteins. These proteins may have a variety of enzy-
at metamorphic climax when plasma TH concentration is matic activities; for example, three genes isolated from
maximal. The continued rise in TH synthesis by the thyroid X. laevis code for: (1) a cytosolic aldehyde dehydrogenase
gland, paired with a decline in TTR, could result in an increase (ALDH1) which catalyzes the formation of retinoic acid
in the free hormone fraction in the blood. At the same time, the (Yamauchi and Tata, 1994), (2) an M2 pyruvate kinase
rate of clearance of T3 from the circulation would likely (PKM2) (Shi et al., 1994), and (3) protein disulfide isomerase
increase. However, because the thyroid synthetic rate is high (PDI; induced by T3 in tadpole brain (Denver et al., 1997)),
at metamorphic climax, total plasma T3 concentration which catalyzes disulfide bond formation, and human PDI
continues to rise. Thus, one would predict that, not only does has been shown to bind TH with high affinity (Cheng et al.,
the hormone production rate increase at metamorphic climax, 1987; Yamauchi et al., 1987). These CTHBPs may transport
but so does the proportional availability of T3 to the target THs within the cytoplasm to the nucleus to bind to TRs, or
tissues. To my knowledge, T3 or T4 clearance rates have not alternatively, they may serve as chelators to limit the cellular
been calculated in tadpoles at different stages of development. free TH concentration (Shi, 2000a). It is also possible that
Based on TTR expression profiles one would predict that clear- TH may regulate the enzymatic activity of these proteins
ance rates would be lower during prometamorphosis (Ashizawa and Cheng, 1992). Choi et al. (2015a) recently
compared with premetamorphosis or metamorphic climax. showed that forced expression in tadpole tail muscle cells of
Furthermore, given the lower affinity of TTR for T4 compared PKM2 and another CTHBP that has not been previously char-
with T3, one would predict that the clearance rate for T4 would acterized in amphibians, the mu-crystallin homolog CRYM
be higher than that of T3. accelerated T3-induced cell loss, supporting that these
proteins facilitate TH action.
2.09.2.2.5 Cellular Uptake of Thyroid Hormone
Tadpole cells have the capacity to actively take up TH (see Krain 2.09.2.2.6 Mechanisms of Thyroid Hormone Action
and Denver, 2004), and this activity may be regulated during Tadpoles become competent to respond to exogenous TH at
metamorphosis. Saturable, carrier-mediated uptake of THs the time of hatching (Tata, 1968). This establishment of
Endocrinology of Complex Life Cycles: Amphibians 153

competence to respond to the hormone likely depends on the by the inability to delete, or knockdown, the TRs. Recent tech-
expression of TRs (Shi et al., 1996). TRs are ligand-activated nological developments using transcription activator-like
transcription factors that belong to the steroid hormone effector nucleases (TALENS) for genome editing have shed light
receptor superfamily (Mangelsdorf et al., 1995). There are on the role of TRa in tadpole development and metamor-
two TR genes, termed a and b, in all vertebrates studied to phosis. Two groups recently used TALENS to introduce muta-
date; owing to its pseudotetraploidy, X. laevis possesses four tions into the TRa locus in Xenopus tropicalis (Choi et al.,
TR genes, two as and two bs (Buchholz et al., 2006). The two 2015b; Wen and Shi, 2015). Mutations in TRa led to enhanced
X. laevis TRa genes each appear to give rise to single, unique growth of premetamorphic tadpoles and the acceleration of the
proteins; whereas, alternative mRNA splicing of TRb transcripts onset of metamorphosis (Wen and Shi, 2015). Although the
can give rise to two different receptor isoforms for each TRb timing of the initiation of metamorphosis was accelerated,
gene (Buchholz et al., 2006). these mutant tadpoles were resistant to exogenous T3 and
The TRa genes are first expressed shortly after hatching in exhibited a delay in spontaneous metamorphosis. Gene expres-
X. laevis, and their expression rises during premetamorphosis sion analyses showed increased baseline mRNA levels, but the
and remains high throughout metamorphosis (Baker and loss of T3 response in TR target genes (Choi et al., 2015b; Wen
Tata, 1990; Yaoita and Brown, 1990; Banker et al., 1991; and Shi, 2015). Together, these findings support that the apor-
Kawahara et al., 1991). It has been hypothesized that the early eceptor normally represses metamorphic genes in larvae, and
expression of TRa establishes hormone responsiveness of when removed shortens the larval period. However, liganded
tadpole tissues (Baker and Tata, 1990; Shi et al., 1996). TRb TRa is required for establishing tissue responsiveness to the
mRNA is not detected until early prometamorphosis, but its hormone during metamorphosis, and so these studies provide
expression increases during prometamorphosis in parallel critical support for the dual function model for TR actions
with TH synthesis (Yaoita and Brown, 1990; Kawahara during metamorphosis (Buchholz et al., 2006; Shi et al.,
et al., 1991; Baker and Tata, 1992; Kanamori and Brown, 2012; Grimaldi et al., 2013a). Future studies using TALENS
1992). Several studies have shown that the TR genes are upre- and CRISPR/Cas9 genome editing should shed further light
gulated by T3 in X. laevis and R. catesbeiana (Yaoita and Brown, on the different roles of the TR subtypes in tadpole
1990; Kawahara et al., 1991; Schneider and Galton, 1991; development.
Helbing et al., 1992) (a phenomenon termed ‘autoinduction’; Thyroid hormone receptors function as dimers; that is, the
see Tata et al., 1993). A thyroid hormone response element to DNA consensus sequences that TRs bind to are six nucleotides
which TRs can bind and regulate transcription has been in length and are referred to as ‘half sites.’ Two of these half
identified in the X. laevis TRbA gene (Ranjan et al., 1994; sites comprise a TRE (Williams and Brent, 1995). These TREs
Machuca et al., 1995). Autoinduction may require the upregu- can be located within the promoter, within the structural part
lation of accessory transcription factors such as the immediate of the gene, or upstream of the transcription start site. Homo-
early, TH-inducible gene Krüppel-like factor 9 (Klf9; dimers of TRa or TRb can form on most TREs, but the preferred
aka basic transcription element binding protein 1; BTEB1) configuration appears to be as a heterodimer with retinoid X
(Bagamasbad et al., 2015; Hu et al., 2016). receptor (RXR) (Wong and Shi, 1995; PuzianowskaKuznicka
A central role for TRs in metamorphosis is supported by et al., 1997). TR-RXR heterodimers bind DNA and
transgenic studies in X. laevis. For example, transgenic expres- transactivate TRE-containing genes much more effectively
sion of a dominant negative TR blocks metamorphosis than TR homodimers. In the unliganded form, the TR-RXR
(Schreiber et al., 2001; Buchholz et al., 2003), while expres- complex functions as a transcriptional repressor (Wong and
sion of a dominant positive TR promotes metamorphic Shi, 1995). The TR-RXR heterocomplex recruits cofactor
changes in the absence of TH (Buchholz et al., 2004). Aran proteins that mediate the repressive or activational actions of
et al. (2014) recently showed that autoinduction of TR the complex (Shi, 2000b; McKenna and O’Malley, 2002). The
a and b and the actions of exogenous T3 on epibranchial unliganded receptor may have important developmental
remodeling were strongly reduced or eliminated in paedomor- functions in the premetamorphic tadpole (discussed below).
phic versus metamorphic populations of the Oklahoma sala- The TR and RXR genes exhibit more or less coordinate
mander Eurycea tynerensis. These findings provide further regulation during metamorphosis, and this coordination may
support for the essential role of TH and TRs in metamorphosis, be essential to the timing of tissue-specific changes (Wong
and their roles in the expression of alternative developmental and Shi, 1995). Current studies are focused on identifying
modes in amphibians. TREs throughout the genome in different tadpole tissues
Specific functions for the different receptor subtypes in using chromatin immunoprecipitation (ChIP) combined
amphibia are poorly understood. Expression studies suggest with high-throughput sequencing (Grimaldi et al., 2013b).
differential roles, as do pharmacological studies with TR Hormone binding to the TR-RXR receptor complex induces
subtype-selective agonists. For example, studies with the TRa- gene expression in target tissues. The TRs cause modifications
selective agonist CO23 support that this receptor subtype is of chromatin through recruitment of coactivator and
involved in cell proliferation (Ocasio and Scanlan, 2006; corepressor proteins (Shi et al., 2012; Grimaldi et al., 2013a).
Denver et al., 2009), while studies with the TRb selective In the unliganded state, TRs function primarily as
agonists GC1 and GC24 support that this subtype is primarily transcriptional repressors, recruiting corepressors such as
involved with tissue resorption (apoptosis) and cell differenti- NCoR and SMRT which then recruit histone deacetylases
ation (Furlow et al., 2004; Ocasio and Scanlan, 2006; Denver (Tomita et al., 2004). This leads to a compact, repressive
et al., 2009). Experimental studies addressing specific chromatin structure (Sachs et al., 2001; Jones and Shi, 2003).
functions for the different TRs in amphibians were limited When TH binds to TRs, corepressors are exchanged for
154 Endocrinology of Complex Life Cycles: Amphibians

coactivators, leading to transcriptional derepression and 2.09.2.3.1 Development of and Hormones Produced
transactivation. Many of the coactivators have intrinsic by Amphibian Interrenal Glands
histone acetyl transferase activity, leading to the addition of The interrenal gland is generally less active in early, premeta-
acetyl groups to lysine residues on histone tails. Coactivator morphic developmental stages and more active during prome-
expression and recruitment to target genes is correlated with tamorphosis and metamorphic climax (Dodd and Dodd,
tissue transformation and gene activation (Paul and Shi, 1976). The ultrastructural appearance of X. laevis interrenal
2003; Paul et al., 2005b), and transgenic analysis suggests cells indicates relative inactivity in mid-prometamorphs,
that the recruitment of coactivators by TRs is essential for increasing to peak activity at metamorphic climax (reviewed
metamorphosis to proceed (Paul et al., 2005a, 2007). A role by Dodd and Dodd, 1976; however, see below for contrary
for unliganded TR in tadpole development, whereby it evidence). Activity of the interrenal enzyme, D5-3b-hydroxyste-
represses the expression of adult genes (Sachs et al., 2002; roid dehydrogenase (HSD) is present throughout development
Sato et al., 2007), has led to the development of the dual in R. catesbeiana and X. laevis but increases at metamorphic
function model for TR action during metamorphosis. This climax in R. catesbeiana (Hsu et al., 1980; Kang et al., 1995).
model proposes that unliganded TRs (the aporeceptors) play Carr and Norris (1988) found a similar pattern for plasma
an important role in development by repressing adult genes corticosterone and interrenal HSD activity in the tiger sala-
in the tadpole prior to the onset of thyroid activity, but mander, A. tigrinum.
activate expression of metamorphosis-associated genes when Radioimmunoassays for corticosteroids have been done on
ligand is present (reviewed by Buchholz et al., 2006; plasma samples collected throughout the metamorphic period
Grimaldi et al., 2013a; Shi, 2013). The aporeceptors have for several amphibian species: R. catesbeiana (Jaffe, 1981; Krug
also been implicated in heart development and the et al., 1983; Kikuyama et al., 1986); B. japonicas (Niinuma et al.,
maturation of the brain and other organs prior to the onset 1989); X. laevis (Jolivet-Jaudet and Leloup-Hatey, 1984);
of thyroid gland function in mammals (Bernal and Morte, A. tigrinum (Carr and Norris, 1988). Whole-body measures of
2013). Discussion of the characteristics of the gene regulation corticosteroid content have also been determined throughout
cascades and the functions of the gene products induced by development: S. hammondii (Denver, 1998a); X. laevis (Kloas
TH in different tissues during metamorphosis is beyond the et al., 1997; Krain and Denver, 2004) (Glennemeier and
scope of this chapter. For detailed discussion of these topics Denver, 2002a); Rana pipiens (Glennemeier and Denver,
the reader is referred to the following reviews: Furlow and 2002a). The majority of these studies showed low-to-nonde-
Neff (2006), Buchholz et al. (2006), Brown and Cai (2007), tectable corticosteroids during premetamorphosis, and
and Grimaldi et al. (2013a). a marked increase at metamorphic climax, more or less in
parallel with the rise in THs. The only exception to this rule
is whole-body corticosteroid content in X. laevis. Kloas et al.
2.09.2.3 Corticosteroids
(1997) reported that whole-body corticosterone content in
In addition to TH, corticosteroids (CS), the primary verte- X. laevis increases during premetamorphosis to reach a peak
brate stress hormones, play important roles in amphibian at NK stage 48 and then declines during prometamorphosis
metamorphosis. Like TH, the production of CS changes and is low at metamorphic climax. Kloas et al. (1997) also
with development and li

You might also like