You are on page 1of 630

The Sage Handbook of

Cognitive and
Systems Neuroscience

Neuroscientific Principles, Systems and Methods


This page intentionally left blank
The Sage Handbook of
Cognitive and
Systems Neuroscience

Neuroscientific Principles, Systems and Methods

Edited by
Gregory J. Boyle
with
Georg Northoff
Aron K. Barbey
Felipe Fregni
Marjan Jahanshahi
Alvaro Pascual-Leone and
Barbara J. Sahakian

Associate Editors
Nadia Bolognini and
Anja Soldan
1 Oliver’s Yard Editorial Arrangement © Gregory J. Boyle, Georg Northoff, Aron K. Barbey, Felipe
55 City Road Fregni, Marjan Jahanshahi, Álvaro Pascual-Leone, and Barbara J. Sahakian, 2024
London EC1Y 1SP
Chapter 1 © Gregory J. Boyle, Chapter 17 © Luis M. García Marín,
Georg Northoff, Nadia Bolognini, Zuriel Ceja, Miguel E. Rentería,
2455 Teller Road Aron K. Barbey, Marjan Jahanshahi, Albert Galaburda, 2024
Thousand Oaks, California 91320 Álvaro Pascual-Leone, and Chapter 18 © Giacomo Rizzolatti,
Barbara J. Sahakian, 2024 Leonardo Fogassi, 2024
Unit No 323-333, Third Floor, F-Block Chapter 2 © Georg Northoff, Philipp Chapter 19 © Martin Seeber, Thomas
International Trade Tower Nehru Place Klar, 2024 Koenig, Christoph M. Michel, 2024
New Delhi 110 019 Chapter 3 © Lídia Vaqué-Alcázar, Chapter 20 © Arcady A. Putilov, 2024
Anja Soldan, David Bartrés-Faz, Chapter 21 © Erich Schröger,
2024 Alexandra Bendixen, 2024
8 Marina View Suite 43-053
Chapter 4 © Tiago Lazzaretti, Paulo Chapter 22 © Paul C. J. Taylor, 2024
Asia Square Tower 1 Ricardo de Melo, Anna Marduy, Chapter 23 © Jon H. Kaas, Leah A.
Singapore 018960 Rafaella Rogatto de Faria, 2024 Krubitzer, Hui-Xin Qi, Jamie L. Reed,
Chapter 5 © Joan Y. Chiao, 2024 2024
Chapter 6 © Alexei Verkhratsky, Alexey Chapter 24 © Vishal Rawji, John
Semyanov, Arthur Butt, Rothwell, Marjan Jahanshahi, 2024
Olga Garaschuk, 2024 Chapter 25 © Anna Kristina
Chapter 7 © Jack van Honk, Zhang Li, Hernandez, Laiquan Zou, Thomas
Norihiro Sadato, Joan Y. Chiao, 2024 Hummel, 2024
Chapter 8 © Saana M. Korkki, Goran Chapter 26 © Hugo Critchley, Yoko
Papenberg, Marc Guitart-Masip, Alireza Nagai, Lisa Quadt, 2024
Salami, Nina Karalija, Lars Nyberg, Chapter 27 © Claudio Brozzoli,
Editor: Janka Romero Lars Bäckman, 2024 Nadia Bolognini, Alessandro Zanini,
Editorial Assistant: Benedict Hegarty Chapter 9 © Daniel Osorio-Gómez, Alessandro Farnè, 2024
María-Isabel Miranda, Federico Chapter 28 © Randolph F. Helfrich,
Production Editor: Neelu Sahu
Bermúdez-Rattoni, Kioko Guzmán- Robert T. Knight, 2024
Copyeditor: Martin Noble Ramos, 2024 Chapter 29 © Irina G. Skotnikova,
Proofreader: Lawrence Baker Chapter 10 © María-Isabel Miranda, Alexei N. Gusev, Alexandr M.
Indexer: KnowledgeWorks Global Ltd Kioko Guzmán-Ramos, Chernorizov, 2024
Marketing Manager: Camille Richmond Daniel Osorio-Gómez, Chapter 30 © John Bickle, David J.
Cover Design: Ginkhan Siam Federico Bermúdez-Rattoni, 2024 Parker, 2024
Typeset by KnowledgeWorks Global Ltd Chapter 11 © Jiang-Fan Chen, Yan Chapter 31 © Jorge F. Mejías, Katrin
Printed in the UK Li, 2024 Amunts, Jan G. Bjaalie, Sander M.
Chapter 12 © Linda Becker, Nicolas Bohté, Alain Destexhe, Lars Muckli,
Rohleder, 2024 Pier S. Paolucci, Martin J. Pearson,
Chapter 13 © Anneke Alkemade, Cyriel M. A. Pennartz, 2024
Bernadette C. M. van Wijk, Birte U. Chapter 32 © Sicong Tu, William
Forstmann, 2024 Huynh, Matthew Kiernan, 2024
Chapter 14 © Mario Manto, 2024 Chapter 33 © Matt Tharp, Shannon
Chapter 15 © Hans Joachim L. Risacher, Paula Bice, Paul M.
Markowitsch, Jens Borgelt, Angelica Thompson, Andrew J. Saykin,
Staniloiu, 2024 Meichen Yu, 2024
Chapter 16 © Rosanna Olsen, Chapter 34 © Rotem Botvinik-Nezer,
Mrinmayi Kulkarni, 2024 Tor D. Wager, 2024

Apart from any fair dealing for the purposes of research, private study,
or criticism or review, as permitted under the Copyright, Designs and
Patents Act, 1988, this publication may not be reproduced, stored or
transmitted in any form, or by any means, without the prior permission
in writing of the publisher, or in the case of reprographic reproduc-
tion, in accordance with the terms of licences issued by the Copyright
Licensing Agency. Enquiries concerning reproduction outside those
At Sage we take sustainability seriously. terms should be sent to the publisher.
Most of our products are printed in the
UK using responsibly sourced papers and Library of Congress Control Number: 2023946932
boards. When we print overseas, we ensure
sustainable papers are used as measured British Library Cataloguing in Publication data
by the Paper Chain Project grading system. A catalogue record for this book is available from the British Library
We undertake an annual audit to monitor
our sustainability. ISBN 978-1-5297-5355-4
In loving memory of my dear wife Dolores
– GJB
This page intentionally left blank
Contents

The Editors and Contributors x

1 Cognitive Neuroscience: Basic Principles, Systems, and Methods 1


Gregory J. Boyle, Georg Northoff, Nadia Bolognini, Aron K. Barbey,
Marjan Jahanshahi, Álvaro Pascual-Leone, and Barbara J. Sahakian

PART I BACKGROUND CONSIDERATIONS

2 Historical, Empirical, and Philosophical Perspectives on


Cognitive Neuroscience 17
Georg Northoff and Philipp Klar

3 Aging Brain Changes across the Lifespan 30


Lídia Vaqué-Alcázar, Anja Soldan, and David Bartrés-Faz

4 Innovation Pathways, Real-life Neuroscience Startups, and


Applications 48
Tiago Lazzaretti, Paulo S. de Melo, Anna Marduy, Rafaella Rogatto de Faria,
Marcel Simis, and Felipe Fregni

PART II NEUROSCIENTIFIC SUBSTRATES AND PRINCIPLES

5 Neural Processes and Activity: Cultural Neuroscience 65


Joan Y. Chiao

6 Role of Neuroglia in Cognition 79


Alexei Verkhratsky, Alexey Semyanov, Arthur Butt, and Olga Garaschuk

7 Cognitive Neurogenetics 102


Jack van Honk, Zhang Li, Norihiro Sadato, and Joan Y. Chiao

8 Dopamine System and Cognitive Function across the Adult Life Span 114
Saana M. Korkki, Goran Papenberg, Marc Guitart-Masip,
Alireza Salami, Nina Karalija, Lars Nyberg, and Lars Bäckman

9 Neurotransmission and Neuromodulation of Recognition Memory:


Novelty vs. Familiarity 133
Daniel Osorio-Gómez, María-Isabel Miranda, Federico Bermúdez-Rattoni,
and Kioko Guzmán-Ramos
viii THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

10 Neurotransmission and Neuromodulation of Memory: Intensity and Valence 151


María-Isabel Miranda, Kioko Guzmán-Ramos, Daniel Osorio-Gómez,
and Federico Bermúdez-Rattoni

11 Neuromodulator Adenosine A2A Receptor Control of Cognition 171


Jiang-Fan Chen and Yan Li

12 Hormonal Influences on Cognition 185


Linda Becker and Nicolas Rohleder

PART III NEUROANATOMICAL BRAIN SYSTEMS

13 Modeling of Basal Ganglia Structure and Function 197


Anneke Alkemade, Bernadette C. M. van Wijk, and Birte U. Forstmann

14 Cerebellum: Advances in Understanding of Cerebellar Functions 212


Mario Manto

15 Amygdaloid Complex 227


Hans J. Markowitsch, Jens Borgelt, and Angelica Staniloiu

16 Hippocampal Formation 242


Rosanna K. Olsen and Mrinmayi Kulkarni

17 Cerebral Lateralization and Hemispheric Specialization 259


Luis M. García-Marín, Zuriel Ceja, Miguel E. Rentería, and
Albert M. Galaburda

PART IV NEURAL DYNAMICS AND PROCESSES

18 Mirror Mechanism in Cognition 279


Giacomo Rizzolatti and Leonardo Fogassi

19 Spatio-temporal Dynamics of Brain Rhythms 305


Martin Seeber, Thomas Koenig, and Christoph M. Michel

20 Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 318


Arcady A. Putilov

PART V SENSORY-PERCEPTUAL SYSTEMS AND COGNITION

21 Auditory Processing 337


Erich Schröger and Alexandra Bendixen

22 Vestibular Processing in Cognition 351


Paul C. J. Taylor
Contents ix

23 Somatosensory Processing 365


Jon H. Kaas, Leah A. Krubitzer, Hui-Xin Qi, and Jamie L. Reed

24 Motor Control: Response Preparation, Initiation, and Inhibition 379


Vishal Rawji, John C. Rothwell, and Marjan Jahanshahi

25 Olfactory and Gustatory Sensation-Perception 398


Anna Kristina Hernandez, Laiquan Zou, and Thomas Hummel

26 Interoception and Thermoreception 414


Hugo D. Critchley, Yoko Nagai, and Lisa Quadt

27 Peripersonal Space Representation: Neural Bases, Properties,


and Functional Significance 435
Claudio Brozzoli, Nadia Bolognini, Alessandro Zanini, and
Alessandro Farnè

PART VI METHODOLOGICAL ADVANCES

28 Neural Recordings and Time Series Analyses 453


Randolph F. Helfrich and Robert T. Knight

29 Advances in Psychophysical Techniques 472


Irina G. Skotnikova, Alexei N. Gusev, and Alexandr M. Chernorizov

30 Revolutions in “Wet” Neurobiology 495


John Bickle and David J. Parker

31 Human Brain Project and Beyond 511


Jorge F. Mejías, Katrin Amunts, Jan G. Bjaalie, Sander M. Bohté,
Alain Destexhe, Lars Muckli, Pier S. Paolucci, Martin J. Pearson,
and Cyriel M. A. Pennartz

32 Advances in Brain Imaging 533


Sicong Tu, William Huynh, and Matthew C. Kiernan

33 Novel Approaches to Large-scale Data in Neuroimaging 553


Matthew Tharp, Shannon L. Risacher, Paula Bice,
Paul M. Thompson, Andrew J. Saykin, and Meichen Yu

34 Advancing Cognitive Neuroscience 569


Rotem Botvinik-Nezer and Tor D. Wager

Index 590
The Editors and Contributors

EDITOR-IN-CHIEF

Gregory J. Boyle is an Hon. Professorial Fellow at the University of Melbourne. He earned separate
PhDs from both the University of Delaware, and the University of Melbourne. Also, he earned a DSc from
the University of Queensland, for his sustained contributions to psychological science. He is a Fellow of
both the Association for Psychological Science, and the Australian Psychological Society, and recipient
of the Buros Institute of Mental Measurements Distinguished Reviewer Award. Dr. Boyle has produced
some 300 scholarly publications and has served as lead editor of several international handbooks includ-
ing: The SAGE Handbook of Personality Theory and Assessment (2 Vols.); Measures of Personality and
Social Psychological Constructs (Academic Press); as well as the SAGE Benchmarks in Psychology
Series: Work and Organisational Psychology (5 Vols.); Psychological Assessment (4 Vols.); and
Psychology of Individual Differences (4 Vols.). He is also Editor-in-Chief of the current volumes.

EDITORS

Aron K. Barbey is Professor of Psychology, Neuroscience, and Bioengineering at the University of


Illinois at Urbana-Champaign. He is chair of the Intelligent Systems Research Theme, leader of the
Intelligence, Learning, and Plasticity Initiative, and director of the Decision Neuroscience Laboratory at
the Beckman Institute for Advanced Science and Technology. Dr. Barbey’s research investigates the
neural mechanisms of human intelligence and decision making, with particular emphasis on enhancing
these functions through cognitive neuroscience, physical fitness, and nutritional intervention. His research
has been supported by the Office of the Director of National Intelligence, the Department of Defense, the
White House BRAIN Initiative, the National Institutes of Health, the National Science Foundation, and
private industry (Abbott Nutrition, Google Brain, and PepsiCo). Dr. Barbey has received multiple
research awards and is editor of The Cambridge Handbook of Intelligence and Cognitive Neuroscience
and the forthcoming Oxford Handbook of Cognitive Enhancement and Brain Plasticity. He earned his
doctorate in Psychology from Emory University and completed a research fellowship in Cognitive
Neuroscience at the National Institutes of Health.

Felipe Fregni is Professor of Physical Medicine and Rehabilitation, Harvard Medical School, Professor
of Epidemiology, Harvard School of Public Health, and Director of the Spaulding Neuromodulation
Center. He is also the course director for the HMS continuing medical education course, Principles and
Practice of Clinical Research, a six-month distance learning course. Currently, his research is focused on
understanding neuroplastic changes associated with conditions such as chronic pain, Parkinson’s disease,
and stroke, using non-invasive brain stimulation as an investigative tool for such aims. In addition, his
laboratory comprises about 15 research fellows and staff and is a training center for clinical research and
neuromodulation methodology. Dr. Fregni’s laboratory is funded by several sponsors including NIH, the
Christopher and Dana Reeve Foundation, CIMIT, and the RJG Foundation.

Marjan Jahanshahi is Professor Emeritus of Clinical Neuropsychology at the UCL Queen Square
Institute of Neurology. She is a Fellow of the British Psychological Society. She trained as a clinical
psychologist and neuropsychologist. She was a principal investigator at the Sobell Department of Motor
Neuroscience & Movement Disorders, UCL Queen Square Institute of Neurology; where she led the
The Editors and Contributors xi

Cognitive-Motor Neuroscience Group. She was also a founding member and the honorary consultant
neuropsychologist at the Functional Neurosurgery Unit, the National Hospital for Neurology &
Neurosurgery for 15 years. Her research focuses on movement disorders such as Parkinson’s disease and
dystonia, including assessment of the impact of surgical treatments such as deep brain stimulation for
these disorders on cognition and movement and investigation of the psychosocial impact of these chronic
disorders on the quality of life of the patients and their families. She has published 350 papers in peer-
reviewed journals with an h-index of 90 and also 2 books.

Georg Northoff is Professor of Neurosciences and Mental Health and holds a Canada Research Chair for
Mind, Brain Imaging and Neuroethics at the University of Ottawa/Canada. Dr. Northoff has made major
contributions in neuroscience regarding the neural correlates of mental features such as consciousness,
self, mind wandering and mental disorders, having discovered their spatiotemporal mechanisms bridging
the gap of neural and mental activity. This led him to develop an integrated brain-mind model for which
Spatiotemporal Neuroscience is the key discipline (www.georgnorthoff.com).

Álvaro Pascual-Leone is Professor of Neurology and Director of the Berenson-Allen Center for
Noninvasive Brain Stimulation at Beth Israel Deaconess Medical Center and Harvard Medical School,
where he also serves as Program Director of the Harvard-Thorndike Clinical Research Center. His
research aims at understanding the mechanisms that control brain plasticity across the lifespan to be able
to modify them for the subject’s optimal behavioral outcome. Pascual-Leone combines various brain
imaging and brain stimulation methodologies to establish a causal relationship and a precise chronometry
between regional brain activation and behavior, and uses noninvasive brain stimulation techniques to
modulate brain plasticity, suppressing some changes and enhancing others, to gain a clinical benefit and
behavioral advantage for a given individual. Such non-invasive approaches can lead to clinically relevant
therapeutic effects in neuropsychiatry and neurorehabilitation, and serve as proof-of-principle prior to
more invasive neuromodulatory interventions. A major interest of current work aims at translating
insights from cognitive neuroscience into clinical interventions (h-index 192).

Barbara J Sahakian is Professor of Clinical Neuropsychology in the Department of Psychiatry at the


University of Cambridge. She is a Fellow of the British Academy and a Fellow of the Academy of
Medical Sciences. She is also a Fellow of Clare Hall. She is a past president of the International
Neuroethics Society and of the British Association for Psychopharmacology. According to Research.com
she is among the very top researchers worldwide in the fields of neuroscience and psychology. Dr.
Sahakian has contributed over 550 publications (h-index 162) in scientific journals, including Science,
Nature and The Lancet. She has also been a leader in Government Policy on neuroscience and mental
health, including the UK Government Foresight Project. She is co-Inventor of the Cambridge Cognition
CANTAB computerized cognitive tests used worldwide, the PEAK Wizard Memory Game, and the
Decoder Attention and Concentration Game. Cambridge Cognition is a prominent neuroscience technol-
ogy organization that seeks to optimize cognitive assessment. Dr. Sahakian is also co-inventor of the
EMOTICOM computerized tests of social and emotional cognition. She is co-author of two popular sci-
ence books, entitled Bad Moves and Sex, Lies and Brain Scans.

ASSOCIATE EDITORS

Nadia Bolognini is Professor of Psychobiology and Physiological Psychology at the University of


Milano Bicocca and Director of the Laboratory of Neuropsychology at the I.R.C.C.S. Istituto
Auxologico Italiano (Milan, Italy). She is also currently coordinator of the Bachelor of Science in
Psychological Science and Techniques at the University of Milano Bicocca. She received her PhD in
Clinical and Experimental Psychology from the University of Bologna (Italy). Dr. Bolognini has been a
visiting researcher at the Department of Neurobiology and Anatomy of Wake Forest University, North
Carolina, at Spaulding Rehabilitation Hospital, and at Beth Israel Deaconess Medical Center, Harvard
Medical School. She has published more than 120 peer-reviewed scientific papers specifically within
the field of cognitive neuroscience, has edited two books, and has contributed 16 national and interna-
tional book chapters.
xii THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Anja Soldan is an Associate Professor of Neurology at Johns Hopkins University School of Medicine.
She holds PhD in Cognitive Neuroscience from New York’s Columbia University. She completed a post-
doctoral fellowship in Neuropsychology and Cognition in Aging at Columbia University Medical Center
and was an Assistant Professor of Psychology at the University of Massachusetts – Dartmouth, prior to
joining the faculty at Johns Hopkins University in 2011. Dr. Soldan’s research focuses on the cognitive
changes that occur with age and Alzheimer’s disease and their relationship to brain structure and function.
Her research has focused on characterizing the earliest cognitive and brain changes associated with
Alzheimer’s disease to identify early markers for cognitive decline. Additionally, she is investigating
individual differences in modifiable risk and protective factors for cognitive decline (such as cognitive
reserve and vascular risk) and their relationship to genetic factors.

CONTRIBUTORS

Anneke Alkemade is an expert in human neuroanatomy and Assistant Professor working at the Brain and
Cognition, Psychology Department, at the University of Amsterdam. In her work she aims to uncover the
functional neuroanatomy in health and disease, and to translate post-mortem histological findings into
information relevant for clinicians as well as other fields of neuroscience. Her work contributes to
increase the understanding of the pathogenesis underlying neurological disease and is focused on increas-
ing insight in the human subthalamic nucleus, which is targeted by deep brain stimulation in patients with
Parkinson’s disease. Dr. Alkemade’s work is published in journals from the fields of neuroscience, neu-
roanatomy, psychiatry, biochemistry, and endocrinology.

Katrin Amunts is Neuroscience Professor at the Cécile and Oskar Vogt Institute for Brain Research at
the University Hospital of Düsseldorf, and is also Director of the Institute of Neuroscience and Medicine
in INM-1 in Forschungszentrum Jülich and the Scientific Research Director of the Human Brain Project.
Her research focus is on brain mapping, particularly on creating a three-dimensional multi-level brain
atlas (including the cytoarchitectonic Julich-Brain atlas and the “Big Brain”) to allow the complex con-
figuration of brain regions and functions to be imaged and understood microscopically. This would
advance current abilities to combat diseases or disorders such as depression, addiction, dementia, and
Parkinson’s disease.

Lars Bäckman has been a Professor in the Cognitive Neuroscience of Aging since 2002 at the Aging
Research Center at Karolinska Institute, Sweden. He has previously held professorships at Gothenburg
University (1993–1998) and Uppsala University (1998–2002), Sweden. He is a cognitive psychologist by
training and received his PhD from Umeå University, Sweden, in 1984. He is a member of the Royal
Swedish Academy of Sciences and the European Academy of Sciences. Lars Bäckman’s primary research
area is cognition in normal and pathological aging, with special focus on memory. His research activities
range from large-scale epidemiological studies to experimental brain-imaging work.

David Bartrés-Faz is Professor of Medical Psychology at the University of Barcelona. He received train-
ing in neuropsychology, brain imaging and non-invasive brain stimulation and performed postdoctoral
stages at the INSERM (France) and at Harvard Medical School (USA). His research focus on modifiable
factors and neural mechanisms that explain maintenance of brain and mental health in advanced age. PI
of consecutive Spanish National grants since 2004 and PI of the University of Barcelona Center for
European Programs FP7 and Horizon 2020. PI of the prospective study Barcelona Brain Health initiative
(BBH), Honorary member of the Department of Clinical Educational and Health Psychology, University
College London (2022-2027) and chair (2001-2003) of the “Reserve, Resilience and Protective Factors”
Professional Interest Area of the Alzheimer International Association (ISTAART). He sits on the
Director’s board of the Institute of Neuroscience at the University of Barcelona and has been awarded
with a Ramon y Cajal and an ICREA Academia fellowships by the Spanish and Catalan Governments.

Linda Becker received her diploma degrees (equivalent to Master of Science) in physics and in psychol-
ogy in 2007 from the University of Bremen (Germany). In 2016, she received her PhD in psychology from
the Friedrich-Alexander University Erlangen-Nürnberg (FAU), Germany. Her research focusses on the
relationships between peripheral physiological and cognitive processes. Her main research interests are
The Editors and Contributors xiii

the associations between biological stress responses and cognitive functions. In her research, she is using
a variety of bio-psychological and neuroscientific methods such as electroencephalography (EEG), mag-
netencephalography (MEG), as well as the measurement of stress hormones and inflammatory markers.

Alexandra Bendixen is a professor of Structure and Function of Cognitive Systems at the Institute of
Physics at Chemnitz University of Technology. Her main scientific contributions are in the field of audi-
tory perception and cognition, auditory scene analysis, sensory ambiguity, and multistability, as well as
resource allocation and interference.

Federico Bermúdez-Rattoni obtained his M.D. from UNAM and a Ph.D. from the University of
California, Los Angeles. He is a Guggenheim Fellow and External Fellow of the Center for the
Neurobiology of Learning and Memory, University of California, Irvine. He obtained the Mexican National
Science Prize, UNAM prize and Syntex Prize. Currently, he is an Emeritus Professor and Head of the
Cognitive Neuroscience Department of the Cellular Physiology Institute, UNAM, Mexico City Campus.

Paula J. Bice has a broad background in Neuroscience, with specific training and expertise in neurobiol-
ogy of addiction. Her earlier research in the Alcohol Research Center in the Indiana University School of
Medicine (IUSM) focused on identifying and characterizing genes underlying addiction. Additionally,
she has taught Neuroscience and other related courses, such as Neurobiology of Addiction and Drugs in
the Nervous System. Currently, as a faculty member in the Indiana Alzheimer’s Disease Research Center
in the IUSM, she utilizes her background in neuroscience to assist faculty and trainees with scientific
writing, research project development, grant development, statistics, graphics, media, and metrics.

John Bickle is Professor of Philosophy and Shackouls Honors College Faculty at Mississippi State
University, and Scientist Educator in the Department of Advanced Biomedical Education at the University
of Mississippi Medical Center. His research areas are the philosophy of neuroscience and the philosophy
of science-in-practice. He is the author of four academic books, Psychoneural Reduction: The New Wave
(MIT Press, 1998), Philosophy and Neuroscience: A Ruthlessly Reductive Account (Kluwer, now
Springer, 2003), Understanding Scientific Reasoning, 5th Ed. (co-authors Ronald Giere and Robert
Mauldin, Cengate, 2005), and Engineering the New Revolution in Neuroscience (co-authors Alcino J.
Silva and Anthony Landreth, Oxford University Press, 2014), and around 100 papers and chapters in
philosophy of science and neuroscience journals and book volumes. He has edited two collections of
essays, including The Oxford Handbook of Philosophy and Neuroscience (2009), and most recently The
Tools of Neuroscience Research: Philosophical and Scientific Perspectives (co-editors Carl F. Craver and
Ann-Sophie Barwich, Routledge, 2021).

Jan Bjaalie is Professor at the Institute of Basic Medical Sciences, University of Oslo, Norway,
Infrastructure Director of the EU Human Brain Project, leader of the EBRAINS Data services, and Head
of the Norwegian Neuroinformatics Node. His research group joined the Centre for Molecular Biology
and Neuroscience, a Norwegian Centre of Excellence appointed by The Research Council of Norway, in
2002. His laboratory has discovered fundamental principles of sensory map transformations in large
projection systems of the brain and has performed novel regional and whole brain atlasing and histologi-
cal mapping. Professor Bjaalie has served as co-Chair of the International Brain Initiative since 2019,
Head of the Institute of Basic Medical Sciences at the University of Oslo, Chair of the Governing Board
and founding Executive Director of the International Neuroinformatics Coordinating Facility (INCF), and
member of the Neuroinformatics Committee of the Society for Neuroscience.

Sander M. Bohté is Principal Investigator in the CWI Machine Learning group and also a part-time
Professor of Cognitive Computational Neuroscience at the University of Amsterdam. He received his PhD
in 2003 at CWI on the topic of “Spiking Neural Networks.” He then did a postdoc with Michael Mozer
at the University of Colorado in Boulder. In 2004, he rejoined CWI as junior permanent staff to work on
distributed spiking neural network models and multi-agent systems. In 2016, he co-founded the CWI
Machine Learning group, where his research bridges the field of neuroscience with applications thereof
as advanced neural networks. His work has been pioneering in the development of advanced and efficient
spiking neural networks, including seminal work on supervised learning with spike-time coded networks.
Recent work has focused on large-scale and effective spiking neural networks next to biologically plau-
sible deep learning and deep reinforcement learning models for cognition.
xiv THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Jens Borgelt completed his doctoral studies at the University of Konstanz, Germany, in the field of func-
tional brain imaging and clinical psychophysiology, investigating neural correlates of altered information
processing in PTSD via MEG (magnetoencephalography). Dr. Borgelt has also studied and worked in the
Department of Psychiatry, Oxford University, and in the Department of Psychiatry and Behavioral
Sciences, Stanford University. Later he became a licensed psychotherapist in cognitive behavior therapy
and has worked in several clinical fields including neurorehabilitation and the treatment of severe psychi-
atric disorders and is now chief psychologist at the private Oberberg Clinic, Black Forest, Germany.

Rotem Botvinik-Nezer is a postdoctoral researcher in Psychological and Brain Sciences at Dartmouth


College and incoming faculty at the Psychology Department at the Hebrew University of Jerusalem. With
background in decision-making, behavioral change, and neuroplasticity, Dr. Botvinik-Nezer’s current
research focuses on beliefs and expectations, in the context of the placebo effect and beyond. Dr.
Botvinik-Nezer studies how beliefs and expectations are formed and updated given new information, and
how they affect mental, physiological, and social processes, with diverse tools from behavioral experi-
mental manipulations to computational modeling to neuroimaging. Dr. Botvinik-Nezer is also proactively
conducting and promoting open and reproducible scientific research, with a particular emphasis on repro-
ducibility, transparency, robustness to analytical decisions, and inclusion.

Claudio Brozzoli is an INSERM senior researcher at the IMPACT team of the Centre de Recherche en
Neuroscience de Lyon (CRNL). He received his master degree in Psychology at the University of
Bologna (Italy) and his PhD in Cognitive Neuroscience from the Neuroscience and Cognition doctoral
school of the University Claude Bernard of Lyon. He is currently leading several research programs
focusing on the multisensory representation of the body and the space near the body (peripersonal space),
their role in action planning and execution and how these multisensory and motor processes support
human cognition for language and social interactions.

Arthur Butt is a Professor of Biology and has been a glial cell biologist for over 25 years. He received
his PhD from King’s College London in 1986, working with Joan Abbott, a leader in blood-brain barrier
research. After a postdoctoral position in North Carolina with Ed Lieberman, he was awarded a Grass
Fellowship to work at the world-famous Wood’s Hole Marine Biology Laboratories. Next, he moved to
Yale University to work with Bruce Ransom, where he began his work on optic nerve glia, a line of
research he has pursued ever since. On return to King’s College in the UK, he received a Royal Society
Fellowship to work at the Marine Laboratories in Plymouth, UK, before obtaining his first independent
position at Guy’s and St Thomas’ Hospitals in 1990. After gaining a personal Chair at King’s College in
2000, he moved to the University of Portsmouth in 2005, where he was Director of the Institute of
Biomedical and Biomolecular Sciences. Much of his work has focused on oligodendrocyte cell biology
and the factors that regulate their regeneration.

Zuriel Ceja is a PhD student at The University of Queensland and QIMR Berghofer Medical Research
Institute in Brisbane. He holds a degree in Psychology from the University of Guanajuato in Mexico. His
research interests focus on behavioral science and psychiatric genetics. He has previously held a research
internship at the Instituto de Neurobiología of the Universidad Nacional Autónoma de México in
Juriquilla, Mexico. He was a Summer Research Intern sponsored by the Mexican Academy of Sciences,
was a Mitacs Globalink research intern in Toronto, Canada, and served as a Delegate at the Canada-
Mexico Youth Lab.

Jiang-Fan Chen at Wenzhou Medical University, China undertakes research into the neuromodulator
adenosine A2A receptor control of cognition within the Molecular Neuropharmacology Lab, School of
Optometry and Ophthalmology, Wenzhou, China. Dr. Chen also undertakes research into cognition within
the State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou, China.

Joan Y. Chiao is the Director of the International Cultural Neuroscience Consortium. She earned her PhD
from Harvard University in Psychology and B.S. with Honors from Stanford University in Symbolic
Systems. She is the Director of the Laboratory for Social and Affective Neuroscience. She is a Fellow of
the American Psychological Society, Society for Personality and Social Psychology, and the Society for
Experimental Social Psychology. She is an editor of peer-reviewed journals and book series such as The
Oxford Handbook of Cultural Neuroscience. She is a recipient of the Distinguished Scientist Award and
Fields Prize from the International Cultural Neuroscience Society.
The Editors and Contributors xv

Hugo Critchley is Professor of Psychiatry at Brighton and Sussex Medical School. Hugo’s research
focuses on brain mechanisms supporting normal and abnormal emotion, with particular emphasis on
bidirectional brain–body interactions. This work encompasses basic and clinical investigation of emo-
tions, autonomic control, interoception and their disorders. Hugo studied Physiology and Medicine in the
University of Liverpool before completing a Doctorate in Psychological Studies at the University of
Oxford. He trained in Psychiatry and clinical neuroscience at the Kings College London Institute of
Psychiatry and University College London (UCL) Institute of Neurology, developing his own research
programme through research fellowships. Hugo moved to Sussex from UCL as foundation Professor of
Psychiatry in 2006. He was also a founding co-director of the Centre for Consciousness Science at the
University of Sussex in 2010. Hugo works clinically as a neuropsychiatrist at the Neurodevelopmental
Services at Sussex Partnership NHS Foundation Trust.

Alain Destexhe is Director of Research at CNRS, Vice Director of NeuroPSI and Director of the
European Institute for Theoretical Neuroscience in Paris, France. His research activity in computational
neuroscience is at the interface between physics (dynamic systems) and neuroscience (electrophysiol-
ogy). His current research projects include the study of collective dynamics of neural networks, the
genesis of brain states and sensory responses, as well as sensory perception.

Alessandro Farnè is Inserm Research Director, Head of the Impact team of the Lyon Neuroscience
Research Center (CRNL) and of the Neuro-immersion CRNL facility. He is currently member of the
Direction Committee of the CRNL. After obtaining his PhD degree (1999) at the University of Bologna,
a Post-Doc (2000) at Rice University in Houston, and Assistant Professorship (2001) at the faculty of
Psychology in Cesena, he got an INSERM research position (2005) in Lyon. His research interests con-
cern the multisensory integration processes for perception and action in the peripersonal space, the sen-
sorimotor control of hands and hand-held tools, body representation and somatosensory plasticity,
combining methods from experimental psychology and cognitive neuroscience.

Birte U. Forstmann is a Professor for Cognitive Neurosciences and Director of the Amsterdam Brain and
Cognition Center at the University of Amsterdam. She earned her PhD in 2006 at the Max Planck Institute
for Human Cognitive and Brain Sciences in Leipzig, Germany. After completing her postdoc in 2008 at
the University of Amsterdam, she became a tenured Research Fellow at the Cognitive Science Center
Amsterdam with the focus of model-based cognitive neurosciences. Since then she has contributed to a
range of topics in cognitive neuroscience, experimental psychology, mathematical psychology, and lately
also in quantitative neuroanatomy. Despite its diversity, her work is motivated by a single strong convic-
tion, namely that behavioral data and brain measurements need to make contact with psychological theory
via concrete mathematical models of latent cognitive processes. A recurrent theme in her work is the
development and testing of quantitative models of cognition.

Leonardo Fogassi has a master’s degree in biology and a PhD in Neuroscience. He was trained in
Neurophysiology at the University of Parma in the lab of Professor Rizzolatti. He also spent one year in
the Richard Andersen’s neurophysiology lab at the Department of Brain and Cognitive Sciences of MIT
(Cambridge, MA). He is now Professor of Physiology in the Department of Medicine and Surgery. His
research focused on the investigation of high order sensory-motor properties of fronto-parietal areas of
non-human primates and humans. In particular, he studied, by using electrophysiological, behavioral and
neuroimaging technique, the processes of visuomotor transformations and the cognitive functions of the
motor system, among which the properties of the mirror neuron system, of which he is co-discoverer. His
research is testified by numerous publications on prestigious international journals. He received, together
with Giacomo Rizzolatti and Vittorio Gallese, the Grawemeyer Award for Psychology (2007).

Albert M. Galaburda is a renowned neuroscientist and Emeritus Professor of Neurology at Harvard


Medical School. He has served as Co-Director of the Mind, Brain, and Behavior Interfaculty Initiative at
Harvard University and as Chief of the Division of Cognitive Neurology at the Beth Israel Deaconess
Medical Center (BIDMC) in Boston. Originally from Chile, he received his medical degree from Boston
University’s School of Medicine. Throughout his career, he has directed the Laboratory for the
Neuroscience of Cerebral Lateralization and Dyslexia at BIDMC and has published extensively in his
field, with over 230 original articles, reviews, book chapters, and books to his credit. He has received
numerous awards and honors, including the Pattison Prize in Neuroscience, the IPSEN Prize in Neural
xvi THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Plasticity, the American Academy of Neurology Decade of the Brain keynote speaker award, the
Behavioral Neurology Society of the American Academy of Neurology’s Lifetime Achievement Award,
and the International Dyslexia Association’s Samuel T. Orton Award. He currently lives in Cambridge,
Massachusetts with his wife, Susana.

Olga Garaschuk is an internationally recognized scholar in the field of neuronal and microglial physiol-
ogy/neuroscience, best known for establishing the techniques for in vivo calcium imaging of neuro-glial
networks at single-cell resolution and the discovery of neuronal as well as microglial hyperactivity during
ageing and in mouse models of Alzheimer’s disease. Garaschuk’s laboratory was the first to study in vivo
calcium signaling of microglia and to reveal that midlife represents the first turning point in microglial
homeostasis. Moreover, she has significantly contributed to understanding the mechanisms of rhythmo-
genesis of the developing cortical and hippocampal networks and the role of sleep-associated rhythmic
activity in this process.

Luis M. García-Marín is a doctoral candidate at The University of Queensland and QIMR Berghofer
Medical Research Institute in Brisbane, Australia. He holds a B.Sc. in Biotechnology Engineering from
the Instituto Tecnológico y de Estudios Superiores de Monterrey (ITESM) in Guadalajara, Mexico. His
research focuses on identifying genetic biomarkers for complex neurological and psychiatric disorders.
He has been recognized for his work with the Lindon Eaves Award from the Institute of Behavioral
Genetics at the University of Colorado Boulder, and the Enrico Greppi Award from the Italian Society for
the Study of Headaches and the European Headache Federation.

Erhan Genç obtained his diploma in psychology at the University of Mannheim and completed after-
wards his PhD at the Max Planck Institute for Brain Research in Frankfurt am Main. Subsequently, he
was a postdoc and principal investigator at the Ruhr University Bochum. Currently, he is a group leader
at the Department of Psychology and Neurosciences at the Leibniz Research Centre for Working
Environment and Human Factors at Technical University of Dortmund (IfADo). He is highly interested
in the neural basis of intelligence and personality. In his research he uses a multimodal approach to link
behavior to brain structure and function. His research has been supported by the German Research
Foundation (DFG). He is a member of the editorial board of the journal Intelligence.

Marc Guitart-Masip is an Associate Professor at the Aging Research Center at Karolinska Institute,
Sweden. In parallel, he works as a consultant psychiatrist at the Affective disorder unit at Psykiatri
Nordväst, Stockholm, Sweden. He has a degree in Medicine (2002) and a PhD in Neuroscience (2006)
from Universitat Autònoma de Barcelona, Spain. During his PhD, he performed experiments in the field
of Behavioral and Molecular Pharmacology using animal models of vulnerability to addiction. After his
PhD, his interests expanded to Cognitive Neuroscience and between 2008 and 2012 he was a post-doc-
toral fellow first at the Institute of Cognitive Neuroscience, and thereafter at the Wellcome Trust Centre
for Neuroimaging, at University College London (UCL), UK.

Alexei N. Gusev is Professor of Faculty of Psychology of Lomonosov Moscow State University. In 1981,
he graduated from the Faculty of Psychology of Lomonosov Moscow State University and specialized in
the fields of psychophysics and cognitive psychophysiology. The main areas of scientific interests are
psychophysics, cognitive psychology, computer psychodiagnostics. The research results are presented in
more than 180 articles, 21 handbooks, monographs and collective monographs (Psychophysics of sensory
tasks, 2002, Sensation and Perception, 2007, 2009, Psychological Measurements: Theory and Methods,
2011, Measurement in psychology, 1997-2005). Alexei N. Gusev - Laureate of the Lomonosov Prize for
Pedagogical Activity, (1998), Awardee of the Russian Government Prize in Education (2010), Winner of
the Moscow Innovator competition (2022).

Kioko Guzmán-Ramos (Universidad Autónoma Metropolitana) completed her PhD at Department of


Neuroscience at the Instituto de Fisiología Celular at the Universidad Nacional Autónoma de México
(UNAM) focusing her research on molecular processes of learning and memory and the dopaminergic
influence on the cognitive impairment developed in Alzheimer’s Disease models. She is currently
Principal Investigator at Universidad Autónoma Metropolitana and Head of Health Sciences Department
investigating the effect of obesogenic environments on cognitive performance.
The Editors and Contributors xvii

Randolph F. Helfrich is a clinician-scientist at the Center for Neurology and the Hertie-Institute for
Clinical Brain Research at the University of Tübingen, Germany. His lab studies large-scale network
mechanisms underlying human cognition and sleep physiology using intracranial electrophysiology. He
is the recipient of an Emmy Noether Award of the German Research Foundation and the Career
Development Award of the Ernst Jung Foundation.

Anna Kristina Hernandez is a researcher at the Smell and Taste Clinic in the Department of
Otorhinolaryngology, Technische Universität Dresden, Dresden, Germany and an Otorhinolaryngologist –
Head and Neck Surgeon at the Philippine General Hospital and Asian Hospital and Medical Center in the
Philippines. Dr. Hernandez graduated with a degree in Psychology from the University of the Philippines –
Diliman and subsequently completed her medical education at the College of Medicine, University of the
Philippines - Manila. She finished her residency training in Otorhinolaryngology at the University of the
Philippines – Philippine General Hospital, with subsequent completion of fellowships for Olfaction and
Gustation at the Technische Universität Dresden and Head and Neck Oncologic Surgery at Erasmus MC
in Rotterdam, Netherlands. Her research interests include olfactory and trigeminal dysfunction, as well as
psychophysical testing.

Thomas Hummel received his medical education at the University of Erlangen-Nürnberg, Germany.
There, he also went through a special program in Pharmacology and Toxicology (“Habilitation”) which
was guided by Dr. Gerd Kobal. As a post-doc, in 1994, he stayed at the Department of Pharmacology at
the University of Iowa, Iowa City, USA. From 1997 to 1998, Dr. Hummel was an Assistant Professor at
the Department of Otorhinolaryngology of the University of Pennsylvania, Philadelphia, USA where he
worked at the Smell and Taste Center under the guidance of Dr. Richard L. Doty. In 1998, Dr. Hummel
started a Smell and Taste Clinic at the Department of Otorhinolaryngology, Technische Universität
Dresden, where he sees approximately 1000 patients per year. He is the author/co-author of more than
700 peer-reviewed, original publications, more than 80 reviews in the chemosensory area, over 50 book
chapters and is the editor of three books about the chemical senses.

William Huynh is a clinical academic and translational researcher dividing his time between clinical
neurology and neurophysiology, and research, as well as teaching. He holds appointments as a Consultant
Neurologist and Clinical Neurophysiologist at the Prince of Wales Tertiary Teaching Hospital in Sydney,
Australia; Associate Professor with the Faculty of Health and Medicine at University of Sydney. His
research has been focused on multiple areas along the spectrum of neurodegenerative disorders.

Jon H. Kaas is a Distinguished Centennial Professor of Psychology at Vanderbilt University, Nashville,


TN, USA. He received his Ph.D. training in comparative studies of forebrain organization in mammals in
the laboratory of Dr. I. T. Diamond at Duke University and studied cortical organization during his post-
doctoral training in the comparative neurophysiology laboratory of Dr. C. N. Woolsey at the University
of Wisconsin. He is an elected member of the National Academy of Sciences and the American Academy
of Arts and Sciences. Dr. Kaas is a recipient of the Javits Neuroscience Investigator Award from the
National Institute of Neurological Disorders and Stroke, the Karl Spencer Lashley Award from the
American Philosophical Society, the Krieg Cortical Discoverer Award from the Cajal Club, and the Ralph
W. Gerard Prize in Neuroscience, the highest honor from the Society for Neuroscience.

Nina Karalija is an Associate Professor at the Department of Integrative Medical Biology at Umeå
University, and a researcher at Umeå Center for Functional Brain Imaging (UFBI), Sweden. She has an
MSc in Chemical Engineering (2008), and a Ph.D.in Medical Science (2013) with orientation towards
histology and cell biology. Her past and present research has focused on the dopamine system, with the
aim to understand how dopamine decline in disease and healthy aging affects motor and cognitive func-
tions. She also studies how other brain changes, e.g., cerebrovascular dysfunction and neuroinflammation,
affect dopamine decline and cognition in aging. These research questions have been studied via experi-
mental neuroscience methods and brain imaging (PET and MRI).

Matthew Kiernan is the Bushell Chair of Neurology at the University of Sydney, Consultant Neurologist
at Royal Prince Alfred Hospital and Co-Director of the Brain and Mind Centre. He is Chair of the World
Federation of Neurology ALS/MND Specialty Group and established the Pan-Asian Consortium for
Treatment and Research in ALS (PACTALS). He is President of the Brain Foundation and currently
serves as Editor-in-Chief of the Journal of Neurology, Neurosurgery & Psychiatry.
xviii THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Philipp Klar is graduate student for Ph.D. in neuroscience at the University of Duesseldorf/Germany. His
main interest is neurophilosophy and especially consciousness with its neural mechanisms and correlates.

Robert T. Knight is a neurologist and professor of Neuroscience and Psychology at UC Berkeley. He has
received the Jacob-Javits-Award from the National Institute of Neurological Disorders and Stroke, the
Distinguished Career Contribution Award from the Cognitive Neuroscience Society and the Howard-
Crosby-Warren-Medal for Distinguished Career Contributions to Psychological research. He is a member
of the American Association for the Advancement of Science and a Fellow of the American Academy of
Science. His laboratory records intracranial EEG to understand the role of prefrontal cortex in goal-
directed behavior.

Thomas Koenig is a professor of psychiatric neurophysiology and head of the EEG research laboratories
of the University Hospital of Psychiatry of the University of Bern. His research has been focused on link-
ing mental processes and experiences to EEG data, including the development of theoretical models and
the corresponding analysis tools, as well as empirical studies in healthy and mentally ill subjects. Methods
and theory-wise, He has been one of the key promotors of the EEG microstate methodology that is receiv-
ing increasing attention in the EEG community, and where he contributed a series of software tools to
conduct such analyses. He also developed novel statistical procedures for the analysis of ERPs and makes
a continuously evolving open-source toolbox implementing those tools openly available. Regarding
empirical studies, he has a steady track of contributions to the psychological correlates of EEG micro-
states during resting and to the neurophysiology of auditory verbal hallucinations and experience (or lack
thereof) of agency in the context of psychosis.

Saana M. Korkki is a post-doctoral researcher at the Aging Research Center at Karolinska Institute,
Sweden. Prior to joining Karolinska Institute, she completed a PhD in Psychology (2020) and held a post-
doctoral position at the University of Cambridge, UK. Her research interests concern the cognitive and neural
mechanisms underpinning episodic memory decline in aging. To address these questions, she uses a range
of methods including functional, structural, and molecular neuroimaging and behavioral experiments.

Leah A. Krubitzer is an American neuroscientist, Professor of Psychology, and head of the Laboratory
of Evolutionary Neurobiology in the Center for Neuroscience at the University of California, Davis, USA.
She earned her Ph.D in Psychology at the laboratory of Dr. Jon H. Kaas at Vanderbilt University,
Nashville, TN, USA. She continued her training in the laboratory of Dr. M.B. Calford at the University
of Queensland, Brisbane, Australia. Her research interests center on how complex brains in mammals
evolve from simpler forms, as well as the roles of environment and sensory experience on the functional
organization of the cortex. Among her awards is the MacArthur “genius grant” Fellowship from the John
D. and Catherine T. MacArthur Foundation.

Mrinmayi Kulkarni is a post-doctoral researcher in the labs of Dr. Rosanna Olsen and Dr. Bradley
Buchsbaum, at the Rotman Research Institute, Baycrest Health Sciences. She completed her Master’s
degree in Neuroscience at the University of York, UK, and received her Ph.D. from the University of
Wisconsin Milwaukee. Mrinmayi is interested in studying the brain networks underlying learning and
memory. She uses multimodal techniques such as structural and functional neuroimaging, and eye-
tracking to examine how long-term memory interacts with other systems such as the attention and cogni-
tive control system to support behaviour. She aims to examine how these processes change with age and
disease, with an emphasis on identifying factors that may alleviate age-related cognitive decline.

Tiago Lazzaretti is a Collaborative Professor, Researcher and Advisor of the Post Graduate Program of
Science in Musculoskeletal System from the Faculty of Medicine, University of São Paulo. Founder and
Director of the program Scholars in Medical Innovation since 2020. Founder and CEO of SPERO Science
Innovation company. Member of INOVAHC, Center of Innovation and Entrepreneurship of Hospital das
Clínicas, University of São Paulo. Post doctorate program (2017-2019) related to Tissue Engineering &
Cartilage Regeneration - University of São Paulo, Medical School with research fellow at Harvard
Medical School (CAPES 88881.171651/2018-01). International Society of Knee Surgery and Sports
Medicine (ISAKOS) Osteoarthritis Award - 2017. Doctorate Sandwich Program at Harvard Medical
School (CAPES, 2014). Scientific Coordinator of Tissue Engineering of Bone and Cartilage, Sports
Medicine Division and Site Director of PPCR (Principles and Practice on Clinical Research Harvard
The Editors and Contributors xix

Medical School / Harvard T.H. Chan) at Sírio-Libanês Hospital. Physician assistant of Sports Medicine
Division at University of São Paulo Medical School and collaborator of FIFA Medical Center of
Excellence (IOT-FMUSP).

Yan Li is an associate Professor of Neurology at the 2nd Affiliated Hospital of Wenzhou Medical
University, Wenzhou, China. He received his PhD from Wenzhou Medical University in 2015, following
Jiang-fan Chen, a leading researcher in adenosine neurobiology and received further education as Visiting
Professor from 2019-2020 at the Beth Israel Deaconess Medical Center (BIDMC) of Harvard Medical
School, Boston, USA. Currently, his research focuses on executive dysfunctions of Parkinson’s Disease
and the role of adenosine neuromodulator in cognition.

Zhang Li is a professor who is conducting research into cognitive neurogenetics within the Department
of Psychology at Capitol Normal University, Beijing, China.

Anna Marduy is a medical student and completed a Research Fellowship at the Spaulding
Neuromodulation Center, Boston-MA. She started medical school in 2017 at the University União
Metropolitana de Educação e Cultura, UNIME, Lauro De Freitas, Brazil; where she had positions as
Teaching assistant on Anatomy and Histology, and Medical Semiology. She completed a research intern-
ship at Neuromodulation Research Center – Spaulding Rehabilitation Hospital/Harvard Medical School –
in 2021 and was certified by the Principles and Practice in Clinical Research course – Harvard T.H. Chan
School of Public Health, USA in 2020, a course that he worked after as Boston Local Site Monitor in
2021 and a Teacher Assistant I in 2022. Her interest are focus on neuromodulation, chronic pain in post-
surgical settings, and surgical interventions for cardiovascular disease.

Mario Manto is Professor of Neuroanatomy at the University of Mons (Mons, Belgium). He also teaches
pathophysiology of the nervous system. He is Head of the Department of Neurology at the CHU-
Charleroi (Charleroi, Belgium). He is Editor of The Cerebellum (Springer Nature).

Hans J. Markowitsch was Professor of Physiological Psychology at the University of Bielefeld. He


studied psychology and biology at the University of Konstanz, was studying and working in the Medical
Faculty of the University of Copenhagen and in the Faculty of PPP of Oxford University and had profes-
sorships for biopsychology and physiological psychology at the Universities of Konstanz, Bochum, and
Bielefeld and was offered chairs of psychology and neuroscience at Australian and Canadian Universities.
His research centers on the neural and psychic bases of memory disorders and on interactions between
memory and emotion. He is author, co-author or editor of about 30 books and has written about
700 scientific articles and book chapters.

Jorge Mejias is Assistant Professor of computational neuroscience at the University of Amsterdam in the
Netherlands. He also serves as external faculty at the Carlos I Institute of Theoretical Physics in Spain and
at the European Institute for Theoretical Neuroscience in Paris, France. His team in Amsterdam conducts
theoretical and computational research into the mechanistical principles of perception and memory, with a
particular focus on modeling brain networks as a multiscale system and using such models to understand the
emergence of cognitive functions such as working memory, multisensory integration, and predictive coding.

Paulo S. de Melo began medical school in 2017. He is currently in the sixth year of his graduate course
at Escola Bahiana de Medicina e Saúde Pública (EBMSP), where he had experience as a biomorphology
monitor in 2017 and neuroanatomy monitor in 2018/2019. He completed a research internship at
Neuromodulation Research Center, Spaulding Rehabilitation Hospital/Harvard Medical School, in 2021
and was certified by the Principles and Practice in Clinical Research program, Harvard T.H. Chan School
of Public Health, USA in 2020, working after as the Boston Local Site Monitor in 2021 and as Teacher
Assistant I in 2022. He had experience as a junior scientific consultant at Neurive Inc in 2022.

Christoph Michel has studied Behavioral Biology at the Swiss Federal Institute of Technology (ETH)
in Zurich. He is now Full Professor at the Neuroscience Department of the Medical Faculty of the
University Geneva and Head of the HUG/UNIGE-EEG core of the Biomedical Imaging Center
Lausanne-Geneva. He is the Past-President of the Swiss Society for Neuroscience. He has authored over
260 articles and book chapters and is first author of the book “Electrical Neuroimaging” published by
xx THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Cambridge University Press in 2009. He is Editor-in-Chief of the journal Brain Topography, Associate
Editor of the European Journal of Neuroscience, and Associate Editor of Frontiers in Behavioral
Neuroscience. His group is applying electrical neuroimaging tools to cognitive as well as clinical studies
with the aim to unravel the spatio-temporal properties of large-scale brain network communication in
health and disease.

Alexandr M. Chernorizov – is Professor and Head of the Psychophysiology Department Faculty of


Psychology, Lomonosov Moscow State University (MSU), Russia. Dr. Chernorizov’s main research inter-
est is undertaking fundamental research in comparative psychophysiology of color perception in humans
and animals. The main provisions of “vector psychophysiology” are verifying experimentally, describing
information processing in neural networks. In neurophysiological experiments a generalized spherical
model of light and color perception mechanisms was substantiated in humans and animals. This model
allows combining of psychophysical (behavioral) and neurophysiological results of color perception
study, within a single metric system. Dr. Chernorizov’s research results are presented in more than 150
peer-reviewed articles and summarized in the 2019 monograph, “Vector psychophysiology: From behav-
ior to neuron”.

María-Isabel Miranda earned her PhD in Neuroscience at the Instituto de Fisiología Celular of the
Universidad Nacional Autónoma de México (UNAM), where she focused on cortical cholinergic system
function during memory formation. Dr. Miranda completed a postdoctoral fellowship at at the Center for
Neurobiology of Learning and Memory in Dr. James McGaugh´s laboratory at the University of
California, Irvine,. In 2004, she joined the Instituto de Neurobiología, UNAM as a Principal Investigator,
where she is currently working on several projects related to neurochemical and anatomical interactions
during learning and memory.

Lars Muckli is Professor of Visual and Cognitive Neurosciences, Director of fMRI at the Centre for
Cognitive Neuroimaging (CCNI) in Glasgow, and Co-chair of 7T-Imaging Center of Excellence (ICE)
MRI. He has worked for 24 years in the field of fMRI and multi-modal brain imaging. His work focuses
on brain imaging of cortical feedback, investigation of layer specific fMRI, and multi-level cross –species
computational neuroscience. The Muckli-lab was previously funded by ERC consolidator grant on “Brain
reading of contextual feedback and predictions.” Since 2016, Lars is member of the Human Brain Project
(HBP), leading a work package on rodent and human neuroscience on “Context-sensitive multisensory
object recognition a deep network model constrained by multi-level, multi-species data.”

Yoko Nagai is a Senior Lecturer at Brighton and Sussex Medical School. She is a translational neurosci-
entist with interest in mind-body interaction and autonomic function. Her research over the last 20 years
has centred on investigating and developing a non-pharmacological treatment for epilepsy. This pioneer-
ing work attracted funding for clinical trials from various grant bodies. Two clinical trials involving
patients with drug resistant epilepsy demonstrated successful outcome and this non-drug therapy is
undergoing translation for clinical dissemination. More recently, her research interests expanded from
human wellbeing to social wellbeing, integrating her educations in science, business and law.

Lars Nyberg serves as Professor of Psychology and Neurosciences at Umeå University, Sweden. He has
been active in the field of functional neuroimaging of memory since 1994. He is the director of Umeå
Center for Functional Brain Imaging (UFBI), and a principal investigator of the Betula longitudinal pro-
ject on aging, memory, and dementia. Since 2008, he is a member of the Royal Swedish Academy of
Sciences. Lars Nyberg’s research is focused on the identification of genetic, brain, and life-style predic-
tors of heterogeneity in cognitive-aging profiles.

Rosanna K. Olsen is a Senior Scientist at the Rotman Research Institute, Baycrest Health Sciences and
an assistant professor in the Department of Psychology at the University of Toronto. She completed her
undergraduate degree at the University of California, San Diego and her Ph.D. at Stanford University.
Dr. Olsen studies how the brain supports human memory and how these memory-related brain regions
change as we age. She uses neuroimaging as well as eye-tracking to better understand how memories are
formed and later recalled. She also leads an international working group on the standardization of neuro-
imaging methods.
The Editors and Contributors xxi

Daniel Osorio-Gómez received his BS in Psychology, a MS, and PhD in Biochemistry from the
Universidad Nacional Autónoma de México (UNAM). In 2018, He was incorporated as an Associated
Researcher at Instituto de Fisiología Celular, UNAM. His research focuses on the involvement of the
brain circuits associated with emotional learning and memory with a special interest in appetitive and
aversive gustatory memories. He is a member of the National System of Investigators under the National
Council of Science and Technology of Mexico (CONACyT).

Kevin Pacheco-Barrios holds a medical degree, a master’s degree in neuroscience, and a master’s
degree in public health with a concentration in quantitative methods and artificial intelligence from the
Harvard T.H. Chan School of Public Health. Currently, he is a senior research associate in the
Neuromodulation Center & Center for clinical research learning, at Spaulding Rehabilitation Hospital
and Massachusetts General Hospital, Harvard Medical School. He also is an active member in the
COCHRANE and GRADE methodological working groups. He has published over 100 papers in
indexed journals and presented in several international conferences. His main research interests are pain
medicine, neuromodulation, e­ vidence-based neurology, global neurology, patient-empowerment
­methods, and adaptive neurotechnology.

David J. Parker is a lecturer at the Department of Physiology, Development and Neuroscience,


University of Cambridge. His research interests are in nervous system plasticity and neural circuits. This
work started during his PhD and continued in his postdoctoral work on the lamprey locomotor network.
Has has examined the synaptic organization of the locomotor network, and more recently changes in
spinal cord function after injury.

Goran Papenberg is a Principal Researcher at the Aging Research Center at Karolinska Institute,
Sweden. He conducted his PhD in Psychology at the Max Planck Institute for Human Development
(Berlin, Germany), awarded in 2012 by the Free University in Berlin. His main field of research is the
molecular contributions to cognitive aging, with a special focus on dopamine and neuroinflammation. He
uses genetic, epigenetic, lifestyle, and neuroimaging techniques (MRI and PET) to understand the hetero-
geneity in brain and cognitive aging.

Martin J. Pearson is a Senior Lecturer at the University of the West of England, Bristol and theme leader
for Neurorobotic and Biomimetic research at the Bristol Robotics Laboratory. He holds a BEng. In
Electronic and Electrical Engineering from the University of Manchester and MSc. and PhD. from
University of the West of England, Bristol. His research interest lies at the intersection of robotics, animal
behaviour, neuroscience, and AI and has published this work in over 50 peer reviewed articles.

Cyriel Pennartz is Professor of Cognitive and Systems Neuroscience at the Swammerdam Institute for
Life Sciences and the University of Amsterdam, the Netherlands. His current research is focused on
neural mechanisms of memory, perception, sleep, and consciousness. His group uses a multidisciplinary
combination of techniques to understand the relationships between distributed neural activity and cogni-
tion, including in vivo electrophysiology and in vivo two-photon imaging, animal behavior, and compu-
tational modeling. A specialty of this group is to perform high-density parallel recordings from neuronal
ensembles in task-performing animals and analyze population codes, rhythmicity and coherence of neural
activity, and correlate neuronal firing patterns to behavioral and cognitive events. In the field of con-
sciousness research, he developed a theory called Neurorepresentationalism, which uses predictive coding
circuits as building blocks for multi-level perceptual representations.

Arcady A. Putilov is an expert on scientific study of rhythmic phenomena in living nature and individual
variation in the fields of chronophysiology, psychology, and psychiatry. Dr. Putilov is the head of inde-
pendent research group for Mathematical Modeling of Biomedical Systems (the Research Institute for
Molecular Biology and Biophysics of the Federal Research Centre for Fundamental and Translational
Medicine, Novosibirsk). He is the first and/or corresponding author of 95% of more than a hundred
research papers on chronobiology, somnology, personality, biological psychiatry, etc. published in peer
reviewed international journals.

Hui-Xin Qi is a Research Associate Professor of Psychology at Vanderbilt University, Nashville, TN,


USA. She received her Ph.D. in Neurophysiology from the laboratory of Dr. Marie-Claude
xxii THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Hepp-Reymond in the Brain Research Institute, University of Zurich (Zurich, Switzerland). Her current
research interest is on understanding the mechanisms of recovery in the somatosensory and motor systems
of primates after sensory loss by using of anatomical, behavioral, and functional approaches. With long-
standing collaboration with Dr. J. H. Kaas, they continue to make contributions toward fundamental and
translational neuroscience.

Lisa Quadt is a neurodivergent researcher at the Department of Clinical Neuroscience, Brighton and
Sussex Medical School. She applies her research interest in the central and autonomic nervous system
to the understanding of the unique strengths and challenges of neurodivergent populations. She attained
her PhD in Philosophy, taking an interdisciplinary approach to characterizing social cognition before
transitioning into neuroscience. She has led the implementation and testing of a large randomized clini-
cal trial using interoceptive training to decrease anxiety in autistic adults, which was subsequently pub-
lished in EClinicalMedicine.

Vishal Rawji completed his medical training and MBPhD at University College London with Professor
John C. Rothwell, Professor Marjan Jahanshahi and Professor Tom Foltynie. He used non-invasive brain
stimulation techniques and reaction time paradigms to study motor physiology and movement in healthy
individuals and patients with neurological disorders such as Tourette syndrome and Parkinson’s disease.
His research interests include human motor control, motor neuroscience in neurological disorders and
brain stimulation techniques. He is currently a post-doctoral research fellow working with Professor
Dario Farina at Imperial College London and is also working as a medical doctor at St Thomas’
Hospital, London.

Jamie L. Reed is a Research Assistant Professor of Psychology at Vanderbilt University, Nashville, TN,
USA. She was trained in the laboratory of Dr. J. H. Kaas at Vanderbilt, whose work in comparative neu-
roanatomy and neurophysiology has been foundational in understanding the evolution and functional
organization of sensory and motor systems. There she has continued her long-time research collaborations
to characterize contributions to the receptive field properties of neurons in primary somatosensory cortex
and the effects of sensory loss and therapeutic interventions.

Miguel E. Rentería leads the Computational Neurogenomics Laboratory at the QIMR Berghofer
Medical Research Institute in Brisbane, Australia. He has received recognition for his work, including the
2023 Rosenstrauss Fellowship from the Rebecca L. Cooper Medical Research Foundation in Sydney,
Australia. With a background in human genetics, Miguel holds a PhD from the University of Queensland,
Australia, a Master of Public Policy from Oxford University, and a BSc in Genomic Sciences from the
Universidad Nacional Autónoma de México. He has further advanced his expertise through postdoctoral
research at the Brigham and Women’s Hospital and Harvard Medical School in Boston, USA, supported
by an early career fellowship from Australia’s National Health and Medical Research Council. His
research focuses on using clinical, genetic, and neuroimaging data from global cohorts to understand and
identify biomarkers for complex psychiatric and neurological disorders, including depression, self-harm,
chronic pain, migraine, Parkinson’s disease, and sleep apnea. Additionally, he is an Atlantic Global
Fellow of the Global Brain Health Institute, an initiative from the University of California, San Francisco,
and Trinity College Dublin.

Shannon L. Risacher is trained in medical neuroscience with a focus on neuroimaging of Alzheimer’s


disease and related dementias (ADRD), as well as other neurodegenerative diseases. Her publications
focus on imaging and other biomarkers of ADRD, primarily structural and functional magnetic resonance
imaging (MRI) and positron emission tomography (PET). She has evaluated structural and functional
MRI scans with automated and semi-automated programs, as well as PET scans with multiple tracers (i.e.,
amyloid, tau, etc) from local cohorts (i.e., Indiana Memory and Aging Study (IMAS), Indiana Alzheimer’s
Disease Research Center (IADRC)) and national initiatives (i.e., Alzheimer’s Disease Neuroimaging
Initiative (ADNI)). She also works collaboratively to integrate neuroimaging measures with genetics,
cognitive and clinical variables, medical history, and environmental factors. Dr. Risacher also investigates
novel sensory biomarkers of ADRD, including vision, olfaction, and auditory function in older adults at
risk for ADRD. Finally, she focuses on the impact of lifestyle and co-morbid medical conditions, as well
as medication use, on neuroimaging biomarkers of ADRD.
The Editors and Contributors xxiii

Giacomo Rizzolatti graduated in Medicine and Neurology in Padua. He has been Professor of Human
Physiology in Parma since 1975. He is member of Accademia Nazionale dei Lincei, Academia Europaea,
Académie Francaise des Sciences, American Academy of Arts and Sciences, National Academy of
Science, Royal Society and Istituto Lombardo. He received the Prize Feltrinelli for Medicine, Grawmeyer
Prize for Psychology, the Prize Prince of Asturias and the Brain Prize for Neuroscience from Lundbeck
Foundation. He has been awarded with Honorary Degrees from Universities of Claude Bernard Lion,
St. Petersburg, K U of Leuven, Sassari, San Martin of Buenos Aires and Universidad de Montevideo.

Rafaella Rogatto de Faria is a Biomedical Engineer graduated from PUC-SP and a Doctoral student in
sciences of the musculoskeletal system at the Faculty of Medicine, University of São Paulo. She works
as a researcher at Cultivare and with projects in the field of tissue engineering and healthcare innovation
at the startup Spero Science Innovation. She won 1st place in the Cândido de Pinto Melo Award, granted
by SBEB at the Brazilian Congress of Biomedical Engineering.

Nicolas Rohleder graduated from University of Trier in Germany, in 2003. He is a psychologist with a
focus on biological and health psychology. His main research interest is on the pathways between Central
Nervous System (CNS) states such as stress, depression, and trauma and pathophysiological changes in
the organism. He currently directs the chair of Health Psychology at Friedrich-Alexander University
Erlangen-Nürnberg.

John C. Rothwell is currently Emeritus Professor of Human Neurophysiology at UCL Queen Square
Institute of Neurology and Fellow of the Academy of Medical Sciences. His work has provided the theo-
retical rationale and methodological developments underpinning the use of transcranial magnetic stimula-
tion (TMS) as a novel therapy in stroke and depression. He has been a primary contributor to our
understanding of how TMS interacts with ongoing brain activity, and has devised techniques to probe
synaptic connections between brain areas that are now used as biomarkers in neurological disease and
movement disorders. He has pioneered methods of repetitive stimulation that modulate synaptic plasticity,
the basis of behavioural learning. This provides a unique opportunity to influence activity in human cer-
ebral cortex and has opened up new therapeutic opportunities in neurology and psychiatry. He is an author
on over 700 publications with an h-index of 159.

Norihiro Sadato is Professor at the National Institute for Physiological Sciences (since 1999) in Japan.
Following completion of his MD, he subsequently earned a PhD at Kyoto University. Dr Sadato’s
research interests include understanding the human brain’s plastic change mechanisms, accompanied by
learning, sensory deprivation, and development of social cognition and its neural correlates (explored via
functional MRI). Other multimodal measures employed by Dr Sadato include the Electroencephalogram
(EEG), Magnetoencephalography (MEG), and Near-infrared spectroscopy (NIRS), as appropriate. In
addition, he utilizes hyper-scanning fMRI (with two MRI scanners) in exploring the neural substrates of
interpersonal cognitive interactions (e.g., during joint attention and eye contact).

Alireza Salami is an Associate Professor and a group leader at the Wallenberg Centre for Molecular
Medicine at Umeå University, Sweden, and a Principal Investigator at the Aging Research Center at
Karolinska Institute, Sweden. He received his PhD in Physiology (Neuroscience) in 2012 from Umeå
University. The main aim of his research is to better understand molecular and functional basis of cogni-
tive decline in normal and pathological aging, using advanced functional magnetic resonance imaging
(fMRI) and positron emission tomography (PET) techniques.

Andrew J. Saykin is the Raymond C. Beeler Professor of Radiology and Imaging Sciences, and
Professor of Medical and Molecular Genetics, Neurology and Psychiatry, at Indiana University School of
Medicine. He serves as director of the IU Center for Neuroimaging as director of the NIA-designated
Indiana Alzheimer’s Disease Research Center. Dr. Saykin leads the Genetics Core of the Alzheimer’s
Disease Neuroimaging Initiative (ADNI) and co-leads several other research consortia including
CLEAR-AD, AI4AD and KBASE. Dr. Saykin’s research program focuses on precision medicine for early
detection of AD and related dementias and identification of disease mechanisms and potential diagnostic
and therapeutic targets. He co-leads a collaborative group of investigators and trainees using integrative
analysis strategies to study the relationship among clinical phenotypes, genetic susceptibility, and
xxiv THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

molecular signatures of Alzheimer’s and other complex diseases. Computational approaches to address
key questions include artificial intelligence (deep learning, machine learning) and network science (brain
connectomics, systems biology and social network analysis). Dr. Saykin participates in multiple pre- and
post-doctoral training programs, is an author or co-author of over 550 publications, and is the founding
Editor-in-Chief of Brain Imaging and Behavior, a Springer-Nature journal.

Martin Seeber obtained his Ph.D. at Graz University of Technology, studying neural correlates of move-
ment including gait. Afterward, he joined the basic neuroscience department at Campus Biotech in
Geneva, Switzerland, to investigate intrinsic, spontaneous brain dynamics by combining intracranial and
scalp electroencephalography. Currently, he is an Assistant Researcher at UCLA, where he aims to better
understand neural oscillations related to movement trajectories and the formation of episodic knowledge
with intracranial EEG and full-body motion capture.

Erich Schröger is a Professor of Cognitive and Biological Psychology at the Wilhelm Wundt Institute of
Psychology at Leipzig University. His fields of research include perception, voluntary and involuntary
attention, memory, and the perception-action cycle, all of which he mainly studies in the auditory domain.

Alexey Semyanov is a Professor of Neurophysiology known for his pioneering work in the field of extra-
synaptic signaling in the brain. He also described local synaptic and perisynaptic signaling mediated by
potassium. His most recent work is related to astrocytic physiology. Semyanov revealed spatiotemporal
properties of calcium activity in single astrocytes and astrocytic networks. In the serious of recent papers,
he described spatiotemporal remodeling of astrocytes in neurodegenerative diseases and ageing.

Marcel Simis is a Neurologist, Neurophysiologist and Physiatrist. He is a Collaborative Professor at


Faculty of Medicine, University of São Paulo (USP). He is the head of the Neuromodulation Laboratory
at Institute of Physical Medicine and Rehabilitation at USP.

Irina G. Skotnikova has a PhD in Psychological Science and is Leading Researcher at the Institute of
Psychology Russian Academy of Sciences, laboratory of Cognitive Processes and Mathematical
Psychology. In 1972 she graduated from the Faculty of Psychology of Lomonosov Moscow State
University, where she specialized in the fields of Psychology of Cognitive and Emotional Processes. The
main fields of research are: psychophysics, visual discrimination, time and face perception, decision
making, individual differences in humans and animals, cognitive styles, confidence, mathematical mod-
eling, subject-oriented psychophysics, animal psychophysics and cognitions. The role of a subject’s activ-
ity structure (of his/her tasks goals and conditions, individual differences and mental states) was revealed
while performing sensory and cognitive tasks in uncertain situations. Thus, a subject-oriented approach
in psychophysics was justified. The research results are presented in more than 140 articles, and results
in the field of psychophysics in 11 handbooks, monographs, and collective monographs.

Angelica Staniloiu is a senior lecturer at the Universities of Bielefeld (Germany) and Bucharest
(Romania). She studied medicine and psychology at Universities in Bucharest, Boston, and Bielefeld and
has certificates and licenses for practicing medicine and psychiatry from the Massachusetts Board of
Registration in Medicine, the Royal College of Physicians and Surgeons (Ontario, Canada), the Medical
Council of Canada, the American Board of Psychiatry and Neurology, and the Government of North-
Rhine Westphalia and the Ärztekammer Münster (for Germany and the European Union). She is staff
psychiatrist at Oberberg Clinic in the Black Forest. She has a number of publications on memory and
memory disorders, consciousness, and emotion, including articles in Lancet on “Amnesic disorders” and
in Lancet Psychiatry on “Dissociative amnesia.”

Pier S. Paolucci is Researcher at INFN (Istituto Nazionale di Fisica Nucleare) in Rome, Italy. His
­interests are the modeling of the beneficial cognitive and energetic effects of sleep during incremental
learning and the development of tools for the characterization and comparison of the spatio-temporal
features of cortical waves expressed by actual brains and brain models. Between 1998 and 2009, he served
as CTO of the design center of a leading semiconductor manufacturer, directing the design of the Diopsis
family of Multi-Processor Systems-on-Chip. Since 1984, he has been a member of the INFN APE
parallel/­distributed computing laboratory, where he has contributed to the application-hardware-software
The Editors and Contributors xxv

co-design essential for several generations of APE parallel and distributed systems. He is inventor and
co-inventor of several international patents, numerical algorithms, and hardware-software co-design
solutions.

Francesca Starita is a Junior Assistant Professor at the Department of Psychology of the University of
Bologna and researcher at Center of Studies and Research in Cognitive Neuroscience and the Motivation,
Decision and Learning Group. Her research concerns the psychophysiological, cognitive, and neural
bases of human reinforcement learning, decision making and affective processing, with a particular focus
on aversive motivation and fear. She uses a multimodal approach, combining behavioral methods, psy-
chophysiological (e.g. skin conductance, electromyography, eye tracking) and electrophysiological
(EEG) techniques, and non-invasive brain stimulation (e.g. TMS), both in healthy participants and in
patients with brain injury.

Paul C. J. Taylor is a cognitive neuroscientist. After initial graduate and postgraduate work at Oxford
and London he has held positions at LMU Munich in the medicine, philosophy, and psychology faculties.
Paul and his group research into attention, vestibular cognition, and perception during movement. They
often use transcranial magnetic stimulation (TMS) in order to investigate the causal roles of cortical areas
in perceptual cognition. They also contribute to the development of new approaches, such as combining
EEG online with TMS, tACS, eye movements and locomotion, looking for new ways to find out what our
attentional systems do as we explore the world around us.

Matthew Tharp received their B.S. in biomedical engineering from Purdue University where they devel-
oped an interest in data methods in neuroscience while assisting in the development of mathematical
models to study central auditory processing under Dr. Edward Bartlett. They since joined the Center for
Neuroimaging (CfN) and Indiana Alzheimer’s Disease Research Center (IADRC) to provide support for
data projects with a focus in preprocessing methods for neuroimaging and novel analytical approaches in
statistics, dimensionality reduction, and machine learning in addition to developing web applications to
streamline large-scale neuroimaging data management.

Paul M. Thompson is a Professor of Neurology, Psychiatry, Radiology, Pediatrics, and Engineering,


at the University of Southern California (USC) where he directs the Imaging Genetics Center, and is
Associate Director for the Stevens Neuroimaging & Informatics Institute. Professor Thompson is
also PI and Co-founder of the ENIGMA Consortium (http://enigma.ini.usc.edu). ENIGMA has coop-
eratively analyzed data from over 45 countries to publish the largest worldwide neuroimaging studies
of over 15 brain diseases and conditions, including Parkinson’s disease, epilepsy, ataxia and brain
injury, PTSD, substance use disorder, bipolar disorder, and major depression, and neurodevelopmen-
tal conditions including OCD, ADHD and ASD. In parallel, the ENIGMA Consortium has led world-
wide imaging genetics studies that discovered over 500 common and rare genomic variants that
affect brain structure, disease risk, and brain connectivity (Grasby et al., Science, 2020). Recent
efforts have discovered major factors influencing brain development and disease worldwide, in popu-
lations of diverse ancestry. Dr Thompson also co-leads AI4AD - an $18M NIH Initiative developing
AI methods for automatic disease subtyping, genetic target discovery and drug discovery in
Alzheimer’s disease.

Sicong Tu is a cognitive neuroscientist and Lenity Fellow for neurodegenerative disorders at the Brain
and Mind Centre, University of Sydney. He was awarded his PhD from the University of New South
Wales, examining the neural bases of episodic memory in young-onset dementia using multi-modal mag-
netic resonance imaging (MRI). Findings were translated as novel clinical screening assessments now
implemented across multi-national research programs and clinical trials in Alzheimer’s disease. He com-
pleted his post-doctoral studies at the Wellcome Centre for Integrative Neuroscience, University of
Oxford, performing advanced clinical MRI at 3T and ultra-high 7T magnetic field strength to capture
neurodegenerative change. Dr Tu’s research currently specializes in characterizing neural substrates of
cognitive dysfunction across neurodegenerative conditions for the development of novel longitudinal
imaging biomarkers of disease.

Jack van Honk is Professor of Social Neuroscience at Utrecht University as well as at the Neuroscience
Institute, University of Cape Town. He has investigated causal research methodologies in social, affective,
xxvi THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

cultural and clinical neuroscience, using techniques such as EEG, ECG, eye-tracking, fMRI, rTMS, as
well as manipulation of steroid and peptide hormones. He leads the combined University of Cape Town/
University of Stellenbosch research program into the South African variant of Urbach-Wiethe disease
(UWD), a rare genetic disorder resulting in selective calcification of the basolateral amygdala, the social-
emotional center of the brain.

Bernadette C.M. van Wijk earned her PhD in human movement sciences at the Vrije Universiteit
Amsterdam. She focused beta oscillations in the sensorimotor system of healthy individuals using elec-
troencephalography (EEG), magnetoencephalography (MEG), and electromyography (EMG). Since
2013, she has held postdoctoral positions at University College London and the Charité – University
Medicine Berlin, where she investigated LFP recordings from deep brain stimulation electrodes in
patients with movement disorders. Using advanced signal processing techniques (such as cross-frequency
coupling and dynamic causal modeling), she has investigated physiological and pathological neural oscil-
lations within the cortico-basal ganglia network with the aim of improving deep brain stimulation treat-
ment for Parkinson’s disease patients.

Lídia Vaqué-Alcázar obtained her Biomedical Sciences degree from the University of Barcelona. In
2020, she acquired the PhD in Medicine and Translational Research from the University of Barcelona.
As a predoctoral researcher, her main scientific interest has been the study of brain mechanisms that
support preserved cognition in aging. Specifically, she has used multimodal magnetic resonance imag-
ing and non-invasive brain stimulation tools to better understand the structural and functional brain
changes related to the aging processes, the associations of these neural substrates with cognition, and
how cognitive reserve acts as a modulator of these factors and their associations. In 2022, she joined the
Sant Pau Memory Unit as a postdoctoral researcher, where she develops research on reserve and resil-
ience mechanisms in Alzheimer’s disease in adults with Down’s syndrome applying multimodal neuro-
imaging approaches.

Alexei Verkhratsky is an internationally recognized scholar in the field of cellular neurophysiology, best
known for his contributions to our knowledge of the physiology and pathophysiology of neuroglia.
Dr. Verkhratsky also pioneered the emerging field of neurogliopathology, having characterized astroglial
asthenia and paralysis as key pathogenetic steps in age-dependent neurodegenerative diseases. In a series
of conceptual papers and monographs he has outlined basic principles of glial physiology and pathophysi-
ology, which have greatly influenced this rapidly developing area of neuroscience.

Tor Wager is the Diana L. Taylor Distinguished Professor in Neuroscience at Dartmouth College, and
the Director of Dartmouth’s Cognitive and Affective Neuroscience laboratory, the Dartmouth Brain
Imaging Center, and the Dartmouth Center for Cognitive Neuroscience. Professor Wager’s research cent-
ers on the neurophysiology of affective processes – pain, emotion, stress, and empathy – and how they
are shaped by cognitive and social influences. One focus area is the impact of thoughts and beliefs on
learning, brain function, and brain–body communication. Another focus is the development of brain bio-
markers that track and predict affective experience, including pain and other clinical symptoms. A third
focus is on statistical, machine learning, and computational techniques that provide a foundation for new
models of the affective brain. Professor Wager’s laboratory conducts basic research in these focus areas
and applies the resulting techniques and models to collaborative, translational research on clinical disor-
ders and interventions. In support of these goals, Professor Wager and his group have developed several
publicly available software toolboxes (see http://canlab.github.io). He also teaches courses and workshops
on fMRI analysis and has co-authored a book, Principles of fMRI. More information about Dr. Wager and
his lab’s activities, publications, and software can be found at http://canlab.science.

Meghan Whalen is currently a PhD student at Tufts University studying Psychology. Her research
focus is fear learning and affect labeling as a potential treatment for post-traumatic stress disorder
(PTSD), anxiety, and other emotional disorders. Prior to her doctoral studies, Meghan worked in clinical
trials for neuromodulation of chronic pain conditions at Spaulding Rehabilitation Hospital’s
Neuromodulation Center.
The Editors and Contributors xxvii

Meichen Yu is an Assistant Professor at the Department of Radiology and Imaging Sciences at Indiana
University School of Medicine. Dr. Yu’s research expertise is in the areas of multimodal neuroimaging
(MRI and EEG/MEG) and human connectome analyses in dementia and mood disorders. He worked as
a Postdoc Fellow in Computational Neuroscience at the Department of Psychiatry at the University of
Pennsylvania, Philadelphia. He obtained a Ph.D. in Computational Neuroscience from the Amsterdam
University Medical Center (location VUmc), The Netherlands. He earned his master’s degree in
Theoretical Physics from Lanzhou University in China. Dr. Yu has been serving on the editorial boards of
several peer-reviewed journals, including Brain Imaging and Behavior, Frontiers in Psychiatry, and
Frontiers in Psychology. Dr. Yu’s research has been supported by several grants from the Alzheimer’s
Association and National Institute on Aging. Since 2013, Dr. Yu has published several research articles in
top-tier peer-reviewed journals, such as Nature Reviews Neurology, PNAS, Brain, Trends in Cognitive
Sciences, Neuroimage, Neuroimage: Clinical, Human Brain Mapping, Molecular Psychiatry, Lancet
Psychiatry, Chaos, and Neurobiology of Aging.

Alessandro Zanini completed his undergraduate degree in Clinical Psychology and Neuropsychology at
the University of Milano-Bicocca (Italy), with a master thesis focused on the effects of tDCS on motor
planning in both healthy-brain participants and apraxic patients. He subsequently received his PhD at the
Claude Bernard University of Lyon (France) in 2021, where he studied the behavioral effects and fMRI
responses induced by the presentation of multisensory stimuli within the peripersonal space in humans.
His current research as postdoctoral associate at the Centre for Functional and Metabolic Mapping
(Western University, London, Ontario) focuses on the behavioral, cognitive, and MRI repercussions of
alpha-synuclein propagation in marmosets’ brain.

Laiquan Zou completed his Ph.D. in the Institute of Psychology, Chinese Academy of Sciences,
Beijing, China. He was also a visiting scholar at the Interdisciplinary Center for Olfaction and Gustation,
Technische Universität Dresden, Germany. Currently, Dr. Zou is an Associate Professor in the
Department of Psychology, Southern Medical University, Guangzhou, China. His research is focused on
multimodal brain imaging technology, neurophysiological and psychophysical methods in relation to the
processing mechanism of olfaction in neuropsychiatric diseases such as Anorexia Nervosa, Parkinson’s
Disease, and Schizophrenia.
This page intentionally left blank
1
Cognitive Neuroscience: Basic
Principles, Systems, and Methods
Gregory J. Boyle, Georg Northoff,
Nadia Bolognini, Aron K. Barbey,
Marjan Jahanshahi, Álvaro Pascual-Leone,
and Barbara J. Sahakian

INTRODUCTION processes can offer not only relevant fundamental


insights into brain–behavioral relations, but pos-
Cognitive neuroscience is the interdisciplinary sibly also lead to actionable knowledge that can
study of how cognitive/intellectual functions are lead to impactful translational clinical applica-
processed and represented within the brain (e.g., tions and novel therapeutic approaches to patients
Braun et al., 2015; Buzsáki, 2019, 2020; Carrion- with various brain-related disabilities.
Castillo et al., 2020; Chen et al., 2019; Costanzo
et al., 2015; Donoso et al., 2014; Douw et al.,
2016; Gazzaniga, 2018; Gotts et al., 2013; Gross,
1995; McAvoy et al., 2016; Northoff, 2018, 2022;
Picchioni et al., 2007; Poeppel et al., 2020;
HISTORICAL TRACES OF COGNITIVE
Sahakian, 2014; Sahakian and Gottwald, 2016; NEUROSCIENCE
Tandon, 2000; Wang et al., 2014; Winters et al.,
2004; Wu et al., 2021, 2022a; Zhang et al., 2022). Cognitive neuroscience can be traced historically
Cognitive neuroscience is the result of merging to behavioural neurology and clinical neuropsy-
psychology (i.e., the scientific exploration of chology during the nineteenth century wherein
human cognitive mental processes), and neurosci- patients with brain lesions were investigated
ence (i.e., the scientific study of the central (cf. Boyle et al., 2012b, 2023a,b). The study of
nervous system or CNS), in order to study how the brain-injured patients has revealed how cognitive
human brain processes and supports cognitive functions, such as attention (e.g., visual attention;
functions using rather sophisticated technologies Kastner and Ungerleider, 2000), perception
(e.g., fMRI; fNIRS; MEG – see below). Since its (Pomerantz, 2003), working memory (Baddeley,
inception in the early 1990s, cognitive neurosci- 2007; Redondo-Camós et al., 2022; Schacter and
ence has shed substantial light on many such Addis, 2007), awareness, and learning (Terry,
important questions despite the emergence of 2006) are affected by lesions within particular
some critiques (e.g., Uttal, 2011). Essentially, the anatomical areas of the brain. Historically, the
core belief underlying cognitive neuroscience is investigation of brain lesions led French neurolo-
that understanding the neurobiological underpin- gist Paul Broca to discover a distinct brain region
nings of psychological, cognitive, and behavioral in the left inferior frontal cortex that is primarily
2 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

dedicated to speech production – designated as the brain regions and networks is distinguished from
speech motor articulation center (Broca’s area; cf. a more holistic view that assumes global brain
Hagoort, 2014). Likewise, Carl Wernicke discov- activity to be central for cognition (equipotential-
ered the predominant sensory speech reception ism; cf. Halstead, 1947; Hugdahl, 2000). Initially,
center (Wernicke’s area) in the left temporo-parietal equipotentiality had been defined by Karl Lashley
cortex, necessary for speech reception and com- in the 1950s as the capacity of an intact section
prehension (cf. DeWitt and Rauschecker, 2013). of the brain to carry out functions lost by damage
The application of lesion methods to the study of incurred in other areas of the brain (cf. Fancher
large-scale brain networks further enabled the and Rutherford, 2012).
characterization of cognitive control networks for Another historical origin of cognitive neuro-
adaptive behavior and decision making science was behaviorism at the beginning of the
(Barbey et al., 2012, 2013; Hsu et al., 2022; twentieth century wherein psychological func-
Ji et al., 2019; Shine et al., 2016), along with tions were explained in terms of their behavior
neural networks that underlie specific facets of as input–output relations (extended into the mod-
intelligence (cf. Barbey et al., 2014a, 2021; cf. ern cognitive science era wherein such functions
Boyle, 1988; Boyle et al., 2012a; Brody, 1992; are accounted for largely in terms of cognitive
Carroll, 1993; Cattell, 1982, 1987; Horn, 1988; input–output relations). Behavioral scientists rec-
Neisser et al., 1996; Sternberg and Kaufman, ognized the importance of investigating actual
1998; Varshney and Barbey, 2021), including the observable behaviors (e.g., Skinner, 1976; Watson,
ability factors included in the comprehensive 2017; cf. Carpintero, 2004; Malone, 2014; Rilling,
Cattell-Horn-Carroll (CHC) theory of cognitive 2000). Behaviorism did not deny the existence
abilities (Schneider and McGrew, 2018). of subjective experiences but viewed any con-
In this context of lesion-studies and brain– scious or cognitive phenomena (Gage and Baars,
behavior relations, it is important to recall the semi- 2018; Kosslyn and Andersen, 1995) as depending
nal contributions of Constantin von Monakov who on the behavior of the organism. As subjective
emphasized the crucial role of the time domain or experience (e.g., cognition) was defined as inac-
“chronometry” in understanding the neural sub- cessible for scientific inquiry, this shifted the focus
strate on the basis of brain lesions and argued that onto behavioristic stimulus–response relation-
knowing the location and characteristics of a lesion ships. Such stimulus–response relationships have
is necessary but not sufficient in understanding its been studied with different types of tasks such as
impact on behavior (von Monakow and Harris, simple choice, and go- versus no-go reaction times
1969). Instead, von Monakov pioneered the idea that have measured and compared response times
that lesions impact activity across distributed net- to a wide range of incoming stimuli (Jahanshahi,
works of the brain (‘functional localization’) and 2003). In conjunction with the go- no-go reac-
the brain in turn adapts to such functional impact tion time tasks that allow investigation of action
given rise to signs and symptoms (‘symptom restraint, subsequent development of a stop sig-
localization’). Informed by such notions and lev- nal reaction task (Logan and Cowan, 1984) and
eraging the capabilities of modern brain imaging, variants of this have facilitated the study of behav-
resting-state functional magnetic resonance imag- ioural inhibition, which is an important compo-
ing, large data-sets, and sophisticated analytic nent of cognitive control (Jahanshahi et al., 2015).
methodologies, “lesion network mapping” offers In recent decades, there has been extensive
a transformative method to relate patterns of brain examination of the brain regions and neural
activity and the functional impact of brain lesions networks involved in the stop signal reaction time
to cognitive processes, behaviors, and symptoms task. Aron et al. (2003) discovered that a neural cir-
of disease (Boes et al., 2015; Fischer et al., 2016; cuit including the right inferior frontal gyrus was
Joutsa et al., 2022; Kim et al., 2021). responsible for response inhibition. A subsequent
In the development of cognitive neurosci- study demonstrated a reduction in impulsivity by
ence, the early neuroanatomical discoveries by drugs that enhance both dopamine and noradren-
Broca and Wernicke (cf. Raichle, 2009) and the aline activity in the brain, such as methylpheni-
theoretical elaborations by von Monakov served date, but also drugs which were more selective for
as paradigmatic examples that have extended noradrenaline, such as atomoxetine (Chamberlain
into the contemporary connectome mapping et al., 2006). In a functional magnetic resonance
of cognitive, affective, and social functions in imaging (fMRI) study, Chamberlain et al. (2009)
relation to specific anatomical brain regions/ showed that atomoxetine modulated right infe-
networks (cf. Bassett et al., 2020; Bolognini et al., rior frontal activation during inhibitory control,
2013; Glasser et al., 2013; Neumann et al., 2015, thus demonstrating that noradrenaline modulates
2016; Toga et al., 2012; Van Essen et al., 2013). response inhibition by its actions on the right
Localization of cognitive functions to specific inferior frontal gyrus.
Cognitive Neuroscience: Basic Principles, Systems, and Methods 3

Current Status of Cognitive adapted for neuroimaging (cf. Langley et al.,


Neuroscience 2023). For example, during the CANTAB paired
associates learning task (PAL), at lower levels of
Cognitive psychology merged with neuroscience, difficulty, the hippocampi of MCI patients were
leading to the development of cognitive neurosci- activated significantly more than in controls, and
ence as an autonomous discipline in the late significantly less at higher levels of difficulty.
1980s, with many researchers who had been car- Thereby, providing neural validation for the
rying out psychological and behavioral studies CANTAB PAL test in the early detection of
now looking for neuroscientific explanations of Alzheimer’s disease (De Rover et al., 2011).
their findings. During the twentieth century, cog- Using the CANTAB intra extra dimensional set
nitive psychology emerged as a separate field of shifting task (IED), Langley et al. (2021) showed
study, which together with the development of that the frontal striatal circuits involved in
computational models (Churchland and cognitive flexibility were altered in premanifest
Sejnowski, 1988, 2017; Dayan and Abbott, 2001) Huntington’s disease. Moreover, development of
served as a basis for the subsequent emergence of objective standardized neurocognitive psychomet-
cognitive neuroscience (the so-called “cognitive ric testing has also aided in the understanding of
revolution” – Boone and Piccinini, 2016; neuromodulation by neurotransmitters (cf.
Gazzaniga et al., 2018; Mandler, 2002; Miller, Pama et al., 2013; Sahakian and Owen, 1992).
2003; Poeppel et al., 2020). Now cognition was Mehta et al. (2000) using the CANTAB spatial
seen as mediating the behavioral response to sen- working memory task (SWM) showed that meth-
sory input and therefore central to understanding ylphenidate induced improvements in working
the nature of human behavior (cf. Cuzzolin et al., memory performance, which was associated with
2020; Laird et al., 2017; Langley et al., 2022). task-related reductions in regional cerebral blood
Other key ideas from the “cognitive revolution” flow in the dorsolateral prefrontal cortex and pos-
included that while the mind is modular, with terior parietal cortex. This demonstrated that
distinct neural systems dedicated to specific func- methylphenidate improved performance through
tions, orchestrated in the service of complex, goal- greater efficiency of the neural network underly-
directed behavior (Pinker, 2002). ing working memory. In addition, they found that
Emergence of the “cognitive revolution” the beneficial effects of methylphenidate on work-
opened the door for investigating the multifaceted ing memory were greatest in the subjects with
neural underpinnings of cognitive functions such lower baseline working memory capacity (e.g.,
as attention, working memory, and executive func- episodic autobiographical memory recall;
tions, as well as of higher-order processes includ- Roehri et al., 2022).
ing conceptualization of the self and consciousness
(Chan et al., 2008; Chiao et al., 2009a,b; Chiao and
Blizinsky, 2016; Fregni et al., 2005; Gazzaniga,
2018; Northoff, 2014a,b; Northoff et al., 2006; Role of Emotions and Nonconscious
Poeppel et al., 2020; Schaefer and Northoff, Processes
2017), paving the way for the future dialogue with
other disciplines such as social cognition and the However, until 1990s, the role of emotions and
psychological study of personality (cf. Boyle, nonconscious processes remained largely
2019; Boyle and Saklofske, 2004a,b; Boyle et al., neglected by cognitive neuroscientists. At that
2008a,b; 2015; Neumann et al., 2016). time, researchers prioritized the analysis of
mental states that could be translated in terms of
computational operations, namely perception,
learning, and memory, excluding the study of
emotions, affects, and nonconscious processes
CAMBRIDGE NEUROPSYCHOLOGICAL among the research interests of the cognitive
TEST AUTOMATED BATTERY neurosciences (Barbey et al., 2014b, 2014c;
Damasio, 2000; LeDoux, 2000). Recently, com-
In response to gaining a better understanding of puterized batteries for assessing social and emo-
attention, working memory and executive function tional cognition have also been developed. For
processes, a computerized battery incorporating a example, the EMOTICOM battery has not only
wide variety of objective neuropsychological been used in healthy volunteers (Bland et al.,
tests – the Cambridge Neuropsychological Test 2016), including with tryptophan depletion
Automated Battery or CANTAB (camcog.com) – (Kanen et al., 2021), but also in patients with
was constructed (Robbins et al., 1994, 1997, depression (Dam et al., 2019) and Huntington’s
2012). Many CANTAB subtests have since been disease (Scahill et al., 2020).
4 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

METHODOLOGIES AND TECHNIQUES IN possible – later termed magnetic resonance imag-


COGNITIVE NEUROSCIENCE ing or MRI (see NMR Progress Report, 1984).
Using fMRI, the experimenter can administer a
cognitive task that manipulates an independent
In more recent years, leveraging further techno- variable (e.g., the number of items to be main-
logical developments, new tools have emerged tained in working memory) and measure the
that capture behavior and cognitive processes with Blood-Oxygen-Level-Dependent (BOLD) signal
exquisite sensitivity leveraging mobile sensors (dependent variable), while simultaneously moni-
and multimodal signal acquisition (Gomes-Osman toring brain activity (Ogawa et al., 1992) in spe-
et al., 2021; Libon et al., 2022; Souillard-Mandar cific neuroanatomical regions or networks. While
et al., 2021). Digital technologies can offer time- early work using neuroimaging focused on finding
efficient brain assessments that uncover issues not “brain lesions” or other biomarkers of pathology,
otherwise detectable using standard tests. First, contemporary research is focused on detecting
they can capture metrics with a much higher sam- disruptions in the of neurocircuitry connectiv-
pling rate than a human observer (e.g., eye move- ity including subtle “brain signatures” associated
ments, postural and facial expressions, drawing, with certain brain disorders. In addition, high-
writing, and spoken output). Second, they enable resolution, high-density electroencephalographic
the analysis of process by quantifying neurocogni- (EEG) methods along with computational analysis
tive operations in real time as individuals try to of the recorded signals, enables the identification
solve a given task (e.g., Boston Process Approach), of spatio-temporal patterns of brain activity linked
thus quantifying with great accuracy highly to specific cognitive processes, disease symptoms,
nuanced and subtle behaviors previously unob- and behaviors (Michel and Pascual-Leone, 2020).
tainable. Third, data-driven AI-supported analytic
approaches can detect subtle signs of brain decline
and illness with greater sensitivity and specificity
so they can be better managed before they become Non-Invasive Brain Stimulation
clinically evident and disabling for the patient. (Neuromodulation) Techniques
Importantly, such approaches can be deployed in
real-life settings, avoiding the need for the stress- More recently, non-invasive neuromodulation
ful and artificial constraints of formal testing techniques including transcranial electrical stimu-
conditions. lation (tES; Brunoni et al., 2022; Fregni et al.,
Cognitive neuroscience has been greatly facili- 2005; Woods et al., 2016), and transcranial mag-
tated by the emergence of cutting-edge brain netic stimulation (TMS; Caballero-Villarraso
imaging technologies over recent decades, making et al., 2022; Jannati et al., 2023; Pascual-Leone
it possible to investigate the neural correlates of et al., 1991, 1999, 2000, 2005, 2011; Redondo-
various cognitive functions including identifica- Camós et al., 2022), have expanded our knowledge
tion of which regions/networks and frequencies in of brain plasticity (Pascual-Leone et al., 2005) by
the brain’s neural activity are recruited in response probing the causal contribution of brain areas to
to specific cognitive tasks (Bassett and Sporns, behaviour, moving beyond the correlational meth-
2017). Both single photon emission computed ods of fMRI (Boes et al., 2018; Frohlich, 2016)
tomography (SPECT), and positron emission and enabling the dissection of chronometric pro-
tomography (PET) emerged in the 1970s (Portnow cesses of cognition and the mind (Pascual-Leone
et al., 2013). For example, a relatively early study et al., 2000; Walsh et al., 2006; Walsh and Pascual-
combined the spatial resolution of PET with the Leone, 2003). Both tES and TMS can be combined
temporal resolution of movement-related poten- with high spatial and temporal resolution tech-
tials, to compare the neural substrates of temporal niques (MEG1-EEG; EEG-fMRI; EEG-fNIRS2),
decision-making by comparing self-initiated and linking structural and neurochemical data (PET-
externally triggered movements. This compari- CT; PET-MRI), or using one modality to alter
son revealed that the dorsolateral prefrontal cor- neurocircuitry, and another to establish in vivo the
tex mediated volitional decisions about timing of disruptive impact (e.g., fMRI-TMS or tES; TMS-
movements; whereas the activation of the supple- EEG; TMS-fNIRS; EEG-tES; cf. Bolognini and
mentary motor area (SMA) related to preparation Diana, 2021; Bolognini and Ro, 2010; Farzan
(Jahanshahi et al., 1995), and as confirmed in a et al., 2016; Shafi et al., 2012; Wagner et al., 2007).
subsequent PET study of stimulus predictability, Such multi-modal association approaches further
the activation of the rostral SMA relates to prepa- helped to elucidate human brain networks and the
ration whereas the caudal SMA is a motor execu- mechanisms by which they process cognition,
tion area (Jenkins et al., 2000). Subsequently, behavior, and emotion. In an early review of appli-
nuclear magnetic resonance (NMR) became cations of TMS for the study of cognition, the
Cognitive Neuroscience: Basic Principles, Systems, and Methods 5

range of questions that could be addressed with methods and techniques, such as Temporal
TMS included a temporary “lesion” effect to deter- Interference Stimulation (TIS) enable more precise
mine whether the contribution of a target area to and even selective non-invasive stimulation of deep
task performance is essential, identifying the time brain structures (Grossman et al., 2017).
course of cognitive processes (i.e., time window
during which the contribution of a brain area is
essential or the relative timing of the contribution
of two or more brain areas to task performance), DECADE OF THE BRAIN:
what changes in excitability occur in target brain
areas with learning, and investigation of intracorti-
COMPUTATIONAL APPROACHES AND
cal and transcallosal connectivity (Jahanshahi and COGNITIVE NEUROSCIENCE STUDIES
Rothwell, 2000). TMS can be reliably and safely
applied to disrupt the activity of a given brain The decade of the brain (Jones and Mendell,
region transiently and thus provide insights about 1999) has inspired extensive brain mapping efforts
the causality of brain activity in behavior. As an worldwide, including the Human Brain Project in
early application, Pascual-Leone et al. (1991) Europe (HBP; Markram et al., 2011; Sahakian
showed that repetitive TMS can be used to induce et al., 2021), the ENIGMA Consortium (Kong
speech arrests and study language networks in et al., 2018), the Human Connectome Project
health and disease – the Neuronix eXimia System (HCP; Toga et al., 2012; cf. Glasser et al., 2013;
is now cleared by the Food and Drug Administration Van Essen et al., 2013), the China Brain Project
in the United States for this application. Similar (CBP; Poo et al., 2016), and the BRAIN Initiative
application of “temporary lesion approaches” (BI; Litvina et al., 2019). Some of the highlights
using the controlled perturbation induced by TMS of the HBP include the development of the
and guided by brain imaging, has served to clarify, Julich-Brain Atlas (JBA; Amunts et al., 2020) and
for example, the causal role of primary visual the Virtual Epileptic Patient (VEP; Jirsa et al.,
cortex (Kosslyn et al., 1999), and the dynamics of 2017). The European-funded HBP has also cre-
interhemispheric compensatory processes (Sack ated EBRAINS, an open research infrastructure
et al., 2005), during mental imagery; or the need built on the scientific advances and tools devel-
for fast back-projections in visual awareness oped by the project’s research teams, and making
(Pascual-Leone and Walsh, 2001). them available to the scientific community via a
Subsequent electrophysiological research has shared digital platform (https://ebrains.eu/).
employed TMS not only for establishing functional Computational tools have been developed and
specialization, but also for examining functional Big Data has been compiled (Fox and Lancaster,
connectivity, afunctional interaction, and integra- 2002; Yarkoni et al., 2011), as well as the recent
tion within the cognitive domain (Jahanshahi, standardization Neuroimaging Meta-Analysis
2005). Other applications of TMS to the investi- Research Environment (NiMARE)3 endeavor.
gation of cognition have included the pharmacol- The aim has been (1) to link functional and struc-
ogy of cognition through the study of the effects tural brain maps to cognitive tasks from previous
of medication, the impact of surgical or rehabili- studies and (2) to amass and share large data sets.
tative interventions, as well as brain damage, and/ These large-scale collaborations have enabled
or neurological or psychiatric disorders on cogni- application of artificial intelligence techniques
tion (Jahanshahi, 2005; Jahanshahi and Rothwell, using machine learning to predict behavior from
2000). Subsequent applications of TMS as a thera- measures of brain activity (Li et al., 2019). The
peutic tool have included increasing processing combination of Big Data from neuroimaging with
speed, improving selective attention or naming, the human genome project and genetic neuroimag-
improving accessibility to memory, or enhancing ing provide remarkable opportunities for the next
implicit learning (Bolognini and Miniussi, 2018; decade of research and discovery in cognitive neu-
Jahanshahi, 2005; Vallar and Bolognini, 2011). In roscience. Even more powerful is when these Big
addition, in treatment resistant patients with obses- Data cohorts include neuroimaging, cognitive and
sive-compulsive disorder, deep brain stimulation behavioural measures, such as in the IMAGEN,
(DBS) has been shown to improve cognitive flex- ABCD, and UK Biobank research projects.
ibility (Tyagi et al., 2019). The past 25 years has Recent publications from these large cohorts have
brought substantial advances in knowledge about investigated many aspects of brain and behavioral
the mechanisms of action of non-invasive brain function, including sleep (Li et al., 2022), social
stimulation; novel and more precise technologies; isolation (Shen et al., 2022), attention deficit
safety, training, regulatory and ethical guidelines hyperactivity disorder (Shen et al., 2020a,b), OCD
along with research applications and their clinical (Wu et al., 2022b), and the androgynous brain
translation. New non-invasive brain stimulation (Zhang et al., 2021).
6 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Comparative Cognitive Neuroscience cognitive functions and the identification of


Including Studies Into Infra-Human which regions/networks in the brain’s neural
activity are recruited in response to specific cog-
Species nitive tasks (Bassett and Sporns, 2017). The
The study of non-human primates is also an area decade of the brain (Jones and Mendell, 1999)
of interest in cognitive neuroscience that examines has inspired extensive brain mapping efforts
our similarities with respect to cognitive capabili- worldwide, including the Human Brain Project
ties, physiology, neuroanatomy, social complex- in Europe (HBP; Sahakian et al., 2021), the
ity, reproduction, and development. Recent ENIGMA Consortium (Kong et al., 2018), the
developments in magnetic resonance imaging, for Human Connectome Project (HCP; Toga et al.,
instance, have the potential for extracting neuro- 2012), the China Brain Project (CBP; Chang
biological measures that can be compared between et al., 2021), and the BRAIN Initiative (BI;
species within a phylogenetic framework. Litvina et al., 2019). Some of the highlights of
Comparative cognitive neuroscience enriches the the HBP include the development of the Julich-
understanding of how the human brain works and Brain Atlas (JBA; Amunts et al., 2020) and the
the appreciation of its uniqueness by looking to Virtual Epileptic Patient (VEP; Jirsa et al.,
problems the brain evolved to solve and how these 2017). It has also created EBRAINS, an open
relate to its organization (Mars et al., 2014). Both research infrastructure available to the scientific
historical lines of development involving the neu- community via a shared digital platform (https://
roscientific study of the human brain and of the ebrains.eu/). Computational tools have been
brains of non-human primates allows comparative developed and Big Data has been compiled
cognitive neuroscience to share certain empirical (Yarkoni et al., 2011), as well as the recent
features and philosophical assumptions. Neuroimaging Meta-Analysis Research
Environment (NiMARE) endeavor.
Combining Big Data from neuroimaging with
the human genome project and genetic neuro-
Spatiotemporal Cognitive imaging (e.g., Wu et al., 2022b) has enabled
Neuroscience remarkable opportunities for future research and
discovery within cognitive neuroscience. These
However, both the human and non-human primate Big Data cohorts include neuroimaging, cogni-
models of cognitive neuroscience have certain tive and behavioural measures, such as in the
limitations with respect to mental features like self IMAGEN, ABCD, and UK Biobank research
and consciousness (e.g., Chiao et al., 2009a,b) projects. Recent publications from these large
making necessary its extension into what has been cohorts have investigated many aspects of brain
termed “spatiotemporal cognitive neuroscience.” and behavioral function, including sleep (Li et al.,
Most cognitive neuroscientists agree that we need 2022), social isolation (Shen et al., 2022),
to link mental operations to brain functions and to attention-deficit hyperactivity disorder (Shen
do so within an interdisciplinary framework that et al., 2020a,b), obsessive-compulsive disorder
appreciates the importance of both experimental (Wu et al., 2022b), and the androgynous brain
cognitive psychology and neuroscience. We (Zhang et al., 2021).
emphasize the need for approaches that seek to One point we wish to emphasize is that even
integrate experimental psychology with cognitive though the cognitivist paradigm reintroduced the
neuroscience, such as spatiotemporal cognitive notion of “mind” into experimental cognitive psy-
neuroscience (Northoff and Hirjak, 2022; Northoff chology, it led cognitive neuroscience into mutu-
and Huang, 2017; Northoff et al., 2020a,b), as ally exclusive paradigms, both philosophically
well as cognitive computational neuroscience and empirically, which have resurfaced in terms
(Kriegeskorte and Douglas, 2018). of the distinction between the brain’s neuronal
activity versus cognition. Recent developments
have expanded the spatiotemporal framework of
cognitive neuroscience by intrinsically integrating
both neuronal activity and cognition, into the cur-
SUMMARY AND CONCLUSIONS rent approach of “Spatiotemporal Neuroscience.”
Clearly though, we consider that there is need for
Over recent years, cognitive neuroscience has approaches that seek to integrate, for example,
been greatly facilitated by development of cut- cognitive neuroscience with spatiotemporal neu-
ting-edge brain imaging technologies, enabling roscience, along with cognitive computational
investigation of the neural correlates of various neuroscience.
Cognitive Neuroscience: Basic Principles, Systems, and Methods 7

Notes neuroscience. Cambridge, UK: Cambridge Univer-


sity Press.
1  MEG: Magnetoencephalography (a non-invasive Bassett, D. S., Cullen, K. E., Eickhoff, S. B., Farah,
functional neuroimaging technique) measures M. J., Goda, Y., Haggard, P., …Ueda, H. R. (2020).
magnetic fields at the scalp resulting from electri- Reflections on the past two decades of neurosci-
cal brain activity. ence. Nature Reviews Neuroscience, 21, 524–534.
2  fNIRS: functional Near-Infrared Spectroscopy (a Bassett, D. S., & Sporns, O. (2017). Network neuro-
non-invasive optical imaging technique) mea- science. Nature Neuroscience, 20(3), 353–364.
sures brain changes in hemoglobin concentra- Bland, A. R., Roiser, J. P., Mehta, M. A., Schei, T.,
tions upon light being shone through the skull Boland, H., Campbell-Meiklejohn, D. K., … Elliott, R.
and refracted back to a specialized detector (2016). EMOTICOM: A neuropsychological test
(optode). battery to evaluate emotion, motivation, impulsiv-
3  NiMARE provides workflows as command-line ity, and social cognition. Frontiers in Behavioral
interfaces, including ALE meta-analysis, meta- Neuroscience, 10. doi: 10.3389/fnbeh.2016.00025
analytic coactivation modeling (MACM) analysis, Boes, A. D., Prasad, S., Liu, H., Liu, Q., Pascual-Leone,
peaks2maps image reconstruction, and contrast A, Caviness, V. S. Jr., & Fox, M. D. (2015). Network
map meta-analysis. Each workflow generates a localization of neurological symptoms from focal
boilerplate paragraph with workflow details and brain lesions. Brain, 138(10), 3061–3075.
relevant citations. Boes, A. D., Kelly, M. S., Trapp, N. T., Stern, A. P.,
Press, D. Z., & Pascual-Leone, A. (2018). Noninva-
sive brain stimulation: Challenges and opportuni-
ties for a new clinical specialty. Journal of
REFERENCES Neuropsychiatry and Clinical Neurosciences, 30(3),
173–179.
Amunts, K., Mohlberg, H., Bludau, S., & Zilles, K. Bolognini, N., & Diana L. (2021). Multimodal associa-
(2020). Julich-Brain: A 3D probabilistic atlas of the tion of tDCS with electroencephalography. In A.
human brain’s cytoarchitecture. Science, 369, Brunoni, M. Nitsche & C. Loo (Eds.), Transcranial
988–992. direct current stimulation in neuropsychiatric dis-
Aron, A. R., Fletcher, P. C., Bullmore, E. T., Sahakian, orders: Clinical principles and management
B. J., & Robbins, T. W. (2003). Stop-signal inhibi- (pp. 107–126). New York: Springer.
tion disrupted by damage to right inferior frontal Bolognini, N., & Miniussi, C. (2018). Noninvasive
gyrus in humans. Nature Neuroscience, 6(2), brain stimulation of the parietal lobe for improving
115–116. neurologic, neuropsychologic, and neuropsychiat-
Baddeley A (2007). Working memory, thought, and ric deficits. Handbook of Clinical Neurology, 151,
action. Oxford, UK: Oxford University Press. 427–446.
Barbey, A. K., Colom, R., & Grafman, J. (2013). Bolognini, N., & Ro, T. (2010). Transcranial magnetic
Architecture of cognitive flexibility revealed by stimulation: Disrupting neural activity to alter and
lesion mapping. NeuroImage, 82, 547–554. assess brain function. Journal of Neuroscience,
Barbey, A. K., Colom, R., Paul, E. J., & Grafman, J. 30(29), 9647–9650.
(2014a). Architecture of fluid intelligence and Bolognini, N., Rossetti, A., Convento, S., & Vallar, G.
working memory revealed by lesion mapping. (2013). Understanding others’ feelings: The role of
Brain Structure and Function, 219, 485–494. the right primary somatosensory cortex in encod-
Barbey, A. K., Colom, R., & Grafman, J. (2014b). ing the affective valence of others’ touch. Journal
Distributed neural system for emotional intelli- of Neuroscience, 33(9), 4201–4205.
gence revealed by lesion mapping. Social Cogni- Boone, W., & Piccinini, G. (2016). The cognitive neu-
tive and Affective Neuroscience, 9, 265–272. roscience revolution. Synthese, 193, 1509–1534.
Barbey, A. K., Colom, R., Paul, E. J., Chau, A., Solo- Boyle, G. J. (1988). Contribution of Cattellian psy-
mon, J., & Grafman, J. H. (2014c). Lesion mapping chometrics to the elucidation of human intellec-
of social problem solving. Brain, 137, tual structure. Multivariate Experimental Clinical
2823–2833. Research, 8, 267–273.
Barbey, A. K., Colom, R., Solomon, J., Krueger, F., Boyle, G. J. (2019). Overarching personality para-
Forbes, C., & Grafman, J. (2012). An integrative digm: A neo-Cattellian psychometric model. Per-
architecture for general intelligence and executive sonality and Individual Differences, 147, 317–325
function revealed by lesion mapping. Brain, 135, Boyle, G. J., Matthews, G., & Saklofske, D. H.
1154–1164. (2008a). (Eds.), The SAGE handbook of personality
Barbey, A. K., Karama, S., & Haier, R. J. (2021). The theory and assessment: Vol. 1 – Personality theo-
Cambridge handbook of intelligence and cognitive ries and models. Los Angeles: Sage.
8 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Boyle, G. J., Matthews, G., & Saklofske, D. H. Carpintero, H. (2004). Watson’s behaviorism: A com-
(2008b). (Eds.), The SAGE handbook of personality parison of the two editions (1925 and 1930).
theory and assessment: Vol. 2 – Personality meas- History of Psychology, 7(2), 183–202.
urement and testing. Los Angeles: Sage. Carrion-Castillo, A., Pepe, A., Kong, X.-Z., Fisher, S. E.,
Boyle, G. J., & Saklofske, D. H. (2004a). (Eds.), SAGE Mazoyer, B., Tzourio-Mazoyer, N., … Francks, C.
benchmarks in psychology: The psychology of (2020). Genetic effects on planum temporale
individual differences (Vol. 1.): Intelligence. asymmetry and their limited relevance to neurode-
London: Sage. velopmental disorders, intelligence or educational
Boyle, G. J., & Saklofske, D. H. (2004b). (Eds.), SAGE attainment. Cortex, 124, 137–153.
benchmarks in psychology: The psychology of Carroll, J. B. (1993). Human cognitive abilities: A
individual differences (Vol. 3): Cognition, emotion survey of factor-analytic studies. New York: Cam-
and conation. London: Sage. bridge University Press.
Boyle, G. J., Saklofske, D. H., & Matthews, G. Cattell, R. B. (1982). The inheritance of personality
(2012a). (Eds.), SAGE benchmarks in psychology: and ability: Research methods and findings. Paris:
Psychological assessment, Vol. 1: Intelligence Academic.
assessment. London: Sage. Cattell, R. B. (1987). Intelligence: Its structure,
Boyle, G. J., Saklofske, D. H., & Matthews, G. growth and action. Amsterdam: North-Holland.
(2012b). (Eds.), SAGE benchmarks in psychology: Chamberlain, S. R., Muller, U., Blackwell, A. D.,
Psychological assessment, Vol. 3: Clinical neu- Clark, L., Robbins, T. W., & Sahakian, B. J. (2006).
ropsychological assessment. London: Sage. Neurochemical modulation of response inhibition
Boyle, G. J., Saklofske, D. H., & Matthews, G. (2015). and probabilistic learning in humans. Science,
(Eds.), Measures of personality and social psycho- 311, 861–863.
logical constructs. Amsterdam: Elsevier/Academic. Chamberlain, S. R., Hampshire, A., Müller, U., Rubia, K.,
Boyle, G. J., Stern, Y., Stein, D. J., & Sahakian, B. J., Del Campo, N., Craig, K., …Sahakian, B. J. (2009).
Golden, C. J., Lee, T. M.-C., & Chen, S.-H. A. Atomoxetine modulates right inferior frontal acti-
(2023a). (Eds.), The SAGE handbook of clinical vation during inhibitory control: a pharmacological
neuropsychology, Vol. 1: Clinical neuropsychologi- functional magnetic resonance imaging study.
cal disorders. London: Sage. Biological Psychiatry, 65(7), 550–555.
Boyle, G. J., Stern, Y., Stein, D. J., & Sahakian, B. J., Chan, R. C. K., Shum, D., Toulopoulou, T., & Chen, E. Y.
Golden, C. J., Lee, T. M.-C., & Chen, S.-H. A. (2008). Assessment of executive functions: Review
(2023b). (Eds.), The SAGE handbook of clinical of instruments and identification of critical issues.
neuropsychology, Vol. 2: Clinical neuropsychologi- Archives of Clinical Neuropsychology, 23(2),
cal assessment and diagnosis. London: Sage. 201–216.
Braun, U., Schäfer, A., Walter, H., Erk, S., Romanc- Chen, Q., Beaty, R. E., Cui, Z., Sun, J., He, H.,
zuk-Seiferth, N., Haddad, L., … Bassett, D. S. Zhuang, K., … Qiu, J. (2019). Brain hemispheric
(2015). Dynamic reconfiguration of frontal brain involvement in visuospatial and verbal divergent
networks during executive cognition in humans. thinking. NeuroImage, 202. doi: 10.1016/j.
Proceedings of the National Academy of Sciences neuroimage.2019.116065
of the United States of America, 112(37), Chiao, J. Y., Li, Z., & Harada, T. (2009a). Cultural
11678–11683. neuroscience of consciousness: From visual per-
Brody, N. (1992). Intelligence (2nd ed.). New York: ception to self-awareness. Journal of Conscious-
Academic. ness Studies, 15(10–11), 58–69.
Brunoni, A. R., Ekhtiari, H., Antal, A., Auvichayapat, Chiao, J. Y., Harada, T., Komeda, H., Li, Z., Mano, Y.,
P., Baeken, C., Benseñor, I. M., … Fregni, F. (2022). Saito, D., … Iidaka, T. (2009b). Neural basis of
Digitalized transcranial electrical stimulation: A individualistic and collectivistic views of self.
consensus statement. Clinical Neurophysiology, Human Brain Mapping, 30(9), 2813–2820.
143, 154–165. Chiao, J. Y., & Blizinsky, K. D. (2016). Cultural neuro-
Buzsáki, G. (2019). The brain from inside out. New science: Bridging cultural and biological sciences.
York: Oxford University Press. In E. Harmon-Jones & M. Inzlicht (Eds.), Social
Buzsáki, G. (2020). The brain-cognitive behavior neuroscience: Biological approaches to social psy-
problem: A retrospective. eNeuro, 7(4). doi: chology (pp. 247–275). New York: Routledge.
10.1523/ENEURO.0069-20.2020 Churchland, P. S., & Sejnowski, T. J. (1988). Perspec-
Caballero-Villarraso, J., Medina, F. J., Escribano, B. tives in cognitive neuroscience. Science, 242,
M., Agüera, E., Santamaría, A., Pascual-Leone A, 741–745.
& Túnez, I. (2022). Mechanisms involved in neuro- Churchland, P. S., & Sejnowski, T. J. (2017). The com-
protective effects of transcranial magnetic stimula- putational brain (25th ed.). MA: MIT Press.
tion. CNS & Neurological Disorders-Drug Targets, Costanzo, E. Y., Villarreal, M., Drucaroff, L. J., Ortiz-
21(7), 557–573. Villafañe, M., Castro, M. N., Goldschmidt, M., …
Cognitive Neuroscience: Basic Principles, Systems, and Methods 9

Guinjoan, S. M. (2015). Hemispheric specialization Fregni, F., Boggio, P. S., Nitsche, M., Bermpohl, F.,
in affective responses, cerebral dominance for Antal, A., Feredoes, E., … Pascual-Leone, A.
language, and handedness lateralization of emo- (2005). Anodal transcranial direct current stimula-
tion, language, and dexterity. Behavioural Brain tion of prefrontal cortex enhances working
Research, 288, 11–19. memory. Experimental Brain Research, 166,
Cuzzolin, F., Morelli, A., Cîrstea, B., & Sahakian, B. J. 23–30.
(2020). Knowing me, knowing you: Theory of Fröhlich, F. (2016). Noninvasive brain stimulation. In
mind in AI. Psychological Medicine, 50(7), F. Fröhlich (Ed.), Network neuroscience
1057–1061. (pp. 197–210). San Diego: Elsevier/Academic.
Damasio, A. R. (2000). A second chance for emo- Gage, N., & Baars, B. (2018). Fundamentals of cogni-
tions. In R. Lane & L. Nadel (Eds.), Cognitive neu- tive neuroscience: A beginner’s guide (2nd ed.).
roscience for emotion (pp. 12–23). New York: London: Elsevier/Academic.
Oxford University Press. Gazzaniga, M. S. (2018). The consciousness instinct:
Dam, V. H., Stenbæk, D. S., Köhler-Forsberg, K., Ip, C., Unraveling the mystery of how the brain makes
Ozenne, B., Sahakian, B. J., … Frokjaer, V. G. the mind. New York: Farrar, Straus and Giroux.
(2021). Hot and cold cognitive disturbances in Gazzaniga, M. S., Ivry, R. B., & Mangun, G. R. (2018).
antidepressant-free patients with major depressive Cognitive neuroscience: The biology of the mind
disorder: A NeuroPharm study. Psychological (5th ed.). New York: Norton.
Medicine, 51(14), 2347–2356. Glasser, M. F., Sotiropoulos, S. N., Wilson, J. A., Coal-
Dayan, P., & Abbott, L. F. (2001). Theoretical neuro- son, T. S., Fischl, B., Andersson, J. L., … Jenkinson,
science: Computational and mathematical mode- M. (2013). The minimal preprocessing pipelines
ling of neural systems. Cambridge, MA: MIT Press. for the Human Connectome Project. NeuroImage,
de Rover, M., Pironti, V. A., McCabe, J. A., Acosta- 80, 105–124.
Cabronero, J., Arana, F. S., Morein-Zamir, S., … Gomes-Osman, J., Solana-Sánchéz, J., Rogers, E.,
Sahakian, B. J. (2011). Hippocampal dysfunction in Cattaneo, G., Souillard-Mandar, W., Bates, D., …
patients with mild cognitive impairment: A func- Pascual-Leone A. (2021). Aging in the digital age:
tional neuroimaging study of a visuospatial paired Using technology to increase the reach of the clini-
associates learning task. Neuropsychologia, 49(7), cian expert and close the gap between health
2060–2070. span and life span. Frontiers in Digital Health, 3.
DeWitt, I., & Rauschecker, J. P. (2013). Wernicke’s doi: 10.3389/fdgth.2021.755008
area revisited: Parallel streams and word process- Gotts, S. J., Jo, H. J., Wallace, G. L., Saad, Z. S., Cox,
ing. Brain and Language, 127(2), 181–191. R. W., & Martin, A. (2013). Two distinct forms of
Donoso, M., Collins, A. G. E., & Koechlin, E. (2014). functional lateralization in the human brain. Pro-
Human cognition: Foundations of human reason- ceedings of the National Academy of Sciences of the
ing in the prefrontal cortex. Science, 344. doi: United States of America, 110(36), E3435–E3444.
10.1126/science.1252254 Gross, C. G. (1995). Aristotle on the brain.
Douw, L., Wakeman, D. G., Tanaka, N., Liu, H., & Neuroscientist, 1(4), 245–250.
Stufflebeam, S. M. (2016). State-dependent vari- Grossman, N., Bono, D., Dedic, N., Kodandaramaiah,
ability of dynamic functional connectivity between S. B., Rudenko, A., Suk, H. J., … Pascual-Leone,
frontoparietal and default networks relates to A., & Boyden, E. S. (2017). Noninvasive deep brain
cognitive flexibility. Neuroscience, 339, 12–21. stimulation via temporally interfering electric
Fancher, R. E., & Rutherford, A. (2012). Pioneers of fields. Cell, 169(6), 1029–1041.
psychology. New York: Norton. Hagoort, P. (2014). Nodes and networks in the neural
Farzan, F., Vernet, M., Shafi, M. M., Rotenberg, A., architecture for language: Broca’s region and
Daskalakis, Z. J., & Pascual-Leone, A. (2016). Char- beyond. Current Opinion in Neurobiology, 28,
acterizing and modulating brain circuitry through 136–141.
transcranial magnetic stimulation combined with Halstead, W. C. (1947). Brain and intelligence.
electroencephalography. Frontiers in Neural Cir- Chicago, IL: University of Chicago Press.
cuits, 10. doi: 10.3389/fncir.2016.00073 Horn, J. L. (1988). Thinking about human abilities. In
Fischer, D. B., Boes, A. D., Demertzi, A., Evrard, H. C., J. R. Nesselroade & R. B. Cattell (Eds.), Handbook
Laureys, S., Edlow, B. L., Liu, H., Saper, C. B., Pas- of multivariate experimental psychology
cual-Leone, A., Fox, M. D., & Geerling, J. C. (pp. 645–685). New York: Academic.
(2016). A human brain network derived from Hsu, C. L., Manor, B., Travison, T., Pascual-Leone, A.,
coma-causing brainstem lesions. Neurology, & Lipsitz, L. A. (2022). Sensorimotor and frontopa-
87(23), 2427–2434. rietal network connectivity are associated with
Fox, P. T., & Lancaster, J. L. (2002). Mapping context subsequent maintenance of gait speed and epi-
and content: The BrainMap model. Nature sodic memory in older adults. Journals of Geron-
Reviews, Neuroscience, 3(4), 319–321. tology: Series A. doi: 10.1093/gerona/glac193
10 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Hugdahl, K. (2000). Lateralization of cognitive pro- differences in personality. Translational Psychiatry,


cesses in the brain. Acta Psychologica, 105(2–3), 11(1). doi: 10.1038/s41398-020-00880-9
211–235. Kastner, S., & Ungerleider, L. G. (2000). Mechanisms
Jahanshahi, M. (2003). Reaction time as an index of of visual attention in the human cortex. Annual
motor preparation/programming and speed of Review of Neuroscience. 23, 315–341.
response initiation. In M. Hallett (Ed.), Handbook Kim, N. Y., Hsu, J., Talmasov, D., Joutsa, J., Soussand,
of clinical neurophysiology: Movement disorders L., Wu, O., … Fox, M. D. (2021). Lesions causing
(Vol. 1, pp. 203–229). Amsterdam: Elsevier, hallucinations localize to one common brain net-
Jahanshahi, M. (2005). Other cognitive functions. In work. Molecular Psychiatry, 26(4), 1299–1309.
M. Hallett & S. Chokroverty (Eds.), Magnetic Kong, X.-Z., Mathias, S. R., Guadalupe, T., Abé, C.,
stimulation in clinical neurophysiology (2nd ed., Agartz, I., Akudjedu, T. N., … Orhan, F. (2018).
pp. 281–302). Woburn, MA: Butterworth- Mapping cortical brain asymmetry in 17,141
Heinemann. healthy individuals worldwide via the ENIGMA
Jahanshahi, M., Jenkins, H. I., Brown, R. G., Marsden, Consortium. Proceedings of the National Academy
C. D., Passingham, R. E., & Brooks, D. J. (1995). of Sciences of the United States of America,
Self-initiated versus externally-triggered move- 115(22), E5154–E5163.
ments: I. An investigation using regional cerebral Kosslyn, S. M., & Andersen, R. A. (1995). (Eds.), Fron-
blood flow and movement-related potentials in tiers in cognitive neuroscience. Cambridge, MA:
normals and in patients with Parkinson’s disease. MIT Press.
Brain, 118, 913–933 Kosslyn, S. M., Pascual-Leone, A, Felician, O., Cam-
Jahanshahi, M., Obeso, I., Rothwell, J. C., Obeso, J. A. posano, S., Keenan, J. P., Thompson, W. L., Ganis,
(2015). A fronto-striato-subthalamic-pallidal net- G., Sukel, K. E., & Alpert, N. M. (1999). The role of
work for goal-directed and habitual inhibition. area 17 in visual imagery: Convergent evidence
Nature Reviews Neuroscience. doi: 10.1038/ from PET and rTMS. Science, 284,167–170.
nrn4038. Kriegeskorte, N., & Douglas, P. K. (2018). Cognitive
Jahanshahi, M., & Rothwell, J. C. (2000). Transcranial computational neuroscience. Nature Neurosci-
magnetic stimulation studies of cognition: An ence, 21, 1148–1160.
emerging field. Experimental Brain Research, Laird, J. E., LeBiere, C., & Rosenbloom, P. S. (2017). A
131(1), 1–9. standard model of the mind: Toward a common
Jannati, A., Oberman, L. M., Rotenberg, A., & computational framework across artificial intelli-
Pascual-Leone, A. (2023). Assessing the mecha- gence, cognitive science, neuroscience, and robot-
nisms of brain plasticity by transcranial magnetic ics. AI Magazine, 38(4), 13–26.
stimulation. Neuropsychopharmacology. doi: Langley, C., Cîrstea, B.-I., Cuzzolin, F., & Sahakian, B.
10.1038/s41386-022-01453-8 J. (2022). Theory of mind in humans and in
Jenkins, H. I., Jahanshahi, M., Jueptner, M., Passing- machines. Frontiers in Artificial Intelligence, 5. doi:
ham, R. E., Brooks, D. J. (2000). Self-initiated 10.3389/frai.2022.917565
versus externally-triggered movements: II. The Langley, C., Gregory, S., Osborne-Crowley, K.,
effects of stimulus predictability studied with PET. O’Callaghan, C., Zeun, P., Lowe, J., … Sahakian, B. J.
Brain, 123, 1216–1228. (2021). Fronto-striatal circuits for cognitive flexibil-
Ji, J. L., Spronk, M., Kulkarni, K., Repovš, G., Antice- ity in far from onset Huntington’s disease: Evi-
vic, A., & Cole, M. W. (2019). Mapping the human dence from the young adult study. Journal of
brain’s cortical-subcortical functional network Neurology, Neurosurgery & Psychiatry, 92(2),
organization. NeuroImage, 185, 35–57. 143–149.
Jirsa, V. K., Proix, T., Perdikis, D., Woodman, M. M., Langley, C., Sahakian, B. J., & Robbins, T. W. (2023).
Wang, H., Gonzalez-Martinez, J., … Bartolomei, F. Cambridge Neuropsychological Test Automated
(2017). The virtual epileptic patient: Individualized Battery (CANTAB). In G. J. Boyle, Y. Stern, D. J.
whole-brain models of epilepsy spread. NeuroIm- Stein, B. J. Sahakian, C. J. Golden, S.-H. A. Chen, &
age, 145, 377–388. T. M.-C. Lee (Eds.), The SAGE handbook of clinical
Jones, E. G., & Mendell, L. M. (1999). Assessing the neuropsychology, Vol. 2: Clinical neuropsychologi-
decade of the brain. Science, 284, 739–739. cal assessment and diagnosis. Los Angeles: Sage.
Joutsa, J., Corp, D. T., & Fox, M. D. (2022). Lesion LeDoux, J. E. (2000). Emotion circuits in the brain.
network mapping for symptom localization: Annual Review of Neuroscience, 23, 155–184.
Recent developments and future directions. Cur- Li, X., Guo, N., & Li, Q. (2019). Functional neuroim-
rent Opinion in Neurology, 35(4), 453–459. aging in the new era of big data. Genomics, Prot-
Kanen, J. W., Arntz, F. E., Yellowlees, R., Cardinal, R. eomics & Bioinformatics, 17(4), 393–401.
N., Price, A., Christmas, D. M., … Robbins, T. W. Li, Y., Sahakian, B. J., Kang, J., Langley, C., Zhang,
(2021). Serotonin depletion amplifies distinct W., Xie, C., … Feng, J. (2022). The brain structure
human social emotions as a function of individual and genetic mechanisms underlying the nonlinear
Cognitive Neuroscience: Basic Principles, Systems, and Methods 11

association between sleep duration, cognition and approaches. In G. J. Boyle, D. H. Saklofske & G.
mental health. Nature Aging, 2(5), 425–437. Matthews (Eds.), Measures of personality and social
Libon, D. J., Swenson, R., Lamar, M., Price, C. C., psychological constructs (pp. 257–289). San Diego:
Baliga, G., Pascual-Leone, A, … Andersen, S. L. Elsevier/Academic.
(2022). The Boston process approach and digital Neumann, D. L., Chan, R. C. K., Wang, Y., & Boyle,
neuropsychological assessment: Past research and G. J. (2016). Cognitive and affective components
future directions. Journal of Alzheimer’s Disease, of empathy and their relationship with personality
87(4), 1419–1432. dimensions in a Chinese sample. Asian Journal of
Litvina, E., Adams, A., Barth, A., Bruchez, M., Social Psychology, 19, 244–253.
Carson, J., Chung, J. E., … Koroshetz, W. (2019). NMR Progress Report (1984). Review: Federation of
BRAIN Initiative: Cutting-edge tools and resources American Hospitals, 17(3), 36–37, 40–34.
for the community. Journal of Neuroscience, Northoff, G. (2014a) Unlocking the brain. Vol I:
39(42), 8275–8284. Coding. New York: Oxford University Press.
Logan, G. D., & Cowan, W. B. (1984). On the ability Northoff, G. (2014b) Unlocking the brain. Vol II:
to inhibit thought and action: A theory of an act Consciousness. New York: Oxford University Press.
of control. Psychological Review, 91, 295–327. Northoff, G. (2018). The spontaneous brain: From
McAvoy, M., Mitra, A., Coalson, R. S., D’Avossa, G., the mind–body to the world–brain problem.
Keidel, J. L., Petersen, S. E., & Raichle, M. E. Cambridge, MA: MIT Press.
(2016). Unmasking language lateralization in Northoff, G. (2022). From self-continuity to culture
human brain intrinsic activity. Cerebral Cortex, and back: The brain’s scale-free activity and its
26(4), 1733–1746. temporal memory of the world. In D. M. Tucker.,
Malone, J. C. (2014). Did John B. Watson really D. R. Wehrs & S. Nalbantian (Eds.), Cultural
“found” behaviorism? Behavior Analyst, 37(1), memory: From the sciences to the humanities
1–12. (pp. 61–77). Oxford, UK: Routledge.
Mandler, G. (2002). Origins of the cognitive (r)evolu- Northoff, G., Heinzel, A., de Greck, M., Bermpohl, F.,
tion. Journal of the History of the Behavioral Dobrowolny, H., & Panksepp, J. (2006). Self-refer-
Sciences, 38(4), 339–353. ential processing in our brain: A meta-analysis of
Markram, H., Meier, K., Lippert, T., Grillner, S., Frack- imaging studies on the self. NeuroImage, 31(1),
owiak, R., Dehaene, S., … Saria, A. (2011). Intro- 440–457.
ducing the human brain project. Procedia Northoff, G., & Hirjak, D. (2022). Spatiotemporal
Computer Science, 7, 39–42. psychopathology: An integrated brain–mind
Mars, R. B., Neubert, F. X., Verhagen, L., Sallet, J., Miller, approach and catatonia. Schizophrenia Research.
K. L., Dunbar, R. I., & Barton, R. A. (2014). Primate doi: 10.1016/j.schres.2022.10.006
comparative neuroscience using magnetic reso- Northoff, G., & Huang, Z. (2017). How do the brain’s
nance imaging: promises and challenges. Frontiers in time and space mediate consciousness and its dif-
Neuroscience, 8. doi: 10.3389/fnins.2014.00298 ferent dimensions? Temporo-spatial theory of
Mehta, M. A., Owen, A. M., Sahakian, B. J., consciousness (TTC). Neuroscience & Biobehavioral
Mavaddat, N., Pickard, J. D., & Robbins, T. W. Reviews, 80, 630–645.
(2000). Methylphenidate enhances working Northoff, G., Wainio-Theberge, S., & Evers, K.
memory by modulating discrete frontal and pari- (2020a). Is temporo-spatial dynartyhoffmics the
etal lobe regions in the human brain. Journal of “common currency” of brain and mind? In quest
Neuroscience, 20(6), RC65–RC65. of “spatiotemporal neuroscience.” Physics of Life
Menon, V. (2011). Large-scale brain networks and Reviews, 33, 34–54.
psychopathology: A unifying triple network model. Northoff, G. Wainio-Theberge, S., & Evers, K.
Trends in Cognitive Sciences, 15(10), 483–506. (2020b). Spatiotemporal neuroscience: What is it
Michel, C. M., & Pascual-Leone, A. (2020). Predicting and why we need it? Physics of Life Reviews, 33,
antidepressant response by electroencephalogra- 78–87.
phy. Nature Biotechnology, 38(4), 417–419. Ogawa, S., Tank, D. W., Menon, R., Ellermann, J. M.,
Miller, G. A. (2003). The cognitive revolution: A his- Kim, S. G., Merkle, H., & Ugurbil, K. (1992). Intrin-
torical perspective. Trends in Cognitive Sciences, sic signal changes accompanying sensory stimula-
7(3), 141–144. tion: Functional brain mapping with magnetic
Neisser, U., Boodoo, G., Bouchard Jr, T. J., Boykin, A. resonance imaging. Proceedings of the National
W., Brody, N., Ceci, S. J., …Urbina, S. (1996). Academy of Sciences of the United States of
Intelligence: knowns and unknowns. American America, 89(13), 5951–5955.
Psychologist, 51(2), 77–101. Pama, E. A. C., Colzato, L. S., & Hommel, B. (2013).
Neumann, D. L., Chan, R. C. K., Boyle, G. J., Wang, Y., Optogenetics as a neuromodulation tool in cogni-
& Westbury, H. R. (2015). Measures of empathy: tive neuroscience. Frontiers in Psychology, 4. doi:
Self-report, behavioural, and neuroscientific 10.3389/fpsyg.2013.00610
12 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Pascual-Leone, A, Gates, J. R., & Dhuna, A. (1991). related to working memory and reasoning in
Induction of speech arrest and counting errors middle-aged adults. Frontiers in Psychology, 13.
with rapid-rate transcranial magnetic stimulation. doi: 10.3389/fpsyg.2022.813444
Neurology, 41(5), 697–702. Rilling, M. (2000). How the challenge of explaining
Pascual-Leone, A., Bartres-Faz, D., & Keenan, J. P. learning influenced the origins and development
(1999). Transcranial magnetic stimulation: Study- of John B. Watson’s behaviorism. American Jour-
ing the brain–behaviour relationship by induction nal of Psychology, 113(2), 275–301.
of “virtual lesions.” Philosophical Transactions of Robbins, T. W., James, M., Owen, A. M., Sahakian, B. J.,
the Royal Society of London. Series B: Biological McInnes, L., & Rabbitt, P. (1994). Cambridge
Sciences, 354, 1229–1238. Neuropsychological Test Automated Battery
Pascual-Leone, A., Walsh, V., & Rothwell, J. (2000). (CANTAB): A factor analytic study of a large
Transcranial magnetic stimulation in cognitive neu- sample of normal elderly volunteers. Dementia,
roscience: Virtual lesion, chronometry, and func- 5(5):266–281.
tional connectivity. Current Opinion in Robbins, T. W., James, M., Owen, A. M., Sahakian,
Neurobiology, 10(2), 232–237. B. J., McInnes, L., & Rabbitt, P. (1997). A neural
Pascual-Leone, A., & Walsh, V. (2001). Fast backpro- systems approach to the cognitive psychology of
jections from the motion to the primary visual area ageing using the CANTAB battery. In P. Rabbitt
necessary for visual awareness. Science, 292, (Ed.), Methodology of frontal and executive func-
510–512. tion (pp. 215–235). Hove, UK: Erlbaum.
Pascual-Leone, A., Amedi, A., Fregni, F., & Merabet, Robbins, T. W., James, M., Owen, A. M., Sahakian,
L. B. (2005). The plastic human brain cortex. B. J., Lawrence, A. D., McInnes, L., & Rabbitt, P. M.
Annual Review of Neuroscience, 28, 377–401. A. (2012). A study of performance on tests from
Pascual-Leone, A., Freitas, C., Oberman, L., Horvath, the CANTAB Battery sensitive to frontal lobe dys-
J. C., Halko, M., Eldaief, M., … Rotenberg, A. function in a large sample of normal volunteers:
(2011). Characterizing brain cortical plasticity and Implications for theories of executive functioning
network dynamics across the age-span in health and cognitive aging. In G. J. Boyle, D. H. Saklofske
and disease with TMS-EEG and TMS-fMRI. Brain & G. Matthews (Eds.), SAGE benchmarks in psy-
Topography, 24, 302–315. chology: Psychological assessment, Vol. 3: Clinical
Picchioni, M., Matthiasson, P., Broome, M., Giampi- neuropsychological assessment (pp. 309–341).
etro, V., Brammer, M., Mathes, B., … McGuire, P. London. Sage.
(2007). Medial temporal lobe activity at recogni- Roehri, N., Bréchet, L., Seeber, M., Pascual-Leone, A.,
tion increases with the duration of mnemonic & Michel, C. M. (2022). Phase-amplitude coupling
delay during an object working memory task. and phase synchronization between medial tem-
Human Brain Mapping, 28(11), 1235–1250. poral, frontal and posterior brain regions support
Pinker, S. (2002). The blank slate: The modern denial episodic autobiographical memory recall. Brain
of human nature. New York: Viking. Topography, 35, 191–206.
Poeppel, D., Mangun, G. R., & Gazzaniga, M. S. Sack, A. T., Camprodon, J. A., Pascual-Leone, A., &
(2020). (Eds.), The cognitive neurosciences Goebel, R. (2005). The dynamics of interhemi-
(6th ed.). Cambridge, MA: MIT Press. spheric compensatory processes in mental
Pomerantz, J. R. (2003): Perception: Overview. In L. imagery. Science, 308, 702–704.
Nadel (Ed.), Encyclopedia of cognitive science Sahakian, B. J. (2014). Commentary: What do
(Vol. 3., pp. 527–537). London: Nature experts think we should do to achieve brain
Publishing. health? Neuroscience & Biobehavioral Reviews,
Poo, M.-M., Du, J.-l., Ip, N. Y., Xiong, Z.-Q., Xu, B., & 43, 240–258.
Tan, T. (2016). China Brain Project: Basic neurosci- Sahakian, B. J., & Gottwald, J. (2016). Sex, lies, and
ence, brain diseases, and brain-inspired comput- brain scans: How fMRI reveals what really goes on
ing. Neuron, 92(3), 591–596. in our minds. Oxford, UK: Oxford University Press.
Portnow, L. H., Vaillancourt, D. E., & Okun, M. S. Sahakian, B. J., Langley, C., & Amunts, K. (2021) The
(2013). The history of cerebral PET scanning: From Human Brain Project: Six achievements of Europe’s
physiology to cutting-edge technology. Neurol- largest neuroscience programme. The Conversa-
ogy, 80(10), 952–956. tion (Oct. 11). https://theconversation.com/the-
Raichle, M. E. (2009). A brief history of human brain human-brain-project-six-achievements-of-europ
mapping. Trends in Neurosciences, 32(2), es-largest-neuroscience-programme-169184
118–126. (Retrieved Nov. 17, 2022)
Redondo-Camós, M., Cattaneo, G., Perellón-Alfonso, R., Sahakian, B. J., & Owen, A. M. (1992). Computerized
Alviarez-Schulze, V., Morris, T. P., Solana-Sanchez, assessment in neuropsychiatry using CANTAB:
J., … Pascual-Leone, A., & Bartres-Faz, D. (2022). Discussion paper. Journal of the Royal Society of
Local prefrontal cortex TMS-induced reactivity is Medicine, 85, 399–402.
Cognitive Neuroscience: Basic Principles, Systems, and Methods 13

Scahill, R. I., Zeun, P., Osborne-Crowley, K., Johnson, Tandon, P. N. (2000). The decade of the brain: A brief
E. B., Gregory, S., Parker, C., … Tabrizi, S. J. (2020). review. Neurology India, 48(3), 199–207.
Biological and clinical characteristics of gene carri- Terry, W. S. (2006). Learning and memory: Basic
ers far from predicted onset in the Huntington’s principles, processes, and procedures. Boston,
disease young adult study (HD-YAS): A cross-sec- MA: Pearson.
tional analysis. Lancet Neurology, 19(6), 502–512. Toga, A. W., Clark, K. A., Thompson, P. M., Shattuck,
Schacter, D. L., & Addis, D. R. (2007). The cognitive D. W., & Van Horn, J. D. (2012). Mapping the
neuroscience of constructive memory: Remember- human connectome. Neurosurgery, 71(1). doi:
ing the past and imagining the future. Philosophi- 10.1227/NEU.0b013e318258e9ff
cal Transactions of the Royal Society of London. Tyagi, H., Apergis-Schoute, A. M., Akram, H.,
Series B, Biological Sciences, 362, 773–786. Foltynie, T., Limousin, P., Drummond, L. M., …
Schaefer, M., & Northoff, G. (2017). Who am I? The Joyce, E. M. (2019). A randomized trial directly
conscious and the unconscious self. Frontiers in comparing ventral capsule and anteromedial sub-
Human Neuroscience, 11. doi: 10.3389/fnhum. thalamic nucleus stimulation in obsessive-compul-
2017.00126 sive disorder: Clinical and imaging evidence for
Schneider, W. J., & McGrew, K. S. (2018). The Cat- dissociable effects. Biological Psychiatry, 85(9),
tell–Horn–Carroll theory of cognitive abilities. In D. 726–734.
P. Flanagan & E. M. McDonough (Eds.), Contem- Uttal, W. R. (2011). Mind and brain: A critical
porary intellectual assessment: Theories, tests, and appraisal of cognitive neuroscience. Cambridge,
issues (pp. 73–163). New York: Guilford. MA: MIT Press.
Shafi, M. M., Westover, M. B., Fox, M. D., & Pascual- Vallar, G., & Bolognini, N. (2011). Behavioural facilita-
Leone, A. (2012). Exploration and modulation of tion following brain stimulation: Implications for
brain network interactions with noninvasive brain neurorehabilitation. Neuropsychological Rehabili-
stimulation in combination with neuroimaging. tation, 21(5), 618–649.
European Journal of Neuroscience, 35(6), 805–825. Van Essen, D. C., Smith, S. M., Barch, D. M., Behrens,
Shen, C., Luo, Q., Chamberlain, S. R., Morgan, S., T. E. J., Yacoub, E., & Ugurbil, K. (2013). The WU–
Romero-Garcia, R., Du, J., … Sahakian, B. J. Minn Human Connectome Project: An overview.
(2020a). What is the link between attention- NeuroImage, 80, 62–79.
|deficit/hyperactivity disorder and sleep distur- Varshney, L. R., & Barbey, A. K. (2021). Beyond IQ:
bance? A multimodal examination of longitudinal The importance of metacognition for the promo-
relationships and brain structure using large-scale tion of global wellbeing. Journal of Intelligence,
population-based cohorts. Biological Psychiatry, 9(4). doi: 10.3390/jintelligence9040054
88(6), 459–469. von Monakow, C., & Harris, G. (1969). Die lokalisa-
Shen, C., Luo, Q., Jia, T., Zhao, Q., Desrivières, S., tion im grosshirn: Und der abbau der funktion
Quinlan, E. B., … IMAGEN consortium. (2020b). durch kortikale herde. In K. H. Pribam (Ed.). Brain
Neural correlates of the dual-pathway model for and behavior I: Mood states and mind
ADHD in adolescents. American Journal of (pp. 27–36). Baltimore, MD: Penguin.
Psychiatry, 177(9), 844–854. Wagner, T., Valero-Cabre, A., & Pascual-Leone, A.
Shen, C., Rolls, E. T., Cheng, W., Kang, J., Dong, G., (2007). Noninvasive human brain stimulation.
Xie, C., … Feng, J. (2022). Associations of social Annual Review of Biomedical Engineering, 9,
isolation and loneliness with later dementia. 527–565.
Neurology, 99(2), E164–E175. Walsh, V., & Pascual-Leone, A. (2003). Transcranial
Shine, J. M., Bissett, P. G., Bell, P. T., Koyejo, O., magnetic stimulation: A neurochronometrics of
Balsters, J. H., Gorgolewski, K. J., … Poldrack, mind. Cambridge, MA: MIT Press.
R. A. (2016). The dynamics of functional brain Walsh, V., Desmond, J. E., & Pascual-Leone, A.
networks: Integrated network states during cogni- (2006). Manipulating brains. Behavioural Neurol-
tive task performance. Neuron, 92(2), 544–554. ogy, 17(3–4), 131–134.
Skinner, B. F. (1976). About behaviorism. New York: Wang, D., Buckner, R. L., & Liu, H. (2014). Functional
Random House. specialization in the human brain estimated by
Souillard-Mandar, W., Penney, D., Schaible, B., intrinsic hemispheric interaction. Journal of Neuro-
Pascual-Leone, A., Au, R., & Davis, R. (2021). DCT- science, 34(37), 12341–12352.
clock: Clinically-interpretable and automated arti- Watson, J. B. (2017). Behaviorism (Reprint of 1925
ficial intelligence analysis of drawing behavior for book – introduction by G. A. Kimble). Oxford, UK:
capturing cognition. Frontiers in Digital Health, 3. Routledge.
doi: 10.3389/fdgth.2021.750661 Winters, B. D., Forwood, S. E., Cowell, R. A., Saksida,
Sternberg, R. J., & Kaufman, J. C. (1998). Human L. M., & Bussey, T. J. (2004). Double dissociation
abilities. Annual Review of Psychology, 49(1), between the effects of peri-postrhinal cortex and
479–502. hippocampal lesions on tests of object recognition
14 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

and spatial memory: Heterogeneity of function large preteenage population identifies distinct
within the temporal lobe. Journal of Neuroscience, obsessive-compulsive disorder: Like subtypes with
24, 5901–5908. neurocognitive differences. Biological Psychiatry:
Woods, A. J., Antal, A., Bikson, M., Boggio, P. S., Cognitive Neuroscience and Neuroimaging, 7(11),
Brunoni, A. R., Celnik, P., … Nitsche, M. A. (2016). 1078–1089.
A technical guide to tDCS, and related non-inva- Yarkoni, T., Poldrack, R. A., Nichols, T. E., Van Essen,
sive brain stimulation tools. Clinical Neurophysiol- D. C., & Wager, T. D. (2011). Large-scale auto-
ogy, 127(2), 1031-1048. mated synthesis of human functional neuroimag-
Wu, X., Kong, X., Vatansever, D., Liu, Z., Zhang, K., ing data. Nature Methods, 8(8), 665–670.
Sahakian, B. J., Robbins, T. W., … Zhang, J. (2021). Zhang, Y., Luo, Q., Huang, C. C., Lo, C. Y. Z., Lang-
Brain laterality dynamics support human cogni- ley, C., Desrivières, S., … IMAGEN consortium.
tion. bioRxiv. doi: 10.1101/2021.08.05.455350 (2021). The human brain is best described as
Wu, X., Kong, X., Vatansever, D., Liu, Z., Zhang, K., being on a female/male continuum: Evidence from
Sahakian, B. J., Robbins, T. W., … Zhang, J. a neuroimaging connectivity study. Cerebral
(2022a). Dynamic changes in brain lateralization Cortex, 31(6), 3021–3033.
correlate with human cognitive performance. Zhang, H., Yang, S. Y., Qiao, Y., Ge, Q., Tang, Y. Y.,
PLoS Biology, 20(3). doi: 10.1371/journal. Northoff, G., & Zang, Y. F. (2022). Default mode
pbio.3001560 network mediates low-frequency fluctuations in
Wu, X., Yu, G., Zhang, K., Feng, J., Zhang, J., Saha- brain activity and behavior during sustained atten-
kian, B. J., & Robbins, T. W. (2022b). Symptom- tion. Human Brain Mapping. doi: 10.1002/
based profiling and multimodal neuroimaging of a hbm.26024
PART I
Background Considerations
This page intentionally left blank
2
Historical, Empirical, and
Philosophical Perspectives on
Cognitive Neuroscience
Georg Northoff and Philipp Klar

INTRODUCTION brain’s neuronal activity is conceived to be a pas-


sive response to exteroceptive input provided by
The development of neuroimaging in the last the environment, e.g., to visual or auditory stimuli
three decades, especially since the dawn of func- by the respective task. The brain is consequently
tional magnetic resonance imaging (fMRI) from treated as a passive or reflexive organ in relation to
1990 to 1992 (Bandettini et al., 1992; Kwong the environment (Northoff, 2018a; Raichle, 2015).
et al., 1992; Ogawa et al., 1992), led to an emerg- Notably, the discovery of the default-mode-
ing novel domain in brain research. This novel network (DMN) of the brain’s intrinsic sponta-
domain allows merging cognitive psychology neous activity in 2001 (Raichle et al., 2001) led
with neuroscience, namely, cognitive neurosci- to additional and intensive resting-state research
ence (Gazzaniga et al., 2018; Passingham, 2016). designs. Contrary to task-based research designs,
The convergence between both scientific disci- where task-evoked or stimulus-induced activity of
plines allowed to introduce cognitive concepts the brain in response to extrinsic stimuli is inves-
from psychology into neuroscience. The opera- tigated, resting-state designs (Northoff, 2014a;
tionalization of cognitive concepts enabled their Raichle, 2015) aim to understand the brain’s con-
utilization for neuroimaging. Cognitive neurosci- stantly ongoing intrinsic spontaneous activity, that
ence was consequently able to map cognitive is, spontaneous activity (BOLD fluctuations) that
functions onto both brain areas and networks, correlate across brain areas independent of a task.
reflecting localizationism. Here, in addition to the passive-reflexive side of
To link the mind with the brain, traditionally, the brain, an additional active side of the brain’s
task-based research designs are utilized in fMRI. intrinsic functional architecture and organization
While lying in the scanner, the subject has to per- comes into play (Northoff, 2018a). What is ulti-
form a task during which the brain’s higher blood- mately measured even in task-based designs, pre-
oxygenation-level-dependent contrast (BOLD) cisely stimulus-induced/task-evoked activity, is
relative to another task and/or baseline level is the result and hybrid mixture of a dynamic inter-
measured in distinct areas of the brain. Such action between the intrinsic spontaneous activity
research paradigm is a partial reminiscence of and extrinsic stimuli (Boly et al., 2007; Braun
behaviorism in a more cognitive way: the measured et al., 2021; Deco et al., 2013; Fox et al., 2007;
18 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

He, 2013; Huang et al., 2017; Raichle, 2015; (Menon, 2011) changes the conceptual and empir-
Wainio-Thberge et al., 2021; Wolff et al., 2021). ical approach.
Subjects are usually instructed to lie in the scanner The shift from areas to networks is rather quan-
without thinking of anything in particular in one of titative than a qualitative change (i.e., a change in
three possible conditions: (1) eyes open, (2), eyes degrees). One can thus speak of a localization-
closed, or (3) eyes open focusing a fixation cross. based approach to the brain and its constitutive
The brief introduction of resting-state and role for experience and behavior. The localization-
task-based paradigms in fMRI research reflects based approach is defined in two principal ways.
a central theoretical aspect in cognitive neurosci- First, it implies the neuropsychological assump-
ence: a fluctuation between the inside of the brain tion that a particular cognitive function identifies
(i.e., the resting-state activity), and the outside with neural activity in a specific brain region or
(i.e., cognition and behavior). This inside-outside network. The methodological localization by
dichotomy has recently been thematized in cog- neuropsychology and cognitive neuroscience in
nitive neuroscience by various authors includ- regions and networks has its roots in the meta-
ing Buszaki (2019, 2020), Northoff (2012, 2014, physical domain. The philosophical assumption
2018b), Poeppel and Adolfi (2020), and Raichle is that of identity or emergence between human
(2010). We introduce the inside-outside trajecto- experience and the brain’s neuronal activity in
ries over the course of the chapter stemming from specific regions and networks. Today, localiza-
the historical origins of cognitive neuroscience. tionism is one of the guiding theoretical forces of
functional magnetic resonance imaging (fMRI).
For this reason, we will elaborate more on locali-
zationism in the following.
FROM LOCALIZATIONISM TO COGNITIVE
NEUROSCIENCE

From regions to modules


Brain function is related to specific
regions Localizationism of cognitive functions via lesion
studies and neuroimaging stems from the theory
One of the main methodological approaches in of modules by cognitive psychology. Cognitive
neurology at the beginning of the twentieth cen- psychology proposed specific functional modules
tury, when neuroscience as a distinct scientific in charge of processing sensory input and that
discipline did not exist, was the investigation of modular-based computational processes would
patients with brain lesions. These patients revealed subsequently lead to phenomenal representations
how cognitive functions, paradigmatically aware- and their associated behavior.
ness, memory, attention, or learning, were affected Historically, the idea of the mind’s rational or
by lesions in particular regions. The investigation modernly speaking cognitive modules traces back
of brain lesions led the French neurologist Paul to the roots of psychology in philosophy. The
Broca (1824–1880) to a famous discovery of a German philosopher Immanuel Kant (1724–1804)
distinct region in the left frontal cortex that cor- developed the idea of a priori categories in one of
responds to speech production, today called his main books, namely the Critique of Pure Reason
Broca’s area. Broca observed that patients with a (Kant, 1781/1996). A priori categories constitute
lesion in the left lateral frontal cortex showed even the most basic aspects of human experience,
major deficits in producing words and language, such as space, time, and causality. Following
hence presenting expressive aphasia. These clini- Kant’s transcendental philosophy, the experience
cal observations led Broca to infer that the regions of space and time derives from the human mind’s a
BA44 and 45 must be in charge of producing priori categories. The latter provide a general point
words, thus localizing speech production in of view that is subsequently applied to the world
Broca’s area. Observation of patients with lesions for the very possibility of experience. In Kant’s
and their corresponding disturbances has since transcendental philosophy, the status of a priori
been a major tool of insight into brain function. categories is not ontological, i.e., based on the
Besides distinct cognitive functions, conscious- world independent of human experience, but epis-
ness and particularly self-consciousness are cur- temological. The mind’s a priori categories offer
rently investigated in neurological patients who no intuitive information about the thing-in-itself
suffer from various localized lesions in the brain or the noumenal world. In Kant’s terminology,
(Feinberg, 2009; Feinberg and Keenan, 2005). It these terms describe the world apart from human
can be argued that the recent shift from single and experience. Instead, the mind and inherent a pri-
clusters of areas towards widespread networks ori categories reflect the insurmountable window
HISTORICAL, EMPIRICAL, AND PHILOSOPHICAL PERSPECTIVES 19

through which we perceive ourselves in relation to However, recent findings in brain imaging put
the world, hence categories are a priori – prior to such modular organization into a wider context
and shaping experience. Consequently, the mind’s of a hierarchical whole-brain organization (i.e.,
a priori categories concern limitations of principal topography). For instance, there is a topographic
human knowledge about the world beyond shap- distinction of core and periphery: core regions
ing immediate and pre-reflective phenomenology. (like default-mode network regions) are strongly
The philosophical-epistemological idea of connected among each other while peripheral
categories of human perception and cognition regions (like sensory regions) are less connected
emerged in the famous psychological school of with each other and the core regions (Golesorkhi
Gestalt Psychology (Koffka, 1935; Lersch, 1956; et al., 2021; Margulies et al., 2016, Smallwood
Wertheimer, 2017). At the beginning of the twenti- et al., 2021). Hence, single modules such as par-
eth century, Gestalt psychology schools appeared ticular sensory systems (e.g., auditory or visual)
in central Europe, mainly from the Austrian and are integrated within a hierarhical-topographic
German universities (in Graz, Leipzig, and Berlin). organization of the whole brain.
The German word “Gestalt” means form or pat-
tern. The Gestalt psychologists proposed gestalt
principles of human perception such as proxim-
ity, similarity, closure, continuity, or common Cognitive ontology
fate (Köhler, 1920, 1935). The Gestalt psycholo-
gists chose this term to refer to their idea that the Localizationism converges with the more recent
perception of the whole is more or different from idea of “cognitive ontology” from cognitive neu-
the sum of its single constituents. Concerning the roscience (Bilder et al., 2009, 2013; Poldrack
formation of human experience, the mind contrib- et al., 2011). Cognitive ontology holds a broad
utes intrinsic ingredients to sensory input from the definition of cognition. In cognitive ontology,
environment. The philosophical reminiscence is cognition refers to all psychological contents and
obvious: the principal idea of Kantian categories processes. While the term “cognition” appears
reappeared in scientific psychology. Kant was harmless, it lacks a profound conceptual defini-
probably among the first philosophers who, in tion. What is missing is a unified theoretical
modern terms, introduced the notion of cogni- framework about cognition that could converge
tive functions as distinct from sensory inputs. The the epistemological dualism between the psychol-
philosophical idea of categories and their trans- ogy of human experience and physiological pro-
lation into psychology as modules is one of the cesses in the nervous system. In cognitive
factors that laid the theoretical background for psychology and neuroscience, the term cognition
the so-called “cognitive revolution” which started is broadly equated with mental, rational, complex
in the second half of the twentieth century. The phenomena, or, broadly speaking, consciousness.
cognitive revolution represents a paradigm shift in Cognitive ontology reaches even beyond brain
psychology from behaviorism to cognitivism and function, as it assumes that not only cognitive
cognitive science. At the same time, the develop- phenomena can be mapped onto specific brain
ment of modern computer systems provided terms regions and networks, but even on genes (Poldrack
such as “modules” and “information processing” et al., 2011). The modular theory of the mind thus
to cognitive psychology. In analogy to computer perfectly matches cognitive ontology. Cognitive
systems, modules represent solid and geneti- neuroscience proposes the disassembly of the
cally determined neuronal substrates underlying mind into various constituents that would allow
human perception and cognition (Fodor, 1983, human brain mapping. Processes of the brain and
1985, 1998). The abstract and formerly genuine its functions are consequently considered primar-
philosophical idea of the mind’s categories found ily in cognitive terms and models.
application on the empirical level of neuropsy- One ultimate goal of cognitive ontology is the
chology. When cognitive psychology fused with development of a “cognitive atlas” in human brain
neuroscience, the concept of psychological mod- mapping (Poldrack et al., 2011). Inferences of the
ules could perfectly converge with the former neu- functional topography in the brain rest on correla-
rological theory of localizationism (van Eijsden tions between cognitive-behavioral tasks and neu-
et al., 2009). Localizationism holds in cognitive ronal activation patterns. Best possibly, specific
neuroscience: “I take the modular organization of task performances supposedly represent isolated
many brain systems as a well-established fact, and cognitive functions. Respective concepts describ-
discuss only how far fMRI can go in revealing the ing cognitive entities or functions are related to
neuronal mechanisms of behavior by mapping dif- each other. For that reason, cognitive ontology
ferent systems modules and their dynamic inter- distinguishes between different kinds of relations
relationships” (Logothetis, 2008). between concepts that stem from cognitivism. For
20 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

instance, concept A is identical with concept B, or consciousness and corresponding physiological


A could be part of B, hence presenting a mereolog- processes of the nervous system (Northoff et al.,
ical relation. B could result from a transformation 2020). How seemingly two epistemological and
of A, and A could cause B (Poldrack et al., 2011). natural levels of reality, namely the philosophi-
These connections are logical-conceptual relation- cal assumption of an inside level (cognition and
ships between different cognitive concepts. It is consciousness) as opposed to another outside level
questionable to what extend logical relationships (biological and physiological processes of the
correspond to neurobiological processes in the brain), can be overcome remains inconceivable.
brain. Logico-conceptual relationships can demar- Note that the notions of inside and outside take
cate the boundary of unambiguous localizationism in a more epistemological than empirical mean-
in cognitive neuroscience between cognition and ing. Empirically, we can observe that the inside
correlated statistical parametric maps in fMRI. is associated with the brain and the outside with
However, several criticisms of the use of neuro- cognition (Buszaki, 2019, 2020). Taken in a more
imaging techniques in the field of cognitive neu- epistemological context, the inside is associated
roscience are evident. Methodological concerns with cognition or consciousness (i.e., from first-
especially about the use of fMRI arise in relation person perspective). Whereas the brain’s neuronal
to the particular statistical and data analytic opera- activity is considered as outside (i.e., from third-
tions carried out (Sucz and Ionnaides, 2017), as person perspective). The conclusion is that major
well as concerns in relation to physiology (e.g., the traces of empirical and epistemological dualism
neuro-vascular BOLD effect; Logothetis, 2008). resurface in cognitive neuroscience. We discuss
In order to reconstruct what happens in the brain this topic below.
based on how blood is distributed it is necessary to
adopt many intermediate operations to move from
the distribution of blood to the distribution of acti-
vations in the various brain areas. These interme- FROM BEHAVIORISM TO COGNITIVE
diate operations are, in most cases, unknown to the NEUROSCIENCE
researcher who relies on the expertise of physicists
and computer scientists for their execution which
almost always involves “packages” of statistical Input–output relation vs. cognitive
operations applied, more or less blindly. These function
operations are prone to errors (on the incorrect use
of statistical inference in the field of neuroscience; How can we investigate human experience and
Button et al., 2013; Eklund et al., 2016; Szucs and behavior? The answer to this question has been
Ioannidis, 2017). oscillating between inside and outside approaches.
In addition, epistemological issues including An early striking example of the outside approach,
the distinction of instruments (e.g., thermometer) from the beginning to the middle of the twentieth
versus apparatus (e.g., fMRI) need to be consid- century, was the US paradigm of behaviorism.
ered when interpreting fMRI data (Metraux and Behaviorism heavily dominated psychology as
Frisch, 2020). The process of measuring brain well as theories about the functionality of the
activation via fMRI differs greatly from measur- nervous system. After WW2, behaviorism spread
ing temperature using quicksilver thermometers, from the United States to Europe and heavily
or from measuring atmospheric pressure through influenced continental psychology. The American
barographs. The difference is not linguistic, but psychologists John B. Watson (1878–1958) and
rather ontological and epistemological in nature. Burrhus F. Skinner (1904–1990) were two of the
Epistemological issues also arise in the case of most prominent behaviorists. Behaviorism stated
cognitive ontology. Since cognitive ontology that the principal aim of psychology was the
defines human experience in sole cognitive terms, investigation of behavior.
both on the empirical level of conceptualiza- According to behaviorism, to achieve the sta-
tion and operationalization, the epistemological tus of a respected science, psychology must focus
gap and the language barrier between cognition solely on the investigation of observable and exper-
and brain is re-confirmed rather than bridged. imentally influenced behaviors from the objective
Cognitive ontology itself seems to acknowledge “third-person” perspective. The aforementioned
implicitly such a gap when speaking about a extreme physicalist stance was paradigmatic for
“latent unobservable construct” (Poldrack et al., the title of the famous book Psychology of the
2011) and the observables of the brain’s physi- Other-One from the year 1922 by behaviorist
ological processes. Missing in the current frame- psychologist Max F. Meyer (1873–1967) (Meyer,
work of cognitive neuroscience and its ontology 1922). Behaviorism did not deny the exist-
is what we label the “common currency” between ence of subjective experience in the first-person
HISTORICAL, EMPIRICAL, AND PHILOSOPHICAL PERSPECTIVES 21

perspective but regarded any conscious or cog- (i.e., the input–output relationship in the nervous
nitive phenomena as superfluous regarding the system). The authors explicitly favored a paradigm
behavior of organisms. More importantly, subjec- shift from external and passive views on the brain
tive experience, i.e., cognition, was defined to be to an inside and active perspective and investiga-
inaccessible for scientific inquiry. From the per- tion of the brain’s ongoing spontaneous activity.
spective of the philosophy of science, behaviorism The paradigm of behaviorism was further put
can thus be defined to take a rather implicit instru- in doubt by both scientists from the west and the
mentalist stance towards experience. Cognition east. Russian successors of the physiologist and
was considered to be merely epiphenomenal. founder of classical conditioning Ivan Pavlov
Behaviorism consequently defined both human (1849–1936), paradigmatically neuropsycholo-
and animal organisms as “black boxes” or “empty- gist Alexander Luria (1902–1977), argued against
organisms” that lack intentionality and are fully behaviorism. From the beginning of the 1960s,
determined by the environment via input–output even American scientists, notably Edward Tolman,
(stimuli–response) associations. These associa- John Garcia, and Martin Seligman established
tions stem entirely from the organism’s interaction significant empirical foundations against the
with the environment and are learned by linking input–output theory for human and animal behav-
specific stimuli with immediate responses via ior. John Garcia (1917–2012) provided evidence
classical and operant conditioning. In reference to against radical behaviorism in his taste aversion
these ideas, B. F. Skinner labeled one of his main experiment. The “Garcia effect” was named after
works, Beyond Freedom and Dignity (Skinner, Garcia and Koelling. They experimentally showed
1971), whereby the notion of negated free will that specific biological predispositions can
(Skinner, 1971) was based on the behaviorist idea increase the likelihood of associative connections
that behavior is determined solely by external between certain reactions to a priori presented
environmental stimuli. Previously, Skinner had stimuli. Furthermore, and just as strikingly against
speculated on the creation of an optimal society, the perspective of behaviorism, some stimulus-
arising through use of societally induced operant response (S–R) connections could simply not
conditioning (Skinner, 1948/2005). be established in certain organisms such as rats
Early empirical findings that revealed evidence (Garcia et al., 1955; Garcia and Koelling, 1966).
against the behaviorist paradigm emerged in the This finding later was summarized under the term
first half of the twentieth century. One historic “preparedness” (Seligman, 2016; Seligman et al.,
critique by German physiologist Erich von Holst 1970). Certain responses, due to protective biolog-
(1908–1962) was entitled Vom Wesen der Ordnung ical predispositions rooted in phylogenesis, allow
im Zentralnervensystem [On the essence of order no conditioning to occur.
in the central nervous system] (von Holst, 1937). As the cognitive revolution progressed over sev-
This critique challenged the behaviorist notion of eral decades, the powerful behavioristic paradigm
locomotion, the idea that the motor output of an gradually faded. Cognitivism represents the main-
organism is solely determined by a sequence of stream today, hence the term cognitive science.
interconnected stimulus–response (S–R) reflexes. Cognitivism reintroduced the inner perspective of
These muscle contractions are in turn themselves the organism into psychology and ultimately has
initially caused by environmental stimuli. Von largely replaced behaviorism. While cognitivism
Holst severed the corresponding nerve connec- overcame the one-sided outside view of the behav-
tions of an earthworm to show that the motor out- ior of organisms, it reverted into a latent form of
put remained at a constant up to increased speed. behaviorism and Cartesianism. Cognitivism now
The conclusion was that the nervous system repeats the same mistake of behaviorism, albeit
exhibits intrinsic spontaneous activity that does with inverted signs. Instead of completely over-
not require peripheral input from environmen- coming the stimulus-response (S–R) paradigm
tal stimuli; hence, the nervous system can also of behaviorism, cognitivism introduced a cogni-
be characterized by endogenous activity against tive information processing layer in between the
the radical paradigm of a simplified input–output immediate S–R association. Behaviorist and car-
determination of organisms by the environment. A tesian roots in cognitivism become visible when
later and even more famous article was named Das cognitive scientists talk about “sensory inputs,”
Reafferenzprinzip: Wechselwirkungen zwischen which are “processed” and subsequently used for
Zentralnervensystem und Peripherie [The reaffer- the construction of “cognitive representations” of
ence principle: Interactions between the central the environment within the organism. The results
nervous system and the periphery] (von Holst and of internal processing mechanisms in the organ-
Mittelstaedt, 1950). Erich von Holst and his student ism then affect the final behavioral output.
Mittelstaedt argued that physiology previously only Eventually, this paradigm remains in nothing
asked about the afferent and efferent relationship but the S-O-R loop (stimulus–organism–response).
22 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

First, the difference with behaviorism is that the Historically, German psychiatrist and devel-
suggested mediating processes (O) are described oper of electroencephalography (EEG) Hans
in a cognitive language. Second, cognitive rep- Berger (1873–1941) early on observed spontane-
resentations reintroduce a cartesian notion that ous activity in the brain that remained independ-
human experience is principally different from ent of external tasks or stimuli (Berger, 1929). The
the rest of the body and the world. The cartesian investigation into the brain’s spontaneous activ-
Res Cogitans now appears covered as an example ity was further suggested by Bishop (1933) and
of “cognitive representation,” although localized has recently gained more traction in neuroscience
within in the brain (Bennet and Hacker, 2003; with the observation of spontaneous oscillations
Fuchs, 2018). Even though today’s cognitive (Buszaki, 2006; Llinas, 1988; Yuste et al., 2005),
neuroscience rejects ontological cartesian dual- spontaneous coherence or connectivity between
ism, it remains in the inside (cognition/mind/ different regions’ neuronal activities (Biswal et al.,
consciousness)-outside (the brain, body, and the 1995; Greicius et al., 2003), and the default-mode
world) distinction where Descartes’ mind resur- network (DMN) (Greicius et al., 2015; Raichle
faces labeled “cognitive representations” being an et al., 2001; Reiss and Menon, 2003). Historical
epistemological dualism between the former and and modern observations point out the central role
a so-called “external world.” In post-behaviorist of the brain’s spontaneous activity (Huang et al.,
psychology, the term “cognition” was used as a 2015; Northoff, 2014a.b; Northoff et al., 2010,
stopgap able to fill wide-ranging gaps left behind 2012a,b). Research on the brain’s spontaneous
by the anti-mental stance of behaviorism against activity has shifted theories and models about the
all experiential intentional terms. In the cogni- brain. Instead of considering the brain as extrinsi-
tivist paradigm, the term cognition is nowadays cally driven device, spontaneous activity suggests
simply equated to the development of meaning what Raichle described as an “intrinsic model of
(i.e., the semantic content of information). The brain” (Raichle, 2009, 2010). This is reminiscent
result is an imprecise concept to the extent that of a Kantian model of the mind that, applied to the
even unconscious processes can be cognitive since brain, suggests the brain’s spontaneous activity to
neuronal processes that do not correspond with structure and organize its stimulus-induced activ-
conscious experience can carry semantic content. ity of sensory and cognitive functions (Fazelpour
Behaviorism was a reaction against introspect and Thompson, 2015; Northoff, 2012a,b, 2014a,b).
psychology and the assumption of an unobserv- Traditional models of the brain were largely neu-
able level of the “inside” mind. While cognitive rosensory and neurocognitive in that they focus
psychology reintroduced the idea of cognition, on the brain’s sensory and cognitive functions
computational processes, and the mind, it never as mostly mediated by stimulus-induced activity
seriously considered human phenomenology for (Churchland, 2012; Northoff, 2016a, 2018).
scientific inquiries. The observation of the spontaneous activity
may put the brain’s neurosensory and neurocog-
nitive function into a larger empirical context.
The kind of brain model required remains unclear
Spontaneous activity vs. task-related (Klein, 2012; Northoff, 2012, 2018). Concerning
activity brain function, stimulus-induced activity reflects
the traditional external task-driven view on the
How can we characterize the brain by itself inde- brain, while the brain’s intrinsic spontaneous
pendent of cognition? One empirical characteriza- activity may account for an inside view. Hence,
tion of the brain is allowed by what is immediately what the inside in context of function and behav-
observable in neuroscience, namely the brain’s ior (i.e., cognitive function as distinguished from
neuronal activity. The brain’s activity allows two- input–output relations), is the outside in context
fold division: first, the brain exhibits intrinsic of brain activity (i.e., task-evoked activity) The
spontaneous activity, methodologically called brain’s spontaneous activity itself provides the
resting-state or baseline. Second, extrinsic intero- fundamental layer.
ceptive and exteroceptive stimuli, respectively
originating from the body and the environment
(Northoff, 2014a; Raichle, 2015). Cognitive neu-
roscience initially focused heavily on the brain’s Relationship of spontaneous and
extrinsic stimulus-induced activity (task-evoked task-related activity
activity). The central role of the brain’s intrinsic
ongoing activity (i.e., its spontaneous activity, has How are intrinsic and extrinsic brain activity
only recently been considered by the mainstream related? Traditionally, spontaneous and stimulus-
via the utilization of resting-state paradigms). induced activity were assumed to operate
HISTORICAL, EMPIRICAL, AND PHILOSOPHICAL PERSPECTIVES 23

in parallel without interaction. The impact of “Inside–out” vs. “outside–in”


external stimuli or tasks was supposed to be pro- approach
cessed separately from pre-stimulus spontaneous
activity in a purely additive fashion (Arieli et al., The question of the relationship between the
1996; Fox et al., 2006). The additive model is put brain’s neuronal activity and consciousness, para-
in doubt by recent studies that provide evidence of digmatically conceptualized by the philosophical
how spontaneous activity levels modulate the mind–body problem of Western philosophy, has
degree to which external stimuli can elicit task- fascinated philosophers over centuries. Roughly
related activity changes (Braun et al., 2021; He, 20 years ago, the topic of consciousness also
2013; Huang et al., 2017; Northoff et al., 2010; entered neuroscience where, for instance, Buszaki
Wainio-Theberge et al., 2021; Wolff et al., 2021). (2019, 2020) (abbreviated as GB in the following)
Recent studies suggest non-additive or non-linear and Poeppel and Adolfi (2020) (abbreviated as
rather than additive and linear interaction between DP/FA in the following) offer contrasting views
spontaneous and task-related activity. Non- how to research brain and mind. GB opts for the
additive interactions are not only important in primacy of the brain’s inside over cognition and
neuronal terms but influence cognitive function. behavior at its outside – this is reflected in his shift
Both fMRI and EEG studies show that pre-stimu- from outside–in to an inside–out approach
lus resting-state activity levels strongly impact the (Buszaki, 2019, 2020). In contrast, DP/FA suggest
contents of consciousness (Hesselmann et al., that we need to consider both the brain’s inside
2008; Sadaghiani et al., 2010). Based on various and its outside namely behavior/cognition to
findings on consciousness, Northoff and Lamme understand the brain–mind relationship. Put in a
(2020) recently suggested a nested model of spon- nutshell, one emphasizes the priority of the brain’s
taneous and task-related activity. Spontaneous inside over the behavior/cognition at its outside
activity shapes pre-stimulus activity, which in turn (GB) while the other suggest parallelism of both
nests and contains early task-related activity inside and outside (DP/FA). Who is right and
changes. Task-induced neuronal states finally nest which approach shall we follow? Are they mutu-
and contain late task-related activity. ally exclusive? We propose that they are not mutu-
What can dynamic non-additive interactions ally exclusive but can be reconciled on a deeper
between spontaneous and stimulus-induced activ- more fundamental level of the brain’s activity.
ity imply, especially concerning inside and outside Cognitivism and cognitive neuroscience take a
views on brain activity? Non-additive rest–task cognitive view on the brain (Poeppel et al., 2020)
interaction provides a model where the brain’s which also provides the background assumption
ongoing activity strongly shapes task-related for the commentary by DP/FA. Cognitive function
activity. Unlike in the additive model, neuronal is taken as starting point and independent variable
inside and outside are no longer operating in par- to search for its neuronal correlates whose meas-
allel but are dynamic changes of the same process. ures serve as a dependent variable – this is what
The brain’s activity is a hybrid mixture of both GB terms “outside–in approach” (Buszaki, 2019,
intrinsic and extrinsic causes. Wolff et al. (2021) 2020). The cognitive model is contrasted with
showed that early stimulus-induced activity what can be described as “neuronal model” of the
(0–300ms) is strongly impacted by pre-stimulus brain: here the brain and its inside are no longer
activity, whereas only later periods (300–600ms) primarily determined by the outside, i.e., cogni-
are more shaped by external stimuli. These tive function, but by neuronal activity mostly on
and other results (Braun et al., 2021; He, 2013; the cellular and regional level – this amounts to
Huang et al., 2017; Wainio-Theberge et al., 2021) what GB describes as “inside–out” model which
strongly suggest that inside and outside (i.e., spon- has also been emphasized by others (Northoff,
taneous and task-related activity) are integrated in 2014a,b; Northoff et al., 2010a,b; Raichle, 2009).
the brain’s neuronal activity. Rather than operating Rather than opposing the brain’s inside and out-
in parallel, the brain seems to constitute an inside- side, we aim to reconcile GB and DP/FA by point-
outside continuum of neuronal activity. A quanti- ing out their similarities on a fundamental level.
tative inside–outside continuum undermines the Our guiding question is: what is shared between
qualitative historical inside vs. outside debate. The the brain’s spontaneous activity on its inside and
inside vs. outside dichotomy stems from observa- cognition/behavior on the outside? The search for
tional and methodological limitations. Both per- the similarities of inside and outside rather than
spectives are epistemological but not ontological/ for their differences entails the quest to identify
natural (i.e., they are not rooted in the neurobiol- their “common currency,” as we recently put for-
ogy of the brain itself), that is, the way it operates ward (Northoff et al., 2020a,b). Both GB (Buszaki,
and functions independent of our investigation and 2020) and DP/FA (Poeppel and Adolfi, 2020)
methodological approaches. hint upon a possible key role of brain dynamics
24 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

without elaborating it in full detail. Brain dynam- “temporal receptive windows” (Chaudhuri et al.,
ics may consist of the brain’s inner time and space. 2015; Hasson et al., 2012, 2015; Honey et al.,
The spontaneous activity’s inner time and space 2012; Huang et al., 2018; Kiebel et al., 2008;
shape the outside, i.e., task-related activity and Lerner et al., 2011; Raut et al., 2020; Stephens
cognition, in a non-additive fashion. Such shaping et al., 2013; Wasmuht et al., 2018; Watanabe et al.,
of the outside by the inside can be based on the 2019; Wolff et al., 2019) have been introduced to
inside’s temporal and spatial dynamics, which are describe such temporal structuring of input and
thereby linked to spatiotemporal configurations of information processing.
the outside. Hence, the brain’s inside and outside How is such temporal (and spatial) structuring
share temporo-spatial dynamics and topography. of our perception and cognition during the brain’s
This resolves the debate between GB and DP/ task states related to the spontaneous activity and
FA. Both approaches, inside–out and outside–in, its intrinsic neural timescales? A recent study by
are feasible as in both instances one searches for Golesorkhi et al. (2021) investigated intrinsic neu-
what is shared between them, namely their spatial ral timescales (i.e., the autocorrelation window;
topography and temporal dynamics. The brain’s ACW), in rest and task MEG. They observed cor-
dynamics are what is shared inside and outside. relations of rest and task ACW (ranging from 0.7
Temporo-spatial dynamics can be approached to 0.9) including the almost complete preservation
from either direction, that is, from inside–out and of the temporo-spatial core–periphery organiza-
outside–in. tion during different tasks with only a few regions/
networks showing task-specific changes in ACW.
Albeit tentatively, these findings provide a first
bridge between the brain’s inside (GB) and its out-
Brain’s inner time–space shapes the side (DP/AF): they show how the brain’s intrinsic
outer time–space of cognition temporo-spatial dynamics (i.e., the brain’s inside
as emphasized by GB) is carried over and con-
DP/FA consider GB’s focus on the brain’s inside nects to the outside, namely the cognitive function
as insufficient. According to them, the real prob- during task states as they are the focus of DP/AF.
lem is how to connect neuronal and psychological Hence the brain’s temporo-spatial dynamics may
concepts. DP/FA are well aware of this – they call provide a bridge, a “common currency” between
it “alignment or mapping problem” that consists GB’s inside approach and DP/AF’s considera-
in, as they describe it, how to connect or align the tion of an outside approach, and thus between
elementary units or primitives of psychology to the brain’s inside and outside and thereby also
the ones of the brain. The alignment or mapping between brain and world. Temporo-spatial dynam-
problem (AMP) contains two subproblems, the ics may thus be conceived to be shared as “com-
granularity mismatch problem (GMP) and the mon currency” of inside (i.e., brain), and outside
ontological incommensurability problem (OIP): (i.e., cognition and mind), requiring what recently
the GMP refers to the problem that two disciplines has been termed “Spatiotemporal Neuroscience”
like psychology and neuroscience may target the (Northoff et al., 2019, 2020).
same phenomenon but in different resolutions,
while the OIP brings up the potential incompati-
bility of neural and psychological concepts and,
more generally, of brain and mind.
Hasson’s group conducted a series of fMRI SUMMARY AND CONCLUSIONS
(and intra-cranial EEG) task studies showing that
different regions are recruited for the information This chapter has focused on empirical and philo-
processing of different temporal duration (Chen sophical presuppositions of cognitive neurosci-
et al., 2015; Hasson et al., 2015; Honey et al., ence and their historical origin. Localizationism
2012; Regev et al., 2019). For instance, short fea- stemmed from neurology, while the input–cogni-
tures like single words or phonemes recruit early tion–output relationship in cognitive psychology
sensory regions in the auditory and/or visual cor- ultimately derives from behaviorism. The combi-
tex, while longer features like sentences activate nation of the scientific disciplines cognitive psy-
intermediate regions such as the superior temporal chology with neuroscience resulted in a framework
gyrus and inferior frontal gyrus (Stephens et al., that comprises contradictory theories about the
2013). Finally, even longer information like whole mind, brain function, and the relation between
paragraphs activates regions in the default-mode human experience and corresponding physiologi-
network (DMN) (Chen et al., 2015). Departing cal processes in the nervous system. A unified
from these findings, the concepts of “tempo- framework in neuroscience that could contribute
ral receptive fields” (Cavanagh et al., 2016) or toward the solution of controversy around these
HISTORICAL, EMPIRICAL, AND PHILOSOPHICAL PERSPECTIVES 25

themes is lacking. We suggest that neuroscience the National Academy of Sciences of the United
and the science of the mind can profit from States of America, 104, 12187–12192.
Spatiotemporal Neuroscience. This step could Button, K. S., Ioannidis, J. P. A., Mokrysz, C., Nosek,
contribute by intrinsically linking human phenom- B. A., Flint, J., Robinson, E. S. J., & Munafò, M. R.
enology with the brain’s neuronal activity to (2013). Power failure: Why small sample size
advance our understanding of the seeming opposi- undermines the reliability of neuroscience. Nature
tion between the discussed topics. Brain and mind Reviews Neuroscience, 14, 365–376.
could be integrated via their “common currency” Buzsáki, G. (2006). Rhythms of the brain. New York:
on a most fundamental level of time and space. Oxford University Press.
Buzsáki, G. (2019). The brain from inside out. New
York: Oxford University Press.
Buzsáki, G. (2020). The Brain-Cognitive Behavior
REFERENCES Problem: A Retrospective. eNeuro, 7(4). doi:
10.1523/ENEURO.0069-20.2020
Arieli, A., Sterkin, A., Grinvald, A., & Aertsen, A. Castellanos, F. X., Di Martino, A., Craddock, R. C.,
(1996). Dynamics of ongoing activity: exploration Mehta, A. D., & Milham, M. P. (2013). Clinical
of the large variability in evoked cortical responses. applications of the functional connectome. Neuro-
Science, 273, 1868–1871. Image, 80, 527–540.
Bandettini, P. A., Wong, E. C., Hinks, R. S., Tikofsky, Cavanagh, S. E., Wallis, J. D., Kennerley, S. W., &
R. S., & Hyde, J. S. (1992). Time course EPI of Hunt, L. T. (2016). Autocorrelation structure at rest
human brain function during task activation. Mag- predicts value correlates of single neurons during
netic Resonance in Medicine, 25(2), 390–397. reward-guided choice. Elife, 5. doi: 10.7554/
Bay, Y., Nakao, T., Xu, J., Qin, P., Chaves, P., Heinzel, A., … eLife.18937.001
Northoff, G. (2016). Resting state glutamate predicts Chaudhuri, R., Knoblauch, K., Gariel, M. A., Ken-
elevated pre-stimulus alpha during self-related- nedy, H., & Wang, X. J. (2015). A large-scale cir-
ness: A combined EEG-MRS study on the “rest-self cuit mechanism for hierarchical dynamical
overlap.” Social Neuroscience, 11(3), 249–263. processing in the primate cortex. Neuron, 88(2),
Berger, H. (1929). Über das elektrenkephalogramm 419–431.
des menschen. Archiv für Psychiatrie und Nerven- Chen, P. H., Wagner, D. D., Kelley, W. M., & Heath-
krankheiten, 87, 527–570. erton, T. F. (2015). Activity in cortical midline struc-
Bilder, R. M., Sabb, F. W., Parker, D. S., Kalar, D., Chu, tures is modulates by self-construal changes
W. W., Fox, J., Freimer, N. B., & Poldrack, R. A. during acculturation. Culture and Brain, 3(1),
(2009). Cognitive ontologies for neuropsychiatric 39–52.
phenomics research. Cognitive Neuropsychiatry, Churchland, P. (2012). Plato’s camera: How the
14(4-5), 419–450. physical brain captures a landscape of abstract
Bilder, R. M., Howe, A. G., & Sabb, F. W. (2013). universals. Cambridge, MA: MIT Press.
Multilevel models from biology to psychology: Cole, D. M., Smith, S. M., & Beckmann, C. F. (2010).
mission impossible? Journal of Abnormal Psychol- Advances and pitfalls in the analysis and
ogy, 122(3), 917–927. interpretation of resting-state FMRI data. Frontiers
Bishop, G. (1933). Cyclic changes in excitability of in Systems Neuroscience, 4(8). doi: 10.3389%2
the optic pathway of the rabbit. American Journal Ffnsys.2010.00008
of Physiology, 103, 213–224. Cole, M. W., Bassett, D. S., Power, J. D., Braver, T. S.,
Biswal, B., Yetkin, F. Z., Haughton, Haughton, V. M., & Petersen, S. E. (2014). Intrinsic and task-evoked
& Hyde, J. S. (1995). Functional connectivity in the network architectures of the human brain. Neuron,
motor cortex of resting human brain using echo- 83(1), 238–251.
planar MRI. Magnetic Resonance in Medicine, Cole, M. W., Ito, T., Bassett, D. S., & Schultz, D. H.
34(4), 537–541. (2016). Activity flow over resting-state networks
Bijsterbosch, J., Smith, S., & Beckmann, C. (2017). shapes cognitive task activations. Nature Neurosci-
Introduction to resting state fMRI functional con- ence, 19(12), 1718–1726.
nectivity. New York: Oxford University Press. D’Argembeau, A., Collette, F., van der Linden, M.,
Block, N. (2014). Rick conscious perception outside Laureys, S., Del Fiore, G., Degueldre, C., Luxen, A.,
focal attention. Trends in Cognitive Sciences, & Salmon, E. (2005). Self-referential reflective
18(9), 445–447. activity and its relationship with rest: A PET study.
Boly, M., Balteau, E., Schnakers, C., Degueldre, C., NeuroImage, 25(2), 616–624.
Moonen, G., Luxen, A., … Laureys, S. (2007). Davey, C. G., Pujol, J., & Harrison, B. J. (2016). Map-
Baseline brain activity fluctuations predict soma- ping the self in the brain’s default mode network.
tosensory perception in humans. Proceedings of NeuroImage, 132, 390–397.
26 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Deco, G., Jirsa, V. K., & McIntosh, A. R. (2013). Rest- of Sciences of the United States of America,
ing brains never rest: computational insights into 100(1) 253–258.
potential cognitive architectures. Trends in Neuro- Hasson, U., Yang, E., Vallines, I., Heeger, D. J., &
sciences, 36(5), 268–274. Rubin, N. (2008). A hierarchy of temporal recep-
Eklund, A., Nichols, T. E., & Knutsson, H. (2016). tive windows in human cortex. Nature Communi-
Cluster failure: Why fMRI inferences for spatial cations, 28(10), 2539–2550.
extent have inflated false-positive rates. Proceed- Hasson, U., Chen, J., & Honey, C. (2015). Hierarchical
ings of the National Academy of Science of the process memory: memory as an integral compo-
United States of America, 113(28), 7900–7905. nent of information processing. Trends in Cogni-
Fazelpour, S., & Thompson, E. (2015). The Kantian tive Sciences, 19(6), 304–313.
brain: Brain dynamics from a neurophenomeno- Hastings, J., Frishkoff, G. A., Smith, B., Jensen, M.,
logical perspective. Current Opinion in Neurobiol- Poldrack, R. A., Lomax, J., … Martone, M. E.
ogy, 31, 223–229. (2014). Interdisciplinary perspectives on the devel-
Feinberg, T. E. (2009). From axons to identity: Neuro- opment, integration, and application of cognitive
logical explorations of the nature of the self. New ontologies. Frontiers in Neuroinformatics, 8. doi:
York: Norton. 10.3389/fninf.2014.00062
Feinberg, T. E., & Keenan, J. P. (2005). The lost self: He, B. J. (2013). Spontaneous and task-evoked brain
Pathologies of brain and identity. New York: activity negatively interact. Journal of Neuro-
Oxford University Press. sciences, 33(11), 4672–4682.
Ferri, F., Nikolova, Y. S., Perrucci, M. G., Constantini, He, B. J., Zempel, J. M., Snyder, A. Z., & Raichle, M. E.
M., Ferretti, A., Gatta, V., … Northoff, G. (2017). (2010). The temporal structures and functional
A neural “tuning curve” for multisensory experi- significance of scale-free brain activity. Neuron,
ence and cognitive-perceptual schizotypy. Schizo- 66(3), 353–369.
phrenia Bulletin, 43(4), 801–813. He, B. J. (2014). Scale-free brain activity: past, pre-
Fodor, J. A. (1983). The modularity of mind. Cam- sent, and future. Trends in Cognitive Sciences,
bridge, MA: MIT Press. 18(9), 480–487.
Fodor, J. A. (1985). Précis of the modularity of mind. Honey, C. J., Thesen, T., Donner, T. H., Silbert, L. J.,
Behavioral and Brain Science, 8(1), 1–42. Carlson, C. E., Devinsky, O., … Hasson, U. (2012).
Fodor, J. A. (1998). Concepts. Oxford, UK: Slow cortical dynamics and the accumulation of
Clarendon. information over long timescales. Neuron 76(2),
Fox, M. D., Snyder, A. Z., Zacks, J. M., & Raichle, M. E. 423–434.
(2006). Coherent spontaneous activity accounts Howes, O. D., & Murray, R. M. (2014). Schizophre-
for trial-to-trial variability in human evoked brain nia: An integrated sociodevelopmental-cognitive
responses. Nature Neuroscience, 9(1), 23–25. model. Lancet, 383, 1677–1687.
Fox, M. D., Snyder, A. Z., Vincent, J. L., & Raichle, M. E. Hesselmann, G., Kell, C. A., & Kleinschmidt, A.
(2007). Intrinsic fluctuations within cortical sys- (2008). Ongoing activity fluctuations in hMt+ bias
tems account for intertrial variability in human the perception of coherent visual motion. Journal
behavior. Neuron, 56(1), 171–184. of Neuroscience, 28(53), 14481–14485.
Garcia, J., Kimeldorf, D. J., & Koelling, R. A. (1955). Huang, Z., Zhang, J., Wu, J., Qin, P., Wu, X., Wang, Z., …
Conditioned aversion to saccharin resulting from Northoff, G. (2016). Decoupled temporal variabil-
exposure to gamma radiation. Science, 122, ity and signal synchronization of spontaneous
157–158. brain activity in loss of consciousness: An fMRI
Garcia, J. & Koelling, R. A. (1966). Relation of cue to study in anesthesia. NeuroImage, 124, 693–703.
consequence in avoidance learning. Psychonomic Huang, Z., Zhang, J., Wu, J., Liu, X., Xu, J., Zhang, J.,
Science, 4, 123–124. … Northoff, G. (2018). Disrupted neural variability
Gazzaniga, M. S., Ivry, R. B., & Mangun, G. R. (2018). during propofol-induced sedation and uncon-
Cognitive neuroscience: The biology of the mind. sciousness. Human Brain Mapping, 39(11),
(5th ed.). New York: Norton. 4533–4544.
Golesorkhi, M., Gomez-Pilar, J., Zilio, F., Berberian, N., Insel, T. R., & Cuthbert, B. N. (2015). Medicine: Brain
Wolff, A., Yagoub, M. C. E., & Northoff, G. disorders? Precisely. Science, 348, 499–500.
(2021). The brain and its time: Intrinsic neural Kant, I. (1781/1996). Critique of pure reason. Indian-
timescales are key for input processing. Commu- apolis, IN: Hackett.
nications Biology, 4(1). doi: 10.1038/s42003-021- Klein, S. B. (2012). Self, memory, and the self-refer-
02483-6 ence effect: An examination of conceptual and
Greicius, M. D., Krasnow, B., Reiss, A. L., & Menon, V. methodological issues. Personality and Social Psy-
(2003). Functional connectivity in the resting chology Review, 16(3), 283–300.
brain: A network analysis of the default mode Kiebel, S. J., Daunizeau, J., & Friston, K. J. (2008). A
hypothesis. Proceedings of the National Academy hierarchy of time-scales and the brain. Plos
HISTORICAL, EMPIRICAL, AND PHILOSOPHICAL PERSPECTIVES 27

Computational Biology, 4(11). doi: 10.1371/journal. Northoff, G. (2014b). Unlocking the brain. Vol. II:
pcbi.1000209 Consciousness. New York: Oxford University Press.
Koffka, K. (1935). Principles of gestalt psychology. Northoff, G. (2018a). The spontaneous brain. From
London: Routledge & Kegan. the mind-body to the world-brain problem. Cam-
Köhler, W. (1920). Die physischen gestalten in ruhe bridge, MA: MIT Press.
und im stationären zustand: Eine naturphiloso- Northoff, G. (2018b). The brain’s spontaneous activ-
phische untersuchung. Braunschweig, Germany: ity and its psychopathological symptoms – “Spati-
Vieweg. otemporal binding and integration.” Progress in
Kwong, K. K., Belliveau, J. W., Chesler, D. A., Gold- Neuro-Psychopharmacology and Biological Psychi-
berg, I. E., Weisskoff, R. M., Poncelet, B. P., … atry, 80(B), 81–90.
Turner, R. (1992). Dynamic magnetic resonance Northoff, G., & Sibille, E. (2014). Why are cortical
imaging of human brain activity during primary GABA neurons relevant to internal focus in depres-
sensory stimulation. Proceedings of the National sion? A cross-level model linking cellular, bio-
Academy of Sciences of the United States of chemical and neural network findings. Molecular
America, 89(12), 5675–5679. Psychiatry. 19(9), 966–977.
Lerner, Y., Honey, C. J., Silbert, L. J., & Hasson, U. Northoff, G., & Huang, Z. (2017). How do the brain’s
(2011). Topographic mapping of a hierarchy of time and space mediate consciousness and its dif-
temporal receptive windows using a narrated ferent dimensions? Temporo-spatial theory of
story. Journal of Neuroscience, 31(8), 2906–2915. consciousness (TTC). Neuroscience & Biobehavio-
Lersch, P. (1956). Aufbau der Person. München, Ger- ral Reviews, 80, 630–645.
many: Johann Ambrosius Barth. Northoff, G. Wainio-Theberge, S., & Evers, K. (2019).
Liu, X., de Zwart, J. A., Schölvinck, M. L., Chang, C., Is temporo-spatial dynamics the “common cur-
Ye, F. Q., Leopold, D. A. & Duyn, J. H. (2018). rency” of brain and mind? In quest of “spatiotem-
Subcortical evidence for a contribution of arousal poral neuroscience.” Physics of Life Reviews, 33,
to fMRI studies of brain activity. Nature Communi- 34–54.
cations, 9(1). doi: 10.1038/s41467-017-02815-3 Northoff, G. Wainio-Theberge, S., & Evers, K. (2020).
Llinas, R. R. (1988). The intrinsic electrophysiological Spatiotemporal neuroscience – what is it and why
properties of mammalian neurons–insights into we need it. Physics of Life Reviews, 33, 78–87.
central nervous-system function. Science, 242, Ogawa, S., Tank, D. W., Menon, R., Ellermann, J. M.,
1654–1664. Kim, S. G., Merkle, H., & Ugurbil, K. (1992). Intrin-
Logothetis, N. K., Murayama, Y., Augath, M., Stef- sic signal changes accompanying sensory stimula-
fen, T., Werner, J., & Oeltermann, A. (2009). How tion: functional brain mapping with magnetic
not to study spontaneous activity. NeuroImage, resonance imaging. Proceedings of the National
45(4), 1080–1089. Academy of Sciences of the United States of
Margulies, D. S., Ghosh, S. S., Goulas, A., Falkiewicz, America, 89(13), 5951–5955.
M., Huntenburg, J. M., Langs, G., …Smallwood, J. Passingham, R. (2016). Cognitive neuroscience. A
(2016). Situating the default-mode network along very short introduction. Oxford, UK: Oxford Uni-
a principal gradient of macroscale cortical organi- versity Press.
zation. Proceedings of the National Academy of Poepel, D., & Adolfi, F. (2020). Against the epistemo-
Sciences, 113(44), 12574–12579. logical primacy of the hardware: The brain from
Menon, V. (2011). Large-scale brain networks and inside out, turned upside down. eNeuro, 7(4). doi:
psychopathology: A unifying triple network model. 10.1523/ENEURO.0215-20.2020
Trends in Cognitive Sciences, 15(10), 483–506. Poldrack, R. A., Kittur, A., Kalar, D., Miller, E., Seppa,
Métraux, A., & Frisch, S. (2020). Some critical C., Gil, Y., … F. W., Bilder, R. M. (2011). The cogni-
remarks on the epistemology of functional mag- tive atlas: Toward a knowledge foundation for
netic resonance imaging. Science and Philosophy, cognitive neuroscience. Frontiers in Neuroinfor-
8(1), 63–74. matics, 5. doi: 10.3389/fninf.2011.00017
Meyer, M. F. (1922). Psychology of the other-one: An Power, J. D., Schlaggar, B. L., & Petersen, S. E. (2015).
introductory text-book of psychology. Columbia, Recent progress and outstanding issues in motion
MO: Missouri Book. correction in resting state fMRI. NeuroImage, 105,
Morcom, A. M., & Fletcher, P. C. (2007). Does the 536–551.
brain have a baseline? Why we should be resisting Power, J. D., Plitt, M., Laumann, T. O., & Martin, A.
a rest. NeuroImage, 37(4), 1073–1082. (2017). Sources and implications of whole-brain
Northoff, G. (2012). Immanuel Kant’s mind and the fMRI signals in humans. NeuroImage, 146,
brain’s resting state. Trends in Cognitive Sciences, 609–625.
16(7), 356–359. Power, J. D., Plitt, M., Gotts, S. J., Kundu, P., Voon,
Northoff, G. (2014a) Unlocking the brain, Vol. 1: V., Bandettini, P. A., & Martin A. (2018). Ridding
Coding. New York: Oxford University Press. fMRI data of motion-related influences: Removal
28 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

of signals with distinct spatial and physical bases in Szucs, D., & Ioannidis, J. P. A. (2017). When null
multiecho data. Proceedings of the National Acad- hypothesis significance testing is unsuitable for
emy of Sciences of the United States of America, research: A reassessment. Frontiers in Human
115(9), E2105–E2114. Neuroscience, 11. doi: 10.3389/fnhum.2017.
Qin, P., & Northoff, G. (2011). How is our self related 00390
to midline regions and the default-mode network? Tagliazucchi, E., von Wegner, F., Morzelewski, A.,
NeuroImage, 57(3), 1221–1233. Brodbeck, V., Jahnke, K., & Laufs, H. (2013).
Raichle, M. E., MacLeod, A. M., Snyder, A. Z., Breakdown of long-range temporal dependence
Powers, W. J., Gusnard, D. A., & Shulman, G. L. in default mode and attention networks during
(2001). A default mode of brain function. Pro- deep sleep. Proceedings of the National Academy
ceedings of the National Academy of Sciences of of Sciences of the United States of America,
the United States of America, 98(2), 676–682. 110(38), 15419–15424.
Raichle, M. E. (2009). A brief history of human brain Tagliazucchi, E., Chialvo, D. R., Siniatchkin, M.,
mapping. Trends in Neurosciences, 32(2), Amico, E., Brichant, J. F., Bonhomme, V., … Lau-
118–126. reys, S. (2016). Large-scale signatures of uncon-
Raichle, M. E. (2015). The restless brain: how intrinsic sciousness are consistent with a departure from
activity organizes brain function. Philosophical critical dynamics. Journal of the Royal Society, 13.
Transactions of the Royal Society B, 19. doi: doi: 10.1098/rsif.2015.1027
10.1098/rstb.2014.0172 Tellenbach, H. (1990). Aspects of time: Its distortions
Raichle, M. E. (2015). The brain’s default mode net- in psychiatric diseases. Zeitschrift fur Klinische
work. Annual Reviews Neuroscience, 38, Psychologie, Psyhopathologie und Psychotherapie,
433–447. 38(1), 52–61.
Raut, R. V., Snyder, A. Z., & Raichle, M. E. (2020). Tsuchiya, N., Wilke, M., Frässle, S., Lamme, & V. A. F.
Hierarchical dynamics as a macroscopic organizing (2015). No-report paradigms: Extracting the true
principle of the human brain. Proceedings of the neuronal correlates of consciousness. Trends in
National Academy of Sciences of the United States Cognitive Sciences, 19(12), 757–770.
of America, 117(34). doi: 10.1073/pnas.20033 van Eijsden, P., Hyder, F., Rothman, D. L., & Shulman,
83117 R. G. (2009). Neurophysiology of functional imag-
Sadaghiani, S., Hesselmann, G., Friston, K. J., & ing. NeuroImage, 45(4), 1047–1054.
Kleinschmidt, A. (2010). The relation of ongoing von Holst, E. (1937). Vom Wesen der Ordnung im
brain activity, evoked neural responses, and cogni- Zentralnervensystem. Naturwissenschaften, 25,
tion. Frontiers in Systems Neurosciences, 4. doi: 625–631.
10.3389/fnsys.2010.00020 von Holst, E. & Mittelstaedt, H. (1950). Das Reaffer-
Schölvinck, M. L., Maier, A., Ye, F. Q., Duyn, J. H., & enzprinzip: Wechselwirkungen zwischen Zentral-
Leopold, D. A. (2010). Neural basis of global rest- nervesystem und Peripherie. Naturwissenschaften,
ing-state fMRI activity. Proceedings of the National 37, 464–476.
Academy of Sciences of the United States of Wainio-Theberge, S., Wolff, A., & Northoff, G.
America, 107(22): 10238–10243. (2021). Dynamic relationships between spontane-
Seligman, M., Ives, C. E., Ames, H. & Mineka, S. ous and evoked electrophysiological activity.
(1970). Conditioned drinking and its failure to Communications Biology, 4. doi: 10.1038/s42003-
extinguish: avoidance, preparedness, or functional 021-02240-9
autonomy? Journal of Comparative and Physiolog- Wasmuth, D. F., Spaak, E., Buschman, T. J., Miller, E. K.,
ical Psychology, 71(3), 411–419. & Stokes, M. G. (2018). Intrinsic neuronal dynamics
Seligman, M. (2016). Phobias and preparedness. predict distinct functional roles during working
Behavior Therapy, 47(5), 577–584. memory. Nature Communications, 9(1). doi:
Skinner, B. F. (1948/2005). Walden two. Indianapolis, 10.1038/s41467-018-05961-4
IN: Hacket. Watanabe, T., Rees, G., & Masuda, N. (2019). Atypi-
Skinner, B. F. (1971). Beyond freedom and dignity. cal intrinsic neural timescale in autism. eLife, 8.
New York: Knopf. doi: 10.7554/eLife.42256.001
Smallwood, J., Bernhardt, B. C., Leech, R., Bzdok, D., Wertheimer, M. (2017). Max Wertheimer and Gestalt
Jefferies, E., & Margulies, D. S. (2021). The default theory. London: Routledge.
mode network in cognition: A topographical per- Whitfield-Gabrieli, S., Moran, J. M., Nieto-Castanon,
spective. Nature Reviews Neuroscience, 22, A., Triantafyllou, C., Saxe, R., & Gabrieli, J. D.
503–513. (2011). Associations and dissociations between
Stephens, G. J., Honey, C. J., & Hasson, U. (2013). A default and self-reference networks in the human
place for time: the spatiotemporal structure of brain. NeuroImage, 55(1), 225–232.
neural dynamics during natural audition. Journal Wolff, A. Di Giovanni, D. A., Gómez-Pilar, J., Nakao,
of Neurophysiology, 110(9), 2019–2026. T., Huang, Z., Longtin, A., & Northoff, G. (2019).
HISTORICAL, EMPIRICAL, AND PHILOSOPHICAL PERSPECTIVES 29

The temporal signature of the self: Temporal Yuste, R. MacLean, J. N., Smith, J., & Lansner, A.
measures of resting-state EEG predict self-con- (2005). The cortex as a central pattern generator.
sciousness. Human Brain Mapping, 40(3), Nature Reviews Neuroscience, 6, 477–483.
789–803. Zhang, J., Magioncalda, P., Huang, Z., Tan, Z., Hu, X.,
Wolff, A., Chen, L, Tumati, S., Golesorkhi, M., Hu, Z., … Northoff, G. (2018). Altered global
Gomez-Pilar, J., Hu, J., … Northoff, G. (2021). signal topography and its different regional locali-
Prestimulus dynamics blend with the stimulus in zation in motor cortex and hippocampus in mania
neural variability quenching. NeuroImage, 238. and depression. Schizophrenia Bulletin, 45(4),
doi: 10.1016/j.neuroimage.2021.118160 902–910.
3
Aging Brain Changes
across the Lifespan
L í d i a V a q u é - A l c á z a r, A n j a S o l d a n ,
and David Bartrés-Faz

INTRODUCTION Ultimately, such advances shall result in more


individually tailored recommendations to opti-
There is probably no time along the life cycle mize brain and cognitive health across the
where the brain remains “static.” Brain changes lifespan.
occurring during late life were once conceptual- This chapter discusses recent scientific evi-
ized as almost separate in nature, regarding those dence aimed at investigating the origins, factors,
during development. However, current mounting and mechanisms that explain how the brain ages
evidence, including data from both structural and in humans. We focus primarily on findings derived
functional neuroimaging favors a non-linear per- from human neuroimaging evidence, through
spective of lifespan changes, where modes of vari- magnetic resonance imaging (MRI), which is a
ations or patterns of brain changes observed in non-invasive technique that uses a magnetic field
adulthood and aging are already shaped early in and radiofrequency waves to create detailed pic-
life and are associated with inter-individual differ- tures of organs and structures inside the body
ences in cognitive trajectories. Furthermore, in (Mori and Barker, 1999). In recent decades, the
combination with one’s genetic background, life- availability of diverse MRI methods has contrib-
style factors and environmental influences may uted significantly to greater knowledge and under-
impact aspects of cognitive performance across standing of how the brain experiences age-related
the lifespan. Different theoretical models of cog- changes at an anatomical and functional level.
nitive and brain aging have been put forward with Brain MRI can be broadly divided into struc-
a focus on elucidating the factors and neurobio- tural and functional acquisitions. MRI sequences
logical underpinnings associated with the mainte- that assess aspects of brain structure include
nance of cognitive function as we age. Going structural T1- and T2-weighted (T1w and T2w)
forward, the increased access to massive multi- MRI acquisitions that allow for high-­resolution
center longitudinal databases combined with con- imaging of the brain’s tissues, including gray
trolled interventional approaches should propel matter (GM) and white matter (WM), with good
our understanding of the determinants and mecha- anatomic detail. In addition, diffusion-weighted
nisms that account for the lifespan individual dif- imaging (DWI) is increasingly being used to
ferences that influence how our brain ages. assess the microstructural properties of the brain,
Aging Brain Changes across the Lifespan 31

particularly of WM fiber tracts, by measuring the neurobiological processes. For example, cortical
diffusion of water molecules. Functional brain thinning occurring from childhood to adolescence
MRI measures include the T2w* images based might correspond to a progressive reduction in
on the Blood-Oxygen-Level Dependent (BOLD) synaptic and neural density, as well as a possible
signal, which can inform about the brain areas and reduction in the number of cortical glial cells, and
networks that are active during cognitive demands changes in intracortical myelinization, a measure
or can be used to identify resting-state functional that can be estimated through MRI (i.e., using
brain networks. T1w contrast and T1w/T2w ratio). Furthermore,
recently available resources of the RNA transcripts
expressed in the human brain, such as the Allen
Human Brain Atlas (https://portal.brain-map.
Lifespan Changes in Gray Matter org/), or the BrainSpan atlas (www.brainspan.
org/), are providing insight into how maturational
There is a growing corpus of scientific literature inter-regional patterns of changes seen in MRI
investigating the changes in GM structures (e.g., cortical thinning) relate to the gradients of
(volume, surface, and/or thickness) across the glial or neural-specific cell-type gene expression,
lifespan. Broadly, GM increases during develop- and how they may differ across the lifespan (i.e.,
ment and decreases with aging (Pfefferbaum development vs. adulthood; Vidal-Piñeiro et al.,
et al., 2013; Pomponio et al., 2020). At least some 2020). This type of knowledge should be critical
portion of the lifespan changes in cognitive func- to understanding the neurobiological mechanisms
tion and behavior are caused by lifespan-related governing interindividual differences in lifespan
changes in cortical morphology (Fjell et al., trajectories regarding cortical changes and cogni-
2014a; Schmitt et al., 2019). tion, including the neurodevelopmental origins of
Human volume cortical growth arises through major neuropsychiatric conditions.
the complex interplay of several distinct facets of Studies investigating the topographic pat-
cortical anatomy, such as changes in cortical thick- tern of GM changes among older adults, have
ness (CTh), cortical area and gyrification during reported a prominent reduction of the prefrontal
development (Tamnes et al., 2017). Maturation cortex, particularly dorsolateral and dorsomedial
changes are highly heterogeneous across regions regions, with many also implicating lateral pari-
(Shaw et al., 2008). Furthermore, human brain etal and lateral temporal areas (Fjell et al., 2014a;
development is characterized by a great expan- Raz et al., 2005; Salat, 2004). Moreover, related
sion in the surface area of the cerebral cortex, to the negative association between age and
exhibiting rapid increases during infancy, further GM values, there appears to be an exponential
expansion during childhood, and a peak in late increase of ventricular cerebrospinal fluid with
childhood or adolescence, followed by stability age (Pfefferbaum et al., 2013). The lateral ventri-
or subtle decreases in adulthood (Norbom et al., cles show a continuous enlargement throughout
2021). The areas that expand the most during the lifespan (Dima et al., 2022), and is detectable
human development correspond to those that dif- over one-year follow-up even among cognitively
fer most markedly compared to macaque monkey normal older adults at low-risk of AD (Fjell et al.,
brains (Rilling, 2014). There has been a debate in 2013).
the literature on whether CTh “peaks” are observ- Many earlier studies of GM change did not spe-
able in early development, or whether this meas- cifically screen participants at baseline or follow-
ure rather shows monotonic thinning from early up Mild Cognitive Impairment (MCI), an indicator
to late childhood, adolescence, and beyond across of the presence of underlying disease processes,
most regions over the entire lifespan (Walhovd including Alzheimer’s disease (AD) and cer-
et al., 2016). Using a lifespan perspective, a ebrovascular disease. Longitudinal cohort stud-
study including cross-sectional data from 17,075 ies of middle-aged and older adults with normal
healthy individuals aged 3–90 years (Frangou cognition at baseline (as determined by extensive
et al., 2022) revealed that the highest CTh values clinical and cognitive evaluations) have shown
were observed in childhood (between 3 and 10 that rates of GM decline are lower among those
years of age). Additionally, there appeared to be who remain cognitively normal over the follow-
a stronger relationship between increased age and up period compared to those who develop MCI
lower CTh early in life (i.e., up to the second or or dementia over time (Armstrong et al., 2019;
third decade) compared to later in life (Frangou Soldan et al., 2015). These types of studies have
et al., 2022), with the strength of this association also shown that rates of GM and ventricular change
becoming weaker thereafter (Ziegler et al., 2012). among middle-aged and older adults with normal
Macroscopic MRI cortical changes observed cognition at baseline are greater in those with car-
during maturational stages can be linked to specific diovascular risk factors, such as smoking, high
32 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

blood pressure, obesity, and elevated cholesterol relatively more pronounced decline, compared
levels (Armstrong et al., 2019) those at genetic to other subcortical regions (Dima et al., 2022;
risk for AD (Mishra et al., 2018; Reiter et al., Pomponio et al., 2020), potentially related to pre-
2017) and those with more abnormal baseline lev- clinical and early AD-related pathology.
els of amyloid, tau, or neuroinflammation (Halaas In this context of high heterogeneity, a recent
et al., 2020). The greatest and earliest declines in study applying a graph theory approach to analyze
GM among cognitively normal middle-aged and over 6,000 longitudinal MRIs of individuals rang-
older adults are often seen in medial-temporal ing in age from 2 to 89 demonstrated that subcor-
lobe regions, including the entorhinal cortex, hip- tical GM volumetric changes can be grouped into
pocampus, parahippocampal gyrus (Mishra et al., clusters of regions that show correlated change
2018; Reiter et al., 2017). Differentiating GM over time. These clusters tended to follow trends
changes related to pre-clinical disease processes from embryonic brain development, suggesting
from those that reflect non-disease related aging continuity from earlier to later stages of the lifes-
requires extensive follow-up (to determine who pan (Fjell et al., 2021). This consistency in patterns
eventually develops cognitive impairment), and of change of subcortical structures across the lifes-
careful cognitive and clinical evaluations of par- pan identified by the authors can be summarized
ticipants, as well as the collection of biomarkers as three independent trajectories: (1) accelerated
to assess preclinical disease levels. increase for the ventricles; (2) inverted U-shape
In contrast to the monotone thinning of the cer- progression for structures such as the hippocam-
ebral cortex (Storsve et al., 2014), subcortical GM pus and brain steam (Fjell et al., 2013; Walhovd
structures experience age-dependent changes across et al., 2005); and (3) linear decline through adult-
the human lifespan that are more divergent and hood following a peak in childhood (among
complex (Fjell et al., 2021; Narvacan et al., 2017; caudate, pallidum, putamen, amygdala, nucleus
Walhovd et al., 2005), and these changes are related accumbens, and cortical areas). However, for the
to cognitive performance, behavioral skills, and/or third trajectory, differences in shapes of the slopes
the pathophysiology of age-related brain diseases were observed, stressing divergences in the initial
(Narvacan et al., 2017). A recent lifespan study increase in childhood (Fjell et al., 2021).
with structural MRI data from 18,605 individuals Therefore, although cross-sectional studies
aged 3–90 years demonstrated that the volumes of usually find linear relationships between older age
subcortical structures were at their maximum value and smaller regional brain volumes, which sug-
early in life. Striatal and pallidal volumes peaked gests that aging affects the cortex diffusely (Salat,
in childhood (i.e., during the first decade of life) 2004), some studies have reported tendencies of
and the volumes of the thalamus, hippocampus, accelerating or decelerating estimated decline with
and amygdala peaked later (i.e., during the 20s– increasing age during the last decades of life (Fjell
30s; Dima et al., 2022). The basal ganglia volume et al., 2014b). For instance, Frangou et al., (2022),
showed a predominant negative association with showed how the entorhinal and temporo-polar cor-
age, while GM volumes of the thalamus, amygdala, tex are largely preserved across the lifespan until
and hippocampus showed a prolonged period of the 70s–80s, whereafter both regions showed an
stability (Dima et al., 2022; Pomponio et al., 2020). age-related decrease in CTh (also consistent with
However, after the 60s these structures also showed (Sele et al., 2021). In addition, Frangou et al.,
an abrupt negative association with age, potentially (2022) suggested that the interindividual variabil-
related, at least in part, to AD pathology, which ity seemed to be largest in temporal and frontal
tends to have early and pronounced effects on the regions across the lifespan. Interestingly, a recent
hippocampus and amygdala. longitudinal study of older non-demented partici-
As discussed below, the combination of genetic pants (mean baseline age = 70 years) found that
and environmental determinants contributes to cortical surface area showed a clear decline, on
explaining interindividual differences in GM average, over a span of up to seven years that was
changes observed across the lifespan. In any case, greater in magnitude than for CTh (i.e., 1–5% vs.
there is still significant uncertainty about the shape 0–2% annual decline, respectively). However, the
and interindividual variability regarding the asso- correlation pattern of CTh change between brain
ciation between age and GM in specific areas. Prior regions was strong and largely homogenous, sug-
studies have reported linear and nonlinear asso- gesting that cortical thinning with increasing age
ciations (Mills et al., 2016), and there are notable is correlated across regions. For cortical surface
exceptions to the general pattern described above in area, the pattern of between-region change was
structures such as cingulate and insula (Grieve et al., similar but weaker, suggesting the existence of a
2005), the orbitofrontal cortex (Raz et al., 2005), regional heterogeneity (Sele et al., 2021).
and the hippocampus (Grieve et al., 2005; Raz Changes in frontostriatal areas supporting
et al., 2005). The hippocampus seems to suffer a cognitive control and executive functions have
Aging Brain Changes across the Lifespan 33

been proposed to be characteristic hallmarks of abnormally high signal, referred to as WM


aging (Buckner, 2004), emphasizing that late-­ hyperintensities (WMH; also known as leu-
maturing regions are most vulnerable to age- karaiosis, or WM lesions, WML). WMH are
related changes, often referred to as the “last in, presumed to have a vascular origin with advanced
first out” or “retrogenesis” hypothesis (Fjell et al., age and hypertension being the major risk factors
2014a; Grieve et al., 2005). It seems that areas that (Wardlaw et al., 2013). The main cause of WML
take longer to mature because they follow more is thought to be chronic ischemia and they may
complex developmental trajectories (Shaw et al., reflect pathogenic mechanisms, related to demy-
2008), also are more vulnerable to the negative elination, gliosis, edema, apoptosis, and axonal
effects of aging. Thus, the “last in, first out” model atrophy in adjacent fiber tracts (Fazekas et al.,
has many similarities to the traditional “frontal 1998; Sasson et al., 2013). The prevalence and
theory of aging” (West, 1996). In addition, there is extent of WML increase with age (De Leeuw
a growing realization that genetically programmed et al., 2002), and they represent one of the most
neurodevelopmental events may have a lifelong common structural brain changes among older
impact on the organization of the cerebral cor- adults. According to some estimates, they are
tex decades later (Fjell et al., 2013). These find- present in almost 95% of adults over 45 years of
ings reveal the need for more research focused on age (De Leeuw et al., 2002; Habes et al., 2018).
identifying the genetic determinants and environ- WML have been associated with the cognitive
mental aspects that influence age-related cortical profile commonly seen among older adults,
changes. Moreover, the differences in rates of being associated with lower executive function-
regional GM change reported above may reca- ing and slower speed of processing. They have
pitulate distinct patterns of brain aging and in this also been associated with cognitive impairment
regard, it has been challenging to address whether (Raji et al., 2012), and they predict an increased
there are organizing principles underlying the risk of developing dementia. Recent evidence
relatively greater selective vulnerability of some suggests that WML contribute to the neurode-
regions as opposed to others. generation of cholinergic pathways among non-
In this regard, Cox et al., (2021) used multivari- demented older adults (i.e., above and beyond
ate longitudinal analyses to investigate clusters of AD pathology), likely helping to explain its role
regions showing shared morphometric cortical in cognitive function and cognitive deterioration
changes amongst individuals aged 70 and older (Cedres et al., 2022).
and with MRI acquisitions at three occasions. Beyond the study of small vessel disease-
Results revealed that beyond a general factor of related MRI changes reported above, diffusion-
global brain atrophy, two other orthogonal patterns weighted imaging (DWI) has become one of most
emerged, one characterized by more marked atro- popular MRI techniques in brain research (Assaf
phy in fronto-temporal regions and another pre- and Pasternak, 2008). DWI provides maps of
dominantly in occipito-parietal areas, with negative microscopic structural information of brain tis-
correlations between them (i.e., indicating that the sue (Lamar, 2014) and is most frequently used to
participants exhibiting higher decline in one, tend study WM fiber tracts. These images can be used
to express less declining in the other). In another to study the directionality of axons by measuring
study derived from the European Lifebrain pro- the diffusion of water molecules in the brain. The
ject (www.lifebrain.uio.no), Gorbach et al. (2020) most used DWI-derived metrics include fractional
observed a stronger association between hippocam- anisotropy (FA), a measure of directional diffu-
pal volume changes and memory changes amongst sion (Mukherjee et al., 2008), mean diffusivity
Apolipoprotein E (APOE) ε4 carriers compared to (MD), radial diffusivity (RD), and axial diffusiv-
non-carriers (both groups included non-demented ity (AxD). The MD may help characterizing tis-
older adults). Altogether, these findings are helping sue composition; RD appears has been suggested
to elucidate differences in the brain systems and to be related to myelin content; and AxD appears
brain–behavior associations that are more vulner- to be more specific to axonal injury (Bennett and
able for particular groups of individuals. Madden, 2014). Because these diffusion meas-
ures are affected by a variety of factors, including
fiber arrangement, axon density, myelination, and
White Matter Trajectory Over axonal degeneration, their biological interpret-
Lifespan ability is limited (Jones et al., 2013). In general,
however, higher FA and lower RD and MD values
Classically, WM changes as seen with MRI have tend to represent greater WM “integrity” (Bennett
been assessed by Fluid Attenuation Inversion and Madden, 2014).
Recovery (FLAIR) acquisitions, which is an Over the lifespan, the WM trajectory follows
MRI structural sequence able to identify areas of an inverted U-shape curve, detectable across
34 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

modalities (i.e., using both volume analyses from patterns identified is consistent with the “last in,
structural images and DWI methods; (Bennett and first out” hypothesis (also known as “retrogene-
Madden, 2014; Salat et al., 2005, 2011; Sexton sis”). This pattern is characterized by greater sus-
et al., 2014; Sullivan and Pfefferbaum, 2006). That ceptibility of the anterior brain that develops late
is, global FA values on average, peak around the to degeneration in old age, and the relative spar-
30s, followed by a weak yet stable linear decrease ing of posterior brain regions (i.e., occipital lobe)
until approximately 65 years, when there is a sub- that develop late (Bouhrara et al., 2021; Brickman
sequent accelerating decline (Sexton et al., 2014). et al., 2012). This idea (cf. Kiely et al., 2022), is
Studies of neurodevelopment overwhelmingly similar to what has been proposed for GM, pos-
demonstrate regional differences in the rates of tulating that late-myelinated WM fibers are most
WM maturation (Lynch et al., 2020), which are vulnerable to age-related changes (and/or disease-
characterized by FA increases and MD decreases related degeneration), which in turn, mediate
during childhood and adolescence (Lebel et al., cognitive decline (Brickman et al., 2012). Indeed,
2019). Several studies have suggested a posterior-­ available DWI-based studies have provided sup-
to-anterior gradient of WM maturation, with port for the “gain-predicts-loss hypothesis” of
anterior and associative tracts (e.g., the anterior WM tissue maturation and degeneration (Kiely
thalamic radiation) displaying a greater change et al., 2022; Slater et al., 2019), suggesting that the
during childhood (Krogsrud et al., 2016) than more rate of tissue loss at older ages is equal to the rate
posterior tracts. Recent technological advances of tissue gain during maturation at younger ages.
have provided new insights into a range of micro- Studies relating DWI measures to cognitive
structural aspects of WM that may be more closely function have been driven by diverse hypotheses
related to fundamental neurodevelopmental pro- about the effects of WM maturation and disruption
cesses (Lynch et al., 2020) than more traditional in cognition. During development, WM maturation
diffusion measures. For example, neurite orienta- gives rise to greater stability in cognitive function
tion dispersion and density imaging (NODDI) has (Samara et al., 2019), stressing the importance of
been established as a powerful complementary considering heterogeneities in the neurobiologi-
technique able to quantify other WM microstruc- cal basis underlying cognitive development (Bells
tural properties, including the density of neurites et al., 2019; Buyanova and Arsalidou, 2021). In
(both axons and dendrites), measured by the neu- the aging field, alterations in WM integrity have
rite density index (NDI), and the variability of been linked with cognitive disruption (Sang et al.,
neurite orientation (i.e., all parallel vs. randomly 2021). Additionally, some researchers have pro-
oriented), measured by the orientation dispersion posed various forms of “dis-connectivity” hypoth-
index (ODI). NODDI-based studies demonstrate eses to explain age-related cognitive and affective
age-related increases in NDI across tracts in child- impairments (Filley, 2005). These theoretical
hood and adolescences, while ODI remains rela- models suggest that disconnection leads to dys-
tively stable in this period (Mah et al., 2017). function, because WM tracts represent structural
Current findings suggest that during the last connections of the functional networks recruited
decades of life, FA decreases, whereas MD and during task performance, reductions in their integ-
RD increase, indicative of a decrease of WM rity are correlated with declines in cognitive skills
microstructural integrity with age. Additionally, (Salat, 2011; Sullivan and Pfefferbaum, 2006).
there appear to be regional differences in the rate It is worth noting that in parallel with these
and magnitude of WM changes. Initial studies changes in structural connectivity, functional
reported larger age-related decreases in FA in fron- network reorganizations also occur (see below).
tal regions (Kiely et al., 2022; Salat et al., 2005), Although the precise link is not fully understood,
though subsequent investigations suggested that structural and functional connectivity (FC) are
the anterior-posterior gradient may be an over- related (Hermundstad et al., 2013), and this rela-
simplification. For example, Davis et al. (2009) tionship appears to become less cohesive with age
found that WM tracts traversing the frontal cortex (Betzel et al., 2014). In addition, the development
exhibited a monotonic age-related decrease in FA of structural brain networks has been character-
that was stable across the frontal lobe boundary. ized by using different GM-related anatomical
Furthermore, some posterior WM regions exhib- properties such as CTh. The cortical development
ited larger age-related declines in FA compared to of GM structural networks is also strongly corre-
frontal regions (Salat et al., 2005). lated with (1) the acquisition of high-level cogni-
In addition, other researchers have suggested tive skills (Nie et al., 2014); (2) changes in cortical
the existence of a superior-inferior gradient, fiber density in early postnatal stages (Li et al.,
in which superior WM is more susceptible to 2015); and (3) decreased centrality of subcortical
age-related declines relative to inferior regions regions during the FC development in adolescence
(Zahr et al., 2009). Altogether, the diversity of (Sato et al., 2015).
Aging Brain Changes across the Lifespan 35

Lifespan Changes in Functional MRI the first years of life, there is a strong widespread
organization of networks which starts with segre-
Functional MRI (fMRI; Matthews and Jezzard, gation processes followed by a continuous increase
2004) is sensitive to dynamic changes in the in integration (Stevens, 2016); (2) from adoles-
blood (i.e., hemodynamics) and can be used to cence to early adulthood, a refinement of exist-
examine neuronal activity. In its more traditional ing networks in the brain occurs (i.e., integration
application, fMRI has been used to identify areas increases until about the 4th decade); and (3) the
of increased or decreased neuronal activity during middle age period is associated with an inversion
the performance of a task (i.e., task-based fMRI; of the functional brain trajectories and a decrease
Logothetis, 2008). Nevertheless, fMRI is increas- in segregation (Betzel et al., 2014), in conjunc-
ingly being used to explore how functional brain tion with a large-scale reorganization of between
networks that can be identified when individuals network connections. Broadly, the rs-fMR stud-
are not engaged in a task, the so-called resting- ies about brain maturity have reported consist-
state networks (RSN), operate and influence ent changes in the human functional brain over
cognition (Bressler and Menon, 2010; Smith development (Fransson et al., 2007). Analyzing
et al., 2009). data from people aged 7–30 years, Dosenbach
et al., (2010) suggested that the functional matu-
ration growth curve follows a non-linear asymp-
Functional Connectivity Changes totic shape, characterized by the weakening of
Assessed By rs-fMRI short-range functional connections between the
adult brain’s main functional networks. During
Rs-fMRI is used to identify spontaneous fluctua- development, connectivity increases and become
tion in activity among brain regions, which offers more coordinated, particularly along the anterior-
insights into intrinsic brain organization posterior axis (Dosenbach et al., 2010; Grayson
(Bijsterbosch et al., 2017). In the last decades, and Fair, 2017). Furthermore, it has been sug-
the increasing interest in analyzing neural gested that brain networks tend to exhibit a
dynamics with this technique has given rise to positive-to-negative shift when anterior–posterior
the understanding that the human brain is com- and posterior–subcortical nodes are evaluated,
posed of a set of complex functional RSNs whereas anterior-subcortical associations tend to
(Bressler and Menon, 2010) that are continu- involve positive correlations that become less pos-
ously interacting in a dynamic way with each itive across the lifespan (Rosenberg et al., 2020).
other to modulate cognitive performance FC changes with advanced age (Ferreira and
(Bressler and Menon, 2010; Smith et al., 2009). Busatto, 2013). Since RSN represent known
Thus, the network-based approach has provided functional networks (i.e., regions that are known
valuable metrics to understand the functional to support cognitive functions), the study of the
brain changes and their cognitive and behavioral DMN has attracted much interest, owing to its crit-
associations during the lifespan. ical role in human brain function (Buckner, 2012).
Diverse methodological approaches to study FC The structural architecture of the DMN includes
with rs-fMRI have been proposed. One of the most core areas such as the precuneus/posterior cingu-
widely used techniques is independent component late cortex, the medial frontal cortex, and other
analysis (ICA), based on a data-driven approach regions, such as the parietal, angular gyri, and the
(i.e., non-required predefined regions) that identi- hippocampus (Buckner et al., 2008). This cohe-
fies regions with similar patterns of synchronized sive brain network (Greicius et al., 2003) shows
activity (i.e., brain network; Nickerson et al., decreased activity during the performance of a
2017). Using the approach, several main RSNs wide range of tasks (Alves et al., 2019), while is
have been consistently identified across studies typically active during periods of rest or introspec-
(Moussa et al., 2012), including a visual process- tion (Snyder and Raichle, 2012). Convergent find-
ing network, the default-mode network (DMN), ings further demonstrate that the DMN is fairly
and cerebellum, sensorimotor, auditory, executive established from five years of age (Cao et al., 2014)
control, and frontoparietal networks (Damoiseaux and connectivity within DMN regions decreases
et al., 2006; Smith et al., 2009). Another increas- with increasing age (Andrews-Hanna et al., 2007),
ingly popular approach for the study of brain con- showing deactivation deficits. The most important
nectivity is graph theory, a set of mathematical age-related effect seems to entail FC disruption
tools to characterize topology of networks, both between the medial frontal and posterior midline
on a global and local level (Sporns, 2018). structures (Andrews-Hanna et al., 2007). Further,
Edde et al., (2021) recently proposed that FC between cortical and subcortical nodes of the
development of FC changes can be separated into DMN, such as the hippocampus, also changes
three main phases after birth according: (1) during with advanced age (Salami et al., 2014).
36 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

The above-mentioned age-related antero- pos- predicting significant reorganization in multiple


terior alterations within the DMN are consistent brain regions from childhood to adulthood and
with the hypothesis that among older adults, long- going further to the maturational perspective of
range FC is decreased (Andrews-Hanna et al., neural development, which would predict a shift
2007; Cao et al., 2014; Ferreira and Busatto, 2013; from minimal/no-activation to activation, or pro-
Vidal-Piñeiro et al., 2014), while short-range FC gressive age-related activity increases in adults
increases (Cao et al., 2014; Sala-Llonch et al., compared to children (Joseph et al., 2011). One
2014). Notably, an adequate balance between of the processes involved in the correct matura-
long- and short-distance connectivity is rele- tion trajectories during childhood and adolescence
vant for preserved cognition among older adults is the balance of functional networks integration
(Andrews-Hanna et al., 2007; Sala-Llonch et al., and segregation. Importantly, functional networks
2014; Vidal-Piñeiro et al., 2014), and a loss in this interaction throughout development is mediated
balance might boost between network connec- by functional (i.e., highly connected brain areas)
tivity, leading to a less segregated brain (Spreng which take a prolonged maturation course result-
et al., 2016). DMN regions are also critically vul- ing on the regulation of neural activity (Cao et al.,
nerable to aging, with annual rates of decline in 2014). Findings in this field, combining rs-fMRI
cortical volume in DMN areas being higher than and task-based fMRI data, have pointed out that
for most other cortical regions (Fjell et al., 2014a). centrality (i.e., a measure that reveal internetworks
This specific vulnerability in the cortical sys- interactions; Hwang et al., 2013) of cerebellum
tems constituting the DMN was hypothesized by and cingulo-opercular components are key factors
Buckner et al. (2009) to explain the predilection of for the normal acquisition of executive functions
these areas to amyloid-β deposition. One specula- (Kolskår et al., 2018).
tion is that DMN areas are critically vulnerable to In aging, task-based fMRI studies compar-
subtle age-related lesions and pathology accumu- ing brain activity patterns of older adults with
lating through life because these regions are char- those observed among younger counterparts
acterized by a high degree of life-long plasticity have revealed differences at the level of intensity
(Rapoport and Nelson, 2011). (hypo- or hyper- activation) as well as at the level
of the topographical distribution of the affected
areas (Grady, 2008, 2012). The main functional
changes in aging could be summarized in:
Activity Pattern Changes Assessed by (1) hyperactivation in the frontal and parietal
Task-Based fMRI regions, (2) hyperactivation of the DMN during
task demands, and (3) hypoactivation of the medial
Moreover, fMRI acquisitions can provide infor- temporal lobe. One the one hand, decreased brain
mation about the spatial patterns of brain regions activity has typically been interpreted as a reflec-
that activate and deactivate in synchrony under tion of cognitive deficits in older adults and has
different task conditions by subtracting the been linked to structural changes (Nyberg et al.,
BOLD signal between these conditions (Buch­ 2010). On the other hand, increased brain func-
binder, 2016). Task-based fMRI is acquired tional values have been understood as a compen-
while a subject is inside the scanner and satory mechanism (Cabeza, 2002; Reuter-Lorenz
instructed to perform a goal-directed task, afford- and Park, 2014; see below). Compensation is a
ing an identification of areas involved in the task complex phenomenon that states that older adults
performance and the corresponding scores. show more activity in a brain region than younger
A central question in cognitive neuroscience peers whilst they perform a task at the same level
is the characterization of putative developmental as younger adults (Cabeza, 2002; Cabeza and
changes in brain functioning underlying the acqui- Dennis, 2013), or when this increased activity
sition of skills and higher cognitive functions is positively correlated to performance in older
(Johnson, 2001). Most published studies com- adults, but not younger adults (Reuter-Lorenz
pare groups of children vs. adults, stressing that et al., 2000). Thus, the term compensation is con-
fMRI can be used to distinguish different kinds of ditioned by the fact that activity increases would
involvement among brain regions that change over be directly associated with improvements in
development (Booth et al., 2003; Gaillard et al., task performance, and this is not always accom-
2003; Luna et al., 2001). Studies using fMRI have plished. Some investigations have suggested that
shown that both progressive age-related devel- these over-recruitments, typically identified as
opmental changes (i.e., specialization increases) more activity in frontal regions (Cabeza, 2002;
and regressive changes (i.e., specialization Park and Reuter-Lorenz, 2009) are not invariably
decreases) occur (Joseph et al., 2011). This idea linked to better cognitive performance (Rypma
concurs with the interactive specialization theory et al., 2007). In these cases, increased activations
Aging Brain Changes across the Lifespan 37

have been conceived as attempted or unsuc- Despite the results above including GWAS,
cessful compensatory mechanisms (Cabeza and and those using other approaches such as the
Dennis, 2013) or dedifferentiation (Carp et al., epigenome-wide association study (EWAS) based
2011). Finally, it should be noted that within the on characterizing differences in DNA methyla-
fMRI framework, as investigated longitudinally, tion status (Unnikrishnan et al., 2019), it should
increases in brain activity instead of reflecting be noted that when considering brain and cogni-
compensatory mechanisms as described in cross- tive changes in advanced age, the most investi-
sectional approaches (Cabeza, 2002; Reuter- gated genetic variation has been that of the APOE.
Lorenz and Park, 2014), might underlie cognitive APOE (located on chromosome 19q13.2) is a pol-
decline (Pudas et al., 2017; Vaqué-Alcázar et al., ymorphic gene resulting in three different alleles
2020). Maintaining brain health across the lifes- (ε2, ε3, and ε4). The APOE ε4 isoform is the strong-
pan depends on a variety of factors, including est genetic risk factor for sporadic late-onset AD
genetic and lifestyle components, as well as (Koutsodendris et al., 2021; Serrano-Pozo et al.,
their interaction, which predicts substantial vari- 2021). In this regard, it has been shown that car-
ability in developmental and aging trajectories rying an APOE ε4 allele impacts non-pathological
(Valenzuela and Sachdev, 2006). brains (Flowers and Rebeck, 2020). From early
studies, it has been proved that APOE ε4 isoform
negatively influences cognitive status (Bartrés-Faz
et al., 2001) and cognitive decline trajectories in
Genetic Effects on Brain Change healthy aging (Riaz et al., 2021; Yaffe et al., 2009).
Trajectories These effects may be derived from the impact of
this genetic isoform on different biological path-
Genetic factors are important in determining corti- ways and mechanisms (Parhizkar and Holtzman,
cal architecture (Lenroot et al., 2009) and there 2022; Serrano-Pozo et al., 2021), whereas carry-
are common genetic variants that affect age- ing the APOE ε2 isoform (i.e., APOE ε3/ε2 geno-
related brain changes across the lifespan. Further, type) appears to be protective (Serrano-Pozo et al.,
longitudinal cohort studies have indicated that 2021). From a neuroimaging point of view, APOE
genetic differences between individuals have a ε4 isoform is related to multimodal changes
role in interindividual variability in age-related (Cai et al., 2017): reduced glucose metabolism
cognitive changes, although they may be impor- from midlife in AD-prone areas (Paranjpe et al.,
tant in establishing performance levels and less in 2019), higher medial temporal lobe atrophy, dif-
lifespan trajectories (Nyberg and Pudas, 2019). ferential brain activity during cognitive demands
Although the study of genetic influences on brain (Bartrés-Faz et al., 2008), and GM network abnor-
structure and function supports different methodo- malities in those APOE ε4/ε4 even in the absence
logical approaches, from classical twin studies to of amyloid-β deposition (Cacciaglia et al., 2020).
the assessment of specific variations or polymor- From a lifespan perspective, there has been a
phisms. Regarding the latest perspective, an debate as regards the putative antagonistic pleio-
approach that has recently proliferated is the tropic effects of APOE ε4 on brain and cognition,
genome-wide association study (GWAS), which is prompted by some evidence indicating that ε4 car-
based on examining associations between DNA riers would have better cognitive status at younger
phenotypes variations (i.e., single-nucleotide pol- ages in some cognitive domains (Marioni et al.,
ymorphisms; SNPs) and traits or diseases 2015). However, recent empirical and metanalytic
(Manolio, 2010). Although not the focus of this evidence assessing cognitive and brain measures
chapter, it seems suitable to highlight a recent do not support the antagonistic hypothesis (Corder
publication (Brouwer et al., 2022) that analyzed et al., 2022), and rather subtle abnormalities in
longitudinal data from 15,640 individuals using a hippocampus and cortical measures have already
GWAS approach. Broadly, this study showed that been detected in infants and even prenatal MRI
GPR139, DACH1, and APOE genes are associ- findings (Chang et al., 2016). These results indi-
ated with metabolic processes, CDH8 with cogni- cating that the effects of APOE on brain and cog-
tive function, GPR139, SORCS2, and CDH8 with nition in advanced age are partially rooted during
psychiatric traits, and NECTIN2 and APOE with neurodevelopment do not imply that the poten-
AD (Brouwer et al., 2022). Some of the identified tially deleterious impact of this genetic variant on
genetic variants for structural brain changes were brain health amongst older adults is immutable. In
stable throughout life (i.e., independent of age), this regard, for example, another series of investi-
suggesting that these genetic variants are equally gations suggest that APOE ε4 carriers might ben-
crucial for both early brain development and neu- efit more from preventive lifestyle exposures such
rodegenerative processes in the rates of brain as higher cognitive (Ma et al., 2022) or physical
changes. (Perez-Lasierra et al., 2021) activities.
38 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Environmental Effects and Models notable brain pathological changes (Stern and
of Brain Aging Barulli, 2019). These observations, together with
longitudinal population-based investigations indi-
There is ample evidence for the influence of life- cating that engaging in complex mental activi-
style factors on cognition and brain health, par- ties during lifecourse is related to a significant
ticularly in advancing age, suggesting that decrease in the prevalence of dementia (Valenzuela
age-related impact can be viewed in the context of and Sachdev, 2006), resulted in the foundations of
balance between risk and protective elements the cognitive reserve (CR) model, a theoretical
(Mattson and Magnus, 2006). Thus, the different concept proposed to account for the frequent dis-
interactions with the environment have the poten- crepancy between a person’s underlying level of
tiality to move an individual into a more positive age-related brain changes (or brain pathology) and
(or negative) aging trajectory (Lindenberger, the observed cognitive profile that is expected to
2014). Within this context, a growing amount of result of that damage (Stern, 2009). It is relevant
evidence (Mintzer et al., 2019) highlights the to highlight that there are differences across indi-
importance of sustaining healthy habits such as viduals because CR is dynamic and influenced
engaging in cognitively stimulating activities, by the interaction between innate characteristics
social interactions, and good dietary patterns such (genome) and lifetime exposures (Barulli and
as the Mediterranean diet (Guasch-Ferré et al., Stern, 2013; Stern et al., 20). Because there is exten-
2017), adequate and good sleep quality (Leng sive epidemiological evidence for the independent
et al., 2017), weekly moderate/vigorous exercise, association of different lifetime exposures with
not smoking (Yaffe et al., 2009), and engaging in more successful aging (Lövdén et al., 2005), CR is
behavior to maintain mental well-being (Marchant typically measured via a set of potential “proxies”
et al., 2020). including educational attainment, life-time occu-
Considering this view from a lifespan per- pation or estimated premorbid intelligence among
spective appears to be particularly relevant. For others. Contrarily, there are factors associated with
example, the combination of main modifiable an increased vulnerability to developing dementia,
risk and protective factors for dementia seems to such as elevated levels of anxiety and stress or a
have a different weight depending on when, dur- high burden of repetitive negative thinking, a pro-
ing the life course, individuals are exposed (i.e., cess presented across mental diseases that encom-
early, middle, or advanced age; Livingston et al., passes worry and rumination (Schlosser et al.,
2020), and they influence markers of biological 2020). There are different potential mechanisms
brain aging as well (see below). In addition, con- underlying the positive effects on clinical manifes-
verging evidence indicates that early factors (i.e., tations and cognition predicted by the CR theory,
birth weight, polygenetic risk scores, early life mainly functional brain compensatory reorganiza-
sleep patterns; Lokhandwala and Spencer, 2022) tions occurring in the face of pathology to sustain
or those already measurable at neurodevelopmen- cognition (Lee et al., 2019; Stern, 2009). However,
tal stages (i.e., general cognitive ability) contribute recent evidence does not support a consistent asso-
significantly to explain subjects brain differences ciation between CR estimates and longitudinal
across the whole life course, including middle and cognitive changes in healthy aging performance
older ages (Walhovd et al., 2016, 2022). (Vaqué-Alcázar et al., 2020; Wilson et al., 2019).
Nowadays, there are different concepts and Similarly, recent longitudinal findings (based on
theoretical models that roughly directly incor- 4,422 structural MRI observations) suggested that
porate the relevance of modifiable factors to despite GM displaying marked age-related atrophy
explain interindividual differences in brain and in regions such as the cortex and hippocampus,
cognition across the lifespan (Cabeza et al., 2018; years of formal education (i.e., CR) did not influ-
Reuter-Lorenz and Park, 2014; Stern et al., 2020). ence rates of change in this brain structure meas-
Concepts such as “reserve,” “resilience,” “resist- ures (Nyberg et al., 2021), which are more strongly
ance,” “neuroprotection,” or “brain maintenance,” precited by estimations of global cognitive ability
are often used by researchers working in this field (i.e., those with higher general cognitive ability
(Bartrés-Faz et al., 2020) and efforts to develop a experiencing less cortical thinning across the lifes-
framework of operational definitions as investiga- pan; Walhovd et al., 2022).
tions both in human and non-human species to The brain maintenance (BM) model highlights
understand the underlying biological mechanisms that across the life course, and in particular in
of the observed effects have been put in place advancing age, interindividual differences in brain
(Arenaza-Urquijo and Vemuri, 2018; Pascual- and cognition can be partially accounted for by
Leone and Bartres-Faz, 2021). the fact that some people show a relative lack of
Early influential findings indicated that a por- age-related brain changes in neurochemical, struc-
tion of individuals with advanced age can sustain tural, and functional level, allowing performance
Aging Brain Changes across the Lifespan 39

stability by the preservation of neural resources in atrophy rates and/or cognitive decline associ-
(Nyberg and Pudas, 2019). BM concept is consist- ated with age due to the capacity of these systems
ent with the notion that during late life, the neu- to identify groups or subtypes.
ral resources can remain preserved through the
action of protective mechanisms of cellular repair
(Cabeza et al., 2018) and may overlap to a large
degree with mechanisms of brain plasticity in SUMMARY AND CONCLUSIONS
adulthood (Lövdén et al., 2010).
Summarizing, the current evidence focused on
determining brain characteristics that change
Future Perspectives throughout life has shown that this organ experi-
ences numerous modifications over time, being
All of this leads to highlight the importance of
both the first and last decades of life those periods
developing new methodologies to apprehend the
with more abrupt transformations. Processes of
plastic changes experienced by our brain along
maturation and degeneration are in line with the
the life cycle become necessary. For example,
development of cognitive and sensory abilities
the combination of MRI and Non-Invasive Brain
throughout life, as well as their deterioration.
Stimulation (NIBS) suppose an innovative meth-
Interestingly, the changes associated with aging
odology, which allows interrogating and charac-
seem to mimic, to a certain extent, symmetric
terizing in a controlled manner the brain function.
development, and growth processes. In other
Specifically, combining NIBS and fMRI results
words, there is an inversion of developmental
in a powerful approach able to provide novel
processes during aging, which is in accordance
experimental data on the putative neurophysio-
with the developmental models defending that the
logical mechanisms underlying inter-individual
latest matured regions are the first to deteriorate.
differences and may help reformulate the theo-
Therefore, the brain undergoes numerous plastic
retical models proposed within this field by
changes throughout life, which are the result of
exploring functional mechanisms sustaining dif-
the complex interaction between environmental
ferent levels of cognition and brain properties,
factors and genetic determinants, giving rise to a
such as plasticity (Abellaneda-Pérez et al., 2019).
great inter-individual heterogeneity especially
Moreover, the combination of cross-sectional
relevant in aging.
and longitudinal designs to investigate the effect
Overall, the available literature provides evi-
of age on brain changes across the lifespan to
dence that the brain experiences several changes at
identify multidimensional systems-vulnerability
distinct levels (i.e., structural, functional, and met-
models (Walhovd et al., 2014) have already gained
abolic) which can be assessed by MRI. However,
importance in recent years as an optimal solution
we must not ignore that these macroscopic meas-
overcoming the limitations of cross-sectional clas-
ures are reflecting microscopic changes in our
sical research comparing younger versus older
brain (such as changes in vesicle trafficking, mem-
adults (Fjell et al., 2014a, 2014b). Thereby, neuro-
brane receptors expression, immune system reac-
science has experienced unprecedented growth in
tions, glial composition, neural death, etc.). In this
the accessibility of large-scale data sets including
regard, future research has the challenge of com-
massive genetic data (Tibon et al., 2022).
bining these techniques with more sophisticated
Thus, the new technological research frame-
biological and genetic markers to unravel those
work is mainly focused on multicentric consor-
specific factors conferring resilience/vulnerability
tia and large-scale multimodal initiatives such as
to brain disorders. In conclusion, studying these
the UK biobank (Sudlow et al., 2015), Lifebrain
elements and their neural underpinnings becomes
(Carver et al., 2022), the ENIGMA Consortium
vital to improve the quality of life of anyone by
combining brain imaging with genetic data
unrevealing the crucial determinants that could
(Thompson et al., 2022), integrating biomedi-
lead us to develop, promote, and maintain brain
cal databases and multilevel research resources.
health throughout the lifespan.
In this light, the use of artificial intelligence and
complex computational models has gained inter-
est in recent years (Pillow and Sahani, 2019). For
instance, machine learning methods enable neu-
roscientists to discover statistical patterns in large REFERENCES
data sets (Vu et al., 2018). These approaches bring
analyses to the level of individual subjects, which Abellaneda-Pérez, K., Vaqué-Alcázar, L., Solé-
is extremely useful to manage the previously Padullés, C., & Bartrés-Faz, D. (2019). Combining
described extensive interindividual heterogeneity non-invasive brain stimulation with functional
40 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

magnetic resonance imaging to investigate the age-related differences in cognition. Neuroscience,


neural substrates of cognitive aging. Journal of 276, 187–205.
Neuroscience Research. doi: 10.1002/jnr.24514 Betzel, R. F., Byrge, L., He, Y., Goñi, J., Zuo, X. N., &
Alves, P. N., Foulon, C., Karolis, V., Bzdok, D., Mar- Sporns, O. (2014). Changes in structural and func-
gulies, D. S., Volle, E., & Schotten, M. T. De. tional connectivity among resting-state networks
(2019). An improved neuroanatomical model of across the human lifespan. NeuroImage, 102(2),
the default-mode network reconciles previous 345–357.
neuroimaging and neuropathological findings. Bijsterbosch, J., Smith, S. M., & Beckmann, C. (2017).
Communications Biology. doi: 10.1038/s42003- An introduction to resting state fMRI functional
019-0611-3 connectivity. New York: Oxford University Press.
Andrews-Hanna, J. R., Snyder, A. Z., Vincent, J. L., Booth, J. R., Burman, D. D., Meyer, J. R., Lei, Z.,
Lustig, C., Head, D., Raichle, M. E. E., & Buckner, Trommer, B. L., Davenport, N. D., … Mesulam, M. M.
R. L. (2007). Disruption of large-scale brain (2003). Neural development of selective attention
systems in advanced aging. Neuron, 56(5), and response inhibition. NeuroImage, 20(2),
924–935. 737–751.
Arenaza-Urquijo, E. M., & Vemuri, P. (2018). Resist- Bouhrara, M., Kim, R. W., Khattar, N., Qian, W.,
ance vs resilience to Alzheimer disease. Neurology, Bergeron, C. M., Melvin, D., … Spencer, R. G.
90(15), 695–703. (2021). Age-related estimates of aggregate g-ratio
Armstrong, N. M., An, Y., Beason-Held, L., Doshi, J., of white matter structures assessed using quanti-
Erus, G., Ferrucci, L., … Resnick, S. M. (2019). tative magnetic resonance neuroimaging. Human
Predictors of neurodegeneration differ between Brain Mapping, 42(8), 2362–2373.
cognitively normal and subsequently impaired Bressler, S. L., & Menon, V. (2010). Large-scale brain
older adults. Neurobiology of Aging, 75, networks in cognition: emerging methods and
178–186. principles. Trends in Cognitive Sciences, 14(6),
Assaf, Y., & Pasternak, O. (2008). Diffusion tensor 277–290.
imaging (DTI)-based white matter mapping in Brickman, A. M., Meier, I. B., Korgaonkar, M. S.,
brain research: A review. Journal of Molecular Provenzano, F. A., Grieve, S. M., Siedlecki, K. L., …
Neuroscience, 34(1), 51–61. Zimmerman, M. E. (2012). Testing the white
Bartrés-Faz, D., Clemente, L. C., Junqué, C., Valveny, matter retrogenesis hypothesis of cognitive aging.
N., López-Alomar, A., Sánchez-Aldeguer, J., … Neurobiology of Aging, 33(8), 1699–1715.
Moral, P. (2001). APOE and APOC1 genetic poly- Brouwer, R. M., Klein, M., Grasby, K. L., Schnack, H. G.,
morphisms in age-associated memory impairment. Jahanshad, N., Teeuw, J., … Hulshoff Pol, H. E.
Neurogenetics, 3(4), 215–219. (2022). Genetic variants associated with longitudi-
Bartrés-Faz, D., Arenaza-Urquijo, E., Ewers, M., nal changes in brain structure across the lifespan.
Belleville, S., Chételat, G., Franzmeier, N., … Nature Neuroscience, 25(4), 421–432.
Vemuri, P. (2020). Theoretical frameworks and Buchbinder, B. R. (2016). Functional magnetic reso-
approaches used within the reserve, resilience and nance imaging. In J. C. Masdeu & R. Gilberto
protective factors professional interest area of the González (Eds.), Handbook of clinical neurology:
Alzheimer’s Association International Society to Neuroimaging Part 1 (Vol. 135, pp. 61–92).
Advance Alzheimer’s Research and Treatment. Alz- Amsterdam: Elsevier.
heimer’s and Dementia: Diagnosis, Assessment and Buckner, R. L. (2004). Memory and executive func-
Disease Monitoring, 12(1), 2–7. tion in aging and AD: Multiple factors that cause
Bartrés-Faz, D., Serra-Grabulosa, J. M., Sun, F. T., decline and reserve factors that compensate.
Solé-Padullés, C., Rami, L., Molinuevo, J. L., … Neuron, 44(1), 195–208.
D’Esposito, M. (2008). Functional connectivity of Buckner, R. L. (2012). The serendipitous discovery of
the hippocampus in elderly with mild memory the brain’s default network. NeuroImage, 62(2),
dysfunction carrying the APOE ε4 allele. Neurobi- 1137–1145.
ology of Aging, 29(11), 1644–1653. Buckner, R. L., Andrews-Hanna, J. R., & Schacter,
Barulli, D., & Stern, Y. (2013). Efficiency, capacity, D. L. (2008). The brain’s default network: Anat-
compensation, maintenance, plasticity: Emerging omy, function, and relevance to disease. Annals of
concepts in cognitive reserve. Trends in Cognitive the New York Academy of Sciences. doi: 10.1196/
Sciences, 17(10), 502–509. annals.1440.011
Bells, S., Lefebvre, J., Longoni, G., Narayanan, S., Buckner, R. L., Sepulcre, J., Talukdar, T., Krienen,
Arnold, D. L., Yeh, E. A., & Mabbott, D. J. (2019). F. M., Liu, H., Hedden, T., … Johnson, K. A. (2009).
White matter plasticity and maturation in human Cortical hubs revealed by intrinsic functional con-
cognition. Glia, 67(11), 2020–2037. nectivity: Mapping, assessment of stability, and
Bennett, I. J., & Madden, D. J. (2014). Disconnected relation to Alzheimer’s disease. Journal of Neuro-
aging: Cerebral white matter integrity and science, 29(6), 1860–1873.
Aging Brain Changes across the Lifespan 41

Buyanova, I. S., & Arsalidou, M. (2021). Cerebral dimensions of human brain cortical ageing in rela-
white matter myelination and relations to age, tion to cognitive decline across the eighth decade
gender, and cognition: A selective review. Fron- of life. Molecular Psychiatry, 26(6), 2651–2662.
tiers in Human Neuroscience, 15. doi: 10.3389/ Damoiseaux, J. S., Rombouts, S. A. R. B., Barkhof, F.,
fnhum.2021.662031 Scheltens, P., Stam, C. J., Smith, S. M., & Beck-
Cabeza, R. (2002). Hemispheric asymmetry reduction mann, C. F. (2006). Consistent resting-state net-
in older adults: The HAROLD model. Psychology works across healthy subjects. Proceedings of the
and Aging, 17(1), 85–100. National Academy of Sciences of the United States
Cabeza, R., Albert, M., Belleville, S., Craik, F. I. M., of America, 103(37), 13848–13853.
Duarte, A., Grady, C. L., … Rajah, M. N. (2018). Davis, S. W., Dennis, N. A., Buchler, N. G., White, L. E.,
Maintenance, reserve and compensation: The Madden, D. J., & Cabeza, R. (2009). Assessing the
cognitive neuroscience of healthy ageing. Nature effects of age on long white matter tracts using
Reviews Neuroscience, 19. 701–710. diffusion tensor tractography. NeuroImage, 46(2),
Cacciaglia, R., Cacciaglia, R., Cacciaglia, R., 530–541.
Molinuevo, J. L., Molinuevo, J. L., Molinuevo, J. L., De Leeuw, F. E., De Groot, J. C., Oudkerk, M., Witte-
… Gispert, J. D. (2020). APOE-e4 shapes the cer- man, J. C. M., Hofman, A., Van Gijn, J., & Breteler,
ebral organization in cognitively intact individuals M. M. B. (2002). Hypertension and cerebral white
as reflected by structural gray matter networks. matter lesions in a prospective cohort study. Brain,
Cerebral Cortex, 30(7), 4110–4120. 125(4), 765–772.
Cai, S., Jiang, Y., Wang, Y., Wu, X., Ren, J., Lee, M. Dima, D., Modabbernia, A., Papachristou, E., Doucet,
S., … Huang, L. (2017). Modulation on brain gray G. E., Agartz, I., Aghajani, M., … Frangou, S.
matter activity and white matter integrity by APOE (2022). Subcortical volumes across the lifespan:
ε4 risk gene in cognitively intact elderly: A multi- Data from 18,605 healthy individuals aged 3–90
modal neuroimaging study. Behavioural Brain years. Human Brain Mapping, 43(1), 452–469.
Research, 322, 100–109. Dosenbach, N. U. F., Nardos, B., Cohen, A. L., Fair, D. A.,
Cao, M., Wang, J. H., Dai, Z. J., Cao, X. Y., Jiang, L. Power, J. D., Church, J. A., … Schlaggar, B. L.
L., Fan, F. M., … He, Y. (2014). Topological organi- (2010). Prediction of individual brain maturity
zation of the human brain functional connectome using fMRI. Science, 329, 1358–1361.
across the lifespan. Developmental Cognitive Neu- Ducharme, S., Albaugh, M. D., Nguyen, T.-V.,
roscience, 7(16), 76–93. Hudziak, J. J., Mateos-Pérez, J. M., Labbe, A., …
Carp, J., Park, J., Polk, T. A., & Park, D. C. (2011). Age Karama, S. (2016). Trajectories of cortical thick-
differences in neural distinctiveness revealed by multi- ness maturation in normal brain development: The
voxel pattern analysis. NeuroImage, 56(2), 736–743. importance of quality control procedures for the
Carver, R. B., Fredheim, N. A. G., Mowinckel, A. M., brain development cooperative group HHS public
Ebmeier, K. P., Friedman, B. B., Rosness, T. A., … access. NeuroImage, 125, 267–279.
Budin-Ljøsne I. People’s interest in brain health Edde, M., Leroux, G., Altena, E., & Chanraud, S.
testing: Findings from an international, online (2021). Functional brain connectivity changes
cross-sectional survey. Frontiers in Public Health. across the human life span: From fetal develop-
doi: 10.3389/fpubh.2022.998302 ment to old age. Journal of Neuroscience Research,
Cedres, N., Ferreira, D., Nemy, M., Machado, A., 99(1), 236–262.
Pereira, J. B., Shams, S., … Westman, E. (2022). Fazekas, F., Schmidt, R., & Scheltens, P. (1998).
Association of cerebrovascular and Alzheimer dis- Pathophysiologic mechanisms in the development
ease biomarkers with cholinergic white matter of age-related white matter changes of the brain.
degeneration in cognitively unimpaired individu- Dementia and Geriatric Cognitive Disorders,
als. Neurology, 99(15), 1619–1629. 9(S1), 2–5.
Chang, L., Douet, V., Bloss, C., Lee, K., Pritchett, A., Ferreira, L. K., & Busatto, G. F. (2013). Resting-state
Jernigan, T. L., … Ernst, T. (2016). Gray matter functional connectivity in normal brain aging.
maturation and cognition in children with differ- Neuroscience and Biobehavioral Reviews, 37(3),
ent APOE ∈ genotypes. Neurology, 87(6), 384–400.
585–594. Filley, C. M. (2005). White matter and behavioral
Corder, E. H., Saunders, A. M., Risch, N. J., neurology. Annals of the New York Academy of
Strittmatter, W. J., Schmechel, D. E., Gaskell, P. C., Sciences, 1064, 162–183.
… Nyberg, L. H. (2022). The role of DNA methyla- Fjell, A M, McEvoy, L., Holland, D., Dale, A. M., &
tion in epigenetics of aging. NeuroImage, 12(1). Walhovd, K. B. (2013). Brain changes in older
doi: 10.1016/j.pharmthera.2018.11.001 adults at very low risk for Alzheimer’s disease.
Cox, S. R., Harris, M. A., Ritchie, S. J., Buchanan, Journal of Neuroscience, 33(19), 8237–8242.
C. R., Valdés Hernández, M. C., Corley, Fjell, Anders M., Grydeland, H., Wang, Y., Amlien, I.,
J., … Tucker-Drob, E. M. (2021). Three major Bartrés-Faz, D., Brandmaier, A. M., … Walhovd, K. B.
42 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

(2021). The genetic organization of longitudinal Grieve, S. M., Clark, C. R., Williams, L. M., Peduto, A. J.,
subcortical volumetric change is stable throughout & Gordon, E. (2005). Preservation of limbic and
the lifespan running title: Genetics of subcortical paralimbic structures in aging. Human Brain Map-
lifespan change. ELife, 10. doi: 10.7554/ ping, 25(4), 391–401.
eLife.66466 Guasch-Ferré, M., Salas-Salvadó, J., Ros, E., Estruch, R.,
Fjell, Anders M., McEvoy, L., Holland, D., Dale, A. M., Corella, D., Fitó, M., & Martínez-González, M. A.
& Walhovd, K. B. (2014). What is normal in normal (2017). The PREDIMED trial, Mediterranean diet
aging? Effects of aging, amyloid and Alzheimer’s and health outcomes: How strong is the evidence?
disease on the cerebral cortex and the hippocam- Nutrition, Metabolism and Cardiovascular
pus. Progress in Neurobiology, 117, 20–40. Diseases, 27(7), 624–632.
Fjell, Anders M., Westlye, L. T., Grydeland, H., Habes, M., Erus, G., Toledo, J. B., Bryan, N., Janowitz, D.,
Amlien, I., Espeseth, T., Reinvang, I., … Walhovd, Doshi, J., … Davatzikos, C. (2018). Regional tract-
K. B. (2014). Accelerating cortical thinning: Unique specific white matter hyperintensities are associated
to dementia or universal in aging? Cerebral Cortex, with patterns to aging-related brain atrophy via
24(4), 919–934. vascular risk factors, but also independently. Alzhei-
Flowers, S. A., & Rebeck, G. W. (2020). APOE in the mer’s and Dementia: Diagnosis, Assessment and
normal brain. Neurobiology of Disease, 136. doi: Disease Monitoring, 10, 278–284.
10.1016/j.nbd.2019.104724 Halaas, N. B., Henjum, K., Blennow, K., Dakhil, S.,
Frangou, S., Modabbernia, A., Williams, S. C. R., Idland, A. V., Nilsson, L. N. G., … Fjell, A. M.
Papachristou, E., Doucet, G. E., Agartz, I., … (2020). CSF sTREM2 and Tau work together in
Dima, D. (2022). Cortical thickness across the predicting increased temporal lobe atrophy in
lifespan: Data from 17,075 healthy individuals older adults. Cerebral Cortex, 30(4), 2295–2306.
aged 3–90 years. Human Brain Mapping, 43(1), Hasan, K. M., Mwangi, B., Cao, B., Keser, Z., Tusti-
431–451. son, N. J., Kochunov, P., … Soares, J. (2016).
Fransson, P., Skiöld, B., Horsch, S., Nordell, A., Blen- Entorhinal cortex thickness across the human
now, M., Lagercrantz, H., & Åden, U. (2007). lifespan. Journal of Neuroimaging, 26(3),
Resting-state networks in the infant brain. Pro- 278–282.
ceedings of the National Academy of Sciences of Hermundstad, A. M., Bassett, D. S., Brown, K. S.,
the United States of America, 104(39), Aminoff, E. M., Clewett, D., Freeman, S., …
15531–15536. Carlson, J. M. (2013). Structural foundations of
Gaillard, W. D., Sachs, B. C., Whitnah, J. R., Ahmad, Z., resting-state and task-based functional connectiv-
Balsamo, L. M., Petrella, J. R., … Grandin, C. B. ity in the human brain. Proceedings of the National
(2003). Developmental aspects of language pro- Academy of Sciences of the United States of
cessing: fMRI of verbal fluency in children and America, 110(15), 6169–6174.
adults. Human Brain Mapping, 18(3), 176–185. Hwang, K., Hallquist, M. N., & Luna, B. (2013). The
Gorbach, T., Pudas, S., Bartrés-Faz, D., Brandmaier, development of hub architecture in the human
A. M., Düzel, S., Henson, R. N., … Nyberg, L. functional brain network. Cerebral Cortex, 23(10),
(2020). Longitudinal association between hip- 2380–2393.
pocampus atrophy and episodic-memory decline Johnson, M. H. (2001). Functional brain development
in non-demented APOE ε4 carriers. Alzheimer’s in humans. Nature Reviews Neuroscience, 2,
and Dementia: Diagnosis, Assessment and 475–483.
Disease Monitoring, 12(1). doi: 10.1002/ Jones, D. K., Knösche, T. R., & Turner, R. (2013).
dad2.12110 White matter integrity, fiber count, and other fal-
Grady, C. L. (2008). Cognitive neuroscience of aging. lacies: The do’s and don’ts of diffusion MRI. Neu-
Annals of the New York Academy of Sciences, roImage, 73, 239–254.
1124, 127–144. Joseph, J. E., Gathers, A. D., & Bhatt, R. S. (2011).
Grady, C. L. (2012). Trends in Neurocognitive Progressive and regressive developmental changes
Aging. Nature Reviews Neuroscience, 123(3), in neural substrates for face processing: Testing
106–116. specific predictions of the interactive specialization
Grayson, D. S., & Fair, D. A. (2017). Development of account. Developmental Science, 14(2), 227–241.
large-scale functional networks from birth to Kiely, M., Triebswetter, C., Cortina, L. E., Gong, Z.,
adulthood: A guide to the neuroimaging literature. Alsameen, M. H., Spencer, R. G., & Bouhrara, M.
NeuroImage, 160, 15–31. (2022). Insights into human cerebral white matter
Greicius, M. D., Krasnow, B., Reiss, A. L., & Menon, V. maturation and degeneration across the adult
(2003). Functional connectivity in the resting lifespan. NeuroImage, 247. doi: 10.1016/j.
brain: A network analysis of the default mode neuroimage.2021.118727
hypothesis. Proceedings of the National Academy Kolskår, K. K., Alnæs, D., Kaufmann, T., Richard, G.,
of Sciences of the United States of America, Sanders, A. M., Ulrichsen, K. M., … Westlye, L. T.
100(1), 253–258. (2018). Key brain network nodes show differential
Aging Brain Changes across the Lifespan 43

cognitive relevance and developmental trajectories Lövdén, M., Ghisletta, P., & Lindenberger, U. (2005).
during childhood and adolescence. ENeuro, 5(4). Social participation attenuates decline in percep-
doi: 10.1523/ENEURO.0092-18.2018 tual speed in old and very old age. Psychology and
Koutsodendris, N., Nelson, M. R., Rao, A., & Huang, Y. Aging, 20(3), 423–434.
(2021). Apolipoprotein E and Alzheimer’s disease: Luna, B., Thulborn, K. R., Munoz, D. P., Merriam, E. P.,
Findings, hypotheses, and potential mechanisms. Garver, K. E., Minshew, N. J., … Sweeney, J. A.
Annual Review of Pathology: Mechanisms of Dis- (2001). Maturation of widely distributed brain
ease, 17, 73–99. function subserves cognitive development.
Krogsrud, S. K., Fjell, A. M., Tamnes, C. K., Gryde- NeuroImage, 13(5), 786–793.
land, H., Mork, L., Due-Tønnessen, P., … Walhovd, Lynch, K. M., Cabeen, R. P., Toga, A. W., & Clark, K. A.
K. B. (2016). Changes in white matter microstruc- (2020). Magnitude and timing of major white
ture in the developing brain: A longitudinal diffu- matter tract maturation from infancy through
sion tensor imaging study of children from 4 to 11 adolescence with NODDI. NeuroImage, 212. doi:
years of age. NeuroImage, 124, 473–486. 10.1016/j.neuroimage.2020.116672
Lamar, M. (2014). White matter microstructure in Ma, H., Zhou, T., Li, X., Maraganore, D., Heianza, Y.,
brain aging: Human and animal models. American & Qi, L. (2022). Early-life educational attainment,
Journal of Geriatric Psychiatry, 22(2), 99–101. APOE ε4 alleles, and incident dementia risk in late
Lebel, C., Treit, S., & Beaulieu, C. (2019). A review of life. GeroScience, 44(3), 1479–1488.
diffusion MRI of typical white matter development Mah, A., Geeraert, B., & Lebel, C. (2017). Detailing
from early childhood to young adulthood. NMR in neuroanatomical development in late childhood
Biomedicine, 32(4). doi: 10.1002/nbm.3778 and early adolescence using NODDI. PLoS One,
Lee, D. H., Lee, P., Seo, S. W., Roh, J. H., Oh, M., Oh, 12(8). doi: 10.1371/journal.pone.0182340
J. S., … Jeong, Y. (2019). Neural substrates of Manolio, T. A. (2010). Genomewide association stud-
cognitive reserve in Alzheimer’s disease spectrum ies and assessment of the risk of disease. New
and normal aging. NeuroImage, 186, 690–702. England Journal of Medicine, 363(2), 166–176.
Leng, Y., McEvoy, C. T., Allen, I. E., & Yaffe, K. Marchant, N. L., Lovland, L. R., Jones, R., Pichet Binette, A.,
(2017). Association of sleep-disordered breathing Gonneaud, J., Arenaza-Urquijo, E. M., … Villeneuve,
with cognitive function and risk of cognitive S. (2020). Repetitive negative thinking is associated
impairment: A systematic review and meta-analy- with amyloid, tau, and cognitive decline. Alzheimer’s
sis. JAMA Neurology, 74(10), 1237–1245. and Dementia, 16(7), 1054–1064.
Lenroot, R. K., Schmitt, J. E., Ordaz, S. J., Wallace, G. Marioni, R. E., Proust-Lima, C., Amieva, H., Brayne, C.,
L., Neale, M. C., Lerch, J. P., … Giedd, J. N. (2009). Matthews, F. E., Dartigues, J.-F., & Jacqmin-Gadda,
Differences in genetic and environmental influ- H. (2015). Social activity, cognitive decline and
ences on the human cerebral cortex associated dementia risk: A 20-year prospective cohort study.
with development during childhood and adoles- BMC Public Health, 15(1). doi: 10.1186/
cence. Human Brain Mapping, 30(1), 163–174. s12889-015-2426-6
Li, G., Liu, T., Ni, D., Lin, W., Gilmore, J. H., & Shen, D. Matthews, P. M., & Jezzard, P. (2004). Functional
(2015). Spatiotemporal patterns of cortical fiber magnetic resonance imaging. Handbook of Clini-
density in developing infants, and their relation- cal Neurology, 75, 6–12.
ship with cortical thickness. Human Brain Mapping, Mattson, M. P., & Magnus, T. (2006). Ageing and
36(12), 5183–5195. neuronal vulnerability. Nature Reviews Neurosci-
Lindenberger, U. (2014). Human cognitive aging: ence, 7(4), 278–294.
Corriger la fortune? Science, 346, 572–578. Mills, K. L., Goddings, A. L., Herting, M. M., Meuwese,
Livingston, G., Huntley, J., Sommerlad, A., Ames, D., R., Blakemore, S. J., Crone, E. A., … Tamnes, C. K.
Ballard, C., Banerjee, S., … Mukadam, N. (2020). (2016). Structural brain development between
Dementia prevention, intervention, and care: 2020 childhood and adulthood: Convergence across four
report of the Lancet commission. Lancet, 396, longitudinal samples. NeuroImage, 141, 273–281.
413–446. Mintzer, J., Donovan, K. A., Kindy, A. Z., Lock, S. L.,
Logothetis, N. K. (2008). What we can do and what Chura, L. R., & Barracca, N. (2019). Lifestyle
we cannot do with fMRI. Nature, 453, 869–878. choices and brain health. Frontiers in Medicine, 6.
Lokhandwala, S., & Spencer, R. M. C. (2022). Rela- doi: 10.3389/fmed.2019.00204
tions between sleep patterns early in life and Mishra, S., Blazey, T. M., Holtzman, D. M., Cruchaga,
brain development: A review. Developmental C., Su, Y., Morris, J. C., … Gordon, B. A. (2018).
Cognitive Neuroscience, 56. doi: 10.1016/j.dcn. Longitudinal brain imaging in preclinical Alzheimer
2022.101130 disease: Impact of APOE ∈4 genotype. Brain,
Lövdén, M., Bäckman, L., Lindenberger, U., Schaefer, S., 141(6), 1828–1839.
& Schmiedek, F. (2010). A theoretical framework Mori, S., & Barker, P. B. (1999). Diffusion magnetic
for the study of adult cognitive plasticity. Psycho- resonance imaging: Its principle and applications.
logical Bulletin, 136(4), 659–676. Anatomical Record, 257(3), 102–109.
44 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Moussa, M. N., Steen, M. R., Laurienti, P. J., & Haya- Pascual-Leone, A., & Bartres-Faz, D. (2021). Human
saka, S. (2012). Consistency of network modules brain resilience: A call to action. Annals of
in resting-state fMRI connectome data. PLoS One, Neurology, 90(3), 336–349.
7(8). doi: 10.1371/journal.pone.0044428 Perez-Lasierra, J. L., Casajús, J. A., Casasnovas, J.
Mukherjee, P., Berman, J. I., Chung, S. W., Hess, C. A., Arbones-Mainar, J. M., Lobo, A., Lobo, E., …
P., & Henry, R. G. (2008). Diffusion tensor MR Gonzalez-Agüero, A. (2021). Can physical activ-
imaging and fiber tractography: Theoretic under- ity reduce the risk of cognitive decline in apolipo-
pinnings. American Journal of Neuroradiology, protein e4 carriers? A systematic review.
29(4), 632–641. International Journal of Environmental Research
Narvacan, K., Treit, S., Camicioli, R., Martin, W., & and Public Health, 18(14). doi: 10.3390/
Beaulieu, C. (2017). Evolution of deep gray matter ijerph18147238
volume across the human lifespan. Human Brain Pfefferbaum, A., Rohlfing, T., Rosenbloom, M. J.,
Mapping, 38(8), 3771–3790. Chu, W., Colrain, I. M., & Sullivan, E. V. (2013).
Nickerson, L. D., Smith, S. M., Öngür, D., & Beck- Variation in longitudinal trajectories of regional
mann, C. F. (2017). Using dual regression to inves- brain volumes of healthy men and women (ages
tigate network shape and amplitude in functional 10 to 85 years) measured with atlas-based parcel-
connectivity analyses. Frontiers in Neuroscience, lation of MRI. NeuroImage, 65, 176–193.
11. doi: 10.3389/fnins.2017.00115 Pillow, J., & Sahani, M. (2019). Editorial overview:
Nie, J., Li, G., Wang, L., Shi, F., Lin, W., Gilmore, J. H., Machine learning, big data, and neuroscience.
& Shen, D. (2014). Longitudinal development of Current Opinion in Neurobiology, 55, iii–iv.
cortical thickness, folding, and fiber density net- Pomponio, R., Erus, G., Habes, M., Doshi, J., Srini-
works in the first 2 years of life. Human Brain vasan, D., Mamourian, E., … Davatzikos, C.
Mapping, 35(8), 3726–3737. (2020). Harmonization of large MRI datasets for
Norbom, L. B., Ferschmann, L., Parker, N., Agartz, I., the analysis of brain imaging patterns throughout
Andreassen, O. A., Paus, T., … Tamnes, C. K. the lifespan. NeuroImage, 208. doi: 10.1016/j.
(2021). New insights into the dynamic develop- neuroimage.2019.116450
ment of the cerebral cortex in childhood and Pudas, S., Josefsson, M., Rieckmann, A., & Nyberg, L.
adolescence: Integrating macro- and microstruc- (2018). Longitudinal evidence for increased func-
tural MRI findings. Progress in Neurobiology, 204. tional response in frontal cortex for older adults
doi: 10.1016/j.pneurobio.2021.102109 with hippocampal atrophy and memory decline.
Nyberg, L., Salami, A., Andersson, M., Eriksson, J., Cerebral Cortex, 28, 936–948.
Kalpouzos, G., Kauppi, K., … Nilsson, L.-G. (2010). Raji, C. A., Lopez, O. L., Kuller, L. H., Carmichael, O.
Longitudinal evidence for diminished frontal T., Longstreth, W. T., Gach, H. M., … Becker, J. T.
cortex function in aging. Proceedings of the (2012). White matter lesions and brain gray
National Academy of Sciences, 107(52), matter volume in cognitively normal elders.
22682–22686. Neurobiology of Aging, 33(4), E7–E16.
Nyberg, Lars, Magnussen, F., Lundquist, A., Baaré, Rapoport, S. I., & Nelson, P. T. (2011). Biomarkers and
W., Bartrés-Faz, D., Bertram, L., … Fjell, A. M. evolution in Alzheimer disease. Progress in
(2021). Educational attainment does not influence Neurobiology, 95(4), 510–513.
brain aging. Proceedings of the National Academy Raz, N., Lindenberger, U., Rodrigue, K. M., Kennedy,
of Sciences of the United States of America, K. M., Head, D., Williamson, A., … Acker, J. D.
118(18). doi: 10.1073/pnas.2101644118 (2005). Regional brain changes in aging healthy
Nyberg, L., & Pudas, S. (2019). Successful memory adults: General trends, individual differences and
aging. Annual Review of Psychology, 70(1), modifiers. Cerebral Cortex, 15(11), 1676–1689.
219–243. Reiter, K., Nielson, K. A., Durgerian, S., Woodard, J.
Paranjpe, M. D., Chen, X., Liu, M., Paranjpe, I., Leal, L., Smith, J. C., Seidenberg, M., … Rao, S. M.
J. P., Wang, R., … Zhou, Y. (2019). The effect of (2017). Five-year longitudinal brain volume change
ApoE ε4 on longitudinal brain region-specific glu- in healthy elders at genetic risk for Alzheimer’s
cose metabolism in patients with mild cognitive disease. Journal of Alzheimer’s Disease, 55(4),
impairment: A FDG-PET study. NeuroImage: 1363–1377.
Clinical, 22. doi: 10.1016/j.nicl.2019.101795 Reuter-Lorenz, P. A., Jonides, J., Smith, E. E., Hartley,
Parhizkar, S., & Holtzman, D. M. (2022). APOE medi- A., Miller, A., Marshuetz, C., & Koeppe, R. A.
ated neuroinflammation and neurodegeneration (2000). Age differences in the frontal lateralization
in Alzheimer’s disease. Seminars in Immunology. of verbal and spatial working memory revealed by
doi: 10.1016/j.smim.2022.101594 PET. Journal of Cognitive Neuroscience, 12(1),
Park, D. C., & Reuter-Lorenz, P. (2009). The adaptive 174–187.
brain: Aging and neurocognitive scaffolding. Reuter-Lorenz, P. A., & Park, D. C. (2014). How does
Annual Review of Psychology, 60(1), 173–196. it STAC up? Revisiting the scaffolding theory of
Aging Brain Changes across the Lifespan 45

aging and cognition. Neuropsychology Review, tensor imaging. Frontiers in Neuroscience, 7. doi:
24(3), 355–370. 10.3389/fnins.2013.00032
Riaz, M., Huq, A., Ryan, J., Orchard, S. G., Tiller, J., Sato, J. R., Salum, G. A., Gadelha, A., Vieira, G.,
Lockery, J., … Lacaze, P. (2021). Effect of APOE Zugman, A., Picon, F. A., … Bressan, R. A. (2015).
and a polygenic risk score on incident dementia Decreased centrality of subcortical regions during
and cognitive decline in a healthy older population. the transition to adolescence: A functional con-
Aging Cell, 20(6). doi: 10.1111/acel.13384 nectivity study. NeuroImage, 104, 44–51.
Rilling, J. K. (2014). Comparative primate neuroimag- Schlosser, M., Demnitz-King, H., Whitfield, T., Wirth,
ing: Insights into human brain evolution. Trends in M., & Marchant, N. L. (2020). Repetitive negative
Cognitive Sciences, 18(1), 46–55. thinking is associated with subjective cognitive
Rosenberg, B. M., Mennigen, E., Monti, M. M., & decline in older adults: A cross-sectional study.
Kaiser, R. H. (2020). Functional segregation of BMC Psychiatry, 20(1). doi: 10.1186/s12888-020-
human brain networks across the lifespan: An 02884-7
exploratory analysis of static and dynamic resting- Schmitt, J. E., Raznahan, A., Clasen, L. S., Wallace,
state functional connectivity, Frontiers in Neurosci- G. L., Pritikin, J. N., Lee, N. R., … Neale, M. C.
ence, 14. doi: 10.3389/fnins.2020.561594 (2019). The dynamic associations between cortical
Rypma, B., Eldreth, D. A., & Rebbechi, D. (2007). thickness and general intelligence are genetically
Age-related differences in activation-performance mediated. Cerebral Cortex, 29(11), 4743–4752.
relations in delayed-response tasks: A multiple Sele, S., Liem, F., Mérillat, S., & Jäncke, L. (2021). Age-
component analysis. Cortex, 43(1), 65–76. related decline in the brain: A longitudinal study on
Sala-llonch, R., Bartrés-faz, D., & Junqué, C. (2015). inter-individual variability of cortical thickness, area,
Reorganization of brain networks in aging: A volume, and cognition. NeuroImage, 240. doi:
review of functional connectivity studies. Fron- 10.1016/j.neuroimage.2021.118370
tiers in Psychology, 6. doi: 10.3389/fpsyg.2015. Serrano-Pozo, A., Das, S., & Hyman, B. T. (2021).
00663 APOE and Alzheimer’s disease: Advances in genet-
Sala-llonch, R., Junqué, C., Arenaza-urquijo, E. M., ics, pathophysiology, and therapeutic approaches.
Vidal-piñeiro, D., & Bartrés-faz, D. (2014). Neurobi- Lancet Neurology, 20(1), 68–80.
ology of aging changes in whole-brain functional Sexton, C. E., Walhovd, K., Storsve, A. B., Tamnes, C.
networks and memory performance in aging. K., Westlye, L. T., Johansen-Berg, H., & Fjell, A. M.
Neurobiology of Aging, 35(10), 2193–2202. (2014). Accelerated changes in white matter
Salami, A., Pudas, S., & Nyberg, L. (2014). Elevated hip- microstructure during ageing: A longitudinal diffu-
pocampal resting-state connectivity underlies defi- sion tensor imaging study. Journal of Neuroscience,
cient neurocognitive function in aging. Proceedings 34(46), 15425–15436.
of the National Academy of Sciences of the United Shaw, P., Kabani, N. J., Lerch, J. P., Eckstrand, K.,
States of America, 111(49) 17654–17659. Lenroot, R., Gogtay, N., … Wise, S. P. (2008).
Salat, D. H. (2004). Thinning of the cerebral cortex in Neurodevelopmental trajectories of the human
aging. Cerebral Cortex, 14(7), 721–730. cerebral cortex. Journal of Neuroscience, 28(14),
Salat, D. H., Tuch, D. S., Greve, D. N., Van Der Kouwe, 3586–3594.
A. J. W., Hevelone, N. D., Zaleta, A. K., … Dale, A. Slater, D. A., Melie-Garcia, L., Preisig, M., Kherif, F.,
M. (2005). Age-related alterations in white matter Lutti, A., & Draganski, B. (2019). Evolution of
microstructure measured by diffusion tensor imag- white matter tract microstructure across the
ing. Neurobiology of Aging, 26(8), 1215–1227. life span. Human Brain Mapping, 40(7),
Salat, D. H. (2011). The declining infrastructure of 2252–2268.
the aging brain. Brain Connectivity, 1(4), Smith, S. M., Fox, P. T., Miller, K. L., Glahn, D. C., Fox,
279–293. P. M., Mackay, C. E., … Beckmann, C. F. (2009).
Samara, A., Feng, K., Pivik, R. T., Jarratt, K. P., Badger, Correspondence of the brain’s functional architec-
T. M., & Ou, X. (2019). White matter microstruc- ture during activation and rest. Proceedings of the
ture correlates with memory performance in National Academy of Sciences of the United States
healthy children: A diffusion tensor imaging study. of America, 106(31), 13040–13045.
Journal of Neuroimaging, 29(2), 233–241. Snyder, A. Z., & Raichle, M. E. (2012). A brief history
Sang, F., Chen, Y., Chen, K., Dang, M., Gao, S., & of the resting state: The Washington University
Zhang, Z. (2021). Sex differences in cortical mor- perspective. NeuroImage, 62(2), 902–910.
phometry and white matter microstructure during Soldan, A., Pettigrew, C., Lu, Y., Wang, M.-C.,
brain aging and their relationships to cognition. Selnes, O., Albert, M., … Miller, M. I. (2015). Rela-
Cerebral Cortex, 31(11), 5253–5262. tionship of medial temporal lobe atrophy, APOE
Sasson, E., Doniger, G. M., Pasternak, O., Tarrasch, genotype, and cognitive reserve in preclinical Alz-
R., & Assaf, Y. (2013). White matter correlates of heimer’s disease. Human Brain Mapping, 36,
cognitive domains in normal aging with diffusion 2826-2841.
46 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Sporns, O. (2018). Graph theory methods: Applica- brain mapping. Human Brain Mapping, 43(1),
tions in brain networks. Dialogues in Clinical 15–22.
Neuroscience, 20(2), 111–120. Tibon, R., Geerligs, L., & Campbell, K. (2022). Bridg-
Spreng, R. N., Stevens, W. D., Viviano, J. D., & ing the big (data) gap: levels of control in small-
Schacter, D. L. (2016). Neurobiology of Aging and large-scale cognitive neuroscience research.
Attenuated anticorrelation between the default Trends in Neurosciences, 45(7), 507–516.
and dorsal attention networks with aging: Tomasi, D., & Volkow, N. D. (2011). Aging and func-
evidence from task and rest. Neurobiology of tional brain networks. Molecular Psychiatry, 17(5),
Aging, 45, 149–160. 549–558.
Stern, Y. (2009). Cognitive reserve. Neuropsycholo- Unnikrishnan, A., Freeman, W. M., Jackson, J., Wren,
gia, 47(10), 2015–2028. J. D., Porter, H., & Richardson, A. (2019). The role
Stern, Y., Arenaza-Urquijo, E. M., Bartrés-Faz, D., of DNA methylation in epigenetics of aging. Phar-
Belleville, S., Cantilon, M., Chetelat, G., … macology and Therapeutics, 195, 172–185.
Vuoksimaa, E. (2020). Whitepaper: Defining and Valenzuela, M. J., & Sachdev, P. (2006). Brain reserve
investigating cognitive reserve, brain reserve, and and cognitive decline: A non-parametric system-
brain maintenance. Alzheimer’s and Dementia, atic review. Psychological Medicine, 36(8),
16(9), 1305–1311. 1065–1073.
Stern, Y., & Barulli, D. (2019). Cognitive reserve. In S. Vaqué-Alcázar, L., Sala-Llonch, R., Abellaneda-Pérez,
T. Dekosky & S. Asthana (Eds.), Handbook of clini- K., Coll-Padrós, N., Valls-Pedret, C., Bargalló, N.,
cal neurology (Vol. 167, pp. 181–190). Amsterdam: … Bartrés-Faz, D. (2020). Functional and structural
Elsevier. correlates of working memory performance and
Stevens, M. C. (2016). Neuroscience and biobehavio- stability in healthy older adults. Brain Structure
ral reviews: The contributions of resting state and and Function, 225(1), 375–386.
task-based functional connectivity studies to our Vidal-Pineiro, D., Parker, N., Shin, J., French, L.,
understanding of adolescent brain network matu- Grydeland, H., Jackowski, A. P., … Fjell, A. M.
ration. Neuroscience and Biobehavioral Reviews, (2020). Cellular correlates of cortical thinning
70, 13–32. throughout the lifespan. Scientific Reports, 10(1).
Storsve, A. B., Fjell, A. M., Tamnes, C. K., Westlye, L. doi: 10.1038/s41598-020-78471-3
T., Overbye, K., Aasland, H. W., & Walhovd, K. B. Vidal-Piñeiro, D., Valls-Pedret, C., Fernández-Cabello,
(2014). Differential longitudinal changes in cortical S., Arenaza-Urquijo, E. M., Sala-Llonch, R., Solana,
thickness, surface area and volume across the E., … Bartrés-Faz, D. (2014). Decreased default
adult life span: Regions of accelerating and decel- mode network connectivity correlates with age-
erating change. Journal of Neuroscience, 34(25), associated structural and cognitive changes. Fron-
8488–8498. tiers in Aging Neuroscience, 6. doi: 10.3389/
Stuss, D. T., & Knight, R. T. (2002). (Eds.), Principles fnagi.2014.00256
of frontal lobe function. New York: Oxford Vu, M. A. T., Adalı, T., Ba, D., Buzsáki, G., Carlson, D.,
University Press. Heller, K., … Dzirasa, K. (2018). A shared vision for
Sudlow, C., Gallacher, J., Allen, N., Beral, V., Burton, machine learning in neuroscience. Journal of
P., Danesh, J., … Collins, R. (2015). UK Biobank: Neuroscience, 38(7), 1601–1607.
An open access resource for identifying the causes Walhovd, K. B., Fjell, A. M., & Espeseth, T. (2014).
of a wide range of complex diseases of middle and Cognitive decline and brain pathology in aging:
old age. PLoS Medicine, 12(3). doi: 10.1371/ Need for a dimensional, lifespan and systems vul-
journal.pmed.1001779 nerability view. Scandinavian Journal of Psychol-
Sullivan, E. V., & Pfefferbaum, A. (2006). Diffusion ogy, 55(3), 244–254.
tensor imaging and aging. Neuroscience and Walhovd, K. B., Fjell, A. M., Reinvang, I., Lundervold,
Biobehavioral Reviews, 30(6), 749–761. A., Dale, A. M., Eilertsen, D. E., … Fischl, B.
Tamnes, C. K., Herting, M. M., Goddings, A. L., (2005). Effects of age on volumes of cortex, white
Meuwese, R., Blakemore, S. J., Dahl, R. E., … matter and subcortical structures. Neurobiology of
Mills, K. L. (2017). Development of the cerebral Aging, 26(9), 1261–1270.
cortex across adolescence: A multisample study of Walhovd, K. B., Krogsrud, S. K., Amlien, I. K., Bar-
inter-related longitudinal changes in cortical tsch, H., Bjørnerud, A., Due-Tønnessen, P., … Fjell,
volume, surface area, and thickness. Journal of A. M. (2016). Neurodevelopmental origins of
Neuroscience, 37(12), 3402–3412. lifespan changes in brain and cognition. Proceed-
Thompson, P. M., Jahanshad, N., Schmaal, L., ings of the National Academy of Sciences of the
Turner, J. A., Winkler, A. M., Thomopoulos, S. I., United States of America, 113(33), 9357–9362.
… Kochunov, P. (2022). The enhancing neuroim- Walhovd, K. B., Nyberg, L., Lindenberger, U., Amlien,
aging genetics through meta-analysis consor- I. K., Sørensen, Ø., Wang, Y., … Fjell, A. M.
tium: 10 years of global collaborations in human (2022). Brain aging differs with cognitive ability
Aging Brain Changes across the Lifespan 47

regardless of education. Scientific Reports, 12(1). Yaffe, K., Fiocco, A. J., Lindquist, K., Vittinghoff, E.,
doi: 10.1038/s41598-022-17727-6 Simonsick, E. M., Newman, A. B., … Harris, T. B.
Wardlaw, J. M., Smith, E. E., Biessels, G. J., Cordon- (2009). Predictors of maintaining cognitive func-
nier, C., Fazekas, F., Frayne, R., … Dichgans, M. tion in older adults: The health ABC study.
(2013). Neuroimaging standards for research into Neurology, 72(23), 2029–2035.
small vessel disease and its contribution to ageing Zahr, N. M., Rohlfing, T., Pfefferbaum, A., & Sullivan,
and neurodegeneration. Lancet Neurology, 12(8), E. V. (2009). Problem solving, working memory, and
822–838. motor correlates of association and commis-
West, R. L. (1996). An application of prefrontal sural fiber bundles in normal aging: A quantitative
cortex function theory to cognitive aging. Psycho- fiber tracking study. NeuroImage, 44(3),
logical Bulletin, 120(2), 272–292. 1050–1062.
Wilson, R. S., Yu, L., Lamar, M., Schneider, J. A., Ziegler, G., Dahnke, R., Jäncke, L., Yotter, R. A., May,
Bennett, D. A., Wilson, R. S., & Boyle, P. A. (2019). A., & Gaser, C. (2012). Brain structural trajectories
Education and cognitive reserve in old age. over the adult lifespan. Human Brain Mapping,
Neurology. doi: 10.1212/wnl.0000000000007036 33(10), 2377–2389.
4
Innovation Pathways, Real-life
Neuroscience Startups,
and Applications
Tiago Lazzaretti, Paulo S. de Melo,
Anna Marduy, Rafaella Rogatto de Faria,
Marcel Simis, and Felipe Fregni

INTRODUCTION field at its needed speed. Considering the com-


plexity and multilevel management of healthcare
Advances in neuroscience and better knowledge giants, the process of innovation within these
of the brain have leveraged the development of companies undergoes great bureaucratic proce-
neuro-technologies. Important advances are tak- dures, which slows down the process. This is when
ing place in treatment, diagnosis, quality of life, startups have the potential competitive advantage
and performance. In addition, the growing preva- to introduce novel devices and industry models at
lence of neuropsychiatric disorders related to the a faster pace (Freeman and Engel, 2007).
world’s emergent aging population and environ- Startups are entrepreneurial business models
mental exposures leads to evident need of innova- engaged with innovation and surrounded by rapid
tive neurotechnological. This demand has been growth. Since changes in the healthcare system
met with global research and development initia- happen fast, this business model is gaining space
tives focused on the study of brain physiology and in the sector, as they can rapidly find novel solu-
neurological disorders in countries such as the tions and scale products or services, that already
United States, Japan, Israel, China, and the implement innovation in their foundation as a key
European Union. Nonetheless, the initiatives factor of disruption (Spender et al., 2017). This is
encompass a broad range of aims, on the study of especially true in the field of neurotechnology, as
several neurological aspects, which comes with its startups have become attractive channels for nov-
set of challenges. These programs face predica- elty. To their advantage, their small size allows for
ments such as the need for investment, concilia- a larger focus on faster and larger-scale production
tion of basic and translational research, and the of innovation (Freeman and Engel, 2007).
acceleration of neurotech device market imple- Innovative implementations within the neuro-
mentation (Grillner et al., 2016). The private ini- science field are important measures to improve
tiative is deeply involved in the process of trying people’s lifestyles, lower costs, and increase effi-
to overcome these obstacles and advance innova- ciency. And with the advent of an aging popula-
tion practices. tion, unhealthy lifestyle tendencies, or even an
However, big corporations are not always able unexpected pandemic, there is a requirement
to meet the innovation demand of the neurotech for change in the actual care models, especially
INNOVATION PATHWAYS AND REAL-LIFE NEUROSCIENCE STARTUPS 49

considering most of the time, an innovative idea is being developed and why the company exists,
does not correspond to a successful real-world the purpose of the business. The second layer rep-
opportunity (Chen, 2021; Moroni et al., 2015). In resents the how associated with an action plan on
fact, innovation is not only defined by the creation how to conquer the innovation goals to make the
of a new idea. It has to be followed by its success- business succeed. The third and last layer is the
ful implementation in the market (Freeman and what, which is the final product or service. The
Engel, 2007). Golden Circle urges the business to think about
Risk of failure is even more prominent with the value offered by the new creation. Currently,
up-and-coming neurotech startups considering companies that most thrive seek to disseminate not
the fast pace at which the field is growing, and what they do, but why (Sinek, 2009).
the unpredictable nature of innovation in the area This is seen in the healthcare field, as patients
(Freeman and Engel, 2007; Garden et al., 2019). are always seeking new technologies to enhance
Therefore, to advance innovation in this business, the experience and improve quality of life and
companies need to take advantage of startup strat- well-being, therefore UX must be considered at
egies including user experience (UX), biodesign, all stages of the innovation process in this field
pivoting, and attracting investments. In this chap- (Djamasbi and Strong, 2019). About innovations
ter, we discuss these strategies and how they are in the neurotech field, the use of UX has been ini-
being applied by neurotech startups to promote tially seen in the development of Brain-Computer
innovation and provide a comprehensive summary Interfaces (BCIs). BCIs are programs that connect
of the main acting neurotech startups today. the brain to computerized devices that are widely
Furthermore, we provide some examples of used in the recovery of individuals with motor
innovations related to treatment, diagnosis, quality impairments (Lebedev and Nicolelis, 2017). Inci-
of life and performance, including approaches such ting the previously mentioned innovation cycle
as wearable sensors, predictive medicine, preci- of this strategy, incremental BCI changes have
sion medicine, brain computer interface, genetics, been suggested based on UXs of entertainment
non-invasive stimulation, among others. There is technology, which will consecutively bring about
no conflict of interest between the authors or com- more innovation to this therapeutic model (Bram
panies mentioned. Those are examples of potential et al., 2011). Therefore, UX can serve as a trigger
paths to the future and are not necessarily endorsed and instigator for neurotech innovation strategies.
at this stage due to the need for scientific validation. Although UX is a promising cog for the innova-
tion mechanism in neurotechnology startups,
this experience can sometimes be too broad, and
mislead innovative initiatives in the field, wasting
STARTUP STRATEGIES TO ADVANCE needed resources and capital invested in startups,
which requires more focused strategies for suc-
INNOVATION cessful innovation processes to be implemented.

User Experience and Innovation Cycle


Corporations and startups must be able to com- Stanford Innovation Biodesign Model
bine innovation with user experience (UX), a set
of elements that focuses on the user needs, affec- Filling entrepreneurial gap of
tive responses, and behaviors. Essentially, UX has neurotech startups
been shown to engage an innovation cycle, in With a growing focus on patient-centered innova-
which the feedback of users instigates novelty, tion practices, strategies to develop new technol-
which in turn makes users expect further innova- ogy such as Stanford University’s Biodesign
tion to be that incremental to the novel aspect or Model (SBCB, 2022) are a need-based, problem-
completely revolutionary. To be successful, the solving alternative that consider the patient’s per-
incorporation of the user of experience requires a spective and create effective solutions. This tool
methodical approach to optimize user feedback in combines particularities of Design Thinking and
the development of innovation (Djamasbi and Design Sprint in the three phases of this approach:
Strong, 2019). identification, invention, and implementation
Thus, an UX method that should be applied in (SBCB, 2022).
the initial and final steps of the innovation process Succinctly, the identification phase consists
is the well-known Golden Circle created by Sinek of finding and collecting the health needs of the
(2009), consisting of three layers: why, how, and patients. Challenges and opportunities are taken
what. The first layer depicts that what first inspires into consideration through the full cycle of care
the community is the reason why the technology (diagnosis, treatment, recovery, and billing), but
50 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

mostly it is associated with a key aspect of the capital influx, and less spending and wasting of
innovative process: the deep empathy with the resources. This is especially important consider-
end-user. Empathy enables contemplating differ- ing entrepreneurs in the neurotech field are usually
ent points of view, needs, and desires into a prod- clinical researchers that wants to commercialize
uct or service that will impact society (Bazzano their significant findings. However, they lack the
et al., 2017). This observational phase is based on business knowledge to successfully implement
collect hundreds of needs without judgment, then their devices on the market. Biodesign combines
we need to implement rigorous filter of this list tak- the individualized input of patient needs – familiar
ing in consideration the potential that those ideas to the clinical researcher – with the necessities of
must improve healthcare and each need effect on an entrepreneurial model – often foreign to them
the stakeholders. Finally, the two or three needs (Zenios and Yock, 2010).
with the highest potential from those identified In the neurotech field, the first biodesign efforts
are deeply assessed (SBCB, 2022). Taking these were instigated by the needs of the world’s large –
factors into consideration increases the chances of soon to become larger – aging population. With the
successful market implementation of innovative maturing of the world’s population, the demand
practice or device. for innovations that target neurocognitive disor-
The invention phase consists of coming up ders took significant proportions, and thus, the
with ideas and solutions to later filter into a via- neurotech field decided to target the development
ble technology that improves patient care. After of innovative devices focusing on the diagnosis or
acknowledging the real problems faced by real treatment of disorders under the cognitive impair-
people, the ideation process takes place, consist- ment umbrella, opting for a need-based approach
ing of a brainstorming activity to find a creative in a societal scale (Garden et al., 2019).
and viable solution (Caprari et al., 2018). In this
step, it is important to collect different perspec-
tives from people that somehow are connected to
the project: patient relatives, health professionals, Lean Startup Method
hospital managers, and executives. Different types
of solution prototypes are built in a fast “think- A different framework currently used by startups
build-rethink” way, to identify potential prob- including those in the neurotech field is the lean
lems and failures on those solutions. Besides, it startup method. It is a scientific based approach to
is important to consider the intellectual property, create a startup and the desired product faster and
business model, reimbursement, and regulatory with less chance of failure (Blank, 2013; Ries,
aspects (Maher et al., 2019). Significantly, start- 2022). The key principles of this process involve
ups propitiate an adequate scenario for this phase the build-measure-learn feedback loop, the mini-
to be developed. Creativity and innovation walk mal viable product (MVB), pivot, among others.
hand in hand with the entrepreneurial model of Such principles favor the experimentation, cus-
startups, leading to the successful accomplishment tomer feedback, integrative design, over an elabo-
of this step (Freeman and Engel, 2007). rate planning, intuition, or a traditional design
The implementation phase occurs when the development. The method establishes the transi-
prototyping and testing are done, the final step to tion to a quicker implementation and exposition of
translate an idea into a final technology to reach the business model to learn from the feedback
the market. Prototyping aims to provide a sense provided.
of a real experience to detect possible problems of The build–measure–learn process consists in
the innovation through an iterative improvement. repeated learning cycles, starting with a MVP,
Practical issues as testing to validate a selected pro- with only critical concepts, receiving feedback
totype, patenting, regulatory approval, reimburse- from customers, and then reviewing the MVP.
ment, search the market potential, and explore During the process, lean startups prioritize the
funding take place. This is where, biodesign incor- agile development, that optimize time develop-
porates, in a more goal-oriented way, the use of ing the product and the business model through
UX to make refinements and improve the final interaction and incrementation by the customers
solution (Zenios and Yock, 2010). and the market. The MVP is the minimal version
The key aspect of the use of biodesign for inno- of the product that allows the maximum amount
vation is that it is a need-based system. This comes of learning and information with the least effort.
with several, marketable advantages for start- This concept states that it is more efficient and
ups including more focus on the field’s demand, faster to expose your product to the market and
being able to stipulate values to the innovative customers’ feedback than to wait to build the
technology of model one is trying to implement, perfect product before launching it (Blank, 2013;
which leads to more investments and venture Ries, 2022).
INNOVATION PATHWAYS AND REAL-LIFE NEUROSCIENCE STARTUPS 51

These methods, similar to Stanford’s Innovation purposes. Great strides in neurotechnology are
Biodesign Model (SBCB), are starting to be intro- still overshadowed by the large unknown physi-
duced in different business schools and used by ological factors of brain circuitry, being essential
large companies to induce innovation and as a the initiatives for the study of the central nervous
replacement to the old regimen (Blank, 2013). system (CNS), as described below.
They help the accurate detection of client needs,
being able to change company strategies in order
to better assist the customer and innovate even
more in the process of development, the busi-
ness model, and the final product. Being able to WORLD’S BRAIN RESEARCH PROJECTS
change company’s strategy based on feedback and
customer’s needs is a key tool in the startup envi- Currently, there are several large worldwide initia-
ronment and has been implemented in different tives that seek to improve the understanding of the
situations. CNS, which will allow the generation of impor-
tant insights for the development of innovations in
neurotechnology. The expectations of these pro-
jects are very high, since they involve high-quality
Pivoting researchers from different areas of science, in
addition to a massive investment that exceeds
Although biodesign focuses on a need-based billions of dollars.
approach to construct innovation, the neurotech Among them stand out the US BRAIN initiative
field is considerably malleable and susceptible to is a billion-dollar project, “aimed at revolution-
fast changes. These changes modify patients’ izing our understanding of the human brain. By
needs and require startups in the field to adapt to accelerating the development and application of
them and this is when pivoting takes over. Pivoting innovative technologies, researchers will be able
is the process of conducting strategic changes and to produce a revolutionary new dynamic picture of
reorientations that have a direct impact on the suc- the brain that, for the first time, shows how indi-
cess rate of a business (Kirtley and O’Mahony, vidual cells and complex neural circuits interact in
2020). The term was first introduced with the lean both time and space. Long desired by researchers
startup methodology. According to Failory (2021), seeking new ways to treat, cure, and even prevent
startups that pivot once or two times have a better brain disorders, this picture will fill major gaps in
chance of growth and raise more money than our current knowledge and provide unprecedented
those pivot zero or more than two times (Kotashev, opportunities for exploring exactly how the brain
2022). Thus, pivoting becomes an intrinsic mech- enables the human body to record, process, utilize,
anism to keep startups afloat with the constant store, and retrieve vast quantities of information,
demand evolution of the neurotech and medical all at the speed of thought” (Jorgenson et al., 2015;
industry in general. NIH Brain Initiative, 2022).
Pivoting is known as a strong strategic tactic to The Human Brain Project (HBP) (Amunts
build value and maintain relevance in the business et al., 2019; HBP, 2022) commenced in 2013,
world. Emphatically, it has become a progres- “aiming to make two major innovations: first,
sively frequent tactic in the medical industry. The a new type of science, creating synergy at the
COVID-19 pandemic conveyed a crucial pivoting interface of empirical research on the brain with
point for several medical industry companies that advanced computing, and second, an eco-system
tweaked their business models to accommodate and new culture of collaboration leading to sub-
the global demand, or completely changed their stantial progress in our understanding of the brain,
medical product focus (Laur et al., 2021). Thus, brain medicine, and brain-inspired technologies.”
pivoting allows for relocation of resources that It is a project co-founded by the European Union
were being used toward an unmarketable innova- (EU), with participation of several countries, with
tion, to focus on tangible demands and therefore an estimated budget of US1.3 billion dollars (HBP,
successfully complete the innovation process. 2022; Reardon, 2014).
Pivoting in the neurotechnology field fre- Another example is the China Brain Project
quently involves the switch from therapeutic (CBP; Normile, 2018; Poo et al., 2016) sup-
innovation to diagnostic or detective innovation. port three key research areas. The body of this
An opposite movement may occur. An example is research aims to study the neural basis of cog-
transcranial magnetic stimulation (TMS), which nitive functions, improve, and develop diagno-
was initially developed by Professor Anthony ses and therapies for major brain disorders, and
Barker as a tool for neurophysiological studies develop brain-machine intelligence technologies
but is currently more used for different therapeutic (Poo et al., 2016; Reardon, 2014). The project
52 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

estimates an investment US$157 million per year advances, more opportunities are being created
and states the international cooperation as a prior- for private organizations that sponsor and man-
ity. The center is expected to be a national effort age these initiatives, and lobby for better access to
to bring the country to the top of neuroscience their neuro-innovations.
development, being similar to the US and EU in
the field (Normile, 2018).
The International Brain Initiative (IBI) was
established to catalyze and advance ethical
neuroscience through international collaboration INVESTMENT
and knowledge sharing, uniting diverse ambi-
tions and disseminating discoveries for the benefit Implementing the strategies discussed in this
of humanity (IBI, 2018). It has participation of chapter require not only planning but investments.
some of the world’s major brain research projects, For startups, especially in neurotech, to establish
including the HBP, BRAIN initiative, Australian one’s business in the market and acquire a com-
Brain Alliance (ABA, 2022), Chinese Institute for petitive position in relation to others, it is neces-
Brain Research (Cyranoski, 2018), Japananese sary to attract investors. When it comes to startups,
Brain Mapping by Integrated Neurotechnologies venture capital is a common type of investment at
for Disease Studies (Brain/MINDS, 2021; Okano its initial stage. The capitalists seek an opportu-
et al., 2015), Korean Brain Research Initiative nity that is risky but provides the necessary tools
(KBRI, 2022), and the Canadian Brain Research to grow (Zider, 1998). They motivate the entrepre-
Strategy (CBRS, 2022). neurs to structure their business by considering
Besides the largest initiative, there are many the public and the scalability, taking part in the
smaller projects worldwide contributing to the management activities, and providing strategic
expansion of this important knowledge. One advice (Huse et al., 2005).
example is the São Paulo Research Foundation Efforts for investment in neurotechnology start-
(Brazil), that supports different types of projects ups are made from the private and public sectors
including in neuroscience (FAPESP, 2022) in an international scope. The different neurotech-
Advancing the world’s understanding of the nology ongoing initiatives around the world mutu-
brain has recently started to shift its focus on ally support one another to advance innovation
the investment of neurotechnology and neu- within the field. This combined effort stimulates
roinnovation, with different public and private the participation of the private sector in invest-
organizations investing in neuroscience startups ing, and thus, contributes to faster developments
to stimulate these advancements. An example of that can be made on larger scales, given startups
this investment is the Atlanta-based organization receive these funds (Garden and Winickoff, 2018).
NeuroLaunch, a 50-mentor program that partners An interesting concept in the context of finance
with Atlanta’s most prestigious universities with is the so-called “valley of death,” which is the
strong suits in neuroscience and tech to aid start- startup life cycle period that typically happens
ups on developing products and advancing brain after the company launches a product but it’s still
(Coburn, 2015). This initiative has broadened not making money. A large percentage of startups
its activities to different cities in the US focus- typically fail in this period. Therefore, proper
ing on different product development, such as a financial planning for this period is very important,
more pharmaceutical focus on the east coast and considering strategies that include crowdfunding,
device focus on the west coast, allowing for the joint ventures, incubators, contests, grants, and
rapid expansion of the neuroscience startup field philanthropy (especially for innovation that has
through its investments. significant health impact but with low financial
Another example of an organization joining gain).
public and private forces to advance the neuro- For that reason, a crucial alternative to the
technology field is the Neurotechnology Industry challenging financial period that the startups usu-
Organization (NIO). This is an American, lobby- ally go through is the public–private partnership.
ing-focused institution that encompasses neuro- Different types of public grants inviting companies
science-mediated companies, academic research to develop a new technology is a way to stimu-
centers, and organizations with the aim of promot- late innovation and guarantee investments in what
ing investments, either through federal research may be a win–win situation. National Institute of
funding or private funding to make the neurotech- Health (NIH), a world leader in healthcare inno-
nology field more accredited and established, to vation, offers different opportunities and partners
facilitate product approval through advocacy and with industry and academia to promote medical
promotion of the field (Robinson, 2019). It can research in order to better understand and assist
thus be seen that, as the neurotech startup field human health. Examples of NIH public–private
INNOVATION PATHWAYS AND REAL-LIFE NEUROSCIENCE STARTUPS 53

partnerships are: the Accelerating Medicines interventions for healthy people to increase cogni-
Partnership, that brings together the Foundation tive and motor performance, and to improve emo-
of the National Institute of Health (FNIH), the tional balance. In these cases, products are often
NIH, the Food and Drug Administration (FDA), marketed without adequate scientific evidence and
and 10 different biopharmaceutical companies safety assessment since it is not regulated by the
to increase the new diagnostics and therapies strict criteria such as the US FAD. This situation is
for Alzheimer’s and Parkinson’s disease, type 2 even more worrying when involve the application
diabetes, and autoimmune disorders; the Partner- of electrical brain stimuli and other different not
ship for Accelerating Cancer therapies, that com- innocuous interventions since indiscriminate use
bines the above-cited national institutions and 12 can result in unknow adverse effects.
biopharmaceutical companies to invest in cancer The United Nations Educational, Scientific
immunotherapy, anti-tumor immune response, and Cultural Organization (UNESCO) recently
and imaging approaches to target metastatic published a periodical with the title “Should we
cancers, as well as the Biomarkers Consortium, be afraid of neuroscience?” (UNESCO, 2022). In
that helps to accelerate biomarkers’ development this publication, concerns were expressed regard-
and discovery in neuroscience, cancer, metabolic ing brain–machine interface technology and arti-
disorders, immunity, and inflammation. ficial intelligence algorithms, which in theory can
Promoting this kind of partnership favors monitor, modify, and even manipulate thoughts.
the companies that receive government budget Moreover, discussed the need for specific laws
to develop a new technology and take fewer for these possible future scenarios, to protects
financial risks, and favors the government stimu- the “neurorights” of citizens, including physical
lating the progress, warming the economy, and and mental integrity, mental privacy, freedom of
guaranteeing the development of a national pro- thought, free will, and enjoy the benefits of scien-
duct, that afterwards may yield taxes and tific progress. As stated, the promising neurosci-
economic, environmental, and cultural ben- ence market can be as harmful as beneficial for
efits inducing advancements in several fields, the society, investments by companies and mainly
including neuroscience. the government should take place to audit and
control the development and correct exploitation
of these technologies. Chile is an example of the
first country that started to discuss and implement
RESPONSIBLE INNOVATION AND ITS neurorights in its constitution in order to pro-
tect the integrity of the mind and brain from the
CHALLENGES AMONG NEUROTECH advances in neurotechnology. Moreover, Spain,
STARTUPS USA, France, and Argentina have started to study
the issue (UNESCO, 2022).
Although producing innovation on a large scale Another challenge raised in the document is the
and at a fast past is thought to be a positive aspect inequality in the neuroscience field largely domi-
of neurotechnology startups, recent debates nated by industrialized countries. Regions such as
regarding the neuroethical implications of this Latin America and Caribbean, Central, West, and
rapid innovation have raised significant points. South Asia, Oceania, and Sub-Saharan Africa still
Considering the field of neurotechnology encom- make a lower contribution to the neuroscientific
passes neurological disorders deemed untreatable, literature as compared with their representation
the innovative aspect is rushed as the demands for in the world. Obstacles include limited access
effective therapeutic techniques for these disor- to training, funding, and heavy teaching loads
ders increase. However, there is a lack of regula- (UNESCO, 2022). However, including those
tion that comes with fast-paced innovation, regions in the research map is crucial to under-
especially in the startup setting. Because they are stand the brain and validate research, since they
small, startups have limited resources and choose are responsible for an enormous genetic diver-
not to focus on investing in neuroethical aspects sity and representation of the world’s population.
such as regulatory committees, or assessors that Projects from more developed countries together
can debate the safety or ethical implications of with private institutions may invest and develop
neurotech innovations. This raises concerns such places to acquire better and more valid neu-
toward the quality of innovation and calls for a roscience research.
consideration of neuroethical principles into the With such an emphatic stimulation and invest-
research and development in neurotechnology ment in the neuroscience field in the last couple
startups (Pfotenhauer et al., 2021). of decades, it has been quickly demonstrated how
These concerns are even greater when involving easy it can be to deviate into unethical manage-
neurotechnologies that are not medical devices but ment of this field’s advancement. In fact, when
54 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

asked about the role of neuroethics in the neuro- increase this amount to 37 billion dollars by 2027,
science startup department, neuro-entrepreneurs growing in a compound annual growth rate
have raised important areas where a code of con- (CAGR) of 2.9%. Some sectors might be
duct could be applied. These include data owner- highlighted since they have received more invest-
ship, autonomy, misuse of neurodata, and access ments, also being the ones with more advance-
and justice. Data ownership has been emphasized ments in the last years. Whole-brain imaging, for
as a hard-to-manage topic within the neuroscience example, is the main field recording 2.6% of the
domain because of its significant advancement, CAGR and reaching 7.9 billion by the end of
considering that the findings within the field can 2027. Other segments such as electrophysiology,
be so momentous, it is thought that data obtained brain-computer interfaces, neurostimulation are
through this field should be made public domain expected to have significant increases in their
or be sold to larger companies within the indus- budget (Neuroscience – Global Market Trajectory
try. Taking into account the lack of neuroethical and Analytics; NGMTA, 2021).
implications within neuroscience development
terrain, it is easy for larger companies to pressure
startups into selling their user-data, especially if
it comes with the promise of progress and inno- Brain Imaging
vation through financial incentives (Moss et al.,
2021). Thus, the implementation of neuroethics Brain imaging is also a relatively old technique
in this field could benefit startups in being able to that has significantly advanced in the last few
have more autonomy over their advancement in years, especially due to the great importance in
the field. The seven different principles cited are clinical practice and due to the stimuli arising
fundamental strategies for the startup development from the large investments in neurosciences (as
and innovation as shown in Figure 4.1. described above). Besides the higher costs
involved in developing imaging techniques, this
challenge has been addressed by different startups
in the neuroscience field. Several techniques to
Real-Life Neuroscience Startups visualize the brain have been developed during the
years, like magnetic resonance imaging (MRI)
The brain has been explored as the most complex (Bernasconi et al., 1999, 2000; Cendes et al.,
and mysterious organ of our species. Private 1993; Mamani et al., 2012) and its functional
investments from the industry have advanced neu- form, functional MRI (fMRI) (Poldrack, 2012;
roscience and how we understand and assess brain Rana et al., 2016), functional near-infrared spec-
activity. The neuroscience industry invested 30.3 troscopy (fNIRS) (Chen et al., 2020), and positron
billion dollars in the year 2020 and is planning to emission tomography (PET) (Lu and Yuan, 2015).

Figure 4.1 The seven principles for start up development and innovation chain.
Note: SIBM: Stanford Innovation Biodesign Method; LSUM: Lean Start Up Method.
INNOVATION PATHWAYS AND REAL-LIFE NEUROSCIENCE STARTUPS 55

All these techniques have improved our under- and Wolfram (DIDMOAD) syndrome (cf.
standing of the brain structure and function, as NGMTA, 2021). Furthermore, Verge Genomics
well as their relationships. was founded in 2015 in San Francisco, California
Behind the challenge of the higher costs of (Verge Genomics, 2022). The main activity of this
these techniques in comparison to the other ones startup was using artificial intelligence and
explained here, several companies are aiming to machine learning to develop new drugs for the
explore and improve them, and to create more effi- treatment of Alzheimer’s, AML, Parkinson’s, Sch-
cient ways of analysis to advance the understand- izophrenia, and other neuropsychiatry disorders.
ing of the CNS. Oxford Brain Diagnostics (OBD) Their therapy targets specific genes related to
is a company founded in 2019 in the United those diseases. Amylyx has raised US$134.1
Kingdom. The startup raised US$1.94 million million in funding and has partnerships with 25
to develop a software called Cortical Disarray organizations, including universities, banks, insti-
Measurement (CDM) to help clinicians in the tutes, and other pharmaceutical companies
diagnosis of dementia through MRI (OBD, 2022). (NGMTA, 2021).
Icometrix is another startup in Leuven, Belgium, Gene therapy that initially seemed unattainable
that has raised US$20.2 million to develop imag- is now a reality. Ever since the creation of synthetic
ing, guided and analyzed by artificial intelligence insulin by the startup Genentech, more entrepre-
software. The company was founded in 2011 and neurs have been investing on focusing in the devel-
its main product is the MSmetrix, which detects opment of the gene therapy industry for different
and calculates brain lesions and atrophy volumes conditions. Although faced with some challenges
(Icometrix, 2022). Additionally, PETcoil is a such as delivery method and efficacious manufac-
startup founded in 2018 in Sunnyvale, California turing, gene therapy startups are starting to focus
that combines PET scan with MRI to achieve a on improving these factors to reach complex CNS
cost-effective high-performance PET/MRI. They disorders. Passage Bio (2022) is an example of a
recently raised US$1 million in just one round of company enhancing manufacturing techniques by
funding (PETcoil, 2022). focus on accuracy of gene therapy targets for CNS
conditions such as amyotrophic lateral sclerosis,
frontotemporal dementia, and GM1 gangliodo-
sis, to name a few (Shaffer, 2020). Another inter-
Neuropharmacological Startups esting example is the medically Spinraza from
Biogen, approved by United States Food and Drug
Using pharmacological drugs may be a classical Administration (FDA) to treat spinal muscular
and more common approach when we talk about atrophy (SMA; Prakash, 2017). This medication
neuroscience and neuropsychiatry conditions. controls the mutations caused in the chromosome
However, the progress in the development of phar- 5q, related to SMA, by selectively binds and tar-
macological drugs has helped us to better under- gets RNA and regulates gene expression. The suc-
stand the complex biochemical pathways involved cess of this approach opens an important path for
in brain processing and brain diseases (Geerts treatment of many other genetically caused neu-
et al., 2020). Moreover, several improvements rological disorders. An interesting phenomenon
must be done in the current pharmacotherapies with gene therapy startups is that, being inserted
and a better understanding of the CNS biochemis- in this field and focusing on improving areas such
try may raise for studies testing these types of as delivery and manufacturing rather than solely
interventions and analyzing how they affect the on specific products facilitates their expansion to
nervous system in diverse patterns. Currently, other conditions (Shaffer, 2020).
some companies focus on developing neurophar-
macological interventions for disorders that have
no established treatment or where treatments
might be optimized, bringing more benefits and Electrophysiology Startups
less harm to patients. For example, Amylyx
Pharmaceuticals develops different pharmacologi- The recording of electrophysiologic activity has
cal treatments for neurodegenerative disorders been a key tool in neuroscience with the potential
such as Alzheimer disease (Amylyx, 2022). The to provide a fair understanding of brain activity
company is located in Cambridge, Massachusetts, with a high temporal resolution (Euler and
one of the biggest centers of innovation and Schubert, 2021; Sanchez et al., 2014), allowing to
research in the United States. In fact, Amylyx has identify different stages during the information
raised US$202.2 million in funding involving new process. Furthermore, different approaches can be
pharmacotherapeutic treatment for amyotrophic performed when collecting and analyzing EEG
lateral sclerosis (ALS), Alzheimer’s disease (SD), data, such as resting activity, event-related
56 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

potentials (ERPs), event-related desynchroniza- development of more practical, and easy to


tion (ERDs), and so on (Euler and Schubert, navigate software that eases electrophysiologi-
2021). Additionally, EEG’s potential in neurosci- cal analyses. An example of this opening is the
ence also relies on its portability and low cost, AnyWave software, a data visualizer that makes
being a very feasible tool to apply in research the interpretability of neurophysiological findings
studies and the clinical setting (Krigolson et al., faster and easier for clinicians and methodologists
2017; McWilliams et al., 2021). Moreover, mag- (Colombet et al., 2015).
netic encephalography (MEG) has been also an
important tool to analyze the magnetic field pro-
duced by the brain. With this technique, some-
times even together with the EEG, is possible to Brain–Computer Interfaces Startups
detect patterns in the brain also with great tempo-
ral resolution through event-related magnetic Connecting brains with computers is an emerging
fields (ERFs; Lopes da Silva, 2013). Several com- technique in the neuroscience field that allows
panies have been exploring inG in the last few direct communication of the electrical signals of a
years. For instance, BioSerenety, a company neuron for an external device (Mudgal et al.,
founded in 2014 in Paris, raised US$94 million in 2020). Described here as one specific technology,
funds to develop remote technologies to detect its use is highly dependent on the use of previous
brain signals such as the NeuroNaut, a remote technologies discussed here, such as EEG, MEG,
electroencephalography recording in the testing fMRI, and fNIRS (Mudgal et al., 2020). Therefore,
phase. They have a goal to develop devices that the development of brain–computer interfaces is
can accelerate research in targeted diseases intrinsically related to the development of differ-
NGMTA, 2021). Cerible is another startup that ent types of technologies that help the neurosci-
mainly invests in EEG systems intending to make ence field to understand better than brain behavior
EEG devices more accessible and portable. It has different companies have raised funds to develop
an important role in clinical practice specially to brain–computer interface technologies. The sys-
make easier to detect non-convulsive seizure. The tems can be divided into invasive (with implanted
company commenced in 2014 in Mountain View, devices) or non-invasive.
California and has raised US$121 million in fund- Neuralink (2022) is a startup specialized in
ing to develop their product (Ceribell Rapid developing brain–computer interfaces located
Response EEG, 2022; NGMTA, 2021). in Fremont, California. They focus on helping
Another remote EEG system is the Dreem patients with spinal cord injury in day-to-day
headband, which allows longitudinal and house- activities. Their main product is called “Link”
hold monitoring of important sleep parameters, and captures and interprets neurons activity with
including sleep stages (Dreem, 2022). There are an invasive device. The company has raised
technologies that use EEG signals to monitor US$363 million to develop this technology
cognitive performance and mood. For example, (NGMTA, 2021).
Focus SDK, EEG headband aiming to measure BCI Non-invasive systems are relatively sim-
the engagement and relaxation in educational pler and cheaper, and there are currently many
environments (Focus SDK, 2022). Many of the companies in the area. Some of the examples are
new technologies use the EEG not only to moni- NextMind (2022) that invests in brain–computer
tor, but also to train the brain in the context of the interfaces development, mainly using electro-
brain–machine interface, as described below. encephalography and headset to track real-time
Moreover, neuroinnovation in the field of visual experiences, translating them into digital
electrophysiology also includes the creation of commands in real life through machine learn-
products that facilitate interpretability and analy- ing algorithms (NextMind, 2022). The company
sis of electrophysiological findings. Considering founded in 2017 in Paris, France, has invested
that EEGs and other electrophysiological tests US$4.6 million. The startup’s main product called
are becoming more applicable in the clinical set- Dev Kit has earned the 2020 CES innovation
ting, it benefits clinicians to be able to quantita- award (NGMTA, 2021; NextMind, 2022). Another
tively interpret their findings. This opens space in example is FocusCalm (2022) which proposes use
the market for the development of software that of EEG with neurofeedback to reduce stress and
integrate different aspects of electrophysiology improve one’s sense of well-being (FocusCalm,
analysis in ways that can help further the use of 2022); Another is Bitbrain that use BCI for cog-
these products (Suter et al., 2010). In addition, nitive maintenance and enhancement (“Bitbrain –
the high demand of electrophysiological assess- Advanced neurotechnology,” 2018); and the
ments to understand brain behavior through Emotiv which can even be used for entertainment,
clinical research, has also created space for the but not as medical equipment (Emotiv, 2022).
INNOVATION PATHWAYS AND REAL-LIFE NEUROSCIENCE STARTUPS 57

Neurostimulation Startups Aside from the development of neurostimu-


lation devices, neurotechnology startups have
Neurostimulation devices have been developed in also been tapping into different neurofeedback
the past years targeting the treatment of neuropsy- techniques related to neuromodulation target-
chiatry conditions stimulating the brain with dif- ing different conditions ranging from anxiety
ferent methods. Some examples of neurostimulation to better focus. For instance, several neurotech-
techniques are the transcranial direct current stim- nology startups have focused on developing
ulation (tDCS), transcranial magnetic stimulation devices or phone applications related to mind-
(TMS), transcutaneous vagus nerve stimulation fulness to improve workplace efficiency. Others
(tVNS), between others with different applicability target themselves around improving focus for
in different conditions, such as chronic pain (Costa studying. Nonetheless, startups conveniently tie
et al., 2019; Moisset et al., 2020), depression together lower cost neuro-innovations, such as
(Akhtar et al., 2016; Brunoni et al., 2010), and phone applications and neurotechnology to tar-
stroke (Ting et al., 2021). Development of these get different audiences for neurotech products
techniques, besides increasing the possibilities for (NeurotechJP, 2022). A start up summary by field
the treatment of relevant diseases, helps us to is provided in Table 4.1.
understand better how the CNS works through
research studies that seek the brain response with
biomarkers, imaging, and clinical outcomes.
Among the startups investing in neurostimulation Innovation Translation from
devices, Thync is a company that designs devices Academia to Industry
for non-invasive brain stimulation to help the
improvement of brain function. The startup already Another innovation core component is the acad-
raised US$13 million in funding and was founded emia. Universities are a huge generator of intel-
in 2011 in Los Gatos, California. Their main lectual property and innovative inventions.
product is called FeelZing designed to improve However, translating the innovative invention
the energy and performance of the users from years of effort and research in a profitable
(NGMTA, 2021; Thync, 2022). Additionally, product has been done with restricted efficiency
MicroTransponder is a startup focused in and success by the universities and research
developing neurostimulation devices as therapies organizations (Sanami et al., 2017; Yusuf, 2009).
for specific neurological conditions. The company Different reasons can explain the barriers for this
was founded in 2007 in Dallas, Texas, and aims to translation, as inadequate design to be easily
develop technologies to offer relief for chronic implemented in the health care setting, inadequate
pain, limb mobility, stroke, and tinnitus. Some marketing of the idea, or onerous regulations
US$42.1 million has been raised in funding to (diSessa and Cobb, 2004; Sanami et al., 2017).
invest in the development of these techniques Furthermore, academics researchers usually have
(Microtransponder, 2022; NGMTA, 2021). reservations regarding commercialization, since
Calahealth is another example of a startup in this sometimes the intellectual property protection
field. The bioelectronic company was founded in goes against some knowledge sharing and demo-
2014 in Burlingame, California, and have raised cratic access principles. Therefore, there is a need
US$148.3 million in funding to develop a periph- to balance between sharing knowledge and pro-
eral nerve stimulation device called Cala Trio tecting the intellectual property in order to com-
mainly targeting patients with essential tremor mercialize and make the innovative process
(Cala Health, 2022; NGMTA, 2021). profitable (Sanami et al., 2017).
Table 4.1 Summary of real-life neuroscience start ups
Brain imaging Neuropharmacology Electrophysiology Brain-computer Neurostimulation Cognitive and learning
interface neurotechnologies

Oxford Brain Amylyx BioSerenety, Neuralink, USA Thync, USA MindStrong, USA
Diagnostics, UK Pharmaceuticals, France
USA
Icometrix, Belgium Verge Genomics, Cerible, USA NextMind, MicroTransponder, Neurotrack, USA
USA France USA

PETcoil, USA Passage Bio, USA Dreem, USA FocusCalm, USA Calahealth, USA Virtuleap, Portugal

Biogen, USA BrainCo, USA Bitbrain, Spain


58 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Nowadays, a trend model has been used by practical, easy-to-use smartphone applications
different companies in partnership with research- and devices that aim for memory, and cognitive
ers and universities. The open innovation model improvement of healthy and disease burdened
is based on the collaborative network of trusted individuals.
researchers and companies in order to share An up-and-coming area currently interesting
knowledge and be the middle-term between the neurotech startups is the psychiatric field. With
totally open model usually proposed by the aca- the significant prevalence of anxiety and depres-
demia and the old, closed innovation model, sion amongst today’s society, startups have
proposed by to companies to protect the intel- started to develop softwares aimed at improv-
lectual property (Chesbrough et al., 2006; Trott ing or tracking individuals’ mental health status.
and Hartmann, 2009). In this model, the innova- An example of a startup with this exact aim is
tion process would have benefits from both sides. MindStrong, a company that developed a virtual
The creativity and potentialization of the trust- platform for the assessment of mental health sta-
able sharable knowledge, and the possibility of tus and early detection of traces of psychiatric
commercialization provided by the protection of disorders through an individual’s smartphone
the innovative product. To accomplish this pro- (“MindStrong,” 2022). The company also pro-
posal, academia and industry need to count with vides virtual psychiatric services, for individuals
a multidisciplinary team, composed by academ- with already diagnosed psychiatric conditions.
ics, health care professionals, medical advice and Within the post-pandemic reality, the need for
private sector product design companies, intellec- more services that are practical, and that can be
tual property professionals, innovation managers, done remotely has been elucidated, a reason to
and patients’ groups (Chesbrough et al., 2006; why MindStrong is a good example of the mar-
Glasgow and Emmons, 2007; Sanami et al., 2017). ketable and achievable application of clinically
Different universities and research organizations based neurotechnologies.
have been more focused on commercializing inno- Another significant field catching the interest
vations and participate on the academia-industry of neurotech startups is the area of cognitive and
relationship. Several online platforms can be used memory improvement. Considering brain training
to share knowledge, information, and engage with exercises has become a significant trend within the
industry creating solid relationships to commer- smartphone application realm, neurotech startups
cialize innovation (Sanami et al., 2017). such as Neurotrack and Virtuleap have envisioned
Translating an idea to a product is challenging. how to scientifically optimize these exercises with
Steps involve identifying areas requiring techni- the aim of conjuring memory and cognitive indi-
cal development, manufacture, clinical efficacy, viduals in their product’s users.
regulatory approval, financial viability, as well as a Neurotrack is a Japan-based company with two
large multidisciplinary team to address these differ- different products; an assessment of cognitive
ent areas of development. As Sanami et al. (2017) function and a “brain care program”, both devel-
suggested, a multi-partner approach, besides chal- oped with the goal of preventing Alzheimer devel-
lenging, is required in order to accomplish all the opment (“Neurotrack,” 2022). Whereas Virtuleap
steps of development. During the process, clear is a virtual reality (VR) startup that has devel-
communication and definition of the specific role oped a VR product with the objective of improv-
of each stakeholder is required. As stated, before ing memory function through the development
on this chapter, an intrinsic relationship between of games and tests that can engage motor skills,
the academia, industry, and funding institutions/ attention, problem-solving strategies, informa-
government, this model allows to take advantage of tion processing, and other cognitive functions
each one of the three entities strengths and over- that aid in memory improvement (Kadoi, 2022;
come their limitations, promoting accessible, effec- “Virtuleap,” 2022).
tive, and financially feasible innovative ideas. Considering the linear trend between technol-
ogy development and society advancement, it
makes sense that cognitive improvement is behind
so many neurotech developments to this day
Startups Focusing on Cognitive and (Cinel et al., 2019). Moreover, a large scientific
Learning Neurotechnologies basis is provided for the development of products
aimed at cognitive augmentation given that studies
Given increasing aging of the population and have shown that certain cognitive tasks employed
higher prevalence of decay of mental health status in these neurotech products can activate different
in the past years, a new era of neurotechnology brain areas related to memory and cognition, and
startups is starting to focus on developing tech- thus allow space for the growth of these technolo-
nologies focused on cognitive improvement. gies and their startups in the market (Cinel et al.,
These startups have targeted the development of 2019). The high demand for neurotech aimed at
INNOVATION PATHWAYS AND REAL-LIFE NEUROSCIENCE STARTUPS 59

cognitive development also gives way for future undergoes major bureaucratic procedures, which
projections and more specific directions for tech- therefore slows down the development of new
nologies produced for these goals. technology. Startups are business models engaged
in innovation due to their more flexible and less
bureaucratic management. Since changes in sci-
ence and healthcare knowledge are rapid and
Applications of Neurotechnology ongoing, these types of companies have been
Startups gaining space in the sector developing novel and
innovative cognitive neuroscience solutions faster,
Historically, most of the focus of neuro- as well as scaling this important service and pro-
entrepreneurs has been in generating neuro- ducing new knowledge.
innovations targeted for diagnostic or screening A core concept in translating innovation from
devices or devices designed for the clinical treat- the neuroscience laboratories into population
ment of specific neurological conditions. However, hands is a well-stablished, trustable, and clear
these applications have given way for the advance- partnership between academic research, industry,
ment of neurotech startups and their insertion into and sponsors to promote feasible and effective
the academic field, as clinically oriented neuro- innovative ideas. However, it is important to ana-
innovation has set a precedent for funding neuro- lyze critically the biological plausibility and the
cognitive research and expanding the production scientific basis of the cognitive neuroscience solu-
of their creations. Neuro-innovations have created tions proposed by the plethora of business start-
a new demand for adjunctive devices for diagnosis ups. Moreover, with the rapid advancement and
and treatment of neurological conditions (Moss progress of these startups, serious efforts are being
et al., 2021). Also, a qualitative study conveying made to accredit and substantially advance the
the insight of neuro-entrepreneurs on the role of cognitive neuroscience field by creating adminis-
neuroethics in their field has suggested that neuro- trative opportunities as well as the direct imple-
tech should not replace current practices, but mentation of neuroethics in the management of
instead complement them (Moss et al., 2021). these private initiatives.
Clinical applications of neurotechnology have
vastly contributed to the advancement of startup
development, leading these companies to be able
to expand their products applications to non- REFERENCES
clinical settings, including neurocognition.
With the growth and quick development of Akhtar, H., Bukhari, F., Nazir, M., Anwar, M. N., &
the neurotech startup industry, entrepreneurs Shahzad, A. (2016). Therapeutic efficacy of neuro-
have started to notice how neuro-innovation can stimulation for depression: Techniques, current
be applicable in other areas. Consequently, start- modalities, and future challenges. Neuroscience
ups are now focusing on neurotechnology uses Bulletin, 32(1), 115–126.
other than those involved with neurological dis- Amylyx (2022). AMX0035. www.amylyx.com/our-
eases, such as improving cognitive and memory company (retrieved Oct. 7, 2022).
function, work efficiency, and wellbeing (Moss Amunts, K., Knoll, A. C., Lippert, T., Pennartz, C. M.
et al., 2021). However, there is still some debate A., Ryvlin, P., Destexhe, A., Jirsa, V. K., D’Angelo,
regarding the ethics and applicability of expand- E., & Bjaalie, J. G. (2019). The human brain pro-
ing neurotechnology to non-clinical fields such as ject: Synergy between neuroscience, computing,
cognitive neuroscience. informatics, and brain-inspired technologies. PLoS
Biology, 17(7). doi: 10.1371/journal.pbio.3000344
ABA (2022). Australian Brain Alliance. www.ans.org.
au/ (retrieved Oct. 7, 2022).
SUMMARY AND CONCLUSIONS Bazzano, A. N., Martin, J., Hicks, E., Faughnan, M., &
Murphy, L. (2017). Human-centred design in
Innovative advances in cognitive neuroscience are global health: A scoping review of applications
extremely important to better understand the CNS and contexts. PLoS One, 12(11). doi: 10.1371/
and the conditions that impact it, improving peo- journal.pone.0186744
ple’s overall cognitive function and ultimately Bernasconi, A., Bernasconi, N., Caramanos, Z., Reu-
their quality of life. Large, well-established com- tens, D. C., Andermann, F., Dubeau, F., Tampieri,
panies are not always able to meet innovation D., Pike, B. G., & Arnold, D. L. (2000). T2 relaxom-
demands within the neurotechnology field since etry can lateralize mesial temporal lobe epilepsy in
the management of these corporations has multi- patients with normal MRI. NeuroImage, 12(6),
level complexity, and the process of innovation 739–746.
60 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Bernasconi, N., Bernasconi, A., Andermann, F., Chesbrough, H., Vanhaverbeke, W., & West, J.
Dubeau, F., Feindel, W., & Reutens, D. C. (1999). (2006). Open innovation: Researching a new para-
Entorhinal cortex in temporal lobe epilepsy: a quan- digm. Oxford, UK: Oxford University Press
titative MRI study. Neurology, 52(9), 1870–1876. Cinel, C., Valeriani, D., & Poli, R. (2019). Neurotech-
SBCB (2022). Biodesign innovation process. https:// nologies for human cognitive augmentation: Cur-
biodesign.stanford.edu/about-us/process.html rent state of the art and future prospects. Frontiers
(retrieved Oct. 7, 2022). in Human Neuroscience, 13. doi: 10.3389/
BioSerenity (2022). Care for your patients with smart fnhum.2019.00013
healthcare software. www.bioserenity.com/en/ Coburn, L. (2015). Atlanta eyes startups for starring
(retrieved Oct. 7, 2022). role in neuroscience. www.foxnews.com/science/
Bitbrain (2018). Advanced neurotechnology. www. atlanta-eyes-startups-for-starring-role-in-neurosci-
bitbrain.com/homepage (retrieved Oct. 7, 2022). ence (retrieved Oct. 7, 2022).
Blank, S. (2013). Why the lean start-up changes eve- Colombet, B., Woodman, M., Badier, J. M., & Bénar,
rything. Harvard Business Review, 91(5), 63–72. C. G. (2015). AnyWave: A cross-platform and
NIH Brain Initiative (2022). https://braininitiative.nih. modular software for visualizing and processing
gov/ (retrieved Oct. 7, 2022). electrophysiological signals. Journal of Neurosci-
Brain/MINDS (2022). Brain mapping by integrated ence Methods, 242, 118–126.
neurotechnologies for disease studies. https:// Costa, B., Ferreira, I., Trevizol, A., Thibaut, A., &
brainminds.jp/en/ (retrieved Oct 7, 2022). Fregni, F. (2019). Emerging targets and uses of
Bram, L., Nijboer, F., Gürkök, H., Plass-Oude Bos, D., neuromodulation for pain. Expert Review of Neu-
& Nijholt, A. (2011). User experience evaluation in rotherapeutics, 19(2), 109–118.
BCI: Mind the gap. International Journal of Bioel- Cyranoski, D. (2018). Beijing launches pioneering
ectromagnetism, 13(1), 48–49. brain-science centre. Nature, 556, 157–158.
Brunoni, A. R., Teng, C. T., Correa, C., Imamura, M., diSessa, A. A., & Cobb, P. (2004). Ontological
Brasil-Neto, J. P., Boechat, R., … Fregni, F. (2010). innovation and the role of theory in design experi-
Neuromodulation approaches for the treatment of ments. Journal of the Learning Sciences, 13(1),
major depression: Challenges and recommenda- 77–103.
tions from a working group meeting. Arquivos de Djamasbi, S., & Strong, D. (2019). User experience-
Neuro-Psiquiatria, 68, 433–451. driven innovation in smart and connected worlds.
Cala Health (2022). Retuning the body to transform AIS Transactions on Human-Computer Interaction,
the standard of care for chronic disease with non- 11(4), 215–231.
invasive wearable neuromodulation therapies. Dreem (2022). Merging sleep science & deep-tech to
https://calahealth.com/ (retrieved Oct. 7, 2022). improve health at scale. https://dreem.com/en/
CBRS (2022). Canadian brain research strategy. (retrieved Oct. 7, 2022).
https://canadianbrain.ca/ (retrieved Oct. 7, 2022). Emotiv (2022). EMOTIV EPOC X: The most credible and
Caprari, E., Porsius, J. T., D’Olivo, P., Bloem, R. M., cost-effective mobile EEG Brainwear® device reim-
Vehmeijer, S. B. W., Stolk, N., & Melles, M. (2018). agined. www.emotiv.com/ (retrieved Oct. 7, 2022).
Dynamics of an orthopaedic team: Insights to Euler, M., & Schubert, A.-L. (2021). Recent develop-
improve teamwork through a design thinking ments, current challenges, and future directions in
approach. Work, 61(1), 21–39. electrophysiological approaches to studying intel-
Cendes, F., Andermann, F., Gloor, P., Evans, A., Jones- ligence. Intelligence, 88. doi: 10.1016/J.INTELL.
Gotman, M., Watson, C., … Leroux, G. (1993). 2021.101569
MRI volumetric measurement of amygdala and FAPESP (2022). Biomarcadores, eletroencefalografia:
hippocampus in temporal lobe epilepsy. Neurol- BV FAPESP (Fundação de Amparo à Pesquisa do
ogy, 43(4), 719–725. Estado de São Paulo). https://bv.fapesp.br/en/auxil-
Ceribell Rapid Response EEG (2022). The world’s first ios/99743/inhibitory-deficit-as-a-marker-of-neuro-
brain monitor for point-of-care seizure triage and plasticity-in-rehabilitation/ (retrieved Oct. 7, 2022).
treatment optimization. https://ceribell.com/ FocusCalm (2022). Brain training technology. https://
(retrieved Oct. 7, 2022). focuscalm.com/ (retrieved Oct. 7, 2022).
Chen, W.-L., Wagner, J., Heugel, N., Sugar, J., Lee, Freeman, J., & Engel, J. S. (2007). Models of innova-
Y.-W., Conant, L., … Whelan, H. T. (2020). Func- tion: Startups and mature corporations. California
tional near-infrared spectroscopy and its clinical Management Review, 50(1), 94–119.
application in the field of neuroscience: Advances Garden, H., & Winickoff, D. E. (2018). Issues in neu-
and future directions. Frontiers in Neuroscience, rotechnology governance. OECD Science, Technol-
14. doi: 10.3389/fnins.2020.00724 ogy and Industry working paper. doi: 10.1787/
Chen, L. K. (2021). Population aging and health care c3256cc6-en
services: What governments should do. Archives Garden, H., Winickoff, D. E., Frahm, N. M., & Pfoten-
of Gerontology and Geriatrics, 92. doi: 10.1016/j. hauer, S. (2019). Responsible innovation in neuro-
archger.2020.104296 technology enterprises. OECD Science, Technology
INNOVATION PATHWAYS AND REAL-LIFE NEUROSCIENCE STARTUPS 61

and Industry working paper. doi: 10.1787/ Building health services in a rapidly changing land-
9685e4fd-en scape: Lessons in adaptive leadership and pivots in
Geerts, H., Wikswo, J., van der Graaf, P. H., Bai, J. P. a COVID-19 remote monitoring program. Journal
F., Gaiteri, C., Bennett, D., … Pelleymounter, M. of Medical Internet Research, 23(1). doi: 10.2196/
(2020). Quantitative systems pharmacology for 25507
neuroscience drug discovery and development: Lebedev, M. A., & Nicolelis, M. A. (2017). Brain–
Current status, opportunities, and challenges. CPT: machine interfaces: From basic science to neuro-
Pharmacometrics & Systems Pharmacology, 9(1), prostheses and neurorehabilitation. Physiological
5–20. Reviews, 97(2), 767–837.
Glasgow, R. E., & Emmons, K. M. (2007). How can Lopes da Silva, F. (2013). EEG and MEG: Relevance to
we increase translation of research into practice? neuroscience. Neuron, 80(5), 1112–1128.
Types of evidence needed. Annual Review of Lu, F.-M., & Yuan, Z. (2015). PET/SPECT molecular
Public Health, 28, 413–433. imaging in clinical neuroscience: recent advances
Grillner, S., Ip, N., Koch, C., Koroshetz, W., Okano, in the investigation of CNS diseases. Quantitative
H., Polachek, M., … Sejnowski, T. J. (2016). Imaging in Medicine and Surgery, 5(3), 433–447.
Worldwide initiatives to advance brain research. Maher, S. A., Kyle, R., Morrey, B. F., & Yaszemski, M. J.
Nature Neuroscience, 19(9), 1118–1122. (2019). Translating orthopaedic technologies into
HBP (2022). Human brain project. www.human- clinical practice: Challenges and solutions. Jour-
brainproject.eu/en/ (retrieved Oct. 7, 2022). nal of the American Academy of Orthopaedic
Huse, M., Neubaum, D., & Gabrielsson, J. (2005). Surgeons, 27(1). doi: 10.5435/JAAOS-D-17-
Corporate innovation and competitive environ- 00851
ment. International Entrepreneurship and Man- Mamani, J., Malheiros, J., Cardoso, E., Tannús, A.,
agement Journal, 1, 313–333. Silveira, P., & Gamarra, L. (2012). In vivo magnetic
IBI (2021). International brain initiative. www.interna- resonance imaging tracking of C6 glioma cells
tionalbraininitiative.org/ (retrieved Oct. 7, 2022). labeled with superparamagnetic iron oxide nano-
Brain/MINDS (2022). https://brainminds.jp/en/ (retrieved particles. Einstein, 10(2), 164–170.
Oct 7, 2022). McWilliams, E. C., Barbey, F. M., Dyer, J. F., Islam, M. N.,
Icometrix (2022). Transforming patient care through McGuinness, B., Murphy, B., … Buick, A. R. (2021).
imaging AI. https://icometrix.com/ (retrieved Oct. Feasibility of repeated assessment of cognitive func-
7, 2022). tion in older adults using a wireless, mobile, dry-EEG
Jorgenson, L. A., Newsome, W. T., Anderson, D. J., headset and tablet-based games. Frontiers in Psy-
Bargmann, C. I., Brown, E. N., Deisseroth, K., … chiatry, 12. doi: 10.3389/fpsyt.2021.574482
Wingfield, J. C. (2015). The BRAIN initiative: Microtransponder (2022). A provider of stroke ther-
Developing technology to catalyse neuroscience apy. https://www.microtransponder.com/ (retrieved
discovery. Philosophical Transactions of the Royal Oct. 7, 2022).
Society of London. Series B: Biological Sciences, MindStrong. (2022). Mental health care with true
370. doi: 10.1098/rstb.2014.0164 strength. https://mindstrong.com/ (retrieved Oct.
Kadoi, S. (2022). Five featured startups in cognitive 7, 2022).
assessment & enhancing. https://neurotechjp.com/ Moisset, X., Lanteri-Minet, M., & Fontaine, D. (2020).
blog/5-startups-cognitive-assessment (retrieved Neurostimulation methods in the treatment of
Oct. 7, 2022). chronic pain. Journal of Neural Transmission,
KBRI (2022). Korea brain research initiative. www.kbri. 127(4), 673–686.
re.kr/new/pages_eng/main/ (retrieved Oct. 7, 2022) Moroni, I., Arruda, A., & Araujo, K. (2015). The
Kirtley, J., & O’Mahony, S. (2020). What is a pivot? design and technological innovation: How to
Explaining when and how entrepreneurial firms understand the growth of startups companies in
decide to make strategic change and pivot. Strategic competitive business environment. Procedia Man-
Management Journal, 1(37). doi: 10.1002/SMJ.3131 ufacturing, 3, 2199–2204.
Kotashev, K. (2022). Startup failure rate: How many Moss, A. U., Li, Z. R., & Rommelfanger, K. S. (2021).
startups fail and why? Failory: The all-In-one news- Assessing the perceived value of neuroethics ques-
letter for startup founders. www.failory.com/blog/ tions and policy to neuro-entrepreneurs. Frontiers in
startup-failure-rate#toc-failure-rates-of-all-start- Neuroscience, 15. doi: 10.3389/fnins.2021.702019
ups- (retrieved Oct. 7, 2022). Mudgal, S. K., Sharma, S. K., Chaturvedi, J., &
Krigolson, O. E., Williams, C. C., Norton, A., Hassall, Sharma, A. (2020). Brain computer interface
C. D., & Colino, F. L. (2017). Choosing MUSE: Vali- advancement in neurosciences: Applications and
dation of a low-cost, portable EEG system for ERP issues. Interdisciplinary Neurosurgery, 20. doi: 10.
research. Frontiers in Neuroscience, 11, 109. 1016/j.inat.2020.100694
Laur, C. V., Agarwal, P., Mukerji, G., Goulbourne, E., Neuralink (2022). Breakthrough technology for the
Baranek, H., Pus, L., … Bhattacharyya, O. (2021). brain. https://neuralink.com/ (retrieved Oct. 7, 2022).
62 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

NeurotechJP (2022). The killer applications of Neuro- Sanami, M., Flood, T., Hall, R., Kingscott, F., Jayne,
tech. https://neurotechjp.com/blog/5-startups- D., & Culmer, P. (2017). Translating healthcare
neurofeedback (retrieved Oct. 7, 2022). innovation from academia to industry. Advances
Neurotrack (2022). A modern approach to cognitive in Mechanical Engineering, 9(3). doi: 10.1177/
care. https://neurotrack.com/home (retrieved Oct. 1687814017694114
7, 2022). Sanchez, G., Daunizeau, J., Maby, E., Bertrand, O.,
NextMind (2022). Play, explore, develop with the Bompas, A., & Mattout, J. (2014). Toward a new
power of your mind. www.next-mind.com/ application of real-time electrophysiology: Online
(retrieved Oct. 7, 2022). optimization of cognitive neurosciences hypothe-
NGMTA (2021). Neuroscience: Global market trajec- sis testing. Brain Sciences, 4(1), 49–72.
tory & analytics. www.researchandmarkets.com/ Shaffer, C. (2020). Gene therapy startups extend
reports/5309799 (retrieved Oct. 7, 2022). biotech’s legacy. Genetic Engineering & Biotech-
Normile, D. (2018). Here’s how China is challenging nology News, 40(10), 32, 34, 36.
the U.S. and European brain initiatives: New Sinek, S. (2009). Start with why: How great leaders
research centers move 15-year project closer to inspire everyone to take action. London: Portfolio/
reality. Science. www.science.org/content/article/ Penguin.
heres-how-china-challenging-us-and-european- Spender, J.-C., Corvello, V., Grimaldi, M., & Rippa, P.
brain-initiatives (retrieved Oct. 6, 2022). (2017). Startups and open innovation: A review of
OBD (2022). Oxford brain diagnostics: Rethinking the literature. European Journal of Innovation
brain health. www.oxfordbraindiagnostics.com/ Management, 20(1), 4–30.
(retrieved Oct. 7, 2022). Suter, B., O’Connor, T., Iyer, V., Petreanu, L., Hooks,
Okano, H., Miyawaki, A., & Kasai, K. (2015). Brain/ B., Kiritani, T., … Shepherd, G. (2010). Ephus:
MINDS: Brain-mapping project in Japan. Philo-
Multipurpose data acquisition software for neuro-
sophical Transactions of the Royal Society of
science experiments. Frontiers in Neural Circuits, 4.
London: Series B, Biological Sciences, 370. doi.
doi: 10.3389/fncir.2010.00100
10.1098/rstb.2014.0310
Thync (2022). Neurostimulation for everyone. (2022).
PassageBio (2022). Pipeline: New hope for patients
https://thync.com/ (retrieved Oct. 7, 2022).
with previously untreatable central nervous system
Ting, W. K.-C., Fadul, F. A.-R., Fecteau, S., & Ethier,
disorders. www.passagebio.com/our-science/pipe-
C. (2021). Neurostimulation for Stroke Rehabilita-
line/default.aspx (retrieved Oct. 7, 2022).
tion. Frontiers in Neuroscience, 15. doi: 10.3389/
PETcoil (2022). We make multimodality molecular
fnins.2021.649459
imaging more accurate, sensitive, and affordable.
Trott, P., & Hartmann, D. (2009). Why ‘open innova-
www.petcoil.com (retrieved Oct. 7, 2022).
tion’ is old wine in new bottles Internatio-
Pfotenhauer, S. M., Frahm, N., Winickoff, D., Benrimoh,
D., Illes, J., & Marchant, G. (2021). Mobilizing the nal Journal of Innovation Management, 13(4),
private sector for responsible innovation in neuro- 715–736.
technology. Nature Biotechnology, 39(6), 661–664. UNESCO (2022). Should we be afraid of neurosci-
Poldrack, R. A. (2012). The future of fMRI in cognitive ence? www.unesco.org/en/articles/should-we-be-
neuroscience. NeuroImage, 62(2), 1216–1220. afraid-neuroscience (retrieved Oct. 7, 2022).
Poo, M. M., Du, J. L., Ip, N. Y., Xiong, Z. Q., Xu, B., & Verge Genomics (2022). We are building the next
Tan, T. (2016). China brain project: Basic neurosci- enduring biotechnology company of the digital
ence, brain diseases, and brain-inspired computing. age. www.vergegenomics.com (retrieved Oct. 7,
Neuron, 92(3), 591–596. 2022).
Prakash, V. (2017). Spinraza: A rare disease success Virtuleap (2022). Virtuleap is a health and education
story. Gene Therapy, 24(9), 497–497. VR startup: We aim to elevate the cognitive assess-
Rana, M., Varan, A. Q., Davoudi, A., Cohen, R. A., ment and training industry with the help of
Sitaram, R., & Ebner, N. C. (2016). Real-time fMRI emerging technologies such as virtual reality and
in neuroscience research and its use in studying artificial intelligence. https://virtuleap.com/ (retri-
the aging brain. Frontiers in Aging Neuroscience, eved Oct. 7, 2022).
8. doi:10.3389/fnagi.2016.00239 Yusuf, S. (2009). From creativity to innovation. Tech-
Reardon, S. (2014). Brain-mapping projects to join nology in Society, 31(1), 1–8.
forces. Nature. doi: 10.1038/nature.2014.14871 Zenios, S. A. M. J., & Yock, P. G. (2010). Biodesign:
Ries, E. (2022). The lean startup methodology. http:// The process of innovating medical technologies.
theleanstartup.com/principles (retrieved Oct. 7, 2022) Cambridge, UK: Cambridge University Press.
Robinson, M. D. (2019). The market in mind. Cam- Zider, B. (1998). How venture capital works. Harvard
bridge, MA: MIT Press. Business Review, 76(6), 131–139.
PART II
Neuroscientific Substrates
and Principles
This page intentionally left blank
5
Neural Processes and Activity:
Cultural Neuroscience
J o a n Y. C h i a o

INTRODUCTION networks contributes to the functional properties


of cognitive and sensory processes. The study of
The human brain abounds within the structure and cognitive systems in neuroscience entails under-
function of neurons (Koch and Poggio, 1987). standing the functional specialization of the path-
With approximately 105 neurons within each ways or streams of cortical processing across
cubic millimeter of cortical tissue, the central various levels of processing. At network levels,
nervous system (CNS) comprises more than 1012 patterns of cortical activity consist of the rep-
neurons, 1013 synapses and up to 500 distinct resentation and transformation of sensory input
types of cortical neurons (Churchland and and its perceptual interpretation into responses
Sejnowski, 2017). Neuronal activity occurs within of specific motor output or behavior. The struc-
milliseconds and within a spatial scale of neuro- tural and functional architecture of the cortex of
anatomical organization (100μm). Organization of the brain is linked to cognition and behavior (cf.
the cortex of the brain is comprised of the struc- Hubel and Wiesel, 1968; O’Reilly and Munakata,
tural composition of the dense population of cells 2000). Investigation of higher-level function of
located within the CNS. cognitive systems contributes to the understand-
Neural communication involves the electrical ing of the functional expression of the brain and
signaling of the cellular components of neurons its correspondence to behavior. Understanding
(Edelman et al., 1987). Synaptic transmission cellular and molecular mechanisms of the brain
consists of the differences in synaptic efficiency contributes to understanding the cultural variation
and its inhibitory or excitatory neurotransmission of human populations and phenotypic expression
impact (Kuffler et al., 1984; Shepherd, 2004). (Chaio, 2018; Cheon et al., 2018).
Neuronal activation involves approximately 10-15 The brain’s structural neuroanatomy comprises
joules of energy per action potential. Operation cortical and subcortical brain areas demarcated
of neurons and neuronal networks within spe- within the major gyri and fissures of the occipi-
cific cortical areas comprises the functional basis tal, temporal, parietal and frontal lobes (Angevine
of higher-level cognitive processes (Churchland and Cotman, 1981; Damasio and Damasio, 1989;
and Sejnowski, 2017; Hopfield, 1982). Sensory Huttenlocher, 1990). Cortical and subcortical
input depends on the neuroanatomical basis of areas are demarcated conceptually via the tra-
information processing. Excitatory and inhibi- ditional Brodmann designated areas (cf. Geyer
tory interaction in the connectivity of neural et al., 2011; Judaš et al., 2012).
66 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

STRUCTURAL PRINCIPLES specialization of function for specific perceptual,


cognitive, affective, and social processes. Cortical
General and specific characteristics of the neuro- specialization of functional modules for special-
anatomical structures of cognitive systems arise ized processes ensures the automatic, encapsu-
from the structural and functional principles of lated and innate processing of core capacities. The
cortical organization. For instance, the structural physical instantiation of functional modules as
components of primary sensory modalities, such cortical localization entails that the functional
as the visual system, consist of neuroanatomical architecture of the brain enables the specialized
pathways of brain structures interconnected functional processing of the organism.
through long fiber tracts. Each neuroanatomical Response properties of populations of neurons
location of the visual system comprises part of the involve the levels of population activity of neurons
sequence of information processing of sensory and their functional correspondence to higher-
inputs into the streams of visual pathways, which level processes. Encoding of sensory input into the
detail the areas of the specific cortical areas neural code of the response properties of neurons
responsible for encoding and recognition of the represents a structural and statistical problem to
visual world. determine the functional correspondence of neural
Hierarchical levels of information processing of response. Observation of neural codes and their
cortical organization range from molecular to sys- interpretation as brain function relies on the decod-
tems level neuroscience. The information process- ing of the information that is in the representation
ing hierarchy demonstrates the flow of information of the response properties of neurons. Information
across levels of processing mechanisms from processing of the response properties of neurons
molecular and cellular mechanisms of neurons to details the patterns of physiological signaling that
the cortical systems and pathways of higher-level correspond to the sensory stimuli and their trans-
processes and behavior. Multilevel processing of mission of information from neural responses.
the CNS demonstrates multiple levels of imple- Information flow of interconnected cortical
mentation of the functional components of mental pathways reflects the aggregation of the neu-
processes and their physical instantiation in the ronal processing of populations of neurons that
structural and functional organization of the CNS comprise the cortical brain regions of neural net-
and its biophysical mechanisms. works. Feedforward and feedback connectivity of
The structural principles of CNS organization cortical pathways demonstrates the unidirectional
consist of the understanding of the hierarchical and bidirectional information flow of the neural
structure of information processing across levels processing of cortical networks across the inter-
of implementation. The levels of information pro- connected functional pathways of cortical organi-
cessing of biophysical mechanisms illustrate the zation. Information processing of interconnected
stages of sensory processing of the environment cortical networks shows patterns of the functional
into representational content and its transforma- activation and inhibition of networks of neurons
tion and interpretation into patterns of motor out- and their functional correspondence to mental pro-
put and behavior. Structural properties of sensory cesses and behavior.
representation consist of encoding and decoding Functional patterns of cortical activation are
of sensory input into the representational con- comprised of the response of emergent properties
tent of higher-level processes. Transformation of in response to the effortful processing during func-
sensory input into cortical patterns of functional tional task performance. Other cortical patterns of
properties comprises the embedded distribution of activation reflect the intrinsic processing of cortical
representational content across multilevel process- mechanisms during resting state activation that cor-
ing mechanisms. responds with the structural properties of intercon-
nected cortical networks. Functional integration
of the cortical organization of the CNS consists of
the intrinsic and extrinsic properties of patterns of
functional activation from the basic mechanisms
FUNCTIONAL PRINCIPLES of resting state activity and effortful processing.

Mental and neural processes and their correspond-


ence reflect the functional principles of CNS corti-
cal organization. Functional localization of mental
constructs and functional mechanisms illustrates DISTINCT TYPES OF NEURONS
the functional specialization of such states for
specific higher-level processes. Functional locali- The neuroanatomical evidence suggests approxi-
zation of mental and cortical modules involves mately 50 to 500 distinct types of neurons within
NEURAL PROCESSES AND ACTIVITY 67

the CNS cortical organization. Different types of interpretation that leads to patterns of cognitive-
neurons show distinct patterns of neuronal firing behavioral response.
or firing rate ranging up to about 50 spikes per Encoding of distinct patterns of information
second. Some types of neurons show multiple processing across cortical layers contributes to
firing patterns that are based on the activity pat- the processes of pattern completion that facilitate
tern of the cellular mechanism of a specific neuro- production of cognitive-behavioral response. The
anatomical location or neurotransmitter system. amplification of specific neural activation patterns
Most neurons show spiking patterns of neuronal of information processing mechanisms facilitates
activity that is spontaneous and intrinsic. Regular- the completion of the activity pattern to a specific
spiking neurons demonstrate the output of a neural activation state. The constraint satisfac-
high-frequency spiking pattern of neuronal activ- tion of functional patterns of neural activation is
ity that declines to a sustained lower frequency of responsible for the transformation of the represen-
activity. Repetitive-busting neurons demonstrate tational content of sensory input into an adaptive
the intrinsic pattern of neuronal firing activity that behavioral response. The transformation of the
is in response to a prolonged stimulus. The varia- neural activation pattern into an adaptive behav-
bility of neuronal activity pattern based on the ioral response comprises the multitude of neural
response properties of neurons illustrates the responses underlying the higher-level processes of
multiple patterns of intrinsic and extrinsic activity cognition and sensation.
of neurons. The functional properties of neurons involve
Cortical specificity of neurons further consists response properties that are part of the intrinsic
of neurons dedicated to the neurochemical com- and extrinsic properties of the activity of neu-
munication of specific neurotransmitters located rons. Functional properties of neurons encom-
within the limbic system and its interconnected pass the intrinsic properties of neural activity
cortical projections. Neurotransmission involves that is spontaneous and deterministic as well as
the norepinephrine system of the locus coeruleus the extrinsic properties of the activity of neural
of the brainstem, the serotonin system located networks that is effortful and task-based process-
within the raphe nucleus of the midbrain, the ing. Emergent properties of the activity of neural
dopamine system located within the substan- networks characterizes the patterns of functional
tia nigra of the midbrain, acetylcholine located activity in response to environmental demands.
within the nucleus basalis of the basal forebrain, Functional patterns of neural activity detail the
and the GABA neurotransmitter system located bidirectional processing of neural networks that
within specific groups of cells of the mammil- constitutes the effortful processing of cognitive
lary part of the hypothalamus. The neurotransmit- and sensory processing mechanisms to environ-
ter neuron (e.g., serotonin neuron) shows distinct mental stimulus and its transformation into adap-
neurochemical communication of the specific tive behavioral response. Emergent properties of
neurotransmitter system. Different types of neu- neural activation patterns consist of the level of
rons that originate within the specific regions neural activity that consists of the electrophysi-
of the limbic system, such as the brainstem and ological response to the environment and its func-
hypothalamus, project widely across the cortical tional correspondence to cognitive and sensory
and subcortical regions of cortical brain streams states and observable behavior. Functional and
and perform specific types of neurotransmis- emergent properties of neural networks involve
sions involved in the regulation of higher-level patterns of electrophysiological signaling of cor-
function processing. tical networks and interconnectivity that encom-
Interconnectivity of neural networks is com- passes the informational complexity of CNS
prised of excitatory and inhibitory neurons respon- processing mechanisms.
sible for the electrophysiological signaling of The level of activity of networks of neurons
networks of neurons (Schmidt, 2012). Excitatory comprises interconnected neural networks that
neurons are important for feedforward process- consist of patterns of neural activation that show
ing of neural networks; inhibitory neurons are functional correspondence to cognitive and sen-
important for feedback and inhibitory processing sory processes responsible for observable behav-
of neural networks. Bidirectionality of informa- ior. The cellular and molecular basis of neural
tion processing of neural networks demonstrates transmission emphasizes the notion that the
the excitatory and inhibitory connectivity charac- response properties of single neurons are neces-
teristic of cortical neurons located as input, hid- sary and sufficient to ensure the characteristic
den and output layers of cortical organization. The properties of networks of neurons and their inter-
interconnectivity of neural networks is important connected responsivity. The response properties
for the electrophysiological signaling of sensory of populations of neurons within specific corti-
input and its representational content into an cal regions further demonstrate the conditions
68 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

responsible for the functional specialization of SENSATION AND PERCEPTION


sensation and cognition.
The anatomical and physiological properties of
the visual system highlight the structural and
functional properties of visual processing streams
and the representation of the world within specific
DISTINCT TYPES OF REPRESENTATION neurons distributed across cortical organization
(Le Vay and Nelson, 1991; Van Essen, 1979).
Different types of neurons constitute cellular Physiological processing of the visual world
mechanisms with distinct types of representations describes the transformation of signal processing
of a sensory stimulus. Local representation or from the retina to the primary visual cortex
coding of a sensory stimulus in the cortical (Hubel, 1998; Maunsell and Newsom, 1987).
representations of the environment describes the Processing of visual information into the func-
selectivity of the cellular response of neurons for tional organization of primary visual cortex entails
a specific category of stimulus. For instance, local signal transduction of the image on the retina into
coding of neurons illustrates the functional use of the representational content of the information
one type of neuron for one type of representation processing streams of primary visual cortex. The
(e.g., “grandmother cell”). The distinct type of main neurons that carry processing signals of
neuron acts as a detector of the feature of the sen- visual information from the retina to the primary
sory stimulus encoded in the level of activity of visual cortex consist of photoreceptors of the
the cellular mechanism. The notion of the neuron retina, while the ganglion cells that comprise the
as a feature detector reflects the specificity of optic tract are located within the lateral geniculate
response selectivity of cells as a unit of processing. nucleus (LGN) of the thalamus and subsequently
Scalar coding of a sensory stimulus describes project onto the primary visual (occipital) cortex.
the response properties of neurons as encoding Both principal pathways of visual processing
the representation of a sensory stimulus. The sca- involve two types of neurons located within the
lar representation of a sensory stimulus consists LGN of the thalamus that project onto the primary
of the coding of features of the sensory stimulus visual cortex and its interconnected brain regions.
as a particular firing rate of the neuron. Coding Parvocellular and magnocellular layers of the
of features of the stimulus into scalar representa- LGN detail the distinct layers of cortical process-
tions demonstrates the cellular selectivity of the ing within the thalamus that subsequently project
response properties of the neuron. Selectivity of into distinct streams of processing within primary
the firing rate of the neuron refers to the variability visual cortices. Functional segregation of the two
of the timing and magnitude of electrophysiologi- processing streams of the visual system consists of
cal response of neurons to encode the representa- the information processing streams of the “what”
tion of a particular stimulus. pathway that projects from primary visual cortex
Distributed representation of cortical organi- into the inferior temporal cortex, and the “where”
zation refers to the coding of representational pathway that projects from primary visual cortex
content across multiple areas of the brain. The into the posterior parietal cortex (Ungerleider and
vector coding of neurons depicts the selectivity Haxby, 1994). Response properties of cortical
of a multitude of neurons to encode the represen- neurons of the ventral visual form pathway dem-
tation of the sensory stimulus. The response of onstrate the basic function of the detection of vis-
many neurons encodes the features of the sensory ual form and color (e.g., “face cells”), while other
stimulus into representational content. The dis- response properties of cortical neurons of the dor-
tributed pattern of response selectivity of neurons sal pathway are responsible for spatial location
illustrates the encoding of content across popula- (e.g., “place cells”).
tions of neurons. Summation of activity level of Neurons within the primary visual cortex show
populations of neurons comprises the encoding of response selectivity that encodes the orientation of
features into distinct cortical representation. The an edge. Response properties of simple neurons
pattern of activation of populations of neurons demonstrate a cellular basis of perceptual mecha-
involves the configuration of relative weighing of nisms based on the elemental unit of edge orien-
neuronal activity in the multidimensional or state tation (Figure 5.1). Other response properties of
space representation of features within the cortical neurons located within distinct structural regions of
organization. The many-to-one mapping of neu- primary visual cortex show response selectivity for
ronal representation consists of the level of activ- the other elemental units of perceptual features, such
ity of populations of neurons as a representation as color and textures (Hubel and Livingston, 1987;
into state space. Ungerleider and Desimone, 1986; Zeki, 1983).
NEURAL PROCESSES AND ACTIVITY 69

Figure 5.1a–b Stimulus-selective response of the receptive fields of single neurons in the
cat’s striate cortex.
Source: Adapted from Hubel and Wiesel, 1959.

FACIAL RECOGNITION (IT) cortex shows selectivity to faces (Desimone,


1991; Desimone et al., 1984). The pattern of neu-
Face recognition is the perceptual and cognitive ronal activity of the STS neurons shows response
process that underlies the capacity to recognize selectivity of a specific firing pattern that is spe-
the identity of others from faces. The concept of cialized for faces (Figure 5.2). The pattern of
face recognition comprises a fundamental ele- neuronal activity shows the encoding of response
ment of the recognition of complex visual infor- selectivity for the detection of intact faces com-
mation. The cellular basis of cortical mechanisms pared with facial stimulus without parts of faces,
of facial recognition builds on the notion of objects, and other complex visual stimuli (Allison
single neurons that show response selectivity to et al., 1994; Bentin et al., 1996; Hoffman and
the specific visual category of facial information Haxby, 2000; Kanwisher et al., 1997; McCarthy
or to a specific facial identity. Functional spe- et al., 1997; Puce et al., 1995). Response proper-
cialization of cellular mechanisms for face rec- ties of single neurons located within STS high-
ognition demonstrates the cortical specificity of light the cellular basis of the cortical mechanisms
response selectivity to specific categories of of face recognition.
complex visual information that are important
for behavioral adaptation.
Identification of the cellular mechanisms of
face recognition demonstrates the functional
localization of specific cognitive processes SOCIAL PERCEPTION
within the “what” pathway of visual processing.
For instance, the response properties of neurons The social capacity to recognize conspecifics is
(e.g., “face cells”) located along the superior important to the evolutionary adaptation of social
temporal sulcus (STS) within inferior temporal species. The perception and recognition of self
70 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 5.2a–b Response properties of inferior temporal neurons to faces in the macaque.
Source: Adapted from Desimone et al., 1984.

and others is a basic mechanism of the cognitive STS neurons show response selectivity to the
processes of social capacities (Chiao et al., 2009, face and gaze direction of facial expression and
2010). Social capacity to perceive and recognize social attention ((Perrett et al., 1985, 1992; Puce
the social information of self and others is a fun- et al., 1998). Response selectivity of STS neurons
damental component of the cognitive processes of to the implied motion of face and gaze direction
social capacities. Inferential reasoning of the illustrates the perceptual cues of facial and bodily
shared goals and intentions of others from the movement in the social inferences of dominance
social perception of the shared or joint attention of and submission. Selectivity of response patterns
others comprises a fundamental element of social to gaze direction contributes to the inferences of
cognition (cf. Amodio and Frith, 2006). Implied shared attention. Patterns of response selectivity
or actual motion of others contributes to the social of STS neurons illustrate the activity of cellular
inferences of the shared goals and intentions of mechanisms that are important for social percep-
social capacities. tion and cognition (Perrett et al., 1982). The func-
Cellular mechanisms of the STS demonstrate tional specialization of the cellular mechanisms
the response properties of functional specific- located within the STS illustrates the response
ity for social cognition. The pattern of neuronal properties of neurons responsible for social cog-
activity of cellular mechanisms with the STS dem- nition (i.e., cultural neuroscience; Chiao, 2016,
onstrates the response selectivity for the social 2017a,b, 2018; Chiao and Ambady, 2007; Chiao
perception of social dimensions (Allison et al., and Blizinsky, 2016; Chiao and Immordino-Yang,
2000). Response properties of STS neurons of 2013; Chiao and Mathur, 2016; Chiao et al.,
both hemispheres demonstrate the electrophysi- 2010a,b, 2013, 2021a,b,c).
ological pattern of activity that shows response Other brain regions, such as the amygdala,
selectivity to the implied or actual motion of facial play an important role in the cellular and molecu-
and bodily movement. Increased pattern of activity lar basis of the cortical mechanisms of social
in response to the perception of implied or actual cognition. The cortical regions of STS show
motion of facial and bodily movement suggests structural and functional interconnectivity to the
the importance of the facial and bodily orienta- amygdala receiving feedforward and feedback
tion in the cellular mechanisms of social cogni- projections. The amygdala consists of the cel-
tion. Electrophysiological patterns of activity of lular mechanisms that are responsive to facial
NEURAL PROCESSES AND ACTIVITY 71

and bodily movements, such as facial and bodily faces illustrates the specificity of activity pattern
expressions (Rolls, 1984). The amygdala which that shows the functional specialization of cellular
receives bidirectional projections from the STS mechanisms for specific cognitive and emotional
shows in the response properties of populations processes of social capacities. The increased fir-
of neurons selectivity of activation patterns in ing rate of neurons located within the amygdala in
response to the movement of faces. Response response to the detection of the threat expressions
selectivity of neurons located within the amyg- of faces shows the category specificity of activity
dala serve to enhance the electrophysiological patterns in the cellular mechanisms of social per-
signaling of STS neurons that facilitates the per- ception and cognition.
ceptual and social salience of facial and bodily Multilevel mechanisms of cortical organiza-
movements. Social salience of facial and bodily tion facilitate the cellular and molecular bases of
movement demonstrates the facilitation of per- social perception. Neuronal mechanisms of social
ceptual salience of sensory input from the inter- perception show the firing pattern of activity of
connected projection of the cellular mechanisms cells that are important to the perception and rec-
located within the STS and the amygdala regions ognition of social identity and social expression.
of cortical processing. Functional pattern of circuit-based mechanisms
Cellular mechanisms of the amygdala show the demonstrates the specificity of activity pattern
response properties of neurons that are specific of social perception across interconnected brain
to the detection of the expression and identity of areas (Chiao, 2010; Pineda et al., 1994; Puce
faces as complex visual stimuli. The pattern of and Perrett, 2003). Neural communication across
activity of neurons of the amygdala of monkeys distinct types of neurons illustrates the capac-
demonstrates the response selectivity to face iden- ity of sensory and cognitive processes for the
tity and facial expression (Gothard et al., 2007). bidirectional processing of social information.
Response properties of amygdala neurons fur- Communication of electrophysiological signal-
ther show the specificity of response selectivity ing of circuit-based mechanisms communicates
to the identity or expression of faces suggesting information of the response properties of the
the distinct activity patterns of specific neurons stimulus of the social environment. The cortical
differentiate the featural characteristics of faces mechanisms of social perception demonstrate the
(Figure 5.3). Increased electrophysiological sign- distinct patterns of activity of neurons that facili-
aling of amygdala neurons that demonstrates the tate the sensory and cognitive processing of the
identity-selectivity or expression-selectivity of social environment.

Figure 5.3a–b Stimulus-response properties of amygdala neurons in response to facial


expressions of macaques.
Source: Adapted from Gothard et al., 2007.
72 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

CULTURAL NEUROSCIENCE culture. Functional activity of neuronal mecha-


nisms of interconnected brain areas demonstrates
Culture consists of the processes and mechanisms the communication of information of distinct pat-
that are of importance to the shared meaning of terns of activity of electrophysiological signaling.
systems of values, practices, and beliefs of groups The functional pattern of activity of interconnected
of people defined by geography, ethnicity, ances- areas of cortical networks illustrates the sensory
try, language, and customs among others. and cognitive processing of culture. Bidirectional
Multilevel processes of cortical mechanisms facil- activation of cortical networks facilitates the func-
itate the distinct neuronal patterns of cells and the tional processing of the information of the cultural
aggregation of the level of activity of populations environment. The specificity of response selectiv-
of neurons located within interconnected brain ity of neural activity within interconnected brain
areas of cortical networks that comprise the iden- regions shows the functional activity of circuit-
tity and expression of culture within the organiza- based mechanisms that contribute to the sensory
tion of the CNS (Chiao et al., 2008; Harada et al., and cognitive basis of culture.
2020). Variability in the timing and the rate of the The cognitive and sensory mechanisms of corti-
activity of neurons distinguishes the patterns of cal specialization support the functional and emer-
activation of cortical mechanisms that are sponta- gent properties of cultural processes. Response
neous and intrinsic to the identity and expression properties of neuronal activity within specialized
of culture. Activity of distinct types of neurons cortical mechanisms demonstrate the distinct pat-
show variability in the timing and rate of neuronal terns of response selectivity to cultural identity
activity that encodes and transforms the stimulus and cultural expression. Functional selectivity of
of the cultural environment into cultural patterns neural mechanisms in response to cultural identity
of behavior. Functional specialization of cortical and cultural expression arises in the level of activ-
mechanisms contributes to the functional and ity and its aggregation in cellular and molecular
emergent properties of the cognitive and neural mechanisms. Differential patterns of neurotrans-
basis of culture. mission of distinct systems of neuronal mecha-
Multilevel mechanisms of cortical organization nisms across levels of processing facilitates the
support the cognitive and neural basis of culture. cognitive and neural basis of culture. From single-
Neuronal mechanisms underlying the processes neuron responses to populations of neurons, the
of cognition and sensation demonstrate functional activity pattern of response properties of cellular
expression in response to the cultural environment. and molecular mechanisms formulates the cogni-
Cognitive capacity for the perception and recogni- tive and neural basis of culture (i.e., cultural neu-
tion of cultural identity and cultural expression roscience; Chiao and Immordino-Yang, 2013).
serves as a foundation for the characterization of
the multilevel mechanisms of the fundamental
dimensions of culture. Functional and emergent
properties of neuronal mechanisms of specific
brain regions show response selectivity to the COGNITIVE NEURAL NETWORKS
cultural environment. Cortical specialization of
neuronal mechanisms contributes to the response Interconnectivity of neuronal networks consists of
properties of cellular mechanisms underlying the the excitatory and inhibitory connectivity of spe-
cognitive and sensory processes of culture. The cific brain areas of cortical organization. Excitatory
distinct electrophysiological signaling of cellular and inhibitory neural connectivity demonstrates
mechanisms show functional specialization in the bidirectional information processing of the encod-
differential activity patterns in the cortical net- ing and decoding of sensory input across layers of
works of culture. Functional specialization of cor- cortical organization. Cortical responsivity of
tical mechanisms shows the response selectivity of neural networks consists of the encoding of infor-
the perceptual and cognitive processes of the facial mation of the stimulus in the populations of neu-
identity and facial expression of culture. Response rons and the decoding of such information from
selectivity of the cortical specialization of specific the activity of neuronal populations (Pouget and
brain areas illustrates the distinct patterns of activ- Zemel, 2007). Decoding of the features of a sen-
ity that distinguish the higher-level processing and sory stimulus from the cortical representation in
functional expression of distinct racial and ethnic the pattern of activity of populations of neurons
groups that comprise ethnocultural groups. relies on the selectivity of the response properties
The level of activity of circuit-based mecha- of cellular mechanisms. Intrinsic connectivity of
nisms illustrates the neural transmission of inter- neural networks reflects the spontaneous and
connected brain areas of the cortical networks of autonomous dynamics of neuronal activity,
NEURAL PROCESSES AND ACTIVITY 73

describing the intrinsic properties of cortical of interconnected brain regions located along
organization that arise from the structural brain the cortical midline structures, such as the pos-
components. Spontaneous and intrinsic activity of terior cingulate cortex and ventral portions of the
neural networks constitute the endogenous and medial prefrontal cortex (Fox and Raichle, 2007;
autonomous dynamic activity across nodes of net- Raichle et al., 2001). Activation of the default
works, reflecting the spontaneous activity of neural mode network corresponds with the cognitive
networks important to the maintenance of active functions that are important (He et al., 2008). The
and itinerant cognitive representations. activation pattern of the default mode network
Structural connectivity of cortical net- shows decreased functional responsivity to cogni-
works involves the white matter fiber tracts. tive tasks that are not relevant to the recognition
Functional organization of cortical networks and evaluation of the self. Intrinsic activity of the
depicts the specific brain regions that comprise default mode network is important to the func-
networks of cortical function. Functional spe- tional processes and basic mechanisms of social
cialization of cortical networks allows specific and cultural capacities.
sensory and cognitive processes to contribute The extrinsic properties of networks of neu-
to higher-level cognitive processing (Menon, rons shows the interconnectivity of specific brain
2012). Interconnectivity of brain regions reflects regions and its exogenous activity in response
the bidirectional processing of information to the effortful processing of functional task
flow across the specific brain regions of corti- demands. One core neural network that is respon-
cal networks. Functional integration of cortical sible for the processing of salience consists of
networks facilitates the excitatory and inhibitory the interconnected brain regions of the anterior
processing is of particular importance for the insula and dorsal anterior cingulate cortex. The
transformation and interpretation of sensory and functional processing of the salience network is
cognitive representations across multiple brain important to the detection of the emotional and
regions of cortical networks. motivational salience of the sensory environ-
ment (Menon and Uddin, 2010). The activation
pattern of the salience network facilitates the
processing of emotional and motivation salience
of sensory stimulus Adolphs, 2002; Davis and
DISTINCT TYPES OF NEURAL NETWORKS Whalen, 2001). Another core neural network that
is important to the regulation and control of execu-
Core neural networks describe distinct types of tive function consists of the interconnected brain
networks of neurons that are interconnected across regions of frontoparietal cortices, such as the dor-
brain regions to serve specific higher-level func- sal lateral prefrontal cortex and the posterior pari-
tion. Patterns of activation of core neural networks etal cortex. The functional pattern of activation
contribute to the intrinsic and extrinsic properties of the core neural network of executive function
of the functional mechanisms of networks of neu- contributes to the effortful processing of cortical
rons. Activation patterns of neural networks are in mechanisms that are responsible for the regula-
functional correspondence to the default mode of tory function of inhibitory control and executive
resting state activity or the task-based processing function (Miller, 2000; Miller and Cohen, 2001;
of functional tasks (Raichle, 2015). Intrinsic prop- Pornpattananangkul et al., 2016). Across networks
erties of core neural networks show correspond- of neurons, the distinct pattern of activation of
ence with the resting state activity of the default core neural networks demonstrates the functional
mode network. Activation patterns of the intrinsic correspondence of the effortful processing of
properties of core neural networks characterize cognition and the functional patterns of neuronal
the spontaneous and autonomous activity of net- activation that are important to the emergent prop-
works of neurons that are based on structural erties of the neural basis of cognition (Cabeza and
properties. Extrinsic properties of core neural Nyberg, 1997, 2000). The distinct patterns of the
networks consist of the interconnected brain exogenous activity of cognitive neural networks
regions that show functional patterns of activation is important to the functional processes and basic
in response to the effortful processing of func- mechanisms of the social and cultural environment
tional tasks. (cultural neuroscience; Chiao and Immordino-
Intrinsic properties of networks of neurons Yang, 2013).
illustrates the importance of the interconnectiv- Other neural networks are important to the
ity of specific brain regions and the spontaneous higher-level cognitive brain processes. The acti-
activity of populations of neurons located within vation patterns of neural networks contribute
these cortical mechanisms. The default mode to the higher-level processes of social (Blais
network is a core neural network that consists and Caldara, 2021; Chiao and Mathur, 2016;
74 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

de Gelder and Huis in’t Veld, 2016; Gianola and exploration of the functional correspondence of
Losin, 2021) and cultural capacities (Cheon and neural processes and mechanisms and the link to
Hong, 2016; Chiao et al., 2021). Functional acti- higher-level processes of cognition and observa-
vation of social neural networks demonstrates the ble behavior (Fodor, 1983; Marr 1982).
neuronal processing of social information within Multilevel processing entails correspondence
specific brain areas of occipitotemporal, parietal of the level of neural activity to the functional
and frontal cortices. Localization of the neuronal activation pattern of cortical systems (Arcaro
processing of social information of specific brain and Livingstone, 2021). The convergence of
areas demonstrates the functional specialization multimethod techniques in cognitive neurosci-
of social processing. Interconnectivity of specific ence facilitates the multilevel observation of the
brain areas further illustrates the regulatory con- anatomy, physiology and connectivity of neurons
trol of social capacities. Functional activation of across spatiotemporal timescales (Friston, 1994;
cultural neural networks shows the neuronal pro- Hillyard and Picton, 1987). The response prop-
cessing of cultural information within specific erties of neuronal activity illustrate the response
brain areas of cortical and subcortical structures. selectivity of cellular mechanisms to the envi-
Functional localization of the neuronal processing ronment. From single-cell recording techniques
of cultural information illustrates the materializa- to electrophysiological recording and functional
tion of functional processes and basic mechanisms magnetic resonance imaging, the observation and
for the processing of the identity and expression measurement of the response properties of neu-
of culture (Chiao et al., 2008; Harada et al., 2020). ronal mechanisms ensures understanding of the
Interconnectivity of specific brain areas contrib- functional significance of the activity of neurons
utes to the regulatory control of cultural capaci- across levels of processing and its link to observ-
ties. Functional patterns of activation of social and able behavior.
cultural neural networks illustrate the cognitive Thus, the neuroscientific study of higher-level
and neural basis of social and cultural capacities cognitive processes involves systematic investi-
(cultural neuroscience; Chiao and Immordino- gation of the multilevel mechanisms of cognition
Yang, 2013). and its link to observable behaviors. The role of
the response selectivity of single neurons in the
encoding and decoding of the cortical representa-
tions of the environment illustrates the importance
of cellular and molecular mechanisms in the neu-
COGNITION AND HIGHER-LEVEL ral basis of cognition. The response specificity of
PROCESSES networks of neurons demonstrates the role of the
aggregation of the level of activity of populations
The large-scale CNS organization comprises of neurons as population codes of the representa-
structural and functional principles that character- tional content of the environment. Response selec-
ize the multilevel mechanisms of cortical organi- tivity of neuronal networks to cognitive change
zation. Structural principles of cortical reflects the adaptability of neuronal mechanisms
organization describe the hierarchical structure of (Goh, 2021; Gutchess et al., 2021; Li, 2021;
functional organization across levels of process- Varnum and Hampton, 2021). Transformation
ing. Functional principles of cortical organization of the population codes of the environment into
depict the functional specialization and integra- patterns of motor output illustrates the functional
tion of cortical networks based on localization and input and output of cortical mechanisms as the
interconnectivity of networks of neurons. physical instantiation of the functional processes
Systematic investigation of the structure and func- and basic mechanisms of cortical organization.
tion of neurons entails understanding of its effect Understanding of the multilevel processing of cor-
within the networks of neurons that comprise the tical mechanisms across the CNS structural and
neural pathways of cognitive systems. From single functional organization is foundational to under-
neurons to local networks of neurons, understand- standing the functional significance of the cortical
ing the electrophysiological signaling of the pathways of the higher-level cognitive processes.
response properties of cortical mechanisms as Understanding of the cellular and molecular
well as the neurotransmission of the functional basis of cognition and behavior contributes to the
pathways of cognitive neural systems is important generation of knowledge on neurons and its func-
in the characterization of the levels of CNS pro- tional significance to behavior. The demonstration
cessing and the link to higher-level cognition and of neurons as the cellular and molecular basis of
observable behavior (Churchland and Sejnowski, the information processing of the CNS contrib-
2017; Haynes and Rees, 2005). The causal-func- utes to cognitive and systems neuroscience. The
tional role of cortical mechanisms entails the theoretical foundations of cognitive and systems
NEURAL PROCESSES AND ACTIVITY 75

neuroscience build from the systematic investiga- populations (cultural neuroscience; Chiao and
tion of neurons and its functional correspondence Immordino-Yang, 2013).
of patterns of activity as cortical representations of Understanding the cellular and molecular basis
the environment. of the human brain is foundational to the study
The role of neuronal mechanisms in cortical of cognitive and sensory systems in neurosci-
brain organization and its higher-level processes ence. Moreover, the system-level approach to
demonstrates the importance of the cellular and the study of the human CNS and its phylogenetic
molecular basis of cognition. Functional spe- and ontogenetic origins enables a comprehensive
cialization of cellular mechanisms and its role in synthesis and analysis of functional processes
cognition and higher-level processes contributes and basic mechanisms of the human brain. Study
to the structural and functional properties of corti- of structural and functional principles of brain
cal organization. Systematic investigation of cel- organization contributes to the foundations of
lular and molecular mechanisms contributes to the cognitive and systems neuroscience. The human
understanding of electrophysiological signaling brain as the neurobiological machinery of the
and neural transmission as neural communication. biological organism is a potent computational
Response properties of cellular and molecular machine with a myriad of causal-functional roles.
mechanisms as representations of the environ- Understanding the causal-functional role of the
ment facilitate the functional processes and basic CNS in health and disease (cultural neuroscience;
mechanisms of cognition as higher-level function. e.g., Chiao and Immordino-Yang, 2013; Chiao and
Characterization of the multilevel mechanisms of Mathur, 2016; Chiao et al., 2008, 2017a,b, 2020,
cortical organization contribute to the understand- 2021a,b,c) is important in order to comprehend
ing of the fundamental dimensions of human brain the broader causal power of the human brain and
organization and its link to observable behavior. cognition in the structure of the natural world.
The patterns of activation of neuronal net-
works contribute to the functional processes and
basic mechanisms of cognition. Distinct activation
patterns of cortical neural networks are respon-
sible for the intrinsic and extrinsic processing of REFERENCES
populations of neurons. The distributed code of
neuronal populations demonstrates the category- Adolphs, R. (2002). Neural systems for recognizing
specific representation of the environment across emotion. Current Opinion in Neurobiology, 12,
interconnected brain areas. Functional integra- 169–177.
tion of cortical neural networks facilitates the Allison, T., Puce, A., & McCarthy, G. (2000). Social
information processing across the structural and perception from visual cues: The role of the STS.
functional connectivity of networks of neurons, Trends in Cognitive Science, 4, 267–278.
illustrating the importance of functional integra- Allison, T., McCarthy, G., Nobre, A., Puce, A., &
tion of cortical mechanisms in the higher-level Belger, A. (1994). Human extrastriate cortex and
cognitive processes. the perception of faces, words, numbers and
colors. Cerebral Cortex, 4(5), 544–554.
Amodio, D. M., & Frith, C. D. (2006). Meeting of
minds: The medial frontal cortex and social cogni-
tion. Nature Reviews Neuroscience, 7(4),
SUMMARY AND CONCLUSIONS 268–277.
Angevine, J. B. Jr., & Cotman, C. W. (1981). Principles
Study of neurons is foundational to the fundamen- of neuroanatomy. Oxford, UK: Oxford University
tals of cognitive and systems neuroscience. Press.
Characterization of the structural and functional Arcaro, M. J., & Livingstone, M. S. (2021). On the
properties of neurons contributes to the under- relationship between maps and domains in infer-
standing of the cellular and molecular basis of the otemporal cortex. Nature Review Neuroscience,
human brain. The cultural and biological variation 22(9), 573–583.
of human populations contributes to the pheno- Bentin, S., Allison, T., Puce, A., Perez, E., & McCarthy,
typic variation of the human brain. Distinct types G. (1996). Electrophysiological studies of face
of patterns of neuronal activity reflect the pheno- perception in humans. Journal of Cognitive Neuro-
typic expression of the functional architecture of science, 8, 551–565.
the human brain of distinct ethnocultural groups. Blais, C., Caldara, R. (2021). Culture shapes face
Systematic investigation of the phenotypic expres- processing. In J. Y. Chiao, S.-C., Li, R. Turner, S. Y.
sion of the human brain advances knowledge of Lee-Tauler & B. A. Pringle (Eds.), The Oxford hand-
the cultural and biological variation of human book of cultural neuroscience and global mental
76 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

health (pp. 309–325). New York: Oxford University individualistic and collectivistic views of self.
Press. Human Brain Mapping, 30(9), 2813–2820.
Cabeza, R., & Nyberg, L. (1997). Imaging cognition: Chiao, J. Y., Harada, T., Komeda, H., Li, Z., Mano, Y.,
An empirical review of PET studies with normal Saito, D., … Iidaka, T. (2010a). Dynamic cultural
subjects. Journal of Cognitive Neuroscience, 9(1). influences on neural representations of the self.
doi: 10.1162/jocn.1997.9.1.1. Journal of Cognitive Neuroscience, 22(1), 1–11.
Cabeza, R., & Nyberg, L. (2000). Imaging cognition II: Chiao, J. Y., Hariri, A. R., Harada, T., Mano, Y.,
Empirical review of 275 PET and fMRI studies. Sadato, N., Parrish, T. B., & Iidaka, T. (2010b).
Journal of Cognitive Neuroscience, 12. doi: Theory and methods in cultural neuroscience.
10.1162/08989290051137585 Social Cognitive and Affective Neuroscience, 5(2–3),
Cheon, B. K., & Hong, Y.-Y. (2016). The cultural neu- 356–361.
roscience of intergroup bias. In J. Y. Chiao, S.-C., Chiao, J. Y., Iidaka, T., Gordon, H. L., Nogawa, J., Bar,
Li, R. Seligman & R. Turner (Eds.), The Oxford M., Aminoff, E., Sadato, N., & Ambady, N. (2008).
handbook of cultural neuroscience (pp. 249–270). Cultural specificity in amygdala response to fear
New York: Oxford University Press. faces. Journal of Cognitive Neuroscience, 20(12),
Cheon, B. K., Tang, R., Chiao, J. Y., & Tang, Y.-Y. 2167–2174.
(2018). In J. Spencer-Rodgers & K. Peng (Eds.), The Chiao, J. Y., Cheon, B. K., Pornpattananangkul, N.,
psychological and cultural foundations of east Mrazek, A. J., & Blizinsky, K. D. (2013). Cultural
Asian cognition: Contradiction, change, and holism neuroscience: Progress and promise. Psychological
(pp. 181–211). New York: Oxford University Press. Inquiry, 24(1), 1–19.
Chiao, J. Y. (2010). Neural basis of social status hier- Chiao, J. Y., Li, S.-C., Turner, R., & Lee-Tauler, S. Y.
archy across species. Current Opinion in Neurobi- (2020). Cultural neuroscience and the research
ology, 20(6), 803–809. domain criteria: Implications for global mental
Chiao, J. Y. (2016). Cultural neuroscience of the health. Neuroscience and Biobehavioral Reviews,
developing brain in childhood. Minnesota Sympo- 116, 109–119.
sium on Child Psychology: Culture and Develop- Chiao, J. Y., Harada, T., Mano, Y., Stein, D. J., &
mental Systems, 38, 1–37. Sadato, N. (2021a). Culture and emotion. In J. Y.
Chiao, J. Y. (2017a). Cultural neuroscience of the Chiao, S.-C. Li, R. Turner, S.Y. Lee-Tauler & B. A.
developing brain in adolescence. In N. Budwig, E. Pringle (Eds.), The Oxford handbook of cultural
Turiel & P. D. Zelazo (Eds.), New perspectives on neuroscience and global mental health
human development (pp. 330–350). Cambridge, (pp. 199–220). New York: Oxford University Press.
UK: Cambridge University Press. Chiao, J. Y., Li, Z., & Sadato, N. (2021b). Culture and
Chiao, J. Y. (2017b). Philosophy of cultural neurosci- genomics. In J. Y. Chiao, S.-C. Li, R. Turner, S. Y.
ence. London: Routledge. Lee-Tauler & B. A. Pringle (Eds.), The Oxford hand-
Chiao, J. Y. (2018). Developmental aspects in cultural book of cultural neuroscience and global mental
neuroscience. Developmental Review, 50, 77–89. health (pp. 144–155). New York: Oxford University
Chiao, J. Y., & Ambady, N. (2007). Cultural neurosci- Press.
ence: Parsing universality and diversity across Chiao, J. Y., Li, S.-C-., Turner, R., Lee-Tauler, S. Y., &
levels of analysis. In S. Kitayama & D. Cohen (Eds.), Pringle, B. A. (Eds.) (2021c). The Oxford handbook
Handbook of cultural psychology (pp. 237–254). of cultural neuroscience and global mental health.
New York: Guilford. New York: Oxford University Press.
Chiao, J. Y., & Blizinsky, K. D. (2016). Cultural neuro- Churchland, P. S., & Sejnowski, T. J. (2017). The com-
science: Bridging cultural and biological sciences. putational brain (25th ed.). MA: MIT Press.
In E. Harmon-Jones & M. Inzlicht (Eds.), Social Damasio, H., & Damasio, A. (1989). Lesion analysis in
neuroscience: Biological approaches to social psy- neuropsychology. Oxford, UK: Oxford University
chology (pp. 247–275). New York: Routledge. Press.
Chiao, J. Y., & Immordino-Yang, M. H. (2013). Modu- Davis, M., & Whalen, P. J. (2001). The amygdala:
larity and the cultural mind: Contributions of cul- Vigilance and emotion. Molecular Psychiatry, 6,
tural neuroscience to cognitive theory. Perspectives 13–34.
on Psychological Science, 8(1), 56–61. De Gelder, B., & Huis in’t Veld, E. (2016). Cultural
Chiao, J. Y. & Mathur, V. A. (2016). Cultural neurosci- differences in emotional expressions and body
ence of pain and empathy. In J. Y. Chiao, S.-C., Li, language. In J. Y. Chiao, S.-C., Li, R. Seligman & R.
R. Seligman & R. Turner (Eds.), The Oxford hand- Turner (Eds.), The Oxford handbook of cultural
book of cultural neuroscience (pp. 271–278). New neuroscience (pp. 223–236). New York: Oxford
York: Oxford University Press. University Press.
Chiao, J. Y., Harada, T., Komeda, H., Li, Z., Mano, Y., Desimone, R. (1991). Face-selective cells in the tem-
Saito, D., … Iidaka, T. (2009). Neural basis of poral cortex of monkeys. Journal of Cognitive
NEURAL PROCESSES AND ACTIVITY 77

Neuroscience, 3. doi: 10.1162/089892900511 He, B. J., Snyder, A. Z., Zempel, J. M., Smyth, M. D.,
37585 & Raichle, M. E. (2008). Electrophysiological cor-
Desimone, R., Albright, T. D., Gross, C. G., & Bruce, relates of the brain’s intrinsic large-scale functional
C. (1984). Stimulus-selective properties of inferior architecture. Proceedings of the National Academy
temporal neurons in the macaque. Journal of Neu- of Sciences of the United States of America,
roscience, 4, 2051–2062. 105(41), 16039–16044.
Edelman, G. M., Gall, W. E., & Cowan, W. M. (1987). Hillyard, S., & Picton, T. W. (1987). Electrophysiology
(Eds.), Synaptic function. New York: Wiley. of cognition. In F. Plumb (Ed.), Handbook of physi-
Fodor, J. (1983). The modularity of mind. Cambridge, ology, Section 1: Neurophysiology (pp. 519–584).
MA: MIT Press. New York: American Physiological Society.
Fox, M., & Raichle, M. (2007). Spontaneous fluctua- Hoffman, E. A., & Haxby, J. V. (2000). Distinct repre-
tions in brain activity observed with functional sentations of eye gaze and identity in the distrib-
magnetic resonance imaging. Nature Reviews uted human neural system for face perception.
Neuroscience, 8, 700–711. Nature Neuroscience, 391, 80-84.
Friston, K. J. (1994). Functional and effective con- Hopfield, J. (1982). Neural networks and physical
nectivity in neuroimaging: A synthesis. Human systems with emergent collective computational
Brain Mapping, 2, 56–78. abilities. Proceedings of the National Academy of
Geyer, S., Weiss, M., Reimann, K., Lohmann, G., & Sciences of the United States of America, 79,
Turner, R. (2011). Microstructural parcellation of 2554–2558.
the human cerebral cortex: From Brodmann’s post- Hubel, D. H. (1998). Eye, brain, and vision New York:
mortem map to in vivo mapping with high-field W. H. Freeman.
magnetic resonance imaging. Frontiers in Human Hubel, D. H., & Livingstone, M. S. (1987). Segrega-
Neuroscience, 5. doi: 10.3389/fnhum.2011.00019 tion of form, color, and stereopsis in primate area
Gianola, M., & Losin, E. A. (2021). The neuroscience 18. Journal of Neuroscience, 7, 3378–3415.
of cultural imitative learning and connections to Hubel, D. H., & Weisel, T. N. (1959). Receptive fields
global mental health. In J. Y. Chiao, S.-C., Li, R. of single neurons in the cat’s striate cortex. Journal
Turner, S. Y. Lee-Tauler & B. A. Pringle (Eds.), The of Physiology, 148, 574–591.
Oxford handbook of cultural neuroscience and Hubel, D. H., & Wiesel, T. N. (1968). Receptive fields
global mental health (pp. 326–363). New York: and functional architecture of monkey striate
Oxford University Press. cortex. Journal of Physiology, 195, 215–243.
Goh, J. O. S. (2021). Acculturation by plasticity and Huttenlocher, P. R. (1990). Morphometric study of
stability in neural processes: Considerations for human cerebral cortex development. Neuropsych-
global mental health challenges. In J. Y. Chiao, S.- ologia, 28, 517–527.
C., Li, R. Turner, S. Y. Lee-Tauler & B. A. Pringle Judaš, M., Cepanec, M., & Sedmak, G. (2012). Brod-
(Eds.), The Oxford handbook of cultural neurosci- man’s map of the human cerebral cortex – or
ence and global mental health (pp. 409–437). Brodman’s maps? Translational Neuroscience, 3(1),
New York: Oxford University Press. 67–74.
Gothard, K. M., Battaglia, F. P., Erickson, C. A., Kanwisher, N, McDermott, J., & Chun, M. M. (1997).
Spitler, K. M., & Amaral, D. G. (2007). Neural The fusiform face area: A module in human extras-
responses to facial expression and face identity in triate cortex specialized for face perception. Jour-
the monkey amygdala. Journal of Neurophysiol- nal of Neuroscience, 17, 4302–4311.
ogy, 97(2), 1671–1683. Koch, C., & Poggio, T. (1987). Biophysics of compu-
Gutchess, A., Mukadam, N., Zhang, W., & Zhang, X. tation: Neurons, synapses, and membranes. In G.
(2021). Influence of aging on memory across Endleman, W. Gall & W. Cohen (Eds.), Synaptic
cultures. In J. Y. Chiao, S.-C., Li, R. Turner, S. Y. function (pp. 637–697). New York: Wiley.
Lee-Tauler & B. A. Pringle (Eds.), The Oxford Kuffler, S. W., Nicholls, J. G., & Martin, A. R. (1984).
handbook of cultural neuroscience and global From neuron to brain: A cellular approach to the
mental health (pp. 262–287). New York: Oxford function of the nervous system (2nd ed.). Sunder-
University Press. land, UK: Sinauer.
Harada, T., Mano, Y., Komeda, H., Hechtman, L. A., Le Vay, S. & Nelson, S. B. (1991). Columnar organiza-
Pornpattanangkul, N., Parrish, T. B., … Chiao, J. Y. tion of the visual cortex. In J. R. Cronly-Dillon (Ed.),
(2020). Cultural influences on neural systems of The neural basis of visual function. London:
intergroup emotion perception: An fMRI study. Macmillan.
Neuropsychologia, 137. doi: 10.1016/j.neuropsycho- Li, S.C. (2021). Developmental plasticity and global
logia.2019.107254 aging. In J. Y. Chiao, S.-C., Li, R. Turner, S. Y. Lee-
Haynes, J.-D., & Rees, G. (2005). Predicting the stream Tauler & B. A. Pringle (Eds.), The Oxford handbook
of consciousness from activity in human visual of cultural neuroscience and global mental health
cortex. Current Biology, 15(14), 1301–1307. (pp. 179–198). New York: Oxford University Press.
78 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Marr, D. (1982). Vision: A computational investigation Bayesian brain: Probabilistic approaches to neural
into the human representation and processing of coding (pp. 115–130). Cambridge, MA: MIT Press.
visual information. New York: W. H. Freeman. Puce, A., Allison, T., Gore, J. C., & McCarthy, G.
Maunsell, J. H., & Newsome, W. T. (1987). Visual (1995). Face-sensitive regions in human extrastri-
processing in monkey extrastriate cortex. Annual ate cortex studied by functional MRI. Journal of
Review of Neuroscience, 10, 363–401. Neurophysiology, 74, 1192–1199.
Menon, V. (2012). Functional connectivity, neurocog- Puce, A., Allison, T., Bentin, S., Gore, J. C., & McCa-
nitive networks and brain dynamics. In M. I. Rabi- rthy, G. (1998). Temporal cortex activation in
novich, K. J. Friston & P. Varona (Eds.), Principles of humans viewing eye and mouth movements. Jour-
brain dynamics: Global state interactions (pp. 27– nal of Neuroscience, 18(6), 2188–2199.
48). Cambridge, MA: MIT Press. Puce, A., & Perrett, D. (2003). Electrophysiology and
Menon, V., & Uddin, L.Q. (2010). Saliency, switching, brain imaging of biological motion. Philosophical
attention and control: A network model of insula Transactions of the Royal Society London B; Bio-
function. Brain Structure and Function, 214(5–6), logical Sciences, 358, 435–445.
655–667. Raichle, M. E. (2015). The brain’s default mode net-
McCarthy, G., Puce, A., Gore, J. C., & Allison, T. work. Annual Review of Neuroscience, 38,
(1997). Face-specific processing in human fusiform 433–447.
gyrus. Journal of Cognitive Neuroscience, 9, Raichle, M. E., MacLeod, A. M., Snyder, A. Z.,
604–609. Powers, W. J., Gusnard, D. A., & Shulman, G. L.
Miller, E. K. (2000). The prefrontal cortex and cogni- (2001). A default mode of brain function. Pro-
tive control. Nature Review Neuroscience, 1(1), ceedings of the National Academy of Sciences of
59–65. the United States of America, 98(2), 676–682.
Miller, E. K., & Cohen, J. D. (2001). An integrative Rolls, E. T. (1984). Neurons in the cortex of the tem-
theory of prefrontal cortex function. Annual poral lobe and in the amygdala of the monkey
Review of Neuroscience, 24, 167–202. with responses selective for faces. Human Neuro-
O’Reilly, R. C., & Munakata, Y. (2000). Computa- biology, 3(4), 209–222.
tional explorations in cognitive neuroscience. Schmidt, R. F. (Ed.). (2012). Fundamentals of neuro-
Cambridge, MA: MIT Press. physiology. (D. Jordan, Translator). Berlin:
Perrett, D. I., Rolls, E. T., & Cann, W. (1982). Visual Springer-Verlag.
neurons response to faces in the monkey temporal Shepherd, G. M. (2004). The synaptic organization of
cortex. Experimental Brain Research, 47(3), the brain (5th ed.). Oxford, UK: Oxford University
329–342. Press.
Perrett, D. I., Hietanen, J. K., Oram, M. W., & Benson, Ungerleider, L. G., & Desimone, R. (1986). Cortical
P. J. (1992). Organization and functions of cells projections of visual area MT in the macaque.
responsive to faces in the temporal cortex. Philo- Journal of Comparative Neurology, 248,
sophical Transactions of the Royal Society London 190–222.
B: Biological Sciences, 335, 23–30. Ungerleider, L. G., & Haxby, J. V. (1994). “What” and
Perrett, D. I., Smith, P. A., Potter, D. D., Mistlin, A. J., Head, “where” in the human brain. Current Opinion in
A. S., Milner, A. D., & Jeeves, M. A. (1985). Visual cells Neurobiology, 4, 157–165.
in the temporal cortex sensitive to face view and gaze Van Essen, D. C. (1979). Visual areas of the mam-
direction. Proceedings of the Royal Society of London: malian cerebral cortex. Annual of Review of Neu-
Biological Sciences, 223, 293–317. roscience, 2, 227–263.
Pineda, J. A., Sebestyen, C., & Nava, C. (1994). Face Varnum, M.E.W., & Hampton, R.S. (2021). Cultural
recognition as a function of social attention in non- changes in neural structure and function. In J. Y.
human primates: An ERP study. Cognitive Brain Chiao, S.-C., Li, R. Turner, S. Y. Lee-Tauler & B. A.
Research, 2(1). doi: 10.1016/0926-6410(94)90015-9 Pringle (Eds.), The Oxford handbook of cultural
Pornpattananangkul, N., Hariri, A. R., Harada, T., neuroscience and global mental health (pp. 387–
Mano, Y., Komeda, H., Parrish, T. B., … Chiao, J. 408). New York: Oxford University Press.
Y. (2016). Cultural influences on neural basis of Zeki, S. (1983). Colour coding in the cerebral cortex:
inhibitory control. NeuroImage, 139, 114–126. The reaction of cells in monkey visual cortex to
Pouget, A., & Zemel, R. S. (2007). Population codes. wavelengths and colours. Neuroscience, 9,
In K. Doya, S. Ishii, A. Pouget & Rao, R. P. N. (Eds.), 741–765.
6
Role of Neuroglia in Cognition
A l e x e i Ve r k h r a t s k y , A l e x e y S e m y a n o v,
Arthur Butt, and Olga Garaschuk

INTRODUCTION system (PNS), both of which are multicellular and do


not only contain neurones. Cell types in the nervous
The concept of Neuroglia (Nervenkitt, or nervous tissue are broadly classified into cells of ectodermal
putty) as a connective tissue of the brain and the spinal and mesodermal origin, according to their develop-
cord was introduced by Rudolf Ludwig Karl Virchow mental roots. The ectoderm derived cells are neural
in 1856–1858 (Virchow, 1860), who advanced earlier cells (the progeny of common neuroepithelial precur-
ideas of Karl von Rokitansky, who in 1854, described sors), represented by neurones and neuroglia, includ-
the connective tissue of the nervous system as a store- ing astroglia and oligodendroglia in the CNS, and
room (Lager), scaffold (Gerüste), and binding sub- Schwann cells, satellite cells, and enteric gliocytes in
stance (Rokitansky, 1854). Neuroglial physiology and the PNS. The mesodermal cells are subdivided into
pathology were investigated in great detail by almost CNS resident microglia and cellular elements of
every prominent neuroanatomist and neurologist of blood vessels, including endothelial cells, pericytes,
the nineteenth century, while the fundamental role of smooth muscle cells, and fibroblasts. In pathological
neuroglia in brain homeostasis, nutritional support, conditions, the nervous tissue parenchyma can also
regulation of blood flow, sleep and conscience, as well be invaded by blood-borne cells, such as platelets,
as in neuropathology, was widely recognized in works leukocytes, and various macrophages. All these cel-
of Camillo Golgi, Santiago Ramon y Cajal, Carl lular populations are intimately interacting at struc-
Ludwig Schleich, Albert Kölliker, Wilhelm His, tural and functional levels, thus creating the active
Ernesto Lugaro, Alois Alzheimer, Franz Nissl, and milieu of nervous tissue (Semyanov and Verkhratsky,
others (Chvatal and Verkhratsky, 2018; Kettenmann 2021; also see Figure 6.1).
and Verkhratsky, 2008).

NEUROGLIA: DEFINITIONS,
MULTICELLULAR NATURE OF CLASSIFICATION, AND NUMBERS
NERVOUS TISSUE
Neuroglia are defined as a heterogeneous popula-
The nervous system of vertebrates consists of the tion of cells of ectodermal and mesodermal origin
central nervous system (CNS) and peripheral nervous providing for homeostasis and defence of the
80 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 6.1 Active milieu of the nervous tissue. A: Multicellular nature of the nervous tissue:
neurones and glia form neural networks. B: The concept of the active milieu of the nervous
tissue integrates multiple theories that address different aspects of local functional organi-
zation of the brain: multipartite synapse, neuro(glio)vascular unit, extrasynaptic signaling
and volume transmission. The active milieu is formed through dynamic interactions between
neuronal elements (somata, axons, dendrites, and spines), non-neuronal cell elements (astro-
cytic and microglial processes), vasculature (capillary), extracellular space (ECS), and extracel-
lular matrix (ECM). In an active milieu, synapses can contact, signal, and be homeostatically
controlled by astrocyte branches (a) or, by single or several leaflets (b). A single astrocytic
branch or leaflet may be contacted by several synapses (c). Dynamic changes in morphol-
ogy of astrocytic processes affect diffusional barriers, neurotransmitter clearance, and K+
dynamics, and supply of glutamine or energy substrates, thus regulating neuronal plasticity.
Astrocytic processes form loop-like structures through reciprocal gap junctions (GJ).
Source: (A) (from Kettenmann et al., 2013). (B) (from Semyanov and Verkhratsky, 2021).

nervous system; neuroglia are the ultimate sup- protection therefore became the function of neuro-
portive cells, which keep nervous tissue in a func- glia (Verkhratsky and Nedergaard, 2016).
tional state (Verkhratsky and Butt, 2013). In Neuroglia in the mammalian nervous system are
physiological conditions glial cells are responsi- classified into PNS and CNS neuroglia (Verkhratsky
ble for housekeeping, whereas in pathology they and Butt, 2013) (Figure 6.2). The peripheral nerv-
undergo functional remodelling to preserve and ous system glia are represented by the Schwann
restore brain homeostasis. Failure in glial function cells (myelinating Schwann cells that myelinate
exacerbates nervous tissue damage and facilitates peripheral axons, non-myelinating Schwann cells
the development of neurological diseases (Pekny that surround multiple non-myelinating axons,
et al., 2016; Verkhratsky et al., 2017). Homeostatic perisynaptic Schwann cells, which enwrap periph-
and protective roles of neuroglia reflect evolution- eral synapses (e.g., neuro-muscular junctions),
ary specialisation of the cellular components of and nociceptive Schwann cells, which, together
the nervous system. Neurones evolved as signal- with neuronal endings form sensory organs in the
ling and information processing elements, which, skin), together with the satellite glial cells that sur-
however, came at a price of losing essential house- round neurones in sensory and sympathetic gan-
keeping functions, as neurones are generally inca- glia, and enteric glia, which are a part of enteric
pable of regulating their own immediate nervous system, the intrinsic nervous system of the
environment and are vulnerable to many kinds of gastrointestinal tract. Also, there are several other
environmental insults. The housekeeping and types of glial cells that are distinguished in organs
Role of Neuroglia in Cognition 81

Figure 6.2 Classification of neuroglia.

of special senses, including olfactory ensheathing integral parts of neural networks (Kettenmann
cells, olfactory sustentacular (or supporting) cells, et al., 2011).
supporting cells of the vestibular organ and organ The total numbers of glial cells and neurones
of Corti, and glia-like taste bud cells. are approximately equivalent in the human brain,
Neuroglia of the brain and the spinal cord which contains about 80 billion (80×109) neuroglia
(the CNS glia) are subdivided into the macro- and ~100 billion (100×109) neurones, but there is
glia of ectodermal (neuroepithelial) origin, and substantial regional variation: in the cerebellum,
the microglia of mesodermal origin (Verkhratsky glia are outnumbered by neurones 1:10, whilst in
and Butt, 2013). Macroglia are further subclassi- the cerebral cortex there are twice as many glia
fied into astroglia and oligodendroglia. Astroglia as neurones (2:1), and the glia to neurone ratio
are primary homoeostatic cells of the grey and rises to ~ 10:1 in the brain stem and ~ 6-7:1 in the
white matter (Verkhratsky and Nedergaard, 2018). human spinal cord (Verkhratsky and Butt, 2018;
Oligodendroglia are the myelinating cells of the von Bartheld et al. 2016). It is worth mentioning
CNS (in both gray and white matter), together that the largest white matter tracts, such as the cor-
with a significant population of adult oligoden- pus callosum of the brain and dorsal (posterior)
drocyte precursors (OPCs, also known as NG2 columns of the spinal cord, contain only glia and
glia, reflecting specific expression of nerve/glial axons being devoid of neuronal cell bodies
antigen 2), and perineuronal or satellite oligoden-
drocytes (Nave and Werner, 2014). Microglia are
myeloid cells that enter the embryonic neural tube
and ultimately transform into surveilling micro- ASTROGLIA
glial cells. Microglia in the CNS play numerous
physiological functions, participate in organisa-
tion of synaptic networks and provide for tissue Diversity and Morphological
defence and tissue innate immunity. Although Appearance
microglial cells are myeloid progeny and retain
multiple features of immune cells, they are pro- Astroglia are a class of CNS neural cells that
foundly adapted to the CNS environment and are are specialised for homeostasis and defence
82 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

(Verkhratsky and Nedergaard, 2018) (Figure 6.3). covers ependymocytes, which include the epend-
Astroglia comprise “true” astrocytes and many ymal cells lining the CNS ventricles, together
other related cells, ranging from radial glia with the choroid plexus cells and retinal pigment
to ependymal cells lining the CNS ventricles epithelial cells. Notably, the brains of humans
(Figure 6.4). As such, astroglial cells are highly and higher primates contain specific astrocytes
heterogeneous in form and function and demon- that are absent in other vertebrates: (i) interlami-
strate remarkable adaptive plasticity that defines nar astrocytes, with small somata (~10 μm in
the functional maintenance of the CNS in devel- diameter) in layer I of the cortex and one or two
opment, adulthood ageing and pathology very long (up to 1 mm) processes that extend
(Figure 6.5). The class of astroglia (Figure 6.4) through the cortex to end in layers III and IV;
includes (i) radial glia of the developing brain, (ii) polarised astrocytes, represented by uni- or
(ii) protoplasmic astrocytes of the grey matter, bipolar cells, the somata of which are located in
(iii) fibrous astrocytes of the white matter, (iv) layers V and VI of the cortex; and (iii) varicose
velate astrocytes of the cerebellum and supraoptic projection astrocytes in layers V–VI of the
nucleus; (v) perivascular, marginal, and surface- cortex, which send several (up to 5) long (up to
associated astrocytes responsible for formation 1 mm) unbranched processes that are endowed
of the glia limitans; and (vi) radial astrocytes, with evenly spaced varicosities and extend in
which include Müller glia of the retina, Bergmann all directions through the deep cortical layers
glia, and Fañanas glia of the cerebellum, radial (Colombo and Reisin, 2004; Oberheim et al.,
astrocytes of the supra-optic nucleus, tanycytes 2009). Human astrocytes are in general much
of the third ventricle and pituicytes of the poste- larger and much more complex compared to
rior pituitary, and radial stem astrocytes (also astrocytes of other mammals (Oberheim et al.,
known as neural stem cells) found within the 2009). Astrocytes account for 20–40% of all glial
neurogenic niches of the subventricular and sub- cells depending on the brain region (Verkhratsky
granular zones. In addition, class of astroglia and Nedergaard, 2018).

Figure 6.3 Functions of astroglia.


Source: Adapted from Verkhratsky and Nedergaard, 2018.
Role of Neuroglia in Cognition 83

Figure 6.4 Classification of astroglia.


Source: Adapted from Verkhratsky and Nedergaard, 2018.

Protoplasmic astrocytes are the main homeo- can possess one to several endfeet, which contact
static glia of the grey matter, providing structure the same or different blood vessels. Astrocytes
to the neuropil, establishing intimate contacts with localised in superficial brain regions form end-
synapses, and contributing to the regulation and feet on the parenchymal side of the pia mater (Liu
maintenance of synaptic transmission (Bushong et al. 2013). Astrocytic endfeet are highly plastic
et al., 2002). A single protoplasmic astrocyte in the structures containing smooth and rough endoplas-
rodent cortex contacts 4 to 8 neurones, surrounds mic reticulum, Golgi apparatus, and mitochondria
~300 to 600 neuronal dendrites and provides cover which support local biosynthesis contributing to
for up to 20 000–120 000 synapses, whereas in the the maturation of membranes, protein secretion
human brain a single protoplasmic astrocyte cov- and endfeet-vascular interactions (Boulay et al.,
ers ~ 2 million synapses (Oberheim et al., 2009). 2017). In addition, endfeet secrete vasoactive sub-
Processes of protoplasmic astrocytes are classified stances, which mediate astrocytic regulation of
(Semyanov and Verkhratsky, 2021) (Figure 6.6), local blood flow (MacVicar and Newman, 2015).
into (i) branches (main or primary, secondary, etc.
astrocytic processes), (ii) leaflets, which are thin
membranous extensions 2–10 μm in length and
~100–200 nm in thickness devoid of intracellular
organelles, and (iii) endfeet, either subpial (ter- ASTROCYTIC NETWORKS OR SYNCYTIA
minate beneath the pia mater) and perivascular
(terminate on blood vessels). The leaflets repre- Astrocytes and oligodendrocytes are intercon-
sent the major part of the surface area of an indi- nected into functional syncytia by gap junctions
vidual astrocyte, accounting for ~ 80% of the total to form a glial reticular network in the CNS. Gap
astroglial plasmalemma, although being exceed- junctions form intercellular channels and several
ingly thin they contribute only a minor fraction hundreds of gap junction channels assemble into
(~ 4–10%) to the cell volume (Reichenbach et al., large crystalline-like structures known as gap
2010). This is also reflected by an extremely high junction plaques. Individual gap junctions are
surface-to-volume ratio of leaflets 2–3 times formed by two aligned and closely apposed con-
larger than that of branches (Gavrilov et al., 2018). nexons of adjacent cell membranes (gap of ~ 2 nm),
Endfeet are specialised terminals of astrocytic to form the intercellular channel, which has
branches that form the glia limitans barrier at the a diameter between 6.5 and 15 Å and allows dif-
pia mater and brain vasculature. A single astrocyte fusion of molecules with M.W. up to 1 KDa
84 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 6.5 Types of astroglial cells. The figure shows protoplasmic astrocytes from
hippocampus, fibrous astrocytes from the optic nerve, Bergmann glia and Fañanas glia
of the cerebellum (cerebellar radial astrocytes), and radial stem astrocytes of the
subgranular zone neurogenic niche.

(Giaume et al. 2021). Each connexon comprises Cx47/Cx43 complexes. Astrocyte-neuronal het-
six subunits, known as connexins (Cx), which erocellular contacts are restricted to developing
comprise a large family of proteins. Homocellular and early postnatal brain.
astrocyte-astrocyte gap junctions are composed The concept of a panglial syncytium does not
from Cx26, Cx30, and Cx43, of which Cx43 presume an existence of a single “panbrain” net-
is the most abundant. Heterocellular gap junc- work connecting all glial cells, rather it envisages
tional channels connecting astrocytes with oli- regional connectivity of astrocytes, oligodendro-
godendrocytes are represented by Cx32/Cx30, cytes, and ependymocytes that brings them into
Role of Neuroglia in Cognition 85

Figure 6.6 Morphological classification of astrocytic processes. Three-dimensional


reconstruction of astrocyte loaded with fluorescent dye Alexa Fluo 594 through a
patch-pipette. The cell consists of soma with optically resolved branches of primary,
secondary, and higher orders. These branches host fine terminal leaflets that cannot
be resolved with diffraction-limited optical imaging and appear as a spongiform cloud.
Astrocytic leaflets occupy most of the astrocyte territory also known as an anatomic
domain. Two insets (left and right) are zooming onto branches and leaflets reconstructed
from serial section transmission electron microscopy (TEM) image stacks. Astrocytic
branches contain organelles, whereas leaflets are organelle-free. Leaflets are interspaced
with dendritic spines, filling the space between them like paper cushions around pears
packed for shipping. Thus, most synapses hosted by dendritic spines interact with
organelle-free leaflets, while individual leaflets may interact with several synapses.
Some dendritic spines, however, reside in the vicinity of astrocytic branches and soma,
and those synapses may have a different effect on astrocytic Ca2+ activity.
Source: Adapted from Semyanov and Verkhratsky, 2021.

a functional unit, for example that which pro- ION DISTRIBUTION, MEMBRANE
vides for K+ buffering in white matter (Giaume POTENTIAL, AND ION CHANNELS
et al., 2021). Panglial networks connecting astro-
cytes, oligodendrocytes, and ependymocytes are
functional in the thalamus, neocortex, and hip- Most mature astrocytes have a strongly negative
pocampus (Griemsmann et al., 2015). Astrocytic resting membrane potential (~ -80 to -90 mV),
syncytia are segregated into anatomical structures, reflecting the predominant resting K+ conduct-
for example, confined to individual barrels of the ance (Verkhratsky and Nedergaard, 2018),
somatosensory cortex, individual glomeruli in the which maintains the membrane potential close
olfactory bulb, or barreloid structures in the thala- to the potassium equilibrium potential (EK). The
mus (Giaume et al., 2010). Glial gap junctional ion content of astroglia differs from neurones in
connectivity may be regarded as a specialised that astroglial cells have substantially higher
“analogue” intercellular signalling system (alter- cytosolic Na+ (15–17 mM vs. ~ 8 mM in neu-
native to “binary” inter-neuronal communication), rones) and Cl- (30–60 mM vs. ~ 10 mM in
which by utilising intercellular diffusion of multi- mature neurones), whereas the concentration of
ple molecules, can provide another level of infor- K+ (~120–140 mM) and Ca2+ (<0.0001 mM) in
mation processing within the CNS. astroglia are generally the same as in neurones
86 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

(Verkhratsky and Nedergaard, 2018). The rever- release responsible for synaptic connectivity.
sal potential for Cl- is ~ –40 mV and activation Astrocytes are electrically non-excitable cells
of Cl- permeable channels (e.g., GABAA recep- that do not generate action potentials in response
tors) triggers Cl- efflux and depolarisation of to stimulation, and yet they possess a specific
astrocytes (whereas in neurones, this results in form of excitability. Astroglial excitability (similar
Cl- influx and hyperpolarisation). Astrocytes to other glial cells) is based on spatio-temporally
express all major types of ion channels, includ- organised fluctuations of intracellular ion con-
ing voltage-gated K+, Na+, and Ca2+ channels centrations and intracellular (second) messen-
(in densities not sufficient to produce an action gers (Verkhratsky et al., 2020). Moreover, the
potential), non-selective cation channels and gap junctional pathway allows propagating
various types of anion channels (Verkhratsky waves of intracellular messengers, ions, and
and Nedergaard, 2018). These channels gener- metabolites, which endows astroglial syncytia
ate ionic fluxes which contribute to astrocyte with a mechanism for intercellular long-range
intracellular signalling (see below). signalling (Bernardinelli et al., 2004; Langer
et al., 2012; Scemes and Giaume, 2006).
Physiological stimulation of astrocytes produces
transient changes in all major ions, including
RECEPTORS TO NEUROTRANSMITTERS, Ca2+ Na+, Cl-, K+, and H+, each of these ions can
mediate relevant signaling. In astrocytes, the
NEUROMODULATORS, AND intracellular signaling role for Ca2+ is well docu-
NEUROHORMONES mented (Bazargani and Attwell, 2016), for Na+ is
becoming generally recognised, for Cl- has been
Astroglia, being neural cells, are potentially capa- proposed, and for K+ is yet to be considered
ble of expressing all receptors to neurotransmitters, (Verkhratsky et al., 2020).
neurohormones, and neuromodulators as neurones Astroglial Ca2+ signal generation is spatially
do, which permits astroglial cells to perceive their segregated (Figure 6.7). In the soma and branches
neurochemical environment and sense signaling of protoplasmic astrocytes, Ca2+ signals primarily
molecules released by neurones and other cells in originate from endoplasmic reticulum Ca2+ release
the nervous tissue (Verkhratsky, 2010). Glia, simi- (Lim et al., 2021). The endoplasmic reticulum is a
lar to neurones, are endowed with both ionotropic dynamic Ca2+ store capable of accumulating Ca2+
and metabotropic receptors. ions through the activity of specific Ca2+ pumps
The complement of receptors expressed in of sarco-endo-plasm reticulum (SERCA) type and
astroglia varies between different brain regions. release Ca2+ through ER Ca2+ channels, which in
As a rule of thumb, the modality of neurotrans- astroglia are mainly represented by inositol 1,4,5
mitter receptors expressed by astroglial cells triphosphate receptor (InsP3R) type II. The activity
matches that of their neuronal neighbors and is of InsP3Rs is controlled by the second messenger
most likely controlled by the local neurotransmit- InsP3 which is produced following the activation
ter environment (Verkhratsky and Nedergaard, of numerous G-protein coupled (also known as
2018). For example, astrocytes express glycine metabotropic) receptors linked to phospholipase
receptors in the spinal cord, where glycine acts C as well as by cytoplasmic Ca2+ concentration.
as a main inhibitory mediator, whereas in basal Depletion of the endoplasmic reticulum Ca2+ store
ganglia, which utilise dopaminergic transmission, triggers plasmalemmal Ca2+ entry through store-
astrocytes express dopamine receptors. Therefore, operated Ca2+ channels (Verkhratsky and Parpura,
the expression of astroglial receptors in vivo is 2014). Changes in cytosolic Ca2+ activate various
regulated by neurochemical input, which makes intracellular enzymes (also known as Ca2+ sen-
astrocytes perceptive to signals specific for each sors) that initiate or regulate cellular responses.
particular region of the brain. Waves of elevated Ca2+ can propagate through the
astroglial syncytium over long distances, either by
intercellular diffusion of InsP3 or by the release of
neurotransmitters such as ATP. These Ca2+ waves
are thought to represent long-range signalling
INTRACELLULAR EXCITABILITY within astroglial networks and thus be function-
OF NEUROGLIA ally analogous to action potential in neurones
(Scemes and Giaume, 2006).
Neurones are universally considered as the only In the leaflets of protoplasmic astrocytes,
excitable cells of the nervous system; they gener- devoid of endoplasmic reticulum, Ca2+ signals
ate fast action potentials, which are conducted originate from plasmalemmal Ca2+ influx through
over large distances and initiate neurotransmitter Ca2+ permeable ionotropic receptors (Palyginet al.,
Role of Neuroglia in Cognition 87

Figure 6.7 Morpho-functional organization of Ca2+ signalling compartments in protoplasmic


astrocyte. Calcium signaling in soma and branches is mainly associated with Ca2+ release
from the endoplasmic reticulum (ER) with subsequent store-operated Ca2+ entry (SOCE). This
Ca2+ release is mediated by InsP3 receptors (InsP3R); InsP3 is synthesized by phospholypase C
(PLC) linked to G-protein metabotropic receptors. Calcium signaling in the leaflets is associ-
ated with Ca2+ entry through ionotropic receptors (NMDA glutamate receptors or P2X puri-
noceptors) or Ca2+-permeable channels (such as, for example, TRPA1 channels). Plasmalemmal
Ca2+ influx can also be mediated by the Na+/Ca2+ exchanger (NCX) operating in reverse mode.
Levels of free Ca2+ in the cytosol are also controlled by Ca2+-biding proteins (CBP), also
known as Ca2+ buffers.
Source: From Verkhratsky et al., 2020.

2010), transient receptor potential (TRP) chan- various types of cationic channels (e.g., iono-
nels (Shigetomi et al., 2012) or reversed Na+/Ca2+ tropic receptors such as AMPA receptors or TRP
exchange (Rose et al., 2020). Calcium signals in channels), or from the activity of Na+-dependent
leaflets, with their high surface-to-volume ratio, transporters, most notably excitatory amino acid
allows larger net plasma membrane Ca2+ influx (e.g., glutamate) transporters. Physiological acti-
and hence relatively larger (Ca2+)i fluctuations vation of Na+ fluxes may increase cytosolic Na+
within these tiny compartments (Semyanov and concentration by 10–20 mM. The transmembrane
Verkhratsky, 2021). Na+ gradient regulates many homeostatic molecu-
Another mechanism of astroglial ionic signal- lar cascades expressed in astrocytes, including
ling is based on rapid fluctuations in cytosolic Na+/K+ ATPase, Na+/Ca2+ exchanger, transporters
Na+ concentration (Rose and Verkhratsky, 2016). for glutamine, glutamate, and GABA, proton and
These (Na+) transients result from activation of bicarbonate transporters, transporters for ascorbic
88 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

acid, Kir4.1 potassium channels, etc. (Verkhratsky (Verkhratsky et al., 2020). In addition, through
and Rose, 2020). Functionally, Na+ signalling is programs of reactive gliosis, astrocytes act as
fundamental for coordination of astroglial homeo- principal cellular elements of CNS defence
static responses with neuronal activity. (Escartin et al., 2021).
The metabotropic branch of astroglial inter- Astrocytes are major elements of extracellu-
cellular excitability is mediated by second mes- lar K+ homeostasis (MacAulay, 2020). Neuronal
sengers, most notably by cyclic AMP (cAMP), activity is associated with influx of Na+ and Ca2+
which mediates astroglial responses to noradrena- (depolarisation) and efflux of K+ (repolarisation);
line released from varicosities of catecholamin- extracellular K+ accumulation may significantly
ergic projections from locus coeruleus neurones alter neuronal excitability. The main system
(Verkhratsky et al., 2020). responsible for K+ removal form the extracellular
space is associated with astrocytes and is repre-
sented by local K+ uptake and spatial K+ buff-
ering (MacAulay, 2020). Local K+ uptake is the
main mechanism of K+ clearance in physiologi-
FUNCTIONS OF ASTROGLIA cal conditions. Extracellular K+ is accumulated
by Na+/K+ ATPase (which in astrocytes contains
The main functions of astrocytes are summarized a specific α2 subunit highly sensitive to extra-
in Table 6.1. Conceptually, astroglial cells are cellular K+ concentration (Larsen et al., 2014));
responsible for every possible homeostatic task after cessation of neuronal activity K+ is released

Table 6.1 Main functions of astrocytes

Development of the CNS • Neurogenesis


• Neural cell migration and formation of the layered gray matter
• Synaptogenesis
Structural support • Parcellation of the gray matter through the process of tiling
• Delineation of the pia matter and vessels by perivascular glia
• Formation of astroglio-vascular unit
Barrier function • Regulation of the formation and permeability of blood-brain and CSF-brain
barriers
• Formation of glial-vascular interface
• Covering brain capillaries with endfeet
• Forming the fenestrated blood-brain barrier in hypothalamus allowing
neurosecretion.
Homeostatic function • Control over extracellular K+ homeostasis through local and spatial buffering
• Control over extracellular pH
• Regulation of water transport
• Removal of neurotransmitters from the extracellular space
Metabolic support • Uptake of glucose; deposition of glycogen
• Providing energy substrate lactate to neurones in activity dependent manner
Synaptic transmission • Regulation of synapses maintenance and assisting in synaptic pruning
• Providing glutamate for glutamatergic transmission (through de novo synthesis
and glutamate-glutamine shuttle)
• Regulating synaptic plasticity
• Integrating synaptic fields
• Providing humoral regulation of neuronal networks through secretion of
neurotransmitters and neuromodulators
Regulation of blood flow • Regulation of local blood supply (functional hyperaemia) through secretion of
vasoconstrictors or vasodilators
Brain defence, neuroprotection • Isomorphic and anisomorphic reactive astrogliosis
and post injury remodeling • Scar formation
• Catabolizing ammonia in the brain
• Immune responses and secretion of pro-inflammatory factors (cytokines,
chemokines, and immune modulators)

Source: Adapted from Verkhratsky and Butt, 2013.


Role of Neuroglia in Cognition 89

Figure 6.8 Glymphatic system. Convective fluxes of cerebrospinal fluid (CSF) and interstitial
fluid (ISF) propel the waste products of neurone metabolism into the paravenous space,
from which they are directed into lymphatic vessels and ultimately return to the general
circulation for clearance by kidney and liver.

from astrocytes through inward rectifying K+ flow in response to a local increase in neuronal
channels and shuttled back to neurones to restore activity (MacVicar and Newman, 2015). These
ionic gradients. Higher K+ loads, mainly associ- changes occur in small vessels within ~200–250 μm
ated with pathology, can be redistributed through from the site of increased neuronal activity.
the astroglial syncytium by diffusion through Glutamate, released during neuronal activity, acts
gap junctions (spatial K+ buffering (Bellot-Saez on astrocytic receptors, and induces Ca2+ signals.
et al., 2017)). Subsequently, excessive K+ ions These pass to astroglial perivascular endfeet and
exit astrocytes into the interstitium or into the initiate the release of vasoactive substances, which
perivascular space from where they are removed regulate the tone of small arterioles and/or capil-
to the blood. laries; astrocytes are capable of secreting both
Astroglial cells are critical for water move- vasoconstrictors and vasodilators (MacVicar and
ments in the CNS: water channels, or aquapor- Newman, 2015).
ins, are concentrated in astroglial endfeet and in Astrocytes are also important elements of
perisynaptic processes; water entering astrocytes metabolic support of neurones during periods
is redistributed through gap junctions connecting of intense activity. Astrocytes are able to syn-
astroglial syncytia (Nagelhus and Ottersen, 2013). thesize glycogen which acts as a CNS energy
Astroglial aquaporins also support the operation reserve (Bak et al., 2018; Matsui et al., 2017).
of the brain glymphatic system (brain analogue of In astrocytes, glucose is converted into pyruvate
the lymphatic system responsible for collection of and then into lactate in the presence of oxygen
waste – see Figure 6.8; Hablitz and Nedergaard, (aerobic glycolysis). Glutamate released during
2021). Endfeet of parenchymal astrocytes cre- synaptic activity is accumulated by perisynaptic
ate the paravascular channels that provide for an astrocytic compartments though Na+-dependent
exchange between the interstitial fluid and the glutamate transporters thus leading to an
cerebrovascular fluid thus providing the drainage increase in cytosolic Na+ concentration. This, in
system for removing waste, which accumulates turn, activates the Na+-K+ ATPase, which expels
in the brain parenchyma as a result of cellular the excess Na+ into the extracellular space, and
metabolism. stimulates phosphoglycerate kinase (PGK) to
Through neuro-glio-vascular units (which inte- trigger aerobic glycolysis that produces lactate.
grate parenchymal cellular elements and blood Lactate is then released into the extracellular
vessels (Kugler et al., 2021)), astrocytes contrib- space and finally taken up by neurones and used
ute to the regulation of local blood flow, being at as an energy substrate, this process being known
least in part responsible for functional hyperae- as astrocyte-neurone lactate shuttle (Pellerin and
mia, represented by a rapid rise in the local blood Magistretti, 2012; Veloz Castillo et al., 2021).
90 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Astroglial Homeostatic Transporters major wave of emergence of glutamatergic syn-


apses in the mammalian brain occurs in early
Astrocytes are actively engaged in the transport of postnatal period, immediately after massive
ions, neurotransmitters, hormones, various trans- astrogliogenesis.
mitter and hormonal precursors, energy substrates, Astrocytic membranes cover the majority
amino acids, etc. Most of this transport is accom- of synapses in the CNS; in the hippocampus,
plished by plasmalemmal transporters, genes for about 60% of all synaptic structures are closely
which are the most represented in the astrocytic enwrapped by astroglia (Bernardinelli et al., 2014;
transcriptome. Membrane transporters are gener- Ventura and Harris, 1999). A large area of peri-
ally classified into (i) ABC transporters (Rees et al., synaptic leaflet membrane provides ample space
2009), (ii) pumps or ATP-dependent transporters for receptors, channels, transporters, and pumps
(Apell, 2004), which hydrolyze ATP and use the that contribute to neuronal-glial signalling and are
released energy to move molecules against their responsible for homeostasis in the cleft (Figure 6.9).
electro-chemical gradients (e.g., Na+-K+-pumps) The perisynaptic membranes contain multiple
and (iii) solute carrier transporters (SLCs) neurotransmitter receptors, which are activated
(Pizzagalli et al., 2021), which can be secondary by neurotransmitters released during synaptic
active, utilising the electrochemical gradients of transmission and create localised transient Ca2+
different ions generated by the activity of pumps to and Na+ signals, also known as microdomains
transport ions or other small molecules (e.g., Na+- (Verkhratsky and Nedergaard, 2014).
Ca2+ exchangers, NCX), or facilitative, in which Astrocytes control the extracellular distribu-
solutes move passively down their concentration tion of glutamate and maintain neuronal pools
gradients (e.g., glucose transporters, GLUT). of glutamate and GABA by supplying glutamine
Transporters can be uniporters, which transports a (Figure 6.10). Glutamate concentration in the
single species of substrate (e.g., GLUT, Ca2+ synaptic cleft is controlled by astrocytes through
pumps), co-transporters or symporters, which Na+-dependent astroglia-specific glutamate trans-
transport two or more different substrates in the porters EAAT1 and EAAT2, which maintain low
same direction (e.g., excitatory amino acid trans- glutamate concentration at rest and rapidly remove
porter 1, EAAT1, responsible for astroglial gluta- glutamate released during synaptic transmission
mate uptake, and Na+/glucose cotransporter, (Vandenberg and Ryan, 2013; Zhou and Danbolt,
SGLT), and antiporters or exchangers, which trans- 2013). After being taken up into astrocytes glu-
port two or more different substrates in opposite tamate is converted to glutamine, which subse-
directions (e.g., NACX). The membrane transport- quently is transported to neurones where it acts as a
ers expressed in astrocytes accomplish various precursor for glutamate and for glutamine (GABA)
functional roles and are represented by: (i) trans- glutamate shuttle (Hertz, 2013)). Astrocytes also
porters responsible for ion homeostasis or ionosta- control the extracellular concentration of gly-
sis; (ii) neurotransmitter transporters; (iii) cine and are primary regulators of adenosine
transporters of neurotransmitter precursors; and levels in the CNS through the astroglial adeno-
(iv) metabolite transporters (Ugbode et al., 2017; sine cycle (Boison et al., 2010; Eulenburg et al.,
Verkhratsky and Rose, 2020). 2010). Adenosine is transported to astroglia by
equilibrate ENT1 or concentrating Na+-dependent
transporters CNT2,3. Adenosine subsequently is
catabolised intracellularly by adenosine kinase
that in the CNS is expressed exclusively in astro-
ASTROGLIA AND SYNAPTIC cytes (Boison et al., 2010). Astrocytes also act as
TRANSMISSION a main sink for catecholamines, accumulated by
Na+-dependent norepinephrine transporter NET;
Synaptic structures lie at the very core of informa- catecholamines subsequently are catabolised by
tion transfer and processing in the nervous system. monoaminoxidase-B highly expressed in astro-
Astrocytes are fundamental elements of synaptic cytes (Saura et al., 1992).
connectivity being involved in synaptic formation,
evolution, and maintenance (Augusto-Oliveira
et al., 2020). These multiple roles are generalized
in the concept of the astroglial cradle (Figure 6.9)
GLIOCRINE SYSTEM: ASTROCYTES AS
(Verkhratsky and Nedergaard, 2014). Astrocytes
contribute to synaptic connectivity through regu- SECRETORY CELLS OF CNS
lation of (i) synaptogenesis; (ii) maturation of the
synapse; (iii) synaptic stabilization and mainte- Astrocytes are capable of releasing numerous
nance; and possibly (iv) synaptic elimination. The neuroactive substances that regulate various
Role of Neuroglia in Cognition 91

Figure 6.9 Astroglial cradle and glial homeostatic support of the CNS synapse. A: The
majority of synapses in the brain and spinal cord are multi-partite being composed of (i) the
presynaptic terminal; (ii) the postsynaptic dendritic compartment; (iii) the perisynaptic
process of the astrocyte; (iv) the process of neighboring microglial cell that periodically
contacts the synaptic structure; and (v) the extracellular matrix (ECM) present in the
synaptic cleft and also extended extra-synaptically. Astroglial perisynaptic membrane
contains numerous transporters that control homeostasis in the synaptic cleft. B: Perisynaptic
glial membrane is densely packed with numerous homeostatic transporters that maintain
synaptic transmission by controlling ionostasis, removing neurotransmitters, and supplying
neuronal terminals with neurotransmitter precursors.
Abbreviations: EAAT – excitatory amino acid transporters 1 (SLC1A3) and 2 (SLC1A2); NKA – the Na+/K+ ATP-ase or
ATP-dependent Na+/K+ pump, the α2 subtype (ATP1A2) is predominantly expressed in astrocytes; NKCC1 – the Na-K-Cl
co-transporter (SLC12A2); NCX – the sodium-calcium exchanger expressed in 3 isoforms (SLC8A1, SLC8A2 and SLC8A3);
NAAT – the Na+-dependent ascorbic acid transporter (SLC23); NBC – the sodium-bicarbonate co-transporter (SLC4A4);
CNT2 – the high-affinity Na+-dependent concentrative adenosine transporter (CNT2); ASCT2 – the alanine-serine-cysteine
transporter 2; MCT-1 – the monocarboxylase transporter 1 (SLC16A1); Kir4.1 – inward rectifier Kir4.1 channels; NHE – the
sodium-proton exchanger 1 (SLC9A1); GAT – GABA transporters 1 (SLC6A1) and 3 (SLC6A11); SN1,2 – Na+/H+ dependent
sodium coupled neutral amino acid transporters 1 (SLC38A3) and 2 (SLC38A5); GlyT1 – glycine transporter 1 (SLC6A9).
Source: A: adapted from Verkhratsky and Nedergaard, 2014.

aspects of neuronal activity and also signal to waves, and finally release extracellular signalling
other cells in the CNS (Verkhratsky et al., 2016). molecules (Araque et al., 2014). The concept of
Astrocytes can release (i) classical neurotrans- regulated neurotransmitter release from astro-
mitters (such as glutamate, ATP, and GABA), cytes is generally known as gliotransmission
(ii) neuromodulators (taurine or kynurenic acid), (Savtchouk and Volterra, 2018). Astrocytes
(iii) neurotransmitter precursors such as glutamine release neuroactive substances through several
or L-serine, (iv) hormones, and (v) trophic and molecular pathways which include Ca 2+-
immunocompetent agents such as growth factors regulated exocytosis, diffusion through several
or cytokines. The discovery of astrocytic release types of membrane channels (connexon
of neuroactive compounds has been of funda- hemichannels or anion channels), membrane
mental importance to our views on the role of transporters and recently discovered ectosome
these cells in the nervous system. Indeed, this shedding, which represents shedding of
implies a more active role for astroglial networks microvesicles from the plasma membrane. The
in the CNS, since they can receive information vesicles contain lipids, cell surface proteins, and
from neurones by neurotransmitter receptors, material from the cytoplasm or nucleus of the
convert this information into an alternative form cell, which all can be employed for intercellular
of long-range excitability in the form of Ca2+ communication (Venturini et al., 2019).
92 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 6.10 Glutamate (GABA)-glutamine shuttle. Astrocytes take up (by glutamate


transporters EAAT1/2) glutamate released during synaptic transmission. Glutamate is
converted to glutamine (by glutamine synthetase, GS, highly expressed in astroglial
cells), which is shuttled to neurons for subsequent conversion into glutamate and
GABA.

OLIGODENDROGLIA AND NG2 GLIA FUNCTIONAL PROPERTIES OF


OLIGODENDROCYTES
Oligodendrocytes are specialised to form the
insulating myelin sheaths around axons in the Oligodendrocytes, similar to all other glial cells,
CNS (Baumann and Pham-Dinh, 2001). The are non-excitable. They maintain a negative rest-
myelin sheath is an insulating layer composed of ing membrane potential of about -80 mV; this
lipids and myelin proteins that provides for the resting potential is mainly set up by inward recti-
rapid conduction of nerve impulses and for the fier K+ channels, which are abundantly expressed
miniaturisation of the vertebrate CNS. in mature oligodendrocytes. Oligodendroglial
Oligodendrocytes in the CNS send multiple pro- precursor cells (OPCs) also express anion chan-
cesses and myelinate up to 30 axons located in nels and voltage-gated ion channels for Na+ and
their vicinity. Along the length of an axon, Ca2+, which makes some of them excitable.
myelin sheaths are interrupted by nodes of Activation of voltage-gated Na+ and Ca2+ chan-
Ranvier, the highly specialised region of the nels in immature oligodendrocytes may contrib-
axons where action potentials are generated. The ute to initiation of myelination by detecting
myelinated segments between nodes are called active axons (the latter release K+ in the process
internodes and are connected to the cell body by of electrical activity and local increases in K+
a process that contacts each internodal myelin may depolarise oligodendroglial processes and
sheath (or myelin segment) at its approximate trigger cationic influx that signals to the cell
midpoint, giving the oligodendrocyte a symmet- interior).
rical appearance (Baumann and Pham-Dinh, Oligodendrocytes and their precursors express
2001; Verkhratsky and Butt, 2013). many types of neurotransmitter receptors including
Role of Neuroglia in Cognition 93

receptors to glutamate, ATP, adenosine, acetylcho- incipient internodal segments, inducing axon radial
line, GABA, glycine, and dopamine (Butt et al., growth and establishes nodes of Ranvier; (4) matura-
2019; Nishiyama et al., 2021). These receptors tion phase, in which axons and myelin sheaths within
are activated by neurotransmitters released from units undergo interdependent growth to establish
axons (which secrete glutamate and ATP in the adult dimensions of axon diameter, myelin sheath
process of action potential propagation), as wells g-ratios, and intermodal lengths; (5) late developing
as by neurotransmitters released from neurones oligodendrocytes fill unmyelinated gaps along axons,
and astrocytes. Oligodendrocytes are also cou- as part of normal growth into adulthood, as well as in
pled to each other and to astrocytes through gap response to remodelling and demyelination or remy-
junctions. These integrate oligodendrocytes into elination. Development myelination starts prenatally
neurochemical assemblies of brain cells (Giaume and lasts well into adulthood (in humans it is com-
et al., 2021). pleted by 30 years of age or even later).

MYELINATION OLIGODENDROCYTE PRECURSORS


(NG2-GLIA)
The main constituents of myelin are lipids (70% of
its dry weight) and proteins (30% of the dry Oligodendrocyte precursors (OPC), also known as
weight); cholesterol is a major component of NG2-glia, are identified by their expression of the
myelin (27%) and lack of cholesterol during devel- chondroitin sulphate proteoglycan NG2 (Butt
opment can cause hypomyelination. Formation and et al. 2005; Nishiyama et al., 2021). These cells
maintenance of nodal structure is a key function of are present in the CNS throughout life. The OPCs
oligodendrocytes, and essential for directing axonal receives direct synaptic contacts from neurones.
potassium and sodium channels, which generate The OPCs bear several primary processes; the
the action potential, at the nodes of Ranvier (Nave process-delineated domains have a certain degree
and Werner, 2014). In addition, oligodendrocytes of overlap. In gray matter, OPCs are distributed as
provide metabolic support for the axons they mye- a “mosaic,” whereas in the white matter, OPCs
linate by releasing lactate into the periaxonal space have an elongated appearance, extending pro-
(Philips and Rothstein, 2017); metabolic support is cesses along the axonal axis. OPCs constitute
coupled to axonal activity through glutamate, 8–10% of total glia.
which activates oligodendroglial NMDA receptors. The primary functional role of OPCs is the life-
The formation of the myelin sheath is a highly long generation of oligodendrocytes, although they
complex process that involves multiple steps, from may also subserve other unresolved functions,
mRNA transcription to protein translation and for example at synapses (Nishiyama et al., 2021).
assembly into the membranes (Nave and Werner, OPCs slowly divide in the adult brain to produce
2014). All of the myelin products have to be trans- daughter cells, one of which provides for self-
ported from the cell body and targeted to the maintenance of the OPC population, and the other
“workface” of the myelin sheath, over hundreds differentiates into an oligodendrocyte under the
and potentially thousands of microns. Notably, control of multifarious factors, helping to ensure
disruption of myelination not only results in the that disruption of any single factor does not result
loss of action potential propagation, but also axonal in their loss of function. OPCs can also be reactive
degeneration. and respond to CNS insults by increased prolifera-
Myelination involves a number of axoglial recog- tion and morphological remodelling, manifested by
nition and adhesion events that regulate the produc- shortening and thickening of cellular processes and
tion of myelin-related gene products and radial axon a strong upregulation of NG2 expression. OPCs
growth. Myelination of axons occurs in a series of generate new oligodendrocytes in the post-lesion
distinct axoglial interdependent phases (Verkhratsky period, thus contributing to tissue regeneration.
and Butt, 2013): (1) axonal contact and recognition
by OPCs; (2) induction phase, in which OPCs extend
initiator processes along receptive axons to form
short ensheathing segments, triggering the differenti-
ation of OPCs into premyelinating oligodendrocytes MICROGLIA
and the initiation of axonal ion channel clustering;
(3) remodeling phase, in which non-myelinating Microglial cells are commonly perceived as resi-
processes within the unit are lost, and radial and dent macrophages of the brain and the spinal cord
longitudinal growth of myelinating processes forms that define the innate immunity of the central
94 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

nervous system (Kettenmann et al., 2011; Tay cells account probably for ~ 10% of all neuroglia,
et al., 2019). This broad designation of microglia and their densities are similar in different CNS
seems to be in need of further elaboration, regions. Migration of microglial precursors is
because, as shall be discussed below, microglial accompanied by a remarkable morphological and
cells undergo specific adaptation to the CNS envi- functional metamorphosis: the myeloid progeni-
ronment and become a legitimate member of the tors, generally similar to monocytes, convert into
brain cellular architecture with numerous physio- microglial cells, which acquire “neural-like”
logical functions that far exceed and complement appearance. Differentiated microglial cells have a
their immune capabilities (Garaschuk and small cell body (~ 4–5 μm) and several thin and
Verkhratsky, 2019a). For multiple functions of long processes with characteristic terminal arbori-
microglia, see Table 6.2 and Figure 6.11. sation represented by numerous small secondary
processes (Augusto-Oliveira et al., 2021).
Microglial processes are in continuous motion
(thus being defined as ‘surveilling’ microglia),
moving through the surrounding CNS tissue at a
MORPHOLOGICAL PROPERTIES OF speed of ~0.7–1.5 μm/min. This movement of
MICROGLIA microglial processes is accompanied by regular
extension/retraction (at 2–3 μm/min) of small pro-
After entering the CNS, microglial precursors trusions. Thus, microglial processes constantly
migrate and disseminate almost homogeneously survey their neighbourhood. Microglial processes
throughout the parenchyma of the brain and spinal also define a territorial domain of a single micro-
cord (Ginhoux et al., 2013). In total, microglial glial cell. There is generally little overlap between

Table 6.2 Diversity of microglial functions

Physiological
CNS development • Early synaptogenesis (?)
• Phagocyting redundant/post-apoptotic neurones
• Pruning unwanted/redundant/silent synapses
• Secreting trophic factors (e.g., cytokines, growth factors or neurotrophins)
Neuronal plasticity • Monitoring synapses
• Regulating synaptic plasticity/connectivity through secretion of cytokines, and/or
other factors
• Secretion of lipoprotein particles for maintenance of cell membranes and synapses
Defensive
Recognition of pathogens • Sensing pathogen-associated molecular patterns (PAMPs) through Toll-like receptors
• Recognition of damage through danger-associated molecular patterns through
purinoceptors
Phagocytosis: • Ingestion and destruction by digestive enzymes in lysosomes of:
(a) damaged cells, e.g., neurones (e.g., neuronophagia or Wallerian degeneration);
(b) micro-organisms (e.g., abscess); (c) virally infected cells (e.g., herpes simplex
encephalitis); (d) erythrocytes following haemorrhage
Antigen presentation • Presentation of pathogens (e.g., in bacterial, fungal, viral infections) bound to the
major histocompatibility complex (MHC) for activation of T lymphocytes
• Recognition of bound antibody (adaptive immune function)
Immune response • Secretion of pro-inflammatory factors e.g., chemokines or interferon-γ
Repair • Remodeling of extracellular matrix; Regulation of stem cell proliferation (e.g., granule
cell of hippocampus)
Pathological
Cytotoxicity • Secretion of glutamate; reactive oxygen species/respiratory burst
Tumour growth promotion • Secretion of matrix metalloproteinase
Demyelination • Myelin destruction/phagocytosis in e.g., multiple sclerosis
Infection • Viral entry into CNS; Hosting of HIV-1; Support mycobacteria, including their
intracytoplasmic survival (e.g., tuberculosis)
Role of Neuroglia in Cognition 95

Figure 6.11 Main physiological functions of microglia. Microglia contribute to many


physiological functions in nervous tissue. Through synaptic pruning, microglia assist in
shaping synaptic connectivity; through secretion of numerous factors, microglia interact
with astrocytes and regulate synaptic transmission; microglia influence myelination in early
development and re-myelination in pathology; microglia support angiogenesis; microglia
phagocytose and clear the nervous tissue of apoptotic neurones and other cellular debris,
and microglia regulate adult neurogenesis.
Abbreviations: CD95L: CD95(Fas/APO-1) ligand, a 40-kd type II membrane protein and member of the tumor necrosis factor
superfamily of cytokines; TNF-α: Tumor necrosis factor-α; VEGF: Vascular endothelial growth factor.

microdomains of microglia. Given the speed of ION SIGNALING


processes movement the microglial domain could
be completely scanned by these processes within
After entering the CNS environment, microglial
several hours. Microglial cells show a remarkable
degree of heterogeneity, both molecular and struc- cells undergo profound physiological remodeling
tural, between and within different brain regions as and acquire a receptive phenotype compatible
well as at different developmental states (Stratoulias with the chemical environment. Arguably, rami-
et al., 2019); some physiological microglial mor- fied microglia is the most “receptive” cells of the
photypes are presented in Figure 6.12. CNS because they possess multiple receptors to
96 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 6.12 Some physiological microglial moprhotypes.


Source: From Augusto-Oliveira et al., 2021.

neurotransmitters and neuromodulators as well as (Verkhratsky et al. 2009). Microglial cells consti-
“immuno-competent” receptors characteristic for tutively express P2X7 receptors that contribute to
myeloid cells (Garaschuk and Verkhratsky, numerous responses to neuropathology. The P2X7
2019b). These latter receptors include P2X7 puri- receptors are a general feature of immune cells
noceptors, receptors to chemokines and cytokines, where they mediate various immune reactions,
and receptors to various tissue mediators such as including the processing and the release of vari-
platelet-activating factor, thrombin, histamine, or ous cytokines (Janks et al. 2018). The P2X7 recep-
bradykinin all being critical for triggering various tors are activated in conditions of massive ATP
types of immune responses. release associated with neuronal damage and are
Most electrophysiological experiments on linked to immune responses of microglia, being
microglia were performed in vitro in culture; particularly important activators of cytokines
these experiments consistently reported resting release. Incidentally, direct over-expression of
potentials of ~ -30 mV and therefore much more P2X7 receptors in microglia is sufficient to trigger
positive than those of neurones and astroglia, but their activation in the in vitro system in complete
similar to that of other immune cells. In healthy absence of any other exogenous factors. Microglia
brain, ramified microglia possess very few ion also constitutively express P2X4 receptors (that
channels, but upon activation the density of func- are activated by low μM ATP concentrations),
tional ion channels increases considerably. which are critically involved in mediating micro-
Microglial cells express almost all types of glial activation in conditions of chronic pain. In
receptors to neurotransmitters and neuromodula- addition, microglia express P2Y2, P2Y6, P2Y12,
tors so far found in the nervous system, includ- and P2Y13 metabotropic purinoceptors, which
ing receptors to glutamate, purines and GABA are mainly linked to Ca2+ signalling (Verkhratsky
(Garaschuk and Verkhratsky, 2019b; Tay et al., et al., 2009). Whereas the UTP-sensitive P2Y6
2019). The purinoceptors (adenosine receptors, receptors regulate microglial phagocytosis,
ionotropic P2X and metabotropic P2Y purinocep- ADP-preferring P2Y12 receptors are fundamen-
tors) are, arguably, the most abundant in microglia tal for initiating the acute reactive microgliosis
Role of Neuroglia in Cognition 97

Table 6.3 Pathophysiology of neuroglia


Pathology/Disease Neuroglial contribution

Traumatic brain/spinal cord injury Activation of astrocytes and microglia; formation of astroglial scar isolating the
area of damage; removal of cellular debris through microglial phagocytosis;
promotion of post-traumatic regeneration.
Ischaemia/Stroke Activation of astrocytes and microglia; metabolic support and neuroprotection in
the penumbra; at the same time overactivated astrocytes and microglia can
contribute to neuronal death and infarct propagation; formation of post-stroke
astroglial scar; facilitation of post-lesion regeneration.
Neurodevelopmental disorders/Autistic spectrum disorders
Rett syndrome In Rett syndrome expression of mutant MECP2 gene in astrocytes disrupt their
supportive function and leads to abnormal development of dendrites.
Fragile X syndrome Fragile X mental retardation protein expressed in astrocytes may alter their
supportive function and contribute to pathological synaptic connectivity.
Infectious diseases
Bacterial meningoencephalities Activation of microglia, which can, depending on the degree of activation, be
either protective (through elimination of pathogens) or toxic (through massive
release of pro-inflammatory factors and toxic substances such as NO and
reactive oxygen species).
HIV and HIV-associated dementia Microglia are the primary target; virus infects exclusively microglial cells; which
release pro-inflammatory factors, that activate astrocytes, which in their turn
induce neurotoxicity.
Demyelinating disorders
Multiple sclerosis Degeneration of oligodendrocytes lies at the core of myelin disruption and
aberrant action potential propagation. Pathologically activated microglia are
thought to induce oligodendroglial damage.
Toxic encephalopathies
Poisoning with metals (Manganese, Astrocytes are primary target; they preferentially accumulate metals via specific
Aluminium, Lead) or transporters, this down-regulates expression of glutamate transporters,
metylmercury glutamine synthetase, and possibly GABA transporter. Failure in glutamate
homeostasis underlies excitotoxic neuronal damage and death.
Wernicke encephalopathy (Korsakoff Astrocytes are primary target and key component of pathology; the disease is caused
syndrome) by severe (up to 70%) down-regulation of astroglial expression of glutamate
transporters, which leads to accumulation of extracellular glutamate followed with
massive neuronal excitotoxicity and neuronal death in thalamo-cortical regions.
Hepatic encephalopathy Liver failure causes hyperammonemia and rapid increase in ammonium
concentration in the brain. Astrocytes are primary target, being the main
system for ammonia removal and utilisation though glutamine synthetase.
Increased accumulation of ammonia in astroglial cells triggers metabolic and
homeostatic failure, cellular oedema, and disruption of glutamate homeostasis
with consequent severe disruption in neurotransmission.
Neuropsychiatric disorders
Schizophrenia Decrease in number of astrocytes and oligodendrocytes; impairment of astroglial
glutamate uptake and glutamine synthesis. Abnormal glutamate homeostasis
together with an increased astroglial production and release of kynurenic acid
(exogenous inhibitor of NMDA and ACh receptors) and altered synthesis of
D-serine (positive modulator of NMDA receptors) contributes to pathological
glutamatergic transmission implicated in pathogenesis of the disease.
Major depressive disorder Decrease in number of oligodendrocytes and astrocytes; microglial asthenia and
microglial apoptosis, abnormal glutamate homeostasis, decreased secretion
of growth factors and cytokines, altered gap junctional connectivity in glial
syncytia which all may contribute to abnormal connectivity in neural networks
and neurotransmission disbalance.
(Continued)
98 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Table 6.3 Pathophysiology of neuroglia (Continued)


Pathology/Disease Neuroglial contribution

Dementia and neurodegenerative disorders


Amyotrophic lateral sclerosis (ALS) Degeneration and atrophy of astrocytes at the early (pre-symptomatic) stages of
the disease. Impaired astroglial glutamate uptake may account for glutamate
excitotoxicity. At later stages activation of microglial and astrocytes may
further contribute to neurotoxicity.
Alzheimer’s disease (AD) Astrocytic asthenia at the early stages of AD progression; mild reactive astrogliosis
of astrocytes associated with plaques. Reactive microgliosis is concomitant to
microglia dystrophy and loss of function (phagocytosis). General decrease in
glial neuroprotection may account for the dynamic and severity of the disease
progression.
Fronto-temporal dementia, Pick’s Combination of astroglial degeneration and astrocytic death with astroglial
disease, fronto-temporal lobar activation; secondary activation of microglia that contribute to
degeneration, thalamic dementia neuroinflammation.
Huntington disease (HD) The primary astroglial pathology in HD is associated with a decrease in expression
of glutamate transporters and impaired glutamate homeostasis; HD-affected
astrocytes have decreased ability to produce and release glutathione and
ascorbic acid, which reduces their neuroprotection through ROS scavenging.
Activation of microglia can be seen at the early, pre-symptomatic stages of this
disease.
Addictive disorders
Alcohol and drug abuse/addiction Decrease in astroglial density and astroglial asthenia at the early stages of the
addictive disorders is complemented by secondary astrogliosis associated with
progression of pathology. Impaired glutamate uptake and release (through
cysteine-glutamate antiporter) may account for aberrant neurotransmission
and behavioural symptoms.

including rapid extension of microglial processes, Neuroglia are the principal homeostatic cells of
morphological alterations, membrane ruffling, the nervous system. Accordingly, they are integral
and chemotaxis while ADP-preferring P2Y13 to homeostatic failures in all neurological diseases
receptors regulate microglial morphology, sur- (Pekny et al., 2016; Verkhratsky et al., 2017).
veillance, and resting levels of interleukin 1β Glial cells are involved in all types of neuropa-
release. Intracellular Ca2+ signalling bridges the thology, although they are not always the primary
surveilling or sensor functions of microglia with targets. In its broadest sense, the function of glia
their effector functions (e.g., directed process is homeostasis – maintaining a state of equilib-
motility, cytokine production). Under physiologi- rium both metabolically within neural cells, the
cal conditions and in conditions of minor tissue nervous tissue and the body as a whole, as well as
dyshomeostasis, most microglial Ca2+ signals are psychologically within the individual. Hence,
compartmentalized and occur only in microglial altered glial homeostatic function invariably con-
processes. Microglial processes vividly react with tributes to CNS neurological diseases, sometimes
an increase in Ca2+ signalling both to hyper- and as a principal element, but often secondary to
hypoactivity in the surrounding neural networks. neural or environmental changes. In addition,
Abundant somatic Ca2+ transients in microglia neuroglia forms CNS defence through intrinsic
reflect pathological conditions like status epilep- evolutionary conserved defensive programmes
ticus, aging, brain injury, amyloid pathology, or generally known as reactive gliosis. Such defence-
peripheral inflammation. related reactive changes are expressed in all types
of glia and are represented by reactive astroglio-
sis, the reactive activation of oligodendroglial
precursors and the reactive microgliosis.
Pathological changes in neuroglia are complex and
PATHOPHYSIOLOGY OF NEUROGLIA diverse; they can be generic or disease-specific
and often vary at different disease stages. In the
All diseases including neurological diseases can context of human pathology, they are affected by
be broadly defined as homeostatic failures. age and comorbidity. Pathological changes in
Role of Neuroglia in Cognition 99

neuroglia can be broadly classified into (i) reac- Bak, L. K., Walls, A. B., Schousboe, A., & Waage-
tive gliosis; (ii) degeneration with loss of function; petersen, H. S. (2018). Astrocytic glycogen metab-
(iii) pathological remodelling, which results in olism in the healthy and diseased brain. Journal of
pathological phenotypes that drive neuropathol- Biological Chemistry, 293, 7108–7116.
ogy; all these pathological reactions may occur Baumann, N., & Pham-Dinh, D. (2001). Biology of oligo-
together or in isolation (Verkhratsky et al., 2017). dendrocyte and myelin in the mammalian central
nervous system. Physiological Reviews, 81, 871–927.
Bazargani, N., & Attwell, D. (2016). Astrocyte cal-
cium signaling: The third wave. Nature Neurosci-
SUMMARY AND CONCLUSIONS ence, 19, 182–189.
Bellot-Saez, A., Kekesi, O., Morley, J. W., & Buskila,
In the human brain glial cells are as abundant as Y. (2017). Astrocytic modulation of neuronal excit-
neurons. The relative number of glial cells has ability through K+ spatial buffering. Neuroscience
increased with increasing complexity of the cen- and Biobehavioral Reviews, 77, 87–97.
tral nervous system (CNS) during evolution. In Bernardinelli, Y., Magistretti, P. J., & Chatton, J. Y.
vertebrates three types of glial cells can be distin- (2004). Astrocytes generate Na+-mediated meta-
guished in the CNS, namely astrocytes, oligoden- bolic waves. Proceedings of the National Academy
drocytes, and microglia. In the peripheral nervous of Sciences of the United States of America, 101,
system glial cells are represented by Schwann 14937–14942.
cells, satellite glial cells, enteric glial cells (EGCs), Bernardinelli, Y., Muller, D., & Nikonenko, I. (2014).
and olfactory ensheathing cells. Astroglia are a Astrocyte-synapse structural plasticity. Neural Plas-
heterogeneous cell population that fulfil different ticity. doi: 10.1073/pnas.0405315101.
supportive and homeostatic tasks such as provid- Boison, D., Chen, J. F., & Fredholm, B. B. (2010).
ing guiding structures during development, con- Adenosine signaling and function in glial cells. Cell
trolling homeostasis of the extracellular space, Death and Differentiation, 17, 1071–1082.
providing energy substrate for neurons, control- Boulay, A. C., Saubamea, B., Adam, N., Chasseigneaux,
ling blood flow, and modulating synaptic trans- S., Mazare, N., Gilbert, A., … Cohen-Salmon, M.
mission. Oligodendrocytes in the central and (2017). Translation in astrocyte distal processes sets
Schwann cells in the peripheral nervous system molecular heterogeneity at the gliovascular interface.
form myelin and thereby enable a high conduction Cell Discovery, 3. doi: 10.1038/celldisc.2017.5.
velocity within the axons. Microglial cells are the Bushong, E. A., Martone, M. E., Jones, Y. Z., &
immune competent cells of the brain and are acti- Ellisman, M. H. (2002). Protoplasmic astrocytes in
vated during any pathologic process. The acti- CA1 stratum radiatum occupy separate anatomical
vated microglial cells can release many factors domains. Journal of Neuroscience, 22, 183–192.
which influence the pathologic process. Taken Butt, A. M., Hamilton, N., Hubbard, P., Pugh, M., &
together brain function is only possible by a con- Ibrahim, M. (2005). Synantocytes: The fifth ele-
certed action of neurons and glial cells. ment. Journal of Anatomy, 207, 695–706.
Butt, A. M., Papanikolaou, M., & Rivera, A. (2019).
Physiology of oligodendroglia. Advances in Experi-
mental Medicine & Biology, 1175, 117–128.
REFERENCES Chvatal, A., & Verkhratsky, A. (2018). Early history of
neuroglial research: Personalities. Neuroglia, 1,
Apell, H. J. (2004). How do P-type ATPases transport 245–281.
ions? Bioelectrochemistry, 63, 149–156. Colombo, J. A., & Reisin, H. D. (2004). Interlaminar
Araque, A., Carmignoto, G., Haydon, P. G., Oliet, S. H., astroglia of the cerebral cortex: A marker of the
Robitaille, R., & Volterra, A. (2014). Gliotransmitters primate brain. Brain Research, 1006, 126–131.
travel in time and space. Neuron, 81, 728–739. Escartin, C., Galea, E., Lakatos, A., O’Callaghan, J. P.,
Augusto-Oliveira, M., Arrifano, G. P., Delage, C. I., Petzold, G. C., Serrano-Pozo, A., … Verkhratsky,
Tremblay, M. E., Crespo-Lopez, M. E., & Verkhrat- A. (2021). Reactive astrocyte nomenclature, defini-
sky, A. (2021). Plasticity of microglia. Biological tions, and future directions. Nature Neuroscience,
Reviews of the Cambridge Philosophical Society. 24, 312–325.
doi: 10.1111/brv.12797 Eulenburg, V., Retiounskaia, M., Papadopoulos, T.,
Augusto-Oliveira, M., Arrifano, G. P., Takeda, P. Y., Gomeza, J., & Betz, H. (2010). Glial glycine transporter
Lopes-Araujo, A., Santos-Sacramento, L., Anthony, 1 function is essential for early postnatal survival but
D. C., … Crespo-Lopez, M. E. (2020). Astroglia- dispensable in adult mice. Glia, 58, 1066–1073.
specific contributions to the regulation of syn- Garaschuk, O., & Verkhratsky, A. (2019a). Microglia:
apses, cognition and behaviour. Neuroscience and The neural cells of nonneural origin. Methods in
Biobehavioral Reviews, 118, 331–357. Molecular Biology, 2034, 3–11.
100 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Garaschuk, O., & Verkhratsky, A. (2019b). Physiology (2014). Contributions of the Na+/K+-ATPase,
of microglia. Methods in Molecular Biology, 2034, NKCC1, and Kir4.1 to hippocampal K+ clearance
27–40. and volume responses. Glia, 62, 608–622.
Gavrilov, N., Golyagina, I., Brazhe, A., Scimemi, A., Tur- Lim, D., Semyanov, A., Genazzani, A., & Verkhratsky, A.
lapov, V., & Semyanov, A. (2018). Astrocytic coverage (2021). Calcium signaling in neuroglia. International
of dendritic spines, dendritic shafts, and axonal bou- Review of Cell and Molecular Biology, 362, 1–53.
tons in hippocampal neuropil. Frontiers in Cellular Liu, X., Zhang, Z., Guo, W., Burnstock, G., He, C., &
Neuroscience, 12. doi: 10.3389/fncel.2018.00248 Xiang, Z. (2013). The superficial glia limitans of
Giaume, C., Koulakoff, A., Roux, L., Holcman, D., & mouse and monkey brain and spinal cord. Ana-
Rouach, N. (2010). Astroglial networks: A step tomical Record, 296, 995–1007.
further in neuroglial and gliovascular interactions. MacAulay, N. (2020). Molecular mechanisms of K+
Nature Reviews Neuroscience, 11, 87–99. clearance and extracellular space shrinkage: Glia
Giaume, C., Naus, C. C., Saez, J. C., & Leybaert, L. cells as the stars. Glia, 68, 2192–2211.
(2021). Glial connexins and pannexins in the MacVicar, B. A., & Newman, E. A. (2015). Astrocyte
healthy and diseased brain. Physiological Reviews, regulation of blood flow in the brain. Cold Spring
101, 93–145. Harbor Perspectives in Biology, 7. doi: 10.1101/
Ginhoux, F., Lim, S., Hoeffel, G., Low, D., & Huber, T. cshperspect.a020388.
(2013). Origin and differentiation of microglia. Matsui, T., Omuro, H., Liu, Y. F., Soya, M., Shima, T.,
Frontiers in Cellular Neuroscience, 7. doi: 10.3389/ McEwen, B. S., & Soya, H. (2017). Astrocytic
fncel.2013.00045 glycogen-derived lactate fuels the brain during
Griemsmann, S., Hoft, S. P., Bedner, P., Zhang, J., von exhaustive exercise to maintain endurance capacity.
Staden, E., Beinhauer, A., … Steinhauser, C. Proceedings of the National Academy of Sciences
(2015). Characterization of panglial gap junction of the United States of America, 114, 6358–6363.
networks in the thalamus, neocortex, and hip- Nagelhus, E. A., & Ottersen, O. P. (2013). Physiologi-
pocampus reveals a unique population of glial cal roles of aquaporin-4 in brain. Physiological
cells. Cerebral Ccortex, 25, 3420–3433. Reviews, 93, 1543–1562.
Hablitz, L. M., & Nedergaard, M. (2021). The glymphatic Nave, K. A., & Werner, H. B. (2014). Myelination of
system: A novel component of fundamental neurobi- the nervous system: Mechanisms and functions.
ology. Journal of Neuroscience, 41, 7698–7711. Annual Review of Cell and Developmental Biology,
Hertz, L. (2013). The glutamate-glutamine (GABA) 30, 503–533.
cycle: Importance of late postnatal development Nishiyama, A., Serwanski, D. R., & Pfeiffer, F. (2021).
and potential reciprocal interactions between bio- Many roles for oligodendrocyte precursor cells in
synthesis and degradation. Frontiers in Endocrinol- physiology and pathology. Neuropathology, 41,
ogy, 4. doi: 10.3389/fendo.2013.00059. 161–173.
Janks, L., Sharma, C. V. R., & Egan, T. M. (2018). A Oberheim, N. A., Takano, T., Han, X., He, W., Lin, J.
central role for P2X7 receptors in human microglia. H., Wang, F., … Nedergaard, M. (2009). Uniquely
Journal of Neuroinflammation, 15. doi: 10.1186/ hominid features of adult human astrocytes. Jour-
s12974-018-1353-8. nal of Neuroscience, 29, 3276–3287.
Kettenmann, H., Hanisch, U. K., Noda, M., & Palygin, O., Lalo, U., Verkhratsky, A., & Pankratov, Y.
Verkhratsky, A. (2011). Physiology of microglia. (2010). Ionotropic NMDA and P2X1/5 receptors
Physiological Reviews, 91, 461–553. mediate synaptically induced Ca2+ signalling in
Kettenmann, H., & Verkhratsky, A. (2008). Neuroglia: cortical astrocytes. Cell Calcium, 48, 225–231.
The 150 years after. Trends in Neurosciences, 31, Pekny, M., Pekna, M., Messing, A., Steinhauser, C.,
653–659. Lee, J. M., Parpura, V., … Verkhratsky, A. (2016).
Kettenmann, H., Kirchhoff, F., & Verkhratsky, A. Astrocytes: A central element in neurological dis-
(2013). Microglia: New roles for the synaptic strip- eases. Acta Neuropathologica, 131, 323–345.
per. Neuron, 77(1), 10–18. Pellerin, L., & Magistretti, P. J. (2012). Sweet sixteen
Kugler, E. C., Greenwood, J., & MacDonald, R. B. for ANLS. Journal of Cerebral Blood Flow and
(2021). The “neuro-glial-vascular” unit: The role of Metabolism, 32, 1152–1166.
glia in neurovascular unit formation and dysfunction. Philips, T., & Rothstein, J. D. (2017). Oligodendroglia:
Frontiers in Cell and Developmental Biology, 9. Metabolic supporters of neurons. Journal of Clini-
doi: 10.3389/fcell.2021.732820. cal Investigation, 127, 3271–3280.
Langer, J., Stephan, J., Theis, M., & Rose, C. R. Pizzagalli, M. D., Bensimon, A., & Superti-Furga, G.
(2012). Gap junctions mediate intercellular spread (2021). A guide to plasma membrane solute car-
of sodium between hippocampal astrocytes in rier proteins. FEBS Journal, 288, 2784–2835.
situ. Glia, 60, 239–252. Rees, D. C., Johnson, E., & Lewinson, O. (2009). ABC
Larsen, B. R., Assentoft, M., Cotrina, M. L., Hua, S. transporters: The power to change. Nature
Z., Nedergaard, M., Kaila, K., … MacAulay, N. Reviews: Molecular Cell Biology, 10, 218–227.
Role of Neuroglia in Cognition 101

Reichenbach, A., Derouiche, A., & Kirchhoff, F. (2019). Exosomes from astrocyte processes: Sign-
(2010). Morphology and dynamics of perisynaptic aling to neurons. Frontiers in Pharmacology, 10.
glia. Brain Research Reviews, 63, 11–25. doi: 10.3389/fphar.2019.01452.
Rokitansky, K. (1854). Über das auswachsen der Verkhratsky, A. (2010). Physiology of neuronal-glial
bindegewebs-substanzen und die beziehung dessel- networking. Neurochemistry International, 57,
ben zur entzündung. Sitzungsberichte der Kaiserlichen 332–343.
Akademie der Wissenschaften. Mathematisch- Verkhratsky, A., & Butt, A. M. (2013). Glial physiol-
Naturwissenschaftliche Classe. Wien 13, 122–140. ogy and pathophysiology. Chichester, UK:
Rose, C. R., & Verkhratsky, A. (2016). Principles of Wiley-Blackwell.
sodium homeostasis and sodium signaling in Verkhratsky, A., & Butt, A. M. (2018). The history of
astroglia. Glia, 64(10), 1611–1127. the decline and fall of the Glial Numbers Legend.
Rose, C. R., Ziemens, D., & Verkhratsky, A. (2020). Neuroglia, 1, 188–192.
On the special role of NCX in astrocytes: Translat- Verkhratsky, A., Krishtal, O. A., & Burnstock, G.
ing Na+-transients into intracellular Ca2+ signals. (2009). Purinoceptors on neuroglia. Molecular
Cell Calcium, 86. doi: 10.1016/j.ceca.2019.102154. Neurobiology, 39, 190–208.
Saura, J., Kettler, R., Da Prada, M., & Richards, J. G. Verkhratsky, A., Matteoli, M., Parpura, V., Mothet, J.
(1992). Quantitative enzyme radioautography P., & Zorec, R. (2016). Astrocytes as secretory cells
with 3H-Ro 41-1049 and 3H-Ro 19-6327 in vitro: of the central nervous system: idiosyncrasies of
Localization and abundance of MAO-A and vesicular secretion. EMBO Journal, 35, 239–257.
MAO-B in rat CNS, peripheral organs, and human Verkhratsky, A., & Nedergaard, M. (2014). Astroglial
brain. Journal of Neuroscience, 12, 1977–1999. cradle in the life of the synapse. Philosophical Transac-
Savtchouk, I., & Volterra, A. (2018). Gliotransmission: tions of the Royal Society of London. Series B, Biologi-
Beyond black-and-white. Journal of Neuroscience, cal Sciences, 369. doi: 10.1098/rstb.2013.0595.
38, 14–25. Verkhratsky, A., & Nedergaard, M. (2016). The homeo-
Scemes, E., & Giaume, C. (2006). Astrocyte calcium static astroglia emerges from evolutionary specializa-
waves: What they are and what they do. Glia, 54, tion of neural cells. Philosophical Transactions of the
716–725. Royal Society of London. Series B, Biological Sci-
Semyanov, A., & Verkhratsky, A. (2021). Astrocytic ences, 371. doi: 10.1098/rstb.2015.0428.
processes: From tripartite synapses to the active Verkhratsky, A., & Nedergaard, M. (2018). Physiology
milieu. Trends in Neurosciences, 44, 781–792. of astroglia. Physiological Reviews, 98, 239–389.
Shigetomi, E., Tong, X., Kwan, K. Y., Corey, D. P., and Verkhratsky, A., & Parpura, V. (2014). Store-operated
Khakh, B. S. (2012). TRPA1 channels regulate astro- calcium entry in neuroglia. Neuroscience Bulletin,
cyte resting calcium and inhibitory synapse efficacy 30, 125–133.
through GAT-3. Nature Neuroscience, 15, 70–80. Verkhratsky, A., & Rose, C. R. (2020). Na+-dependent
Stratoulias, V., Venero, J. L., Tremblay, M. E., & transporters: The backbone of astroglial homeo-
Joseph, B. (2019). Microglial subtypes: diversity static function. Cell Calcium, 85. doi: 10.1016/j.
within the microglial community. EMBO Journal, ceca.2019.102136.
38. doi: 10.15252/embj.2019101997. Verkhratsky, A., Semyanov, A., & Zorec, R. (2020).
Tay, T. L., Carrier, M., & Tremblay, M. E. (2019). Physi- Physiology of astroglial excitability. Function, 1.
ology of microglia. Advances in Experimental doi: 10.1093/function/zqaa016.
Medicine and Biology, 1175, 129–148. Verkhratsky, A., Untiet, V., & Rose, C. R. (2020). Ionic
Ugbode, C., Hu, Y., Whalley, B., Peers, C., Rattray, signalling in astroglia beyond calcium. Journal of
M., & Dallas, M. L. (2017). Astrocytic transporters Physiology, 598(9), 1655–1670.
in Alzheimer’s disease. Biochemical Journal, 474, Verkhratsky, A., Zorec, R., & Parpura, V. (2017).
333–355. Stratification of astrocytes in healthy and diseased
Vandenberg, R. J., & Ryan, R. M. (2013). Mechanisms brain. Brain Pathology, 27, 629–644.
of glutamate transport. Physiological Reviews, 93, Virchow, R. L. K. (1860). Cellular pathology. London:
1621–1657. John Churchill.
Veloz Castillo, M. F., Magistretti, P. J., & Cali, C. (2021). von Bartheld, C. S., Bahney, J., & Herculano-Houzel,
l-Lactate: Food for thoughts, memory and behavior. S. (2016). The search for true numbers of neurons
Metabolites, 11. doi: 10.3390/metabo11080548. and glial cells in the human brain: A review of 150
Ventura, R., & Harris, K. M. (1999). Three-dimensional years of cell counting. Journal of Comparative
relationships between hippocampal synapses and Neurology, 524, 3865–3895.
astrocytes. Journal of Neuroscience, 19, 6897–6906. Zhou, Y., & Danbolt, N. C. (2013). GABA and gluta-
Venturini, A., Passalacqua, M., Pelassa, S., Pastorino, mate transporters in brain. Frontiers in Endocrinol-
F., Tedesco, M., Cortese, K., … Cervetto, C. ogy, 4. doi: 10.3389/fendo.2013.00165.
7
Cognitive Neurogenetics
Jack van Honk, Zhang Li, Norihiro Sadato,
a n d J o a n Y. C h i a o

INTRODUCTION question, “What are the underlying neural/neuro-


scientific circuits/pathways in higher-order cogni-
Neuroscientific study of the phylogenetic and tive processes?”
ontogenetic bases of the central nervous system Encoding and decoding of cortical representa-
(CNS) contributes to the understanding of the tions of the sensory environment of cellular and
structure and functional properties of the cogni- molecular mechanisms clearly demonstrates the
tion and its evolutionary and cultural adaptation important role of genetics in human brain func-
(e.g., Chiao, 2016, 2017a,b; Chiao and Ambady, tioning, including higher-level cognition. The nor-
2007; Chiao and Blizinsky, 2010; Chiao and mal cellular and molecular basis of human brain
Immordino-Yang, 2013; Chiao et al., 2010, 2016, activity corresponds functionally with conscious-
2020, 2021a,b,c; Kandel and Schwartz, 1985; ness and the higher-level processes of cognition
Reinvang et al., 2010; Tandon, 2000). The role of and episodic memory (e.g., Aslaksen et al., 2018;
genetics in the higher-level cognitive processes Bender and Raz, 2012; Bouazzaoui et al., 2013;
involves the functional role of cellular and molec- Crick and Koch, 1990; Golchert et al., 2019;
ular mechanisms in the human brain (cf. Bartley Green et al., 2013). Multilevel processing of neu-
et al., 1997; Boyle et al., 2023a,b; Fossella et al., ronal mechanisms consists of synaptic transmis-
2006; also see Table 7.1). The distinct activation
sion that involves neurochemical signaling of
patterns of neurotransmitter systems relate to the
specific neurotransmitters, as well as enabling
functional role of genes within neurons.
Neurogenetics involved in cognitive neuroscience computational cultural neuroscience; Chiao, 2011;
(cognitive neurogenetics) pertains to how the Churchland and Sejnowski, 2017; O’Reilly and
genetic code impacts on expressed cognitive Munakata, 2000). The neurochemical signaling
attributes (e.g., Green and Dunbar, 2012; Green directly affects the release of various neurotrans-
et al., 2008, 2012; Neumann et al., 2016). In this mitters at the presynaptic and postsynaptic junc-
chapter, we discuss specific neuroscientific find- tion of neurons. Specialization of the presynaptic
ings pertaining to cognitive and systems neurosci- and postsynaptic structures illustrates the response
ence (cf. Boyle and Saklofske, 2004; Eising et al., selectivity of distinct types of neurons and special-
2022) in attempting to address the important ized information processes.
Cognitive Neurogenetics 103

Table 7.1 Genes and brain function


Gene Polymorphism Brain function

Serotonin transporter (5-HTT) Variable number tandem repeat in the promoter region (5-HTTLPR) Amygdala
Dopamine receptor polymorphism Variable number of tandem repeats (VNTR) polymorphism Ventral striatum
(DRD2, DRD4)

INTERFACE OF COGNITIVE levels of analysis. Such multiple-phenotype


NEUROSCIENCE AND GENETICS approaches indicate that individual variation in
genes central to maintaining synaptic integrity,
neurotransmitter function, and synaptic plasticity
Genes influence cellular activity that comprise the are important in affecting age-related changes in
neurochemical signaling within the human brain brain structure and cognition. Investigating pheno-
and its function. The functional specialization of types at multiple levels is recommended as a means
specific brain areas shows the prepotent, auto- to advance understanding of the neural impact of
matic, innate, encapsulation of neuronal mecha- genetic variants relevant to cognitive aging. Further
nisms in the performance of the higher-level knowledge regarding the mechanisms of interac-
function of cognition. Genetic regulation of neu- tion between genetic and preventative procedures
rotransmission in the human brain is important in will in turn help in understanding the ameliorative
cognitive processes and cortical organization. effect of various experiential and lifestyle factors on
The malleability of human brain function to the age-related cognitive decline.
interaction of genes and stimuli highlights the role
of genetic expression and neuronal plasticity in The functional integration of cortical organiza-
neural and cognitive change. Genes are involved in tion demonstrates the importance of the intercon-
neuronal growth and maturation that accompanies nectivity of cortical brain regions in the functional
neurodevelopment. Interaction of genetic expres- processes and basic mechanisms of the nervous
sion and environmental influences contributes to the system. The structural and functional connectivity
maturational processes that facilitate neurodevelop- of cortical organization detail the important role of
mental pathways. Genetic expression of human brain cortical networks and functional pathways in cog-
function contributes to the processes of neurogen- nitive and higher-level processes. The intercon-
esis important in the neuronal growth and functional nectivity of cortical networks provides regulatory
maturation of cortical organization (with regard to mechanisms that facilitate the control and regula-
“neurogenetic effects on cognition in aging brains” tion of the level of activity of cortical mechanisms
by Reinvang et al., 2010; cf. Aghamohammadi- and higher-level processes. The control and regu-
Sereshki et al., 2019; Chadjikyprianou et al., 2021; lation of level of activity of functional pathways
Dixon et al., 2014; Lamar et al., 2003; Lundervold contributes to the bidirectional processes that
et al., 2014; Parasuraman and Jiang, 2014; Villela underlie the complexity of cognition and human
et al., 2018). Indeed, according to the Abstract on brain function. Genetic variation of human brain
the first page of Reinvang et al. (2010), function contributes to the understanding of the
individual and cultural differences that comprise
Knowledge of genetic influences on cognitive aging human variation. The impact of genetic and cul-
can constrain and guide interventions aimed at tural variation on the human brain illustrates the
limiting age-related cognitive decline in older adults. overall adaptive preparedness of the organism
Progress in understanding the neural basis of cogni- (i.e., comprising both human and infra-human
tive aging also requires a better understanding of species) for biological and cultural adaptation
the neurogenetics of cognition. [We review] studies (Cavalli-Sforza et al., 1994; Chiao et al., 2010).
aimed at deriving specific neurogenetic information
from three parallel and interrelated phenotype-
based approaches: psychometric constructs, cogni-
tive neuroscience-based processing measures, and
brain imaging morphometric data. Developments in
NEUROGENETICS OF COGNITIVE
newer genetic analysis tools, including genome NEUROSCIENCE
wide association, are also described. In particular,
we focus on models for establishing genotype–­ Research into the neurogenetics of cognitive neu-
phenotype associations within an explanatory roscience emphasizes the role of genotypes and
framework linking molecular, brain, and cognitive intermediate endophenotypes on the production of
104 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

complex phenotypes. Advances in behavioral cognition. Identification of the functional path-


genetics also have revealed the importance of ways of cognition and behavior contributes to the
higher-level cognitive processes as intermediate understanding of the role of individual and cul-
endophenotypes of complex behavior. Such tural differences on human variation. Functional
higher-level cognitive processes act as mediators expression of specific genes in the neural path-
or moderators of the genetic expression of func- ways of human cognitive brain function dem-
tional pathways that underlie complex phenotypes onstrates the regulatory effect of the specialized
(Figure 7.1). information processing of distinct neurons within
Identification of specific genes that regulate the brain’s cortical organization. In this chapter,
levels of human brain activity highlights the func- we focus on the role of neurogenetics in cognitive
tional significance of genes on the functional and systems neuroscience.
pathways of cognition and behavior (also see Specific brain areas consist of distinct types of
Table 7.1). The interaction of genes with contex- neurons responsible for distinct types of neuro-
tual and environmental influences illustrates the transmission. Patterns of electrophysiological sign-
malleability of functional pathways involved in aling of distinct types of neurons show response

Figure 7.1 The genotype-to-phenotype model of behavior.


(a) The behavioral genetics model describes the genotype-to-behavior pathway.
(b) The neurogenetics model characterizes the genotype-to-phenotype with the brain as an
intermediate endophenotype pathway.
(c) The cognitive neurogenetics model identifies the genotype-to-phenotype pathway with
cognitive brain function as an intermediate endophenotype; the cultural neurogenetics
model characterizes the role of genes and culture in the genotype-to-phenotype pathway.
Cognitive Neurogenetics 105

selectivity to intrinsic and extrinsic processes. the raphe nucleus of the midbrain is responsible
Intrinsic processing of neurons illustrates the for the neurochemical signaling of the serotonin
spontaneous and intrinsic activity of electrophysi- neurotransmitter system and its cortical projec-
ological signaling based on the absence of input. tions. The dopamine system that is located within
Extrinsic processing of neurons arises from the the substantia nigra of the midbrain is responsi-
activity of firing patterns in response to sustained ble for the neurotransmission of the dopamine
or prolonged sensory input, and the selectivity neurotransmitter system and its interconnected
of neuronal mechanisms in response to sensory brain regions. In general, distinct types of neu-
stimuli. Electrophysiological signaling of neuronal rotransmitters reveal the selectivity of neuronal
mechanisms is responsible for the encoding and mechanisms within the neurochemical signaling
decoding of sensory stimuli into cortical percep- of synaptic and neural transmission.
tual representations. The role of genotypes in the
endophenotypes of complex cognitive processes
recognizes the important intermediary role of
functional processes and underlying mechanisms
(Bogdan et al., 2012). GENETIC BASIS OF COGNITIVE BRAIN
FUNCTION

Genes impact on the functional pathways of cog-


nitive brain function and observable cognitive
NEUROTRANSMISSION FROM NEURONS behavior (Heils et al., 1997). In general, the
TO COGNITION genetic variants of specific functional polymor-
phisms show the functional variation of human
Multilevel processing of neuronal mechanisms brain activity in response to task-based processes,
consists of synaptic transmission involving the including various types of cognitive tasks (Canli
neurochemical signaling of specific neurotrans- et al., 2006; Hariri and Weinberger, 2003a).
mitters (Churchland and Sejnowski, 2017; Hille, Genetic variants of specific functional polymor-
1984; O’Reilly and Munakata, 2000). Functional phisms illustrate the distinct types of functional
processes and basic mechanisms of the CNS con- patterns of activation in response to the allelic
sist of the pathways of the neurotransmitter sys- variation of specific genes (Brown et al., 2006;
tems that enable cognitive and higher-level Forbes et al., 2009; Hariri et al., 2002; also see
processes. Neurotransmission of specialized func- Figure 7.3). The functional correspondence of the
tional pathways provides a molecular basis for the allelic variation of specific genes and the distinct
chemical processes that underlie the genetic vari- patterns of functional activation of specialized
ations in human cognition. Synaptic transmission brain regions, such as those involved in cognition
consists of the intracellular changes to the mem- and memory, demonstrates the link of genotype to
brane potential of neurons and its release of spe- endophenotype in the functional processes of
cific neurotransmitters to produce the specific human brain function, including higher-level cog-
effects of on cognition and working memory func- nitive function (Bogdan et al., 2017; Viding et al.,
tions (Ihne et al., 2016; Shepherd, 2004; also see 2006). Thus, the distinct levels of functional acti-
Figure 7.2). vation of specific cortical brain regions in response
The norepinephrine system is located within to effortful cognitive processes in the context of
the locus coeruleus of the brain stem and its inter- genetic variation, illustrate the effect of genes on
connected brain regions. The serotonin system of neural transmission of phenotypic variation
(Bogdan et al., 2016; Brown and Hariri, 2006;
Fisher and Hariri, 2012; Hariri and Weinberger,
2003b; Hariri et al., 2005; Nikolova et al., 2014;
Zhou et al., 2008).

GENETIC AND FUNCTIONAL EXPRESSION


OF HUMAN BRAIN FUNCTION

Figure 7.2 The brain as an intermediate Distinct patterns of genetic and functional expres-
phenotype in the genotype-to-phenotype sion in the functional pathways of neural trans-
pathway. mission illustrate the modifiability of genetic and
106 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 7.3 Genotype modulation of reward activity of the ventral striatum.


Source: From Forbes et al., 2005.

neural mechanisms and its intrinsic and extrinsic based on cognitive and higher-level processes.
properties. The modification of genetic and neural Neurocognitive function describes the level of
mechanisms details the alteration of the functional activity of neuronal mechanisms in response to
expression of basic mechanisms of cortical organ- the effortful cognitive processing. Cognition is
ization and its functional correspondence of geno- comprised of automatic and controlled processes
type to phenotype (Canli et al., 2005). The that show correspondence in the functional activ-
interaction of genes and environmental influences ity of human brain function and observable
enacts the functional processes and basic mecha- behavior (Fodor, 1983; Gazzaniga et al., 2002;
nisms of cortical organization that underlie cogni- Sejnowski and Churchland, 1989). The functional
tion and higher-level processes. The role of response of the higher-level consciousness and
cognition in the functional processes of cortical the regulation processes of mental activity consti-
organization facilitates modulation of the level of tutes the role of cognition in the regulatory
activity of neuronal mechanisms based on intrin- mechanisms of the functional activity of the brain.
sic and extrinsic properties. The modification of Information processing of sensory stimuli illus-
genetic and epigenetic expression contributes to trates the malleability of neuronal plasticity to
the regulation of the functional and behavioral learning and experience. The functional response
expression of cognition and higher-level processes of cellular and molecular mechanisms to learning
(Nikolova and Hariri, 2015). demonstrates the importance of experience in the
The regulatory control of the efficacy of functional processes of neuronal and cognitive
presynaptic and postsynaptic activity of neuro- change, and underscores the importance of the
transmitters provides a molecular basis for the interaction of genes and environmental influences
alteration of the excitatory or inhibitory input of in multilevel mechanisms.
neuronal mechanisms and its interconnection. Cognitive neurogenetics underscore the role of
Clearly, genes contribute to the control and regu- the genotype to endophenotype in understanding
lation of the efficacy of synaptic transmission and human brain activity. Neurogenetic models dem-
its effect on the functional processes of neuronal onstrate the genotype to endophenotype and the
mechanisms. Thus, the regulatory control of genes role of genes in functional expression of the brain.
contributes to the neural activation of human cog- The conceptual model of the genotype to endophe-
nitive and memory function. notype highlights the importance of the functional
expression of genes in the cortical tissue. The role
of genes in the expression of human brain func-
tion shows the modifiability of CNS functional
processes and basic mechanisms.
COGNITIVE NEUROGENETICS The genetic variation of human brain activity
in response to a cognitive task illustrates the effect
The neurogenetics of cognition entails the genetic of genes on the functional pathways of cognition
variation of the functional activity of the brain and higher-level processes. The role of genes in
Cognitive Neurogenetics 107

the distinct functional patterns of activation illus- cognition. The modulation of the level of activ-
trates the multiple patterns of activity of neurons ity of neuronal mechanisms based on the level of
that comprise specific neurotransmitter systems mental activity of cognitive processes describes
and their neurochemical signaling of localized the functional expression of the neuronal mecha-
brain regions. The multiple patterns of activity of nisms that underlie cognition and higher-level
neuronal mechanisms that genes regulate demon- function. The distinct patterns of activation of
strates the role of neural transmission in the distinct interconnected brain regions of cortical neural
types of inter-connectivity that underlie cognitive networks shows the modulation of neural activity
processes. Inhibitory or excitatory connectivity of based on the functional task demand of cognitive
cortical neural networks suggests distinct types of processes (Menon, 2012). The distributed pattern
synaptic input based on the synaptic transmission of information processing of cortical neural net-
of neurotransmitter systems. Neural transmission works details the multiple patterns of activity that
of cellular and molecular mechanisms provides a comprise interconnected brain regions and their
putative mechanism for feedforward and inhibi- functional correspondence with cognition.
tory processing of cortical neural networks. The
bidirectional processes of cortical neural networks
demonstrate a basic mechanism in the control or
regulation of cognition and higher-level processes.
The response selectivity of the distinct acti- GENETIC REGULATION OF COGNITION
vation patterns shows the multiple functional
pathways that are responsible for the control and The genetic regulation of neurotransmission of
regulation of cognition and higher-level processes. interconnected brain regions of cortical neural
The distinct types of neurotransmitter systems networks involves the role of genetic variants in
show functional expression in the distinct func- the level of functional activity of interconnected
tional pathways of cognition and higher-level brain areas and the regulation of the efficacy of
processes. The functional pathways of neurotrans- neurochemical signaling of neuronal mechanisms
mitter systems are comprised of the neurotrans- that underlie cognitive processes (Hariri et al.,
mitters located within specific brain regions and 2003). The genetic regulation of the efficacy of
their interconnected projections to cortical and neurochemical signaling facilitates the synaptic
subcortical systems. The genotype to endopheno- transmission of cortical mechanisms. The genetic
type that is observable in the multiple functional regulation of synaptic transmission constitutes of
pathways of distinct neurotransmitter systems putative mechanism for the amplification or inhi-
illustrates the cellular and molecular basis of the bition of the excitatory and inhibitory connectivity
functional processes and basic mechanisms of the of cortical neural networks. The regulation of the
human brain. bidirectional processing of the cognitive informa-
tion of cortical neural networks contributes to the
regulatory control of genes and its effect on neu-
ronal mechanisms.
GENETIC AND FUNCTIONAL EXPRESSION
OF COGNITION
Genetic and neural basis of cognition
Components of cognitive processes illustrate the
complexity of higher-level functions that com- The genetic and neural basis of cognition details
prise human cognition and memory. Cognitive the mutual interactivity of cellular and molecular
change involves the transformation and interpreta- processes in the cognitive representation of corti-
tion of cognitive representations based on the cal organization (Churchland and Sejnowski,
encoding and maintenance of functional activa- 1988). The role of genes in the functional pro-
tion states that contribute to cognitive and behav- cesses of neuronal mechanisms is multifaceted
ioral adaptation. and illustrates the importance of the structure and
The genetic and functional expression of function of genes and its effect on the synaptic
cortical mechanisms comprise the cellular and transmission. The effect of genes in the functional
molecular basis for the control or regulation of pathways of cognition demonstrates the genotype
functional activity into distinct patterns of func- to endophenotype of cognition and human brain
tional activation of interconnected brain regions. function. The effect of genes on the cortical
The distinct patterns of functional activation of mechanisms of cognition implies a moderation or
interconnected brain regions illustrate the func- mediation role of endophenotypes in the etiology
tional expression of the information processes of of complex traits. The cognitive neurogenetics
108 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

approach underscores the role of functional pro- the control and regulation of the interaction of top-
cesses in the control and regulation of cortical down and bottom-up processes of specific brain
mechanisms and the role of cognitive processes. areas across multiple layers of cortical organiza-
The modulation of the functional activity of neu- tion. Cultural factors contribute to bidirectional
ronal mechanisms based on learning and experi- processing of the information of cortical neural
ence illustrates the importance of the dynamics of networks that control and regulate the perceptual
the expression and regulation of genes in the and motivational salience of sensory stimuli (Blais
control and regulation of neural and cognitive and Caldara, 2021; Chiao and Mathur, 2016; De
change (Figure 7.4). Gelder and Huis in’t Veld, 2016). Cultural factors
Cognitive neurogenetics contributes to the fun- are important in the understanding of the intrinsic
damentals of cognitive and systems neuroscience. and extrinsic properties of functional processes and
Systematic investigation of the role of genes in the basic mechanisms of human brain function and
control and regulation of the neurotransmission cognition. Culture influences the regulation of
of cortical mechanisms that are responsible for the processes of neural and cognitive change
cognition and higher-level function contributes to (Goh, 2021; Gutchess et al., 2021; Varnum and
the understanding of the neurobiological machin- Hampton, 2021). The cultural expression of neu-
ery of biological organisms. The characterization ronal mechanisms controls or regulates the func-
of multilevel mechanisms of the nervous system tional expression of complex cognitive processes.
entails the study of the functional processes and Cross-cultural research into neurogenetics is
basic mechanisms of the cellular and molecular important in understanding of the role of cultural
mechanisms of human brain function. Study of the factors that affect human brain function that con-
endophenotypes and phenotypes of complex traits tribute to the fundamentals of cognitive and sys-
contributes to the understanding of the causal- tems neuroscience. Cultural differences in the
functional role of cognition and human brain func- functional processes and basic mechanisms of
tion in functional and behavioral adaptation. human brain function illustrate the role of culture
in the structure and function of cortical organiza-
tion (Chiao et al., 2016). Cultural differences in
the distinct patterns of functional activation of
CULTURAL NEUROGENETICS: IMPACT ON cortical neural networks show the distinct types
of processing of cultural information in the cog-
COGNITION nitive and higher-level systems of the human
brain. Epigenetic expression of genes contributes
Research into cultural neurogenetics generates to modulation of brain function underlying social
novel knowledge of the role of genes and culture behavior (Connelly and Morris, 2016; McGraw
in human brain/cognitive function (Chiao and and Young, 2010).
Ambady, 2007; Chiao and Blizinsky, 2010; Chiao Processes of cultural transmission contribute to
et al., 2010, 2016). Cultural neurogenetics is the the evolutionary adaptation of human populations
systematic investigation of the functional pro- (Cavalli-Sforza and Feldman, 1981). Biological
cesses and basic mechanisms of neuronal mecha- and cultural transmission or inheritance of char-
nisms that show response selectivity to genetic acteristics illustrates the significance of the
and cultural variation. Research into cultural neu- functional architecture of the mind and brain in
rogenetics investigates how cultural variation behavioral and cultural adaptation. Cultural char-
affects the functional expression of biomarkers acteristics of evolutionary change contribute to the
that underlie health and disease. International malleability of endophenotypes and phenotypes
comparative studies of cultural neurogenetics con- of functional adaptation (Chen et al., 1999, 2016;
tribute to the identification of endophenotypes and Uchiyama and Muthukrishna, 2021). The study of
phenotypes across various cultural settings (Chiao neurogenetics also contributes to the fundamen-
et al., 2021a,b,c). The interaction of genes and tals of cognitive and systems neuroscience (Chiao
cultural environmental influences contributes to et al., 2021).
the control and regulation of the functional pro-
cesses and basic mechanisms of the CNS and its
link to observable behavior (Cheon et al., 2014;
2021; Nomura, 2016; Sasaki et al., 2016; Stein
et al., 2016; Stevenson et al., 2016). SUMMARY AND CONCLUSIONS
The study of cultural neurogenetics contributes
to the understanding of the interactivity of the Evidence-based research into neurogenetics
structural and functional connectivity of human builds knowledge-based resources of genes and
brain function. Cultural variation is important in brain function. Understanding the fundamental
Cognitive Neurogenetics 109

Figure 7.4 Neural correlates of epigenesis.


Source: Canli et al., 2006.
110 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

processes and basic mechanisms of the human Bogdan, R., Hyde, L. W., & Hariri, A. R. (2013). A neu-
brain is foundational for understanding cognitive rogenetics approach to understanding individual
and systems neuroscience. Cross-cultural study of differences in brain, behavior, and risk for psycho-
neurogenetics contributes to the understanding of pathology. Molecular Psychiatry, 18(3), 288–299.
the interaction of gene and environmental influ- Bogdan, R., Pagliaccio, D., Baranger, D. A., & Hariri,
ences on human brain function within different A. R. (2016). Genetic moderation of stress effects
nations, including the promotion of mental health on corticolimbic circuitry. Neuropsychopharmacol-
as well as cross-cultural differences in cognitive ogy, 41(1), 275–296.
and memory dexterity. Bogdan, R., Salmeron, B. J., Carey, C. E., Agrawal, A.,
In summary, research into neurogenetics is Calhoun, V. D., Garavan, H., … Goldman, D.
important in understanding the role of genes in (2017). Imaging genetics and genomics in psychia-
human brain function (including higher-level try: A critical review of progress and potential.
cognitive and memory functions). Broadly, neu- Biological Psychiatry, 82(3), 165–175.
rogenetics research investigates the functional Bouazzaoui, B., Fay, S., Taconnat, L., Angel, L., Van-
genetics of neural pathways involving various neste, S., & Isingrini, M. (2013). Differential
neuroanatomical components of the brain. Genes involvement of knowledge representation and
regulate the functional neurotransmission of executive control in episodic memory performance
molecular and cellular mechanisms within the in young and older adults. Canadian Journal of
brain. Identification of specific genes or func- Experimental Psychology, 67(2), 100–107.
tional polymorphisms that regulate neurotrans- Boyle, G. J., & Saklofske, D. H. (2004). (Eds.), SAGE
mission pathways involved in human cognition benchmarks in psychology: The psychology of
and memory remains an important area of neuro- individual differences (Vol. 3): Cognition, emotion
cognitive research. Such research has the potential and conation. London: Sage.
to advance our understanding of the role of genetic Boyle, G. J., Stern, Y., Stein, D. J., & Sahakian, B. J
regulation within the neural mechanisms underly- Golden, C. J., Lee, T. M.-C., & Chen, S.-H. A.
ing cognition and memory functions as well as the (2023a). (Eds.), The SAGE handbook of clinical
role of neurogenetics in both individual and cul- neuropsychology, Vol. 1: Clinical neuropsychologi-
tural variation, more broadly. cal disorders. London: Sage.
Boyle, G. J., Stern, Y., Stein, D. J., & Sahakian, B. J
Golden, C. J., Lee, T. M.-C., & Chen, S.-H. A.
(2023b). (Eds.), The SAGE handbook of clinical
neuropsychology, Vol. 2: Clinical neuropsychologi-
REFERENCES cal assessment and diagnosis. London: Sage.
Brown, S. M., & Hariri, A. R. (2006). Neuroimaging
Aghamohammadi-Sereshki, A., Hrybouski, S., Travis, studies of serotonin gene polymorphisms: Explor-
S., Huang, Y., Olsen, F., Carter, R., Camicioli, R., & ing the interplay of genes, brain and behavior.
Malykhin, N. V. (2019). Amygdala subnuclei and Cognitive, Affective and Behavioral Neuroscience,
healthy cognitive aging. Human Brain Mapping, 6(1), 44–52.
40(1), 34–52. Brown, S. M., Manuck, S. B., Flory, J. D., & Hariri, A.
Aslaksen, P. M., Bystad, M. K., Ørbo, M. C., & Vang- R. (2006). Neural basis of individual differences in
berg, T. R. (2018). The relation of hippocampal impulsivity: Contributions of corticolimbic circuits
subfield volumes to verbal episodic memory meas- for behavioral arousal and control. Emotion, 6(2),
ured by the California Verbal Learning Test II in 239–245.
healthy adults. Behavioural Brain Research, 351, Canli, T., Omura, K., Haas, B. W., Fallgatter, A., Con-
131–137. stable, R. T., & Lesch, K. P. (2005). Beyond affect:
Bartley, A. J., Jones, D. W., & Weinberger, D. R. A role for genetic variation of the serotonin trans-
(1997). Genetic variability of human brain size and porter in neural activation during a cognitive
cortical gyral patterns. Brain, 120(2), 257–269. attention task. Proceedings of National Academy
Bender, A. R., & Raz, N. (2012). Age-related differ- of Sciences of the United States of America, 102,
ences in episodic memory: A synergistic contribu- 12224–12229.
tion of genetic and physiological vascular risk Canli, T., Qiu, M., Omura, K., Congdon, E., Haas, B. W.,
factors. Neuropsychology, 26, 442–450. Amin, Z., Herrman, M. J., Constable, R. T., & Lesch,
Blais, C., & Caldara, R. (2021). Culture shapes face K. P. (2006). Neural correlates of epigenesis. Pro-
processing. In J. Y. Chiao, S.-C. Li, R. Turner, S. Y. ceedings of the National Academy of Sciences of the
Lee-Tauler, & B. A. Pringle (Eds.), The Oxford hand- United States of America, 103(43), 16033–16038.
book of cultural neuroscience and global mental Cavalli-Sforza, L. L., & Feldman, M. W. (1981). Cul-
health (pp. 309–325). New York: Oxford University tural transmission and evolution. Princeton, NJ:
Press. Princeton University Press.
Cognitive Neurogenetics 111

Cavalli-Sforza, L. L. Menozzi, P., & Piazza, A. (1994). cultural neuroscience to cognitive theory. Perspec-
The history and geography of human genes. tives on Psychological Science, 8(1), 56–61.
Princeton, NJ: Princeton University Press. Chiao, J. Y., & Mathur, V.A. (2016). Cultural neurosci-
Chadjikyprianou, A., Hadjivassiliou, M., Papacostas, S., ence of pain and empathy. In J. Y. Chiao, S.-C. Li,
& Constantinidou, F. (2021). The neurocognitive R. Seligman & R. Turner (Eds.), The Oxford hand-
study for the aging: Longitudinal analysis on the book of cultural neuroscience (pp. 271–278). New
contribution of sex, age, education and APOE ε4 York: Oxford University Press.
on cognitive performance. Frontiers in Genetics, Chiao, J. Y., Hariri, A. R., Harada, T., Mano, Y.,
12. doi: 10.3389/fgene.2021.680531 Sadato, N., Parrish, T. B., & Iidaka, T. (2010). Theory
Chen, C., Burton, M., Greenberger, E., & Dmitrieva, and methods in cultural neuroscience. Social Cog-
J. (1999). Population migration and the variation nitive and Affective Neuroscience, 5(2–3),
of the dopamine D4 receptor (DRD4) allele fre- 356–361.
quencies around the globe. Evolution and Human Chiao, J. Y., Li, S.-C., Seligman, R., & Turner, R. (Eds.)
Behavior 20(5), 309–324. (2016). The Oxford handbook of cultural neurosci-
Chen, C., Moyzis, R. K., Lei, X., Chen, C., & Dong, Q. ence. New York: Oxford University Press.
(2016). The enculturated genome: Molecular evi- Chiao, J. Y., Li, S.-C., Turner, R., & Lee-Tauler, S. Y.
dence for recent divergent evolution in human (2020). Cultural neuroscience and the research
neurotransmitter genes. In J. Y. Chiao, S.-C. Li, R. domain criteria: Implications for global mental
Seligman & R. Turner (Eds.), The Oxford handbook health. Neuroscience and Biobehavioral Reviews,
of cultural neuroscience (pp. 315–338). New York: 116, 109–119.
Oxford University Press. Chiao, J. Y., Harada, T., Mano, Y., Stein, D. J., van
Cheon, B. K., Livingston, R. W., Hong, Y.-Y., & Chiao, Honk, J., Sadato, N., & Iidaka, T. (2021a). Culture
J. Y. (2014). Gene x environment interaction on and emotion. In J. Y. Chiao, S.-C. Li, R. Turner, S.
intergroup bias: The role of 5-HTTLPR and per- Y. Lee-Tauler & B. A. Pringle (Eds.), The Oxford
ceived outgroup threat. Social Cognitive and handbook of cultural neuroscience and global
Affective Neuroscience, 9(9), 1268–1275. mental health (pp. 199–220). New York: Oxford
Cheon, B. K., & Hong, Y. Y. (2016). The cultural neu- University Press.
roscience of intergroup bias. In J. Y. Chiao, S.-C. Li, Chiao, J. Y., Li, Z., Harada, T., Sadato, N., Iidaka, T.
R. Seligman & R. Turner (Eds.), The Oxford hand- (2021b). Culture and genomics. In J. Y. Chiao, S.-
book of cultural neuroscience (pp. 248–270). New C. Li, R. Turner, S. Y. Lee-Tauler & B. A. Pringle
York: Oxford University Press. (Eds.), The Oxford handbook of cultural neurosci-
Chiao, J. Y. (2011). Computational cultural neurosci- ence and global mental health (pp. 144–155).
ence: Implications for augmented cognition. Paper New York: Oxford University Press.
presented at 6th International Conference on Chiao, J. Y., Li, S.-C-., Turner, R., Lee-Tauler, S. Y., &
Foundations of Augmented Cognition, 137–142. Pringle, B. A. (Eds.) (2021c). The Oxford handbook
doi: 10.1007/978-3-642-21852-1_17 of cultural neuroscience and global mental health.
Chiao, J. Y. (2016). Cultural neuroscience of the New York: Oxford University Press.
developing brain in childhood. Minnesota Sympo- Churchland, P. S. & Sejnowski, T. J. (2017). The com-
sium on Child Psychology: Culture and Develop- putational brain (25th ed.). Cambridge, MA: MIT
mental Systems, 38, 1–37. Press.
Chiao, J. Y. (2017a). Cultural neuroscience of the Churchland, P. S. & Sejnowski, T. J. (1988). Perspec-
developing brain in adolescence. In N. Budwig, E. tives in cognitive neuroscience. Science, 242,
Turiel & P. D. Zelazo (Eds.), New perspectives on 741–745.
human development (pp. 330–350). Cambridge, Crick, F. H. C. & Koch, C. (1990). Towards a neuro-
UK: Cambridge University Press. biological theory of consciousness. Seminars in the
Chiao, J. Y. (2017b). Philosophy of cultural neurosci- Neurosciences, 2, 263-275.
ence. London: Routledge. Collins, P. Y., Patel, V., Joestl, S. S., March, D., Insel,
Chiao, J. Y., & Ambady, N. (2007). Cultural neurosci- T. R., Daar, A. S., … Stein, D. J. (2011). Grand chal-
ence: Parsing universality and diversity across lenges in global mental health. Nature, 475,
levels of analysis. In S. Kitayama & D. Cohen (Eds.), 27–30.
Handbook of cultural psychology (pp. 237–254). Connelly, J. J., & Morris, J. P. (2016). Epigenetics of
New York: Guilford. social behavior. In J. Y. Chiao, S.-C. Li, R. Seligman &
Chiao, J. Y., & Blizinsky, K. D. (2010). Culture-gene R. Turner (Eds.), The Oxford handbook of cultural
coevolution of individualism-collectivism and the neuroscience (pp. 299–314). New York: Oxford
serotonin transporter gene. Proceedings of the University Press.
Biological Sciences, 277, 529–537. De Gelder, B. & Huis in’t Veld, E. (2016). Cultural dif-
Chiao, J. Y., & Immordino-Yang, M. H. (2013). Modu- ferences in emotional expressions and body lan-
larity and the cultural mind: Contributions of guage. In J. Y. Chiao, S.-C. Li, R. Seligman & R.
112 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Turner (Eds.), The Oxford handbook of cultural Green, A. E., Kraemer, D. J. M., Fugelsang, J., Gray,
neuroscience (pp. 223–236). New York: Oxford J. R., & Dunbar, K. (2012). Neural correlates of
University Press. creativity in analogical reasoning. Journal of Exper-
Dixon, R. A., DeCarlo, C. A., MacDonald, S. W., Ver- imental Psychology: Learning, Memory, and Cog-
gote, D., Jhamandas, J., & Westaway, D. (2014). nition, 38(2), 264–272.
APOE and COMT polymorphisms are complemen- Green, A. E., Kraemer, D. J. M., DeYoung, C., Fossella,
tary biomarkers of status, stability, and transitions J., & Gray, J. R. (2013). A gene-brain-cognition
in normal aging and early mild cognitive impair- pathway for the effect of COMT on executive
ment. Frontiers in Aging Neuroscience, 6. doi: attention and IQ. Cerebral Cortex, 23, 552–559.
10.3389/fnagi.2014.00236 Green, A. E., Munafo, M. R., DeYoung, C., Fossella,
Eising, E., Mirza-Schreiber, N., de Zeeuw, E. L., Wang, J., Fan, J., & Gray, J. R. (2008). Using genetic data
C. A., Truong, D. T., Allegrini, A. G., … Fisher, S. E. in cognitive neuroscience: From growing pains to
(2022). Genome-wide analyses of individual differ- genuine insights. Nature Reviews Neuroscience, 9,
ences in quantitatively assessed reading- and lan- 710–720.
guage-related skills in up to 34,000 people. Gutchess, A., Mukadam, N., Zhang, W., & Zhang, X.
Proceedings of the National Academy of Sciences (2021). Influence of aging on memory across
of the United States of America, 119(35). doi: cultures. In J. Y. Chiao, S.-C. Li, R. Turner, S. Y.
10.1073/pnas.2202764119 Lee-Tauler & B. A. Pringle (Eds.), The Oxford hand-
Fisher, P. M., & Hariri, A. R., (2012). Linking variability book of cultural neuroscience and global mental
in brain chemistry and circuit function through health (pp. 262–287). New York: Oxford Univer-
multimodal human neuroimaging. Genes, Brain sity Press.
and Behavior, 11(6), 633–642. Hariri, A. R., Drabant, E. M., Munoz, K. E., Kol-
Fodor, J. A. (1983). The modularity of mind. Cam- achana, B. S., Mattay, V. S., Egan, M. F., & Wein-
bridge, MA: MIT Press. berger, D. R. (2005). A susceptibility gene for
Forbes, E. E., Brown, S. M., Kimak, M., Ferrell, R. E., affective disorders and the response of the human
Manuck, S. B., & Hariri, A. R. (2009). Genetic vari- amygdala. Archive of General Psychiatry, 62,
ation in components of dopamine neurotransmis- 146–152.
sion impacts ventral striatal reactivity associated Hariri, A. R., Goldberg, T. E., Mattay, V. S., Kol-
with impulsivity. Molecular Psychiatry, 14(1), achana, B. S., Collicott, J. H., Egan, M. F., & Wein-
60–70. berger, D. R. (2003). Brain-derived neurotrophic
Fossella, J., Green, A. E., & Fan, J. (2006). Evaluation factor val66met polymorphism affects human
of a structural polymorphism in the ankyrin repeat memory-related hippocampal activity and predicts
and kinase domain containing-1 (ANKK1) gene memory performance. Journal of Neuroscience,
and the activation of executive attention net- 23(17), 6690–6694.
works. Cognitive, Affective, & Behavioral Neuro- Hariri, A. R., Mattay, V. S., Tessitore, A., Kolachana,
science, 6, 71–78. B., Fera, F., Goldman, D., Egan, M. F., & Wein-
Gazzaniga, M. S., Mangun, G. R., & Ivry, R. B. (2002). berger, D. R. (2002). Serotonin transporter genetic
Cognitive neuroscience: The biology of the mind. variation and the response of the human amyg-
New York: Norton. dala. Science, 297, 400–403.
Goh, J. O. S. (2021). Acculturation by plasticity and Hariri, A. R., & Weinberger, D. R. (2003a). Functional
stability in neural processes: Considerations for neuroimaging of genetic variation in serotonergic
global mental health challenges. In J. Y. Chiao, S.- neurotransmission. Genes, Brain and Behavior,
C. Li, R. Turner, S. Y. Lee-Tauler & B. A. Pringle 2(6), 341–349.
(Eds.), The Oxford handbook of cultural neurosci- Hariri, A. R., & Weinberger, D. R. (2003b). Imaging
ence and global mental health (pp. 409–437). genomics. British Medical Bulletin, 65, 259–270.
New York: Oxford University Press. Heils, A., Mössner, R., & Lesch, K. P. (1997). The
Golchert, J., Roehr, S., Luck, T., Wagner, M., Fuchs, human serotonin transporter gene polymorphism:
A., Wiese, B., … Riedel-Heller, S. G. (2019). Basic research and clinical implications. Journal of
Women outperform men in verbal episodic Neural Transmission, 104(10), 1005–1014.
memory even in oldest-old age: 13-year longitudi- Hille, B. (1984). Ionic channels in excitable mem-
nal results of the AgeCoDe/AgeQualiDe study. branes. Sunderland, UK: Sinauer.
Journal of Alzheimer’s Disease, 69(3), Ihne, J., Gallagher, N., Sullivan, M., Callicott, J., &
857–869. Green, A. E. (2016). Is less really more: Does a pre-
Green, A.E., & Dunbar, K. (2012). Mental function as frontal efficiency genotype actually confer better
genetic expression: Emerging insights from cogni- performance when working memory becomes dif-
tive neurogenetics. In K. J. Holyoak & R. Morrison ficult? Cortex, 74, 79–95.
(Eds.), The Oxford handbook of thinking and rea- Kandel, E. R., & Schwartz, J. (1985). (Eds.), Principles
soning. Oxford, UK: Oxford University Press. of neural science (2nd ed.). New York: Elsevier.
Cognitive Neurogenetics 113

Kessler, R. C., & Ustun, T. B. (2008). The WHO World Sasaki, J. Y., LeClair, J., West, A. L., & Kim, H. S.
Mental Health Surveys: Global perspectives on the (2016). The gene-culture interaction framework
epidemiology of mental disorders. Cambridge, and implications for health. In J. Y. Chiao, S.-C. Li,
UK: Cambridge University Press. R. Seligman & R. Turner (Eds.), The Oxford hand-
Lamar, M., Resnick, S. M., & Zonderman, A. B. book of cultural neuroscience (pp. 279–298). New
(2003). Longitudinal changes in verbal memory in York: Oxford University Press.
older adults: Distinguishing the effects of age from Scott, K. M. (2021). Cross-national epidemiology of
repeat testing. Neurology, 60(1), 82–86. mental health disorders. In J. Y. Chiao, S.-C. Li, R.
Lundervold, A. J., Wollschläger, D., & Wehling, E. Turner, S. Y. Lee-Tauler & B. A. Pringle (Eds.), The
(2014). Age and sex related changes in episodic Oxford handbook of cultural neuroscience and
memory function in middle aged and older adults. global mental health (pp. 3–22). New York: Oxford
Scandinavian Journal of Psychology, 55(3), University Press.
225–232. Sejnowski, T. J., & Churchland, P. S. (1989). Brain and
McGraw, L. A., & Young, L. J. (2010). The prairie cognition. In M. I. Posner (Ed.), Foundations of
vole: an emerging model organism for under- cognitive science. Cambridge, MA: MIT Press.
standing the social brain. Trends in Neurosciences, Shepherd, G. M. (2004). The synaptic organization of
33(2), 103–109. the brain (5th ed.). Oxford, UK: Oxford University
Menon, V. (2012). Functional connectivity, neurocog- Press.
nitive networks, and brain dynamics. In M. I. Rabi- Stein, D. J., Chiao, J. Y., & van Honk, J. (2016). Cul-
novich, K. J. Friston & P. Varona, P. (Eds.), Principles tural neuroscience in South Africa: Promises and
of brain dynamics: Global state interactions pitfalls. In J. Y. Chiao, S.-C. Li, R. Seligman & R.
(pp. 27–48). Cambridge, MA: MIT Press. Turner (Eds.), The Oxford handbook of cultural
Munafò, M. R., Brown, S. M., & Hariri, A. R. (2008). neuroscience (pp. 143–154). New York: Oxford
Serotonin transporter (5-HTTLPR) genotype and University Press.
amygdala activation: A meta-analysis. Biological Stevenson, S., Bruce, L., Dwosh, E., Beattie, B. L., &
Psychiatry, 63(9), 852–857. Illes, J. (2016). When culture informs neuroscience:
Neumann, D. L., Chang, R. C. K., Wang, Y., & Boyle, Considerations for community-based neurogenetic
G. J. (2016). Cognitive and affective components research and clinical care in a first nation commu-
of empathy and their relationship with personality nity with early onset familial Alzheimer disease. In
dimensions in a Chinese Sample. Asian Journal of J. Y. Chiao, S.-C. Li, R. Seligman & R. Turner (Eds.),
Social Psychology, 19, 244–253. The Oxford handbook of cultural neuroscience
Nikolova, Y. S., & Hariri, A. R. (2015). Can we observe (pp. 171–182). New York: Oxford University Press.
epigenetic effects on human brain function? Tandon, P. N. (2000). The decade of the brain: A brief
Trends in Cognitive Science, 19(7), 366–373. review. Neurology India, 48(3), 199–207.
Nomura, M. (2016). Genes, brain and culture through Uchiyama, R. & Muthukrishna, M. (2021). Cultural
a 5-HTT Lens. In J. Y. Chiao, S.-C. Li, R. Seligman & evolutionary neuroscience. In J. Y. Chiao, S.-C. Li,
R. Turner (Eds.), The Oxford handbook of cultural R. Turner, S. Y. Lee-Tauler & B. A. Pringle (Eds.),
neuroscience (pp. 121–128). New York: Oxford The Oxford handbook of cultural neuroscience
University Press. and global mental health (pp. 23–56). New York:
O’Reilly, R. C., & Munakata, Y. (2000). Computa- Oxford University Press.
tional explorations in cognitive neuroscience. Varnum, M. E. W., & Hampton, R. S. (2021). Cultural
Cambridge, MA: MIT Press. changes in neural structure and function. In J. Y.
Parasuraman, R., & Jiang, Y. (2012). Individual differ- Chiao, S.-C. Li, R. Turner, S. Y. Lee-Tauler & B. A.
ences in cognition, affect, and performance: Pringle (Eds.), The Oxford handbook of cultural
Behavioral, neuroimaging, and molecular genetic neuroscience and global mental health (pp. 387–
approaches. NeuroImage, 59(1), 70–82. 408). New York: Oxford University Press.
Patel, V., Minas, H., Cohen, A., & Prince, M. J. (2014). Viding, E., Williamson, D. E., & Hariri, A. R. (2006).
Global mental health: Principles and practice. New Developmental imaging genetics: Challenges and
York: Oxford University Press. promises for translational research. Developmental
Reinvang, I., Deary, I. J., Fjell, A. M., Steen, V. M., Psychopathology, 18(3), 877–892.
Espeseth, T., & Parasuraman, R. (2010). Neuroge- Villela, D., Suemoto, C. K., Leite, R., Pasqualucci, C.
netic effects on cognition in aging brains: A window A., Grinberg, L. T., Pearson, P., & Rosenberg, C.
of opportunity for intervention? Frontiers in (2018). Increased DNA copy number variation
Aging Neuroscience, 2. doi: 10.3389/fnagi. mosaicism in elderly human brain. Neural Plasticity.
2010.00143 doi: 10.1155/2018/2406170
8
Dopamine System and Cognitive
Function across the Adult Life Span
Saana M. Korkki, Goran Papenberg,
Marc Guitart-Masip, Alireza Salami, Nina Karalija,
Lars Nyberg, and Lars Bäckman

INTRODUCTION an emphasis on molecular-imaging studies that


enable in vivo characterization of various compo-
Following the discovery of dopamine (DA) as an nents of the DA system. Although studies concern-
independent neurotransmitter in the 1950s, DA ing these cognitive functions are largely discussed
has been recognized to regulate motor control, separately, partly overlapping mechanisms likely
higher-order cognitive function, as well as motiva- underpin the influence of DA on behavior across
tional and affective aspects of behavior (Björklund domains (Diederen and Fletcher, 2021; Eriksson
and Dunnett, 2007; Salamone and Correa, 2012; et al., 2015; Gardner et al., 2018; Nyberg and
Tritsch and Sabatini, 2012). DA dysfunction is Eriksson, 2016; Westbrook and Braver, 2016). We
implicated in the cognitive and motor symptoms acknowledge that other neurotransmitter systems,
of several neurodegenerative (e.g., Parkinson’s such as acetylcholine and noradrenaline, also play
disease, Huntington’s disease), psychiatric (e.g., an important role in cognitive function and age-
schizophrenia, obsessive compulsive disorder, related cognitive deficits, as reviewed elsewhere
addiction), and developmental (e.g., attention (Everitt and Robbins, 1997; Holland et al., 2021;
deficit hyperactivity disorder) disorders (Klein Rieckmann and Nyberg, 2020).
et al., 2019), as well as in cognitive impairments
associated with healthy aging (Bäckman et al.,
2006, 2010).
In this chapter, we provide an overview of the DOPAMINE SYSTEM AND
role of DA in higher-order cognition across the
adult life span. Specifically, we focus on the role of DOPAMINERGIC PATHWAYS
DA in learning and memory, cognitive control, and
value-based decision making; cognitive processes In the human brain, the majority of dopaminergic
typically associated with dopaminergic neuro- neurons originate from the substantia nigra (SN)
modulation (Ott and Nieder, 2019; Schultz, 2007; and the ventral tegmental area (VTA) of the mid-
Shohamy and Adcock, 2010). We further discuss brain, from where they project to a range of corti-
evidence for contribution of dopaminergic losses cal and subcortical regions. Midbrain dopaminergic
to age-related impairments in these domains, with projections are traditionally divided into three
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 115

pathways (Björklund and Dunnett, 2007). The striatum are connected to each other via the mid-
nigrostriatal pathway contains dopaminergic neu- brain, with the ascending spiral organization of
rons from the SN projecting to the dorsal striatum, these connections enabling information flow from
the mesolimbic pathway neurons from the VTA the ventral striatum to associative and sensory-
projecting to the ventral striatum and other limbic motor areas of the striatum (Haber et al., 2000;
regions, and the mesocortical pathway contains Haber and Knutson, 2010).
neurons from the VTA projecting to the cortex
(Figure 8.1A) (Björklund and Dunnett, 2007). A
fourth, tuberoinfundibular pathway comprises
dopaminergic neurons from the arcuate nucleus of
the hypothalamus that project to the median emi-
PRE- AND POST-SYNAPTIC
nence and is involved in controlling secretion of DOPAMINERGIC MECHANISMS
prolactin (Vallone et al., 2000).
In addition to receiving dense dopaminergic In contrast to fast-acting neurotransmitters that
innervation from the midbrain, the striatum is mediate direct synaptic transmission, DA and
connected to cortical regions via topographically other monoamines act on a slower timescale that
organized cortico-striato-thalamo-cortical loops ranges from hundreds of milliseconds to minutes
(Alexander et al., 1991; Haber and Knutson, 2010; (Greengard, 2001). DA is synthesized in axon
Parent and Hazrati, 1995). Post-commissural terminals of dopaminergic cells, where the
parts of the dorsal putamen receive input from enzyme tyrosine hydroxylase (TH) converts the
sensory-motor regions, pre-commissural putamen amino acid tyrosine into l-3,4-dihydroxyphenyla-
and dorsal caudate input from associative regions, lanine (L-DOPA), which in turn is converted into
and ventral striatum input from limbic regions. DA by aromatic-L-amino acid decarboxylase
Information from the striatum is relayed back to (Elsworth and Roth, 1997; Klein et al., 2019).
the cortex via the thalamus, with DA playing a Cytosolic DA is packaged into synaptic vehicles
key role in modulating signaling in the “direct” by the vesicular monoamine transporter 2
and “indirect” pathways that facilitate and sup- (VMAT2) (Klein et al., 2019). After release of DA
press thalamo-cortical connections, respectively from the presynaptic terminal, the DA transporter
(Surmeier et al., 2007). Different parts of the (DAT) enables rapid reuptake of DA from the

Figure 8.1 Dopaminergic pathways and pre- and post-synaptic components.


A The major dopaminergic pathways in the human brain.
Source: From Papenberg et al., 2019.
B Pre- and post-synaptic components of the dopaminergic system.
Source: From Cenci, 2014.
116 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

synaptic cleft back into the presynaptic terminal from endogenous DA (Laruelle, 2000), these
(Elsworth and Roth, 1997). DAT is abundant in methods can also be used to examine DA release
the striatum, but expressed at lower levels in by assessing changes in DA receptor binding fol-
extrastriatal regions, where norepinephrine trans- lowing cognitive or pharmacological manipula-
porter (NET) mediates DA reuptake (Borgkvist tions postulated to evoke DA release (Cropley
et al., 2012; Morón et al., 2002). The enzymes et al., 2006).
catechol-O-methyltransferase (COMT) and mon-
oamine oxidase (MAO) facilitate degradation of
DA into inactive metabolites (Elsworth and Roth,
1997).
On the post-synaptic cell, DA acts via G pro- DOPAMINERGIC CONTRIBUTIONS TO
tein-coupled receptors that can be divided into two REWARD LEARNING AND VALUE-BASED
main classes: D1-like receptors (D1DRs; compris- DECISION MAKING
ing D1 and D5 receptors) and D2-like receptors
(D2DRs; comprising D2, D3, and D4 receptors) Human and animal work indicate a critical role for
(Beaulieu and Gainetdinov, 2011). Although the DA in reward-based learning and decision making.
majority of DA receptors are located post-synap- Single-neuron recordings in the monkey midbrain
tically, D2DRs are also found in the presynaptic have shown dopaminergic neurons to fire in
terminal, where they function as autoreceptors to phasic bursts in response to delivery of unex-
regulate the synthesis, release, and reuptake of pected primary rewards, such as food or liquid
DA (Ford, 2014). D1DRs are expressed in high (Schultz, 1998; Schultz et al., 1997). Critically,
concentrations in both striatal and extrastriatal when a reward is consistently predicted by a cue,
regions, whereas D2DRs are found in high lev- dopaminergic activation gradually shifts to the
els in the striatum and in lower concentrations in time of the cue presentation (Schultz, 1998;
extrastriatal regions (Beaulieu and Gainetdinov, Schultz et al., 1993). DA neuron activity signals
2011). The cortical distribution of D1DRs appears the difference between anticipated and obtained
to follow a gradient of increasing density from rewards, thus providing a “reward prediction
primary sensorimotor cortex to higher-order asso- error” (RPE) signal consistent with models of
ciation areas (Froudist-Walsh et al., 2021). In reinforcement learning (Schultz, 2013; Schultz
addition to differences in their regional distribu- et al., 1997). Increased burst firing of dopaminer-
tion, D1DRs and D2DRs differ in their affinity gic neurons is observed when a reward greater
(i.e., strength of receptor binding) to DA (Beaulieu than predicted is received, whereas a dip below
and Gainetdinov, 2011). D1DRs have lower affin- baseline activity levels occurs in response to
ity to DA and are thought to mediate phasic reward omission or a reward smaller than expected
actions of DA, reflecting large transient increases (Bayer and Glimcher, 2005; Bayer et al., 2007;
in DA release that result from burst firing of dopa- Schultz, 2013). Learning from positive and nega-
minergic neurons in response to excitatory inputs tive outcomes is thought to differentially involve
(Goto et al., 2007). The higher affinity D2DRs striatal D1DRs and D2DRs expressed in medium-
may be more important for detecting subtler spiny neurons of the “direct” and “indirect” path-
changes in tonic, or sustained, DA levels related ways (Surmeier et al., 2007). D1DR-expressing
to pacemaker-like baseline firing of dopaminergic neurons in the “direct” pathway are thought to
neurons regulated by inhibitory inputs from the mediate learning from rewards and facilitate
ventral pallidum (Floresco et al., 2003; Goto et al., reward-associated behaviors, whereas D2DR-
2007). Upon binding of DA to the post-synaptic expressing neurons in the “indirect” pathway may
receptor, these two receptor classes are associated be critical for learning from negative outcomes
with different intracellular signaling cascades, and inhibition of behavior (Cox et al., 2015; Frank
with D1DR stimulation generally having excita- et al., 2004).
tory, and D2DR stimulation inhibitory, effects In human functional magnetic resonance imag-
on the post-synaptic neuron (Greengard, 2001; ing (fMRI) studies, blood-oxygen-dependent
Surmeier et al., 2007; Tritsch and Sabatini, 2012). (BOLD) signal differences approximating RPEs
In humans, molecular-imaging methods (e.g., have been observed in the SN/VTA, but also in
positron emission tomography or PET; single pho- mesolimbic regions beyond the midbrain, includ-
ton emission computed tomography or SPECT) ing the ventral striatum and the medial prefrontal
using specific radioligands to target pre- and post- cortex (D’Ardenne et al., 2008; Garrison et al.,
synaptic components of the DA system allow char- 2013). While the involvement of dopaminergic
acterization of the DA system in vivo. Given the mechanisms in the RPE cannot be directly assessed
sensitivity of radioligand binding to competition with fMRI, there is evidence that pharmacological
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 117

manipulations of the dopaminergic system impact thoughts and actions in a goal-oriented manner
the neural expression of RPEs in the human stria- (Miller and Cohen, 2001). In the prefrontal cortex,
tum (Pessiglione et al., 2006). Although lack- midbrain dopaminergic projections innervate both
ing the temporal resolution to isolate RPE-like excitatory pyramidal cells and inhibitory interneu-
responses, PET studies have found increased rons (Ott and Nieder, 2019; Ranganath and Jacob,
ventral striatal DA release during performance 2016). The more abundant D1DRs are expressed
of cognitive tasks that involve unpredicted mon- in all layers of the prefrontal cortex, whereas
etary rewards (Pappata et al., 2002; Zald et al., D2DRs are predominantly located in layer V
2004). Reward-related increases in ventral striatal (Lidow et al., 1991; Ott and Nieder, 2019).
DA release have further been observed to corre- Early evidence for a role of DA in working
late with ventral striatal fMRI activation during memory, a critical component of cognitive control
reward anticipation (Schott et al., 2008). This sug- (Miller and Cohen, 2001), was provided by studies
gests that striatal DA release, and the subsequent showing depletion of DA from the monkey pre-
activation of D1DRs generally leading to excita- frontal cortex to result in similar working memory
tory post-synaptic effects, may be reflected in deficits as seen after surgical removal of prefrontal
increased BOLD signal in the striatum (Knutson cortex tissue (Brozoski et al., 1979). Subsequent
and Gibbs, 2007). human and non-human primate pharmacologi-
DA also appears to influence value-based cal work has demonstrated a role for DA not
choices via motivational processes that direct the only in working memory (Kimberg et al., 1997;
allocation of effort to obtain rewards (Salamone Sawaguchi and Goldman-Rakic, 1991), but also
and Correa, 2012; Westbrook et al., 2021). In ani- in other aspects of cognitive control, such as task
mal studies, DA depletion and DA antagonists switching (Mehta et al., 1999, 2004), and selective
have been observed to increase rats’ preference attention (Noudoost and Moore, 2011; ter Huurne
for low rewards requiring minimal effort in com- et al., 2015). Corresponding effects have been
parison to high rewards that require greater effort observed at the neural level, with D1DR signal-
(Cousins and Salamone, 1994; Salamone and ing being critical for delay-period firing (Williams
Correa, 2012). Similarly, markers of striatal DA and Goldman-Rakic, 1995) and D2DR signaling
have been found to correlate with participants’ for response-related firing (Wang et al., 2004)
willingness to exert physical or cognitive effort to of prefrontal neurons during working memory.
receive monetary rewards in human PET studies Human studies also implicate DA in supporting
(Treadway et al., 2012; Westbrook et al., 2020). the functional dynamics underpinning working
Motivational influences of DA may reflect changes memory. For instance, administration of D1DR
in tonic DA levels (Niv et al., 2005, 2007). Tonic agonists reduces fronto-parietal activation dur-
DA has been proposed to encode the average rate ing working memory in healthy young volunteers
of rewards, signaling the opportunity costs of inac- (Fischer et al., 2010), with individual differences
tion or sloth and impacting the vigor of responding in D1DR availability further correlating with the
(Niv et al., 2007). Indeed, an association between degree of functional connectivity within this net-
minute-to-minute fluctuations in DA levels and work (Rieckmann et al., 2011a; Roffman et al.,
average reward rate has been observed in rodent 2016).
studies (Hamid et al., 2016). In humans, increases Interestingly, several studies have observed
in reward rate decrease response times in a per- opposing effects of dopaminergic drugs on cog-
ceptual discrimination task (Guitart-Masip et al., nitive performance in different individuals (Cools
2011), with such effects being greater after admin- and D’Esposito, 2011). For instance, DA receptor
istration of the DA precursor L-DOPA (Beierholm agonists have been found to enhance cognitive per-
et al., 2013), as well as for individuals with high formance in individuals with low working memory
striatal DA synthesis capacity (and slow perfor- capacity, but impair performance in individuals
mance) (Hofmans et al., 2022). with high working-memory capacity (Kimberg
et al., 1997). Given associations between DA syn-
thesis and working memory (Landau et al., 2009),
such findings have been interpreted to reflect an
DOPAMINERGIC CONTRIBUTIONS TO inverted U-shaped relationship between DA and
cognitive performance, where both insufficient and
COGNITIVE CONTROL AND WORKING excessive DA levels can result in cognitive impair-
MEMORY ments (Cools and D’Esposito, 2011). Consistent
with this proposal, in animal studies, dose-
Beyond reward, DA also modulates prefrontally dependent effects of DA stimulation have been
dependent cognitive control operations that guide observed at the neural level, with low dose D1DR
118 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

agonists improving, and high dose D1DR agonists DA from locus coeruleus neurons (synthesizing
degrading, spatial tuning of prefrontal neurons DA as a precursor for noradrenaline) may provide
during the delay-phase of a working-memory task another source of DA to the hippocampus
(Vijayraghavan et al., 2007). A recent computa- (Duszkiewicz et al., 2019). Animal studies indi-
tional model extended these findings by demon- cate a role for hippocampal DA in modulating the
strating moderate levels of DA release to facilitate persistence, or durability, of episodic memories,
persistent delay-period activity, a putative hall- likely reflecting the influence of DA on memory
mark of working memory (Constantinidis et al., consolidation (Lisman et al., 2011). Consistent
2018), in a distributed network of frontal, pari- with a role in memory consolidation, intra-hip-
etal, and temporal regions, whereas insufficient or pocampal administration of D1DR antagonists
excessive DA release resulted in a transient activ- prior to memory encoding disrupts delayed (6–24
ity pattern in this network (Froudist-Walsh et al., hours), but not short-term (20–30 minutes), spa-
2021). Moreover, recent evidence also suggests an tial-location memory (Bethus et al., 2010;
inverted U-shaped relationship between DA and O’Carroll et al., 2006). Others have reported simi-
brain large-scale functional network organization, lar effects for expression of fear memory when a
with cortical regions showing intermediate levels DA antagonist was administered 12h after encod-
of D1DR availability demonstrating the greatest ing, but not at shorter post-encoding intervals
nodal strength (i.e., strength of functional connec- (Rossato et al., 2009). Indeed, DA-dependent
tions) (Nordin et al., 2022). stimulation of protein synthesis is thought to be
Variation in the effects of DA drug admin- critical for the late phase of hippocampal long-
istration may also in part reflect differences in term potentiation or LTP (Duszkiewicz et al.,
the nature of cognitive processes investigated 2019; Lisman et al., 2011; for dopaminergic
(Floresco, 2013). Biophysically grounded compu- modulation of cortico-striatal LTP, see Calabresi
tational models suggest distinct roles for prefron- et al., 2007). LTP refers to activity-dependent
tal D1DRs and D2DRs in working memory, with increases in synaptic strength and is considered a
a D1DR-dominated state supporting robust main- key cellular substrate of memory (Bliss and
tenance of memory representations and a D2DR- Collingridge, 1993). In rodents, late hippocampal
dominated state facilitating flexible switching LTP is reduced by D1DR antagonists (O’Carroll
between representations (Durstewitz and Seamans, and Morris, 2004), and by genetic knockout of
2008; Seamans and Yang, 2004). Similar roles D1DRs (Granado et al., 2008; Matthies et al.,
have been attributed to prefrontal and striatal DA 1997). Similar effects on LTP following D2DR
(Cools, 2019; Cools and D’Esposito, 2011). While blockage have also been observed, with presynap-
prefrontal DA is thought to be critical for cogni- tic D2DRs potentially playing a role in long-term
tive stability, striatal DA may be more important depression (LTD; activity-dependent decrease in
for mediating cognitive flexibility by gating infor- synaptic strength) (Rocchetti et al., 2015). In addi-
mation in and out of working memory in accord- tion to modulating synaptic plasticity, rodent evi-
ance with current task goals (Cools, 2019; Cools dence suggests that DA may contribute to memory
and D’Esposito, 2011; O’Reilly, 2006). Indeed, consolidation by promoting post-encoding hip-
increased striatal DA release has been observed pocampal reactivation of encoded memory repre-
following a working memory updating training sentations (McNamara et al., 2014). In humans,
protocol (Bäckman et al., 2011b), with genetic post-encoding functional connectivity between
(Persson et al., 2015) and PET-derived (Nyberg the VTA and the hippocampus has been found to
et al., 2009a) evidence further suggesting a similar predict long-term, but not immediate, associative
role for striatal DA in the updating of long-term recall (Tompary et al., 2015), providing indirect
memory (LTM) representations. support for dopaminergic modulation of hip-
pocampal post-encoding processes.
Dopaminergic modulation of LTM is thought to
serve an adaptive function, enhancing the retention
of novel, surprising, and motivationally relevant
DOPAMINERGIC CONTRIBUTIONS TO aspects of prior experiences (Frank and Kafkas,
EPISODIC MEMORY 2021; Miendlarzewska et al., 2016; Shohamy
and Adcock, 2010). Exposure to novelty evokes
The hippocampus, a key region for long-term hippocampal DA release (Moreno-Castilla et al.,
episodic memory (Eichenbaum, 2000; Squire, 2017), promotes memory retention, and induces
1992), receives dopaminergic projections from the hippocampal LTP (Li et al., 2003; Wang et al.,
VTA and, to a lesser extent, the SN via the mes- 2010). Specifically, the CA1 subfield of the hip-
olimbic pathway (Gasbarri et al., 1994). In addi- pocampus facilitates novelty detection by compar-
tion, recent evidence suggests that co-release of ing the sensory input arriving from the cortex to
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 119

memory-based predictions from the CA3 (Lisman 2006), with D1DRs, D2DRs, and DAT similarly
and Grace, 2005). Hippocampal novelty signals exhibiting age-related reductions (de Keyser et al.,
are conveyed to the dopaminergic midbrain via 1990; Erixon-Lindroth et al., 2005; Kaasinen
a polysynaptic pathway involving the nucleus et al., 2000; Seeman et al., 1987) that appear to be
accumbens and the ventral pallidum (Lisman and correlated (Ishibashi et al., 2009; Volkow et al.,
Grace, 2005; Lisman et al., 2011). This results in 1998b). Meta-analytic estimates of age-related
reduced tonic inhibition of midbrain dopaminergic reductions in fronto-striatal DA receptor availabil-
neurons from the ventral pallidum and increased ity and striatal DAT of 8.2–14.0% per decade have
baseline dopaminergic neuron firing (Floresco been reported, with D1DRs slightly more affected
et al., 2003), which in turn may facilitate phasic than D2DRs (Karrer et al., 2017). Findings regard-
burst firing (Lisman and Grace, 2005; Lodge and ing striatal DA synthesis capacity are less consist-
Grace, 2006). Thus, the hippocampus itself can ent, with no meta-analytic evidence for age-related
regulate dopaminergic input from the midbrain to differences (Karrer et al., 2017). It is possible that
the hippocampus. preserved DA synthesis capacity, in combination
DA is also thought to underpin value-related with reduced DAT, may reflect a compensatory
enhancements of LTM seen for motivationally mechanism, prolonging the availability of DA in
relevant stimuli (Knowlton and Castel, 2022; the synaptic cleft (Karrer et al., 2017). However,
Shohamy and Adcock, 2010). FMRI evidence associations between DA synthesis and release
indicates stimuli associated with monetary have not been observed in younger adults (Berry
rewards to result in increased activation of the et al., 2018a), and evidence for relationships
hippocampus, ventral striatum, and SN/VTA between striatal DA synthesis capacity and func-
during memory formation (Adcock et al., 2006; tion is mixed in older age (see section on
Wittmann et al., 2005). Similarly, reward has been Dopaminergic contributions to age-related altera-
shown to impact hippocampal post-encoding tions in cognitive control and working memory).
processes, with the degree of post-encoding con- In addition to differences between pre- and
nectivity between the hippocampus and the VTA postsynaptic components (Karrer et al., 2017),
predicting memory benefits for stimuli learned regional heterogeneity in the rate and trajectory of
in high-reward contexts (Gruber et al., 2016). age-related DA decreases may exist. While many
Consistent with effects seen for DA stimulation studies have reported linear age-related decreases
in rodents (McNamara et al., 2014), reward also in PET markers of the DA system (Karrer et al.,
promoted post-encoding hippocampal reactiva- 2017; Malén et al., 2022; Reeves et al., 2002),
tion of memory representations in this study non-linear patterns have also been observed, often
(Gruber et al., 2016). Beyond more general ben- characterized by accelerated DA receptor losses
efits for reward-associated stimuli, recent work after midlife (Bäckman et al., 2006, 2010). A
has also reported the degree of RPEs experienced recent study using the high-affinity radioligand
18
during memory encoding to predict later episodic F-fallypride for characterization of both stri-
remembering, with this effect being mediated by atal and extrastriatal D2DRs observed regional
RPE-related activation of the ventral striatum heterogeneity in the degree of age-related recep-
(Calderon et al., 2021). tor losses, with the largest decline evident in the
temporal and frontal cortex (Seaman et al., 2019).
Interestingly, several regions also displayed non-
linear associations with adult age, which were
characterized by strong reductions in D2DR avail-
AGE-RELATED CHANGES IN THE ability from young to middle age, followed by
DOPAMINERGIC SYSTEM more modest age-related differences in old age
(Seaman et al., 2019). This pattern contrasts some
Concomitant with decreases in many cognitive prior findings from smaller cohorts, as well as typi-
domains, including episodic and working memory, cal age-related trajectories seen for other measures
cognitive control, and decision making, the dopa- of brain integrity, such as hippocampal volume
minergic system exhibits age-related deterioration (Nobis et al., 2019). Regional variation in aging
(Bäckman et al., 2006, 2010). In contrast to some effects is also suggested by findings indicating
clinical conditions that affect more selective D1DR availability across dopaminergic pathways
aspects of the DA system (Kim et al., 2002; to be correlated in young, but to a lesser extent in
Laruelle et al., 2000), aging is associated with older age (Rieckmann et al., 2011b). An important
relatively widespread declines in DA system caveat to note, however, is that prior studies have
integrity. Loss of dopaminergic neurons in the SN essentially relied on cross-sectional comparisons,
of approximately 3% per decade occurs with which may not map onto longitudinal changes in
aging (Fearnley and Lees, 1991; Bäckman et al., DA-system integrity (Raz et al., 2005). Recently,
120 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

first 5-year longitudinal estimates of D2DR cognitive control have been more mixed, with
declines in older age were reported, suggesting an some studies showing positive (Landau et al.,
overestimation of striatal D2DR decline in previ- 2009) and other negative (Klostermann et al.,
ous cross-sectional studies (Karalija et al., 2022). 2012) associations to brain function. In a subset of
Aging-related losses were evident in the striatum studies, relationships of DA synthesis to behavior
and in several extrastriatal regions (hippocampus, and brain function have also been limited to
anterior cingulate, orbitofrontal cortex) in this younger adults (Berry et al., 2018b; Braskie et al.,
study, with correlated D2DR declines observed 2011), suggesting a potentially age-varying rela-
within associative and reward-related regions tionship (Berry et al., 2016).
(Karalija et al., 2022). Longitudinal data on D1DR Candidate-gene studies have provided con-
availability from a large life span cohort are cur- verging evidence for a role of DA in maintaining
rently underway (Nordin et al., 2022; see Marek cognitive control abilities in later life (Papenberg
et al., 2011 for longitudinal data on DAT). et al., 2015). One of the most commonly stud-
ied genetic polymorphisms in this context is the
COMT Val158Met that influences metabolic degra-
dation of DA in the prefrontal cortex and thereby
endogenous DA levels (Matsumoto et al., 2003).
DOPAMINERGIC CONTRIBUTIONS Carriers of the advantageous Met allele, which is
TO AGE-RELATED ALTERATIONS IN associated with slower DA degradation and higher
COGNITIVE CONTROL AND WORKING endogenous prefrontal DA levels (Chen et al.,
MEMORY 2004; Lotta et al., 1995), display better perfor-
mance on tasks of executive function and work-
ing memory (Nagel et al., 2008; Störmer et al.,
In addition to displaying negative associations
2012), as well as more efficient prefrontal activa-
with age and positive associations with cognitive
tion (Nyberg et al., 2014). Such effects are often
performance, statistical control of PET-derived
magnified in later life (Papenberg et al., 2015).
markers of DA system integrity often accounts for
Critically, carriers of the COMT Val allele have
age-related variation in cognitive performance
also been found to exhibit greater longitudinal
(Bäckman et al., 2000; Erixon-Lindroth et al.,
decline in executive function than Met carriers (de
2005, but see Juarez et al., 2019), suggesting a key
Frias et al., 2005). In addition to COMT, variation
contribution of DA losses to cognitive deficits
in D2DR-related genes has been associated with
associated with aging (Bäckman et al., 2006,
inhibitory control (Colzato et al., 2013), and with
2010). Individual differences in DAT and DA
fronto-striatal working memory activation (Li
receptor availability have been found to positively
et al., 2019) in aging. Thus, converging molecular
correlate with working memory, attention, set-
imaging and genetic evidence indicates a contribu-
shifting, and other measures of cognitive control
tion of DA to deficits in cognitive control in older
in adult life span and older adult samples (Erixon-
age, via impacting cortico-striatal function.
Lindroth et al., 2005; Juarez et al., 2019; Landau
et al., 2009; MacDonald et al., 2012; Volkow
et al., 1998a). These associations may in part
reflect the influence of DA on maintenance of
functional integrity of fronto-parietal and cortico- DOPAMINERGIC CONTRIBUTIONS TO
striatal networks important for goal-oriented AGE-RELATED ALTERATIONS IN EPISODIC
behavior. Older adults typically show diminished
modulation of fronto-parietal BOLD signal in
MEMORY
response to task demands, as well as reduced
functional connectivity within this network PET studies have also demonstrated DA receptor
(Geerligs et al., 2015; Nyberg et al., 2009b; Rieck availability (Bäckman et al., 2000; Cervenka
et al., 2017). Age-related reductions in striatal and et al., 2008) and DAT (Erixon-Lindroth et al.,
extrastriatal D1DR availability in part explain the 2005) to be positively associated with measures of
diminished load-dependent modulation of fronto- episodic memory across the adult life span.
parietal activation during working memory Striatal DAT was further observed to predict lon-
(Bäckman et al., 2011a), with caudate D1DR gitudinal changes in episodic memory independ-
availability further correlating positively with ent of the integrity of striatal resting state
fronto-parietal functional connectivity during functional networks over a period of 2–7 years
working memory in older age (Rieckmann et al., (Rieckmann et al., 2018). In genetic studies, ben-
2011a). Findings regarding the role of striatal DA eficial effects of advantageous DA-related poly-
synthesis capacity in age-related deficits in morphisms have been observed for associative
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 121

recognition (Papenberg et al., 2017), backward (Berry et al., 2019; Lighthall, 2020; Samanez-
serial recall (Li et al., 2013), and updating of LTM Larkin and Knutson, 2015). While the processing
representations (Persson et al., 2015) in older age. of basic rewards appears to be relatively well
Consistent with the role of DA in memory con- preserved in aging, older adults typically display
solidation (Lisman et al., 2011), older individual impaired performance on tasks that require learn-
carriers of advantageous alleles of D2DR and ing probabilistic, or dynamically changing, stimu-
DAT-related genes display enhanced retention of lus-reward contingencies (Lighthall, 2020;
visual information after long (1 week), but not Samanez-Larkin and Knutson, 2015). Age differ-
short (2.5 hours), study-test intervals (Papenberg ences in sensitivity to gains and losses when
et al., 2013). acquiring and updating value representations have
Fewer studies have examined associations been observed (Betts et al., 2020; Simon et al.,
between PET markers of DA system integrity and 2010; Sojitra et al., 2018), with older adults typi-
functional imaging measures of episodic mem- cally demonstrating slower learning of action-
ory processes. Some evidence suggests oppos- outcome contingencies (Cutler et al., 2021; Mell
ing relationships between D1DR availability and et al., 2005). At the neural level, these findings are
BOLD signal in the ventral visual cortex during paralleled by fMRI studies demonstrating age-
face recognition across young and older adults related reductions of RPE-related activity in the
(Turner et al., 2020). Specifically, while D1DR ventral striatum and the medial prefrontal cortex
availability was positively coupled with BOLD (Eppinger et al., 2013; Samanez-Larkin et al.,
activation in younger adults, negative associations 2014). Critically, L-DOPA administration was
were observed in older age, which may reflect found to improve reward-associated learning and
desynchronization of DA and BOLD activity in restore the ventral striatal RPE signal in older
aging (Turner et al., 2020). However, in pharma- adults who displayed drug-related performance
cological fMRI studies, L-DOPA (Chowdhury improvements (Chowdhury et al., 2013), implicat-
et al., 2012), as well as DA receptor agonists and ing dopaminergic mechanisms in these age-related
antagonists (Morcom et al., 2010), have been cognitive impairments.
observed to modulate memory encoding-related A recent multimodal neuroimaging study
activity in medial temporal and fronto-striatal (Dopamine, Aging, and Decision making; DAD)
regions in older age. Administration of DA recep- assessed the role of D1DR availability in age dif-
tor agonists and antagonists further modulated the ferences for various components of value-based
specificity of hippocampal context representa- decision making (de Boer et al., 2017, 2019,
tions in a baseline-dependent manner in one study 2020). One of the decision-making paradigms
(Abdulrahman et al., 2017), aligning with com- included in this study was a two-armed bandit
putational accounts that suggest a contribution of task that required participants to choose between
DA to reduced fidelity of neural representations two stimuli associated with fluctuating reward
in older age (Li et al., 2001). Despite prominent probabilities. Behaviorally, older adults made less
dopaminergic losses, many studies have observed optimal choices in this task, which was accom-
preserved reward-related enhancement of episodic panied by diminished value-related activation
memory performance in older age (Knowlton and of the ventromedial prefrontal cortex (de Boer
Castel, 2022), but also reported age-related reduc- et al., 2017). Importantly, value-related activation
tions in mesolimbic activation during encoding of of the ventromedial prefrontal cortex correlated
stimuli associated with monetary rewards (Geddes with D1DR availability in the nucleus accum-
et al., 2018). Thus, further research is required to bens across young and older adults (de Boer et al.,
assess the extent to which dopaminergic mecha- 2017). Independent of D1DR availability, struc-
nisms may continue to support value-based tural integrity of the white matter tracts connect-
enhancement of episodic memory in aging. ing the nucleus accumbens to the ventromedial
prefrontal cortex also predicted prefrontal value-
related signals (de Boer et al., 2020), highlighting
an interplay of functional, structural and neuro-
DOPAMINERGIC CONTRIBUTIONS TO chemical factors in contributing to age-related dif-
ferences in reward learning. In another paradigm,
AGE-RELATED ALTERATIONS IN REWARD participants were to execute or withhold a motor
LEARNING AND DECISION MAKING response to obtain rewards or avoid punishments
(de Boer et al., 2019). On this task, individual dif-
There is growing interest in the contribution of ferences in dorsal striatal D1DR availability were
altered DA signaling to age-related changes in related to the degree of participants’ motivational
reward-based learning and decision making bias (de Boer et al., 2019), that is, the tendency
122 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

to perform an action to obtain rewards even when episodic memory was further mediated by resting
the optimal choice is to withhold action, and to state functional connectivity between the ventral
withhold an action to avoid punishments event caudate and the medial temporal lobe (Nyberg
when the optimal choice is to perform an action et al., 2016). Interestingly, no linear associations
(Guitart-Masip et al., 2012). Analyses of whole- between D2DR availability and performance on
brain functional network organization during both tests of working memory, or perceptual speed,
rest and decision-making tasks further indicated were detected in this initial analysis (Nyberg
that the regions exhibiting the greatest cross-sec- et al., 2016). Aligning with prior reports of non-
tional age-related reduction in D1DR availability monotonic associations between DA and cog-
also showed the largest age differences in func- nition (Cools and D’Esposito, 2011; Floresco,
tional connectivity (Garzón et al., 2021), which 2013), results of a follow-up latent-profile analy-
is consistent with other evidence linking DA to sis indicated a non-linear multivariate association
maintenance of functional network integrity (Nour between episodic memory, working memory, and
et al., 2019; Rieckmann et al., 2011a; Roffman perceptual speed, and D2DR availability in the
et al., 2016). striatum, hippocampus, and cortex (Lövdén et al.,
2018). While majority of the sample (N = 99)
displayed both high cognitive performance and
high D2DR availability, two smaller subgroups
COGNITION, BRAIN, AND AGING STUDY with poor cognitive performance were detected.
In one of these subgroups (N = 42), reduced cog-
Despite several cross-sectional studies supporting nitive performance was coupled with low D2DR
a role for DA in age-related cognitive impair- availability, whereas the other low-performing
ments, longitudinal data on this topic remains subgroup (N = 40) exhibited high D2DR avail-
limited. This leaves the nature of longitudinal ability, especially in the striatum (Lövdén et al.,
changes in DA system integrity and their relation- 2018). This latter group displayed low educational
ship to cognitive changes in aging unclear. The attainment and high body-mass index, which are
Cognition, Brain, and Aging (COBRA) study is a also factors associated with psychiatric conditions
large (N = 181; 64–68 years at baseline) multi- affecting DA signaling (Lövdén et al., 2018). In
modal neuroimaging study that involves longitu- addition to mean-level cognition, similar subgroup
dinal assessment of D2DR availability, gray- and differences were observed for moment-to-moment
white-matter integrity, brain function, and cogni- variability in reaction time (Korkki et al., 2021).
tion over a 10-year period (Nevalainen et al., Two studies further examined the role of
2015; see also Nordin et al., 2022 for an ongoing D2DRs in maintaining functional integrity of
longitudinal study on D1DRs). Findings from the brain circuits involved in working memory using
first 5-year follow-up in COBRA have been data from an n-back working memory paradigm
recently reported, demonstrating longitudinal that participants completed in the MRI scanner
declines in D2DR availability in several striatal (Salami et al., 2018, 2019). Comparing two sub-
and extrastriatal regions (Karalija et al., 2022). groups of older individuals that differed in terms
Moreover, cross-sectional analyses of the baseline of their in-scanner working-memory performance
data have enabled well-powered investigation (high-performing group N = 113, low-performing
of the replicability of prior cross-sectional group N = 55), Salami et al. (2018) found
DA-cognition findings and the assessment of low-performing older adults to exhibit diminished
potential modifying factors, such as genetic vari- load-dependent modulation of fronto-parietal
ance or lifestyle variables. Several key findings activity, reduced fronto-parietal connectivity
from the baseline COBRA cohort are reviewed during both task and rest, as well as decreased
next. frontal D2DR availability. Individual differ-
The distribution of cortical and subcortical ences in D2DR availability were associated with
D2DR availability within the baseline COBRA thalamo-striato-cortical BOLD signal during the
cohort was found to map onto known anatomi- n-back task, with such associations being strong-
cal and functional organization of the DA system est when performing at maximum capacity limits
(Papenberg et al., 2019a). Consistent with the (i.e., the 3-back condition for the high-perform-
importance of DA for episodic memory (Shohamy ing group, and the 2-back condition for the low-
and Adcock, 2010), D2DR availability in the hip- performing group) (Salami et al., 2019). Nordin
pocampus and the caudate correlated positively et al. (2021) further observed D2DR availability
with episodic memory performance across the and episodic memory performance to be jointly
baseline cohort (Nyberg et al., 2016). The rela- associated with hippocampal functional connec-
tionship between caudate D2DR availability and tivity with medial prefrontal, posterior parietal,
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 123

and striatal regions during rest. Associations of females were also observed in the baseline
between D2DR availability and brain structural sample (Karalija et al., 2021b), whereas women
integrity were also observed in the baseline cohort. exhibited steeper longitudinal decline in D2DR
Specifically, whole-brain white matter lesion bur- availability in the nucleus accumbens (Karalija
den was negatively related to D2DR availability in et al., 2022).
the striatum and the hippocampus (Karalija et al., Given the potentially different pattern of effects
2019b). Individuals with the largest lesions were (Nyberg et al., 2010; Raz et al., 2005), cross-
also characterized by reduced episodic memory sectional observations from the COBRA baseline
and processing speed (Karalija et al., 2019b). assessment need to be corroborated by longitudi-
White-matter lesion progression also predicted nal data from the 5-year follow-up. Indeed, first
longitudinal losses in D2DR availability in asso- longitudinal findings suggest that the magnitude
ciative regions when examining data from the first of age-related changes in D2DR availability
5-year follow-up, whereas reduction in cortical may have been overestimated in previous cross-
perfusion was related to D2DR losses in limbic sectional comparisons (Karalija et al., 2022). The
regions (Karalija et al., 2022). longitudinal data further allow assessment of the
Another set of studies from the baseline relationship between the degree of DA losses and
COBRA cohort focused on identifying potential the degree of cognitive decline, as well as changes
modifying factors impacting D2DR-cognition in other brain parameters (e.g., grey-matter vol-
relationships in older age, possibly contributing ume, white-matter lesions, blood flow, functional
to some inconsistencies in DA-behavior associa- activation patterns) with aging. As Rabbitt (1993)
tions observed previously (Juarez et al., 2019). In queried, “Does it all go together when it goes?”.
terms of genetic factors, Karalija et al. (2019a) This issue is at the heart of the COBRA project,
observed significant D2DR-cognition associa- and the focus of ongoing analyses.
tions in C-carriers, but not T-homozygotes, of the
D2DR-gene polymorphism, C957T; likely reflect-
ing inflated D2DR binding via elevated recep-
tor affinity in the latter group (Hirvonen et al.,
2009). Similarly, positive associations between SUMMARY AND CONCLUSIONS
hippocampal D2DR availability and episodic
memory were restricted to individuals carrying In this chapter, we have presented evidence for a
beneficial alleles of the brain-derived neurotrophic contribution of dopaminergic neurotransmission
factor (BDNF) and the kidney and brain expressed to higher-order cognitive function across the adult
protein (KIBRA) polymorphisms (Papenberg life span. Converging findings from molecular
et al., 2019b), both of which have been associated neuroimaging, genetic, and pharmacological stud-
with hippocampus-dependent memory (Kambeitz ies, support a role for DA in modulating human
et al., 2012; Milnik et al., 2012). COMT was fur- memory, cognitive control, and value-based deci-
ther found to interact with D2DR availability to sion making. These diverse contributions may
influence memory performance, with the high- reflect dopaminergic modulation of partly distinct
est performance observed for individuals whose pathways, with the dorsal striatum and the pre-
D2DR availability matched their endogenous DA frontal cortex implicated in cognitive control and
levels, as indexed by COMT (Papenberg et al., working memory, and the ventral striatum and the
2020). This highlights the importance of an opti- mesolimbic system in reward-related processes
mal balance between endogenous DA levels and and long-term episodic memory. Evidence from
receptor availability. Accordingly, a DAT poly- cross-sectional studies further indicates age-
morphism, rs40184, presumably via regulating related-deterioration of the dopaminergic system,
DA availability, was associated with individual including loss of dopaminergic cells, transporters,
differences in striatal and hippocampal D2DR and receptors, that contributes to age-related cog-
levels, as well as working memory performance nitive deficits in these domains. Recent efforts
(Karalija et al., 2021a). Considering lifestyle fac- from large-scale multimodal neuroimaging
tors, Köhncke et al. (2018) observed the intensity cohorts have begun to elucidate potential modify-
of physical activity engagement to be positively ing factors, such as lifestyle and genetic variation,
associated with caudate D2DR availability and that may influence associations between DA
memory performance, whereas Karalija et al. system integrity and cognition in later life. While
(2019b) observed negative associations between longitudinal evidence on age-related DA declines
elevated cardiovascular disease risk and D2DR remains limited, ongoing longitudinal investiga-
availability in the putamen. Sex differences in tions (Nevalainen et al., 2015; Nordin et al., 2022)
D2DR availability and episodic memory in favor will enable assessment of the cascade of
124 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

neurochemical, structural, and functional brain Reward-motivated learning: Mesolimbic activation


changes associated with aging, and their relative precedes memory formation. Neuron, 50(3),
importance for maintaining cognitive performance 507–517.
in later life. Alexander, G. E., Crutcher, M. D., & DeLong, M. R.
Thus far, the majority of existing molecular (1991). Basal ganglia-thalamocortical circuits: Par-
imaging studies have focused on assessing a single allel substrates for motor, oculomotor, “prefrontal”
pre- or post-synaptic marker of DA system integ- and “limbic” functions. Progress in Brain Research,
rity. Given potential discordance of pre- and post- 85, 119–146.
synaptic components (Karrer et al., 2017), and the Bäckman, L., Ginovart, N., Dixon, R. A., Wahlin, T. B.,
importance of a balanced DA system in regulat- Wahlin, A., Halldin, C., & Farde, L. (2000). Age-
ing behavior (Berry et al., 2019; Papenberg et al., related cognitive deficits mediated by changes in
2020), future studies incorporating assessment of the striatal dopamine system. American Journal of
multiple pre- and post-synaptic markers within Psychiatry, 157(4), 635–637.
the same individuals will be critical for gaining a Bäckman, L., Karlsson, S., Fischer, H., Karlsson, P.,
more mechanistic understanding of DA contribu- Brehmer, Y., Rieckmann, A., MacDonald, S. W. S.,
tions to cognitive function, as well as of the exact Farde, L., & Nyberg, L. (2011a). Dopamine D1
age-related trajectories of different pre- and post- receptors and age differences in brain activation
synaptic components. Moreover, parallel assess- during working memory. Neurobiology of Aging,
ment of DA system integrity along with other 32(10), 1849–1856.
neurotransmitter systems supporting higher-order Bäckman, L., Lindenberger, U., Li, S.-C., & Nyberg, L.
cognition and contributing to age-related cogni- (2010). Linking cognitive aging to alterations in
tive deficits (e.g., acetylcholine, noradrenaline) dopamine neurotransmitter functioning: Recent
will be important for delineating their interplay in data and future avenues. Neuroscience & Biobe-
influencing cognitive function in both health and havioral Reviews, 34(5), 670–677.
disease. Bäckman, L., Nyberg, L., Lindenberger, U., Li, S.-C., &
In summary, the neurotransmitter dopamine is Farde, L. (2006). The correlative triad among
implicated in a variety of functions, ranging from aging, dopamine, and cognition: Current status
motor control to higher-order cognition, as well and future prospects. Neuroscience & Biobehavio-
as motivational and affective aspects of behavior. ral Reviews, 30(6), 791–807.
In this chapter, we have reviewed evidence for Bäckman, L., Nyberg, L., Soveri, A., Johansson, J.,
dopaminergic contributions to cognitive function Andersson, M., Dahlin, E., … Rinne, J. O. (2011b).
across the adult life span, focusing on the domains Effects of working-memory training on striatal
of cognitive control, learning and memory, and dopamine release. Science, 333, 718–718.
value-based decision making. We have discussed Bayer, H. M., & Glimcher, P. W. (2005). Midbrain
the general principles of dopaminergic modula- dopamine neurons encode a quantitative reward
tion of cognition based on integrated findings prediction error signal. Neuron, 47(1), 129–141.
from human pharmacological, neuroimaging, and Bayer, H. M., Lau, B., & Glimcher, P. W. (2007). Sta-
genetic studies, as well as experimental animal tistics of midbrain dopamine neuron spike trains in
work. In addition, we have reviewed evidence for the awake primate. Journal of Neurophysiology,
age-related decreases in DA system integrity and 98(3), 1428–1439.
the impact of such decreases on cognitive deficits Beaulieu, J.-M., & Gainetdinov, R. R. (2011). The
in aging. Taken together, although longitudinal physiology, signaling, and pharmacology of dopa-
evidence is currently limited, the findings to date mine receptors. Pharmacological Reviews, 63(1),
support a dopaminergic account of age-related 182–217.
cognitive deficits. Beierholm, U., Guitart-Masip, M., Economides, M.,
Chowdhury, R., Düzel, E., Dolan, R., & Dayan, P.
(2013). Dopamine modulates reward-related vigor.
Neuropsychopharmacology, 38(8), 1495–1503.
Berry, A. S., Jagust, W. J., & Hsu, M. (2019). Age-
REFERENCES related variability in decision-making: Insights from
neurochemistry. Cognitive, Affective, & Behavioral
Abdulrahman, H., Fletcher, P. C., Bullmore, E., & Neuroscience, 19(3), 415–434.
Morcom, A. M. (2017). Dopamine and memory Berry, A. S., Shah, V. D., Baker, S. L., Vogel, J. W.,
dedifferentiation in aging. NeuroImage, 153, O’Neil, J. P., Janabi, M., Schwimmer, H. D., Marks,
211–220. S. M., & Jagust, W. J. (2016). Aging affects dopa-
Adcock, R. A., Thangavel, A., Whitfield-Gabrieli, S., minergic neural mechanisms of cognitive flexibility.
Knutson, B., & Gabrieli, J. D. E. (2006). Journal of Neuroscience, 36(50), 12559–12569.
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 125

Berry, A. S., Shah, V. D., Furman, D. J., White III, R. indicate functional compartmentalization of the
L., Baker, S. L., O’Neil, J. P., … Jagust, W. J. human striatum. NeuroImage, 40(3), 1287–1295.
(2018a). Dopamine synthesis capacity is associated Chen, J., Lipska, B. K., Halim, N., Ma, Q. D., Matsu-
with D2/3 receptor binding but not dopamine moto, M., Melhem, S., … & Weinberger, D. R.
release. Neuropsychopharmacology, 43(6), (2004). Functional analysis of genetic variation in
1201–1211. catechol-O-methyltransferase (COMT): effects on
Berry, A. S., Shah, V. D., & Jagust, W. J. (2018b). The mRNA, protein, and enzyme activity in postmor-
influence of dopamine on cognitive flexibility is tem human brain. American Journal of Human
mediated by functional connectivity in young but Genetics, 75(5), 807–821.
not older adults. Journal of Cognitive Neurosci- Chowdhury, R., Guitart-Masip, M., Bunzeck, N.,
ence, 30(9), 1330–1344. Dolan, R. J., & Düzel, E. (2012). Dopamine modu-
Bethus, I., Tse, D., & Morris, R. G. M. (2010). Dopa- lates episodic memory persistence in old age.
mine and memory: Modulation of the persistence Journal of Neuroscience, 32(41), 14193–14204.
of memory for novel hippocampal NMDA recep- Chowdhury, R., Guitart-Masip, M., Lambert, C.,
tor-dependent paired associates. Journal of Neuro- Dayan, P., Huys, Q., Düzel, E., & Dolan, R. J. (2013).
science, 30(5), 1610–1618. Dopamine restores reward prediction errors in old
Betts, M. J., Richter, A., de Boer, L., Tegelbeckers, J., age. Nature Neuroscience, 16(5), 648–653.
Perosa, V., Baumann, V., … Krauel, K. (2020). Colzato, L. S., van den Wildenberg, W. P. M., &
Learning in anticipation of reward and punish- Hommel, B. (2013). The genetic impact (C957T-
ment: perspectives across the human lifespan. DRD2) on inhibitory control is magnified by aging.
Neurobiology of Aging, 96, 49–57. Neuropsychologia, 51(7), 1377–1381.
Björklund, A., & Dunnett, S. B. (2007). Dopamine Constantinidis, C., Funahashi, S., Lee, D., Murray, J.
neuron systems in the brain: An update. Trends in D., Qi, X.-L., Wang, M., & Arnsten, A. F. T. (2018).
Neurosciences, 30(5), 194–202. Persistent spiking activity underlies working
Bliss, T. V. P., & Collingridge, G. L. (1993). A synaptic memory. Journal of Neuroscience, 38(32),
model of memory: Long-term potentiation in the 7020–7028.
hippocampus. Nature, 361, 31–39. Cools, R. (2019). Chemistry of the adaptive mind:
Borgkvist, A., Malmlöf, T., Feltmann, K., Lindskog, M., lessons from dopamine. Neuron, 104(1), 113–131.
& Schilström, B. (2012). Dopamine in the hippocam- Cools, R., & D’Esposito, M. (2011). Inverted-u–shaped
pus is cleared by the norepinephrine transporter. dopamine actions on human working memory and
International Journal of Neuropsychopharmacology, cognitive control. Biological Psychiatry, 69(12),
15(4), 531–540. e113–e125. doi: 10.1016/j.biopsych.2011.03.028
Braskie, M. N., Landau, S. M., Wilcox, C. E., Taylor, S. Cousins, M. S., & Salamone, J. D. (1994). Nucleus
D., O’Neil, J. P., Baker, S. L., Madison, C. M., & accumbens dopamine depletions in rats affect
Jagust, W. J. (2011). Correlations of striatal dopa- relative response allocation in a novel cost/benefit
mine synthesis with default network deactivations procedure. Pharmacology Biochemistry and
during working memory in younger adults. Human Behavior, 49(1), 85–91.
Brain Mapping, 32(6), 947–961. Cox, S. M. L., Frank, M. J., Larcher, K., Fellows, L. K.,
Brozoski, T. J., Brown, R. M., Rosvold, H. E., & Gold- Clark, C. A., Leyton, M., & Dagher, A. (2015).
man, P. S. (1979). Cognitive deficit caused by Striatal D1 and D2 signaling differentially predict
regional depletion of dopamine in prefrontal learning from positive and negative outcomes.
cortex of rhesus monkey. Science, 205, 929–932. NeuroImage, 109, 95–101.
Calabresi, P., Picconi, B., Tozzi, A., & di Filippo, M. Cropley, V. L., Fujita, M., Innis, R. B., & Nathan, P. J.
(2007). Dopamine-mediated regulation of corti- (2006). Molecular imaging of the dopaminergic
costriatal synaptic plasticity. Trends in Neuro- system and its association with human cognitive
sciences, 30(5), 211–219. function. Biological Psychiatry, 59(10), 898–907.
Calderon, C. B., de Loof, E., Ergo, K., Snoeck, A., Cutler, J., Wittmann, M. K., Abdurahman, A., Hargi-
Boehler, C. N., & Verguts, T. (2021). Signed reward tai, L. D., Drew, D., Husain, M., & Lockwood, P. L.
prediction errors in the ventral striatum drive epi- (2021). Ageing is associated with disrupted rein-
sodic memory. Journal of Neuroscience, 41(8), forcement learning whilst learning to help others
1716–1726. is preserved. Nature Communications, 12(1). doi:
Cenci, M. A. (2014). Presynaptic mechanisms of 10.1038/s41467-021-24576-w
l-DOPA-induced dyskinesia: The findings, the debate, D’Ardenne, K., McClure, S. M., Nystrom, L. E., &
and the therapeutic implications. Frontiers in Cohen, J. D. (2008). BOLD responses reflecting
Neurology, 5. doi: 10.3389/fneur.2014.00242 dopaminergic signals in the human ventral teg-
Cervenka, S., Bäckman, L., Cselényi, Z., Halldin, C., & mental area. Science, 319, 1264–1267.
Farde, L. (2008). Associations between dopamine de Boer, L., Axelsson, J., Chowdhury, R., Riklund, K.,
D2-receptor binding and cognitive performance Dolan, R. J., Nyberg, L., Bäckman, L., &
126 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Guitart-Masip, M. (2019). Dorsal striatal dopamine Everitt, B. J., & Robbins, T. W. (1997). Central cholin-
D1 receptor availability predicts an instrumental ergic systems and cognition. Annual Review of
bias in action learning. Proceedings of the National Psychology, 48(1), 649–684.
Academy of Sciences, 116(1), 261–270. Fearnley, J. M., & Lees, A. J. (1991). Ageing and
de Boer, L., Axelsson, J., Riklund, K., Nyberg, L., Parkinson’s disease: Substantia nigra regional
Dayan, P., Bäckman, L., & Guitart-Masip, M. selectivity. Brain, 114(5), 2283–2301.
(2017). Attenuation of dopamine-modulated pre- Fischer, H., Nyberg, L., Karlsson, S., Karlsson, P., Brehmer,
frontal value signals underlies probabilistic reward Y., Rieckmann, A., … & Bäckman, L. (2010). Simulat-
learning deficits in old age. ELife, 6. doi: 10.7554/ ing neurocognitive aging: Effects of a dopaminergic
eLife.26424 antagonist on brain activity during working memory.
de Boer, L., Garzón, B., Axelsson, J., Riklund, K., Biological Psychiatry, 67(6), 575–580.
Nyberg, L., Bäckman, L., & Guitart-Masip, M. Floresco, S. B. (2013). Prefrontal dopamine and
(2020). Corticostriatal white matter integrity and behavioral flexibility: Shifting from an “inverted-
dopamine D1 receptor availability predict age dif- U” toward a family of functions. Frontiers in Neu-
ferences in prefrontal value signaling during reward roscience, 7. doi: 10.3389/fnins.2013.00062
learning. Cerebral Cortex, 30(10), 5270–5280. Floresco, S. B., West, A. R., Ash, B., Moore, H., &
de Frias, C. M., Annerbrink, K., Westberg, L., Eriks- Grace, A. A. (2003). Afferent modulation of dopa-
son, E., Adolfsson, R., & Nilsson, L.-G. (2005). mine neuron firing differentially regulates tonic
Catechol O-methyltransferase Val158Met polymor- and phasic dopamine transmission. Nature Neuro-
phism is associated with cognitive performance in science, 6(9), 968–973.
nondemented adults. Journal of Cognitive Neuro- Ford, C. P. (2014). The role of D2-autoreceptors in
science, 17(7), 1018–1025. regulating dopamine neuron activity and transmis-
de Keyser, J., de Backer, J.-P., Vauquelin, G., & Ebin- sion. Neuroscience, 282, 13–22.
ger, G. (1990). The effect of aging on the D1 Frank, D., & Kafkas, A. (2021). Expectation-driven
dopamine receptors in human frontal cortex. Brain novelty effects in episodic memory. Neurobiology
Research, 528(2), 308–310. of Learning and Memory, 183. doi: 10.1016/j.
Diederen, K. M. J., & Fletcher, P. C. (2021). Dopa- nlm.2021.107466
mine, prediction error and beyond. Neuroscientist, Frank, M. J., Seeberger, L. C., & O’Reilly, R. C. (2004).
27(1), 30–46. By carrot or by stick: Cognitive reinforcement learn-
Durstewitz, D., & Seamans, J. K. (2008). The dual- ing in parkinsonism. Science, 306, 1940–1943.
state theory of prefrontal cortex dopamine func- Froudist-Walsh, S., Bliss, D. P., Ding, X., Rapan, L.,
tion with relevance to catechol-O-methyltransferase Niu, M., Knoblauch, K., … Wang, X.-J. (2021). A
genotypes and schizophrenia. Biological Psychia- dopamine gradient controls access to distributed
try, 64(9), 739–749. working memory in the large-scale monkey cortex.
Duszkiewicz, A. J., McNamara, C. G., Takeuchi, T., & Neuron, 109(21), 3500–3520.
Genzel, L. (2019). Novelty and dopaminergic mod- Gardner, M. P. H., Schoenbaum, G., & Gershman, S. J.
ulation of memory persistence: A tale of two sys- (2018). Rethinking dopamine as generalized predic-
tems. Trends in Neurosciences, 42(2), 102–114. tion error. Proceedings of the Royal Society B: Bio-
Eichenbaum, H. (2000). A cortical–hippocampal logical Sciences, 285. doi: 10.1098/rspb.2018.1645
system for declarative memory. Nature Reviews Garrison, J., Erdeniz, B., & Done, J. (2013). Prediction
Neuroscience, 1(1), 41–50. error in reinforcement learning: A meta-analysis of
Elsworth, J. D., & Roth, R. H. (1997). Dopamine syn- neuroimaging studies. Neuroscience & Biobehav-
thesis, uptake, metabolism, and receptors: Rele- ioral Reviews, 37(7), 1297–1310.
vance to gene therapy of Parkinson’s disease. Garzón, B., Lövdén, M., de Boer, L., Axelsson, J.,
Experimental Neurology, 144(1), 4–9. Riklund, K., Bäckman, L., Nyberg, L., & Guitart-
Eppinger, B., Schuck, N. W., Nystrom, L. E., & Cohen, J. Masip, M. (2021). Role of dopamine and gray
D. (2013). Reduced striatal responses to reward pre- matter density in aging effects and individual dif-
diction errors in older compared with younger adults. ferences of functional connectomes. Brain Struc-
Journal of Neuroscience, 33(24), 9905–9912. ture and Function, 226(3), 743–758.
Eriksson, J., Vogel, E. K., Lansner, A., Bergström, F., & Gasbarri, A., Verney, C., Innocenzi, R., Campana, E., &
Nyberg, L. (2015). Neurocognitive architecture of Pacitti, C. (1994). Mesolimbic dopaminergic neu-
working memory. Neuron, 88(1), 33–46. rons innervating the hippocampal formation in the
Erixon-Lindroth, N., Farde, L., Robins Wahlin, T.-B., rat: A combined retrograde tracing and immunohis-
Sovago, J., Halldin, C., & Bäckman, L. (2005). The tochemical study. Brain Research, 668(1-2), 71–79.
role of the striatal dopamine transporter in cogni- Geddes, M. R., Mattfeld, A. T., Angeles, C. de los,
tive aging. Psychiatry Research: Neuroimaging, Keshavan, A., & Gabrieli, J. D. E. (2018). Human
138(1), 1–12. aging reduces the neurobehavioral influence of
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 127

motivation on episodic memory. NeuroImage, Ishibashi, K., Ishii, K., Oda, K., Kawasaki, K., Mizu-
171, 296–310. sawa, H., & Ishiwata, K. (2009). Regional analysis
Geerligs, L., Renken, R. J., Saliasi, E., Maurits, N. M., of age-related decline in dopamine transporters
& Lorist, M. M. (2015). A brain-wide study of age- and dopamine D2-like receptors in human stria-
related changes in functional connectivity. Cere- tum. Synapse, 63(4), 282–290.
bral Cortex, 25(7), 1987–1999. Juarez, E. J., Castrellon, J. J., Green, M. A., Crawford,
Goto, Y., Otani, S., & Grace, A. (2007). The Yin and J. L., Seaman, K. L., Smith, C. T., … Samanez-Larkin,
Yang of dopamine release: A new perspective. G. R. (2019). Reproducibility of the correlative triad
Neuropharmacology, 53(5), 583–587. among aging, dopamine receptor availability, and
Granado, N., Ortiz, O., Suarez, L. M., Martin, E. D., cognition. Psychology and Aging, 34(7), 921–932.
Cena, V., Solis, J. M., & Moratalla, R. (2008). D1 but Kaasinen, V., Vilkman, H., Hietala, J., Någren, K.,
not D5 dopamine receptors are critical for LTP, spa- Helenius, H., Olsson, H., Farde, L., & Rinne, J. O.
tial learning, and LTP-induced arc and zif268 expres- (2000). Age-related dopamine D2/D3 receptor loss
sion in the hippocampus. Cerebral Cortex, 18(1). in extrastriatal regions of the human brain. Neuro-
doi: 10.1093/cercor/bhm026 biology of Aging, 21(5), 683–688.
Greengard, P. (2001). The neurobiology of slow syn- Kambeitz, J. P., Bhattacharyya, S., Kambeitz-Ilanko-
aptic transmission. Science, 294, 1024–1030. vic, L. M., Valli, I., Collier, D. A., & McGuire, P.
Gruber, M. J., Ritchey, M., Wang, S.-F., Doss, M. K., (2012). Effect of BDNF val66met polymorphism on
& Ranganath, C. (2016). Post-learning hippocam- declarative memory and its neural substrate: A
pal dynamics promote preferential retention of meta-analysis. Neuroscience & Biobehavioral
rewarding events. Neuron, 89(5), 1110–1120. Reviews, 36(9), 2165–2177.
Guitart-Masip, M., Beierholm, U. R., Dolan, R., Duzel, Karalija, N., Johansson, J., Papenberg, G., Wåhlin, A.,
E., & Dayan, P. (2011). Vigor in the face of fluctu- Salami, A., Köhncke, Y., … Nyberg, L. (2022).
ating rates of reward: An experimental examina- Longitudinal dopamine D2 receptor changes and
tion. Journal of Cognitive Neuroscience, 23(12), cerebrovascular health in aging. Neurology,
3933–3938. 99(12), e1278–e1289.
Guitart-Masip, M., Huys, Q. J. M., Fuentemilla, L., Karalija, N., Köhncke, Y., Düzel, S., Bertram, L., Pap-
Dayan, P., Duzel, E., & Dolan, R. J. (2012). Go and enberg, G., Demuth, I., … Brandmaier, A. M.
no-go learning in reward and punishment: Interac- (2021a). A common polymorphism in the dopa-
tions between affect and effect. NeuroImage, mine transporter gene predicts working memory
62(1), 154–166. performance and in vivo dopamine integrity in aging.
Haber, S. N., Fudge, J. L., & McFarland, N. R. (2000). NeuroImage, 245. doi: 10.1016/j.neuroimage.
Striatonigrostriatal pathways in primates form an 2021.118707
ascending spiral from the shell to the dorsolat- Karalija, N., Papenberg, G., Wåhlin, A., Johansson, J.,
eral striatum. Journal of Neuroscience, 20(6), Andersson, M., Axelsson, J., … Bäckman, L.
2369–2382. (2021b). Sex differences in dopamine integrity and
Haber, S. N., & Knutson, B. (2010). The reward cir- brain structure among healthy older adults: Rela-
cuit: Linking primate anatomy and human imag- tionships to episodic memory. Neurobiology of
ing. Neuropsychopharmacology, 35(1), 4–26. Aging, 105, 272–279.
Hamid, A. A., Pettibone, J. R., Mabrouk, O. S., Karalija, N., Papenberg, G., Wåhlin, A., Johansson, J.,
Hetrick, V. L., Schmidt, R., Vander Weele, C. M., … Andersson, M., Axelsson, J., … Nyberg, L. (2019a).
& Berke, J. D. (2016). Mesolimbic dopamine sig- C957T-mediated variation in ligand affinity affects
nals the value of work. Nature Neuroscience, the association between 11 C-raclopride binding
19(1), 117–126. potential and cognition. Journal of Cognitive Neu-
Hirvonen, M. M., Laakso, A., Någren, K., Rinne, J. O., roscience, 31(2), 314–325.
Pohjalainen, T., & Hietala, J. (2009). C957T poly- Karalija, N., Wåhlin, A., Ek, J., Rieckmann, A., Papen-
morphism of dopamine D2 receptor gene affects berg, G., Salami, A., … Nyberg, L. (2019b). Car-
striatal DRD2 in vivo availability by changing the diovascular factors are related to dopamine
receptor affinity. Synapse, 63(10), 907–912. integrity and cognition in aging. Annals of Clinical
Hofmans, L., Westbrook, A., van den Bosch, R., and Translational Neurology, 6(11), 2291–2303.
Booij, J., Verkes, R.-J., & Cools, R. (2022). Effects Karrer, T. M., Josef, A. K., Mata, R., Morris, E. D., &
of average reward rate on vigor as a function of Samanez-Larkin, G. R. (2017). Reduced dopamine
individual variation in striatal dopamine. Psychop- receptors and transporters but not synthesis
harmacology, 239(2), 465–478. capacity in normal aging adults: A meta-analysis.
Holland, N., Robbins, T. W., & Rowe, J. B. (2021). The Neurobiology of Aging, 57, 36–46.
role of noradrenaline in cognition and cognitive Kim, Y. J., Ichise, M., Ballinger, J. R., Vines, D., Erami,
disorders. Brain, 144(8), 2243–2256. S. S., Tatschida, T., & Lang, A. E. (2002).
128 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Combination of dopamine transporter and D2 Li, S.-C., Papenberg, G., Nagel, I. E., Preuschhof, C.,
receptor SPECT in the diagnostic evaluation of PD, Schröder, J., Nietfeld, W., … Bäckman, L. (2013).
MSA, and PSP. Movement Disorders, 17(2), Aging magnifies the effects of dopamine
303–312. transporter and D2 receptor genes on backward
Kimberg, D. Y., D’Esposito, M., & Farah, M. J. (1997). serial memory. Neurobiology of Aging, 34(1).
Effects of bromocriptine on human subjects doi: 10.1016/j.neurobiolaging.2012.08.001
depend on working memory capacity. NeuroRe- Li, X., Bäckman, L., & Persson, J. (2019). The relation-
port, 8(16), 3581–3585. ship of age and DRD2 polymorphisms to frontostri-
Klein, M. O., Battagello, D. S., Cardoso, A. R., atal brain activity and working memory performance.
Hauser, D. N., Bittencourt, J. C., & Correa, R. G. Neurobiology of Aging, 84, 189–199.
(2019). Dopamine: Functions, signaling, and asso- Lidow, M. S., Goldman-Rakic, P. S., Gallager, D. W., &
ciation with neurological diseases. Cellular and Rakic, P. (1991). Distribution of dopaminergic
Molecular Neurobiology, 39(1), 31–59. receptors in the primate cerebral cortex: Quantita-
Klostermann, E. C., Braskie, M. N., Landau, S. M., tive autoradiographic analysis using [3H]raclopride,
O’Neil, J. P., & Jagust, W. J. (2012). Dopamine and [3H]spiperone and [3H]SCH23390. Neuro­science,
frontostriatal networks in cognitive aging. Neuro- 40(3), 657–671.
biology of Aging, 33(3). doi: 10.1016/j.neurobio- Lighthall, N. R. (2020). Neural mechanisms of deci-
laging.2011.03.002 sion-making in aging. WIREs Cognitive Science,
Knowlton, B. J., & Castel, A. D. (2022). Memory and 11(1). doi: 10.1002/wcs.1519
reward-based learning: A value-directed remem- Lisman, J. E., & Grace, A. A. (2005). The hippocam-
bering perspective. Annual Review of Psychology, pal-VTA loop: Controlling the entry of information
73, 25–52. into long-term memory. Neuron, 46(5), 703–713.
Knutson, B., & Gibbs, S. E. B. (2007). Linking nucleus Lisman, J., Grace, A. A., & Duzel, E. (2011). A neo-
accumbens dopamine and blood oxygenation. Hebbian framework for episodic memory: Role of
Psychopharmacology, 191(3), 813–822. dopamine-dependent late LTP. Trends in Neuro-
Köhncke, Y., Papenberg, G., Jonasson, L., Karalija, sciences, 34(10), 536–547.
N., Wåhlin, A., Salami, A., … Lövdén, M. (2018). Lodge, D. J., & Grace, A. A. (2006). The hippocampus
Self-rated intensity of habitual physical activities is modulates dopamine neuron responsivity by regu-
positively associated with dopamine D2/3 receptor lating the intensity of phasic neuron activation.
availability and cognition. NeuroImage, 181, Neuropsychopharmacology, 31(7), 1356–1361.
605–616. Lotta, T., Vidgren, J., Tilgmann, C., Ulmanen, I.,
Korkki, S. M., Papenberg, G., Karalija, N., Garrett, D. Melen, K., Julkunen, I., & Taskinen, J. (1995).
D., Riklund, K., Lövdén, M., Lindenberger, U., Kinetics of human soluble and membrane-bound
Nyberg, L., & Bäckman, L. (2021). Fronto-striatal catechol O-methyltransferase: A revised mecha-
dopamine D2 receptor availability is associated nism and description of the thermolabile variant of
with cognitive variability in older individuals with the enzyme. Biochemistry, 34(13), 4202–4210.
low dopamine integrity. Scientific Reports, 11(1). Lövdén, M., Karalija, N., Andersson, M., Wåhlin, A.,
doi: 10.1038/s41598-021-00106-y Axelsson, J., Köhncke, Y., … Lindenberger, U.
Landau, S. M., Lal, R., O’Neil, J. P., Baker, S., & Jagust, (2018). Latent-profile analysis reveals behavioral
W. J. (2009). Striatal dopamine and working and brain correlates of dopamine-cognition asso-
memory. Cerebral Cortex, 19(2), 445–454. ciations. Cerebral Cortex, 28(11), 3894–3907.
Laruelle, M. (2000). Imaging synaptic neurotransmis- MacDonald, S. W. S., Karlsson, S., Rieckmann, A.,
sion with in vivo binding competition techniques: Nyberg, L., & Backman, L. (2012). Aging-related
A critical review. Journal of Cerebral Blood Flow & increases in behavioral variability: Relations to
Metabolism, 20(3), 423–451. losses of dopamine D1 receptors. Journal of Neu-
Laruelle, M., Abi-Dargham, A., van Dyck, C., Gil, R., roscience, 32(24), 8186–8191.
D’Souza, D. C., Krystal, J., … & Innis, R. (2000). Malén, T., Karjalainen, T., Isojärvi, J., Vehtari, A.,
Dopamine and serotonin transporters in patients Bürkner, P.-C., Putkinen, V., … Nummenmaa, L.
with schizophrenia: An imaging study with [(123) (2022). Atlas of type 2 dopamine receptors in the
I]beta-CIT. Biological Psychiatry, 47(5), 371–379. human brain: Age and sex dependent variability in
Li, S., Cullen, W. K., Anwyl, R., & Rowan, M. J. (2003). a large PET cohort. NeuroImage, 255. doi: 10.1016/
Dopamine-dependent facilitation of LTP induction j.neuroimage.2022.119149
in hippocampal CA1 by exposure to spatial novelty. Marek, K., Jennings, D., Lasch, S., Siderowf, A.,
Nature Neuroscience, 6(5), 526–531. Tanner, C., Simuni, T., … Taylor, P. (2011). The
Li, S.-C., Lindenberger, U., & Sikström, S. (2001). Parkinson Progression Marker Initiative (PPMI).
Aging cognition: From neuromodulation to repre- Progress in Neurobiology, 95(4), 629–635.
sentation. Trends in Cognitive Sciences, 5(11), Matsumoto, M., Weickert, C. S., Akil, M., Lipska, B.
479–486. K., Hyde, T. M., Herman, M. M., Kleinman, J. E., &
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 129

Weinberger, D. R. (2003). Catechol O-methyltrans- Nagel, I. E., Chicherio, C., Li, S.-C., von Oertzen, T.,
ferase mRNA expression in human and rat brain: Sander, T., Villringer, A., Heekeren, H. R., Bäck-
Evidence for a role in cortical neuronal function. man, L., & Lindenberger, U. (2008). Human aging
Neuroscience, 116(1), 127–137. magnifies genetic effects on executive functioning
Matthies, H., Becker, A., Schröeder, H., Kraus, J., and working memory. Frontiers in Human Neuro-
Höllt, V., & Krug, M. (1997). Dopamine D1- science, 2. doi: 10.3389/neuro.09.001.2008
deficient mutant mice do not express the late Nevalainen, N., Riklund, K., Andersson, M., Axelsson,
phase of hippocampal long-term potentiation. J., Ögren, M., Lövdén, M., Lindenberger, U., Bäck-
NeuroReport, 8(16), 3533–3535. man, L., & Nyberg, L. (2015). COBRA: A prospec-
McNamara, C. G., Tejero-Cantero, Á., Trouche, S., tive multimodal imaging study of dopamine, brain
Campo-Urriza, N., & Dupret, D. (2014). Dopamin- structure and function, and cognition. Brain
ergic neurons promote hippocampal reactivation Research, 1612, 83–103.
and spatial memory persistence. Nature Neurosci- Niv, Y., Daw, N. D., & Dayan, P. (2005). How fast to
ence, 17(12), 1658–1660. work: Response vigor, motivation and tonic dopa-
Mehta, M. A., Manes, F. F., Magnolfi, G., Sahakian, mine. Advances in Neural Information Processing
B. J., & Robbins, T. W. (2004). Impaired set-shifting Systems, 18, 1019–1026.
and dissociable effects on tests of spatial working Niv, Y., Daw, N. D., Joel, D., & Dayan, P. (2007). Tonic
memory following the dopamine D2 receptor dopamine: Opportunity costs and the control of
antagonist sulpiride in human volunteers. Psy- response vigor. Psychopharmacology, 191(3),
chopharmacology, 176(3-4), 331–342. 507–520.
Mehta, M. A., Sahakian, B. J., McKenna, P. J., & Rob- Nobis, L., Manohar, S. G., Smith, S. M., Alfaro-
bins, T. W. (1999). Systemic sulpiride in young Almagro, F., Jenkinson, M., Mackay, C. E., &
adult volunteers simulates the profile of cognitive Husain, M. (2019). Hippocampal volume across
deficits in Parkinson’s disease. Psychopharmacol- age: Nomograms derived from over 19,700
ogy, 146(2), 162–174. people in UK Biobank. NeuroImage: Clinical, 23.
Mell, T., Heekeren, H. R., Marschner, A., Warten- doi: 10.1016/j.nicl.2019.101904
burger, I., Villringer, A., & Reischies, F. M. (2005). Nordin, K., Gorbach, T., Pedersen, R., Panes Lund-
Effect of aging on stimulus-reward association mark, V., Johansson, J., Andersson, M., … Salami,
learning. Neuropsychologia, 43(4), 554–563. A. (2022). DyNAMiC: A prospective longitudinal
Miendlarzewska, E. A., Bavelier, D., & Schwartz, S. study of dopamine and brain connectomes: A new
(2016). Influence of reward motivation on human window into cognitive aging. Journal of Neurosci-
declarative memory. Neuroscience & Biobehavioral ence Research, 100(6), 1296–1320.
Reviews, 61, 156–176. Nordin, K., Nyberg, L., Andersson, M., Karalija, N.,
Miller, E. K., & Cohen, J. D. (2001). An integrative Riklund, K., Bäckman, L., & Salami, A. (2021).
theory of prefrontal cortex function. Annual Distinct and common large-scale networks of the
Review of Neuroscience, 24(1), 167–202. hippocampal long axis in older age: Links to epi-
Milnik, A., Heck, A., Vogler, C., Heinze, H.-J., de sodic memory and dopamine D2 receptor availa-
Quervain, D. J.-F., & Papassotiropoulos, A. (2012). bility. Cerebral Cortex, 31(7), 3435–3450.
Association of KIBRA with episodic and working Noudoost, B., & Moore, T. (2011). Control of visual
memory: A meta-analysis. American Journal of cortical signals by prefrontal dopamine. Nature,
Medical Genetics Part B: Neuropsychiatric Genet- 474, 372–375.
ics, 159B(8), 958–969. Nour, M. M., Dahoun, T., McCutcheon, R. A., Adams,
Morcom, A. M., Bullmore, E. T., Huppert, F. A., Lennox, R. A., Wall, M. B., & Howes, O. D. (2019). Task-
B., Praseedom, A., Linnington, H., & Fletcher, P. C. induced functional brain connectivity mediates the
(2010). Memory encoding and dopamine in the relationship between striatal D2/3 receptors and
aging brain: A psychopharmacological neuroimag- working memory. ELife, 8. doi: 10.7554/eLife.
ing study. Cerebral Cortex, 20(3), 743–757. 45045
Moreno-Castilla, P., Pérez-Ortega, R., Violante-Soria, Nyberg, L., Andersson, M., Forsgren, L., Jakobsson-
V., Balderas, I., & Bermúdez-Rattoni, F. (2017). Hip- Mo, S., Larsson, A., Marklund, P., … Bäckman, L.
pocampal release of dopamine and norepineph- (2009a). Striatal dopamine D2 binding is related to
rine encodes novel contextual information. frontal BOLD response during updating of long-
Hippocampus, 27(5), 547–557. term memory representations. NeuroImage, 46(4),
Morón, J. A., Brockington, A., Wise, R. A., Rocha, B. 1194–1199.
A., & Hope, B. T. (2002). Dopamine uptake Nyberg, L., Andersson, M., Kauppi, K., Lundquist, A.,
through the norepinephrine transporter in brain Persson, J., Pudas, S., & Nilsson, L.-G. (2014). Age-
regions with low levels of the dopamine trans- related and genetic modulation of frontal cortex
porter: Evidence from knock-out mouse lines. efficiency. Journal of Cognitive Neuroscience,
Journal of Neuroscience, 22(2), 389–395. 26(4), 746–754.
130 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Nyberg, L., Dahlin, E., Stigsdotter Neely, A., & Bäck- polymorphisms. Journal of Cognitive Neurosci-
man, L. (2009b). Neural correlates of variable ence, 31(9), 1422–1429.
working memory load across adult age and skill: Papenberg, G., Karalija, N., Salami, A., Rieckmann,
Dissociative patterns within the fronto-parietal A., Andersson, M., Axelsson, J., … Bäckman, L.
network. Scandinavian Journal of Psychology, (2020). Balance between transmitter availability
50(1), 41–46. and dopamine D2 receptors in prefrontal cortex
Nyberg, L., & Eriksson, J. (2016). Working memory: influences memory functioning. Cerebral Cortex,
Maintenance, updating, and the realization of 30(3), 989–1000.
intentions. Cold Spring Harbor Perspectives in Biol- Papenberg, G., Lindenberger, U., & Bäckman, L.
ogy, 8(2). doi: 10.1101/cshperspect.a021816 (2015). Aging-related magnification of genetic
Nyberg, L., Karalija, N., Salami, A., Andersson, M., effects on cognitive and brain integrity. Trends in
Wåhlin, A., Kaboovand, N., … Bäckman, L. (2016). Cognitive Sciences, 19(9), 506–514.
Dopamine D2 receptor availability is linked to hip- Papenberg, G., Salami, A., Persson, J., Lindenberger,
pocampal–caudate functional connectivity and U., & Bäckman, L. (2015). Genetics and functional
episodic memory. Proceedings of the National imaging: Effects of APOE, BDNF, COMT, and KIBRA
Academy of Sciences, 113(28), 7918–7923. in aging. Neuropsychology Review, 25(1), 47–62.
Nyberg, L., Salami, A., Andersson, M., Eriksson, J., Pappata, S., Dehaene, S., Poline, J. B., Gregoire, M.
Kalpouzos, G., Kauppi, K., … Nilsson, L.-G. (2010). C., Jobert, A., Delforge, J., … Syrota, A. (2002). In
Longitudinal evidence for diminished frontal cortex vivo detection of striatal dopamine release during
function in aging. Proceedings of the National reward: A PET study with [11C]raclopride and a
Academy of Sciences, 107(52), 22682–22686. single dynamic scan approach. NeuroImage, 16(4),
O’Carroll, C. M., Martin, S. J., Sandin, J., Frenguelli, 1015–1027.
B., & Morris, R. G. M. (2006). Dopaminergic Parent, A., & Hazrati, L.-N. (1995). Functional anat-
modulation of the persistence of one-trial hip- omy of the basal ganglia. I. The cortico-basal
pocampus-dependent memory. Learning & ganglia-thalamo-cortical loop. Brain Research
Memory, 13(6), 760–769. Reviews, 20(1), 91–127.
O’Carroll, C. M., & Morris, R. G. M. (2004). Heter- Persson, J., Rieckmann, A., Kalpouzos, G., Fischer, H.,
osynaptic co-activation of glutamatergic and & Bäckman, L. (2015). Influences of a DRD2 poly-
dopaminergic afferents is required to induce per- morphism on updating of long-term memory
sistent long-term potentiation. Neuropharmacol- ­representations and caudate BOLD activity: Magni-
ogy, 47(3), 324–332. fication in aging. Human Brain Mapping, 36(4),
O’Reilly, R. C. (2006). Biologically based computa- 1325–1334.
tional models of high-level cognition. Science, Pessiglione, M., Seymour, B., Flandin, G., Dolan, R. J.,
314, 91-94. & Frith, C. D. (2006). Dopamine-dependent pre-
Ott, T., & Nieder, A. (2019). Dopamine and cognitive diction errors underpin reward-seeking behaviour
control in prefrontal cortex. Trends in Cognitive in humans. Nature, 442, 1042–1045.
Sciences, 23(3), 213–234. Rabbitt, P. (1993). Does it all go together when it
Papenberg, G., Bäckman, L., Nagel, I. E., Nietfeld, W., goes? The nineteenth Bartlett memorial lecture.
Schröder, J., Bertram, L., … & Li, S.-C. (2013). Quarterly Journal of Experimental Psychology Sec-
Dopaminergic gene polymorphisms affect long- tion A, 46(3), 385–434.
term forgetting in old age: Further support for the Ranganath, A., & Jacob, S. N. (2016). Doping the
magnification hypothesis. Journal of Cognitive mind: Dopaminergic modulation of prefrontal cor-
Neuroscience, 25(4), 571–579. tical cognition. Neuroscientist, 22(6), 593–603.
Papenberg, G., Becker, N., Ferencz, B., Naveh-Benja- Raz, N., Lindenberger, U., Rodrigue, K. M., Kennedy,
min, M., Laukka, E. J., Bäckman, L., & Brehmer, Y. K. M., Head, D., Williamson, A., Dahle, C., Ger-
(2017). Dopamine receptor genes modulate asso- storf, D., & Acker, J. D. (2005). Regional brain
ciative memory in old age. Journal of Cognitive changes in aging healthy adults: General trends,
Neuroscience, 29(2), 245–253. individual differences and modifiers. Cerebral
Papenberg, G., Jonasson, L., Karalija, N., Johansson, Cortex, 15(11), 1676–1689.
J., Köhncke, Y., Salami, A., … Bäckman, L. Reeves, S., Bench, C., & Howard, R. (2002). Ageing
(2019a). Mapping the landscape of human dopa- and the nigrostriatal dopaminergic system. Inter-
mine D2/3 receptors with [11C]raclopride. Brain national Journal of Geriatric Psychiatry, 17(4),
Structure and Function, 224(8), 2871–2882. 359–370.
Papenberg, G., Karalija, N., Salami, A., Andersson, Rieck, J. R., Rodrigue, K. M., Boylan, M. A., & Ken-
M., Axelsson, J., Riklund, K., Lindenberger, U., nedy, K. M. (2017). Age-related reduction of
Nyberg, L., & Bäckman, L. (2019b). The influence BOLD modulation to cognitive difficulty predicts
of hippocampal dopamine D2 receptors on epi- poorer task accuracy and poorer fluid reasoning
sodic memory is modulated by BDNF and KIBRA ability. NeuroImage, 147, 262–271.
DOPAMINE SYSTEM AND COGNITIVE FUNCTION 131

Rieckmann, A., Johnson, K. A., Sperling, R. A., Buck- differences in frontostriatal representation of predic-
ner, R. L., & Hedden, T. (2018). Dedifferentiation of tion error but not reward outcome. Cognitive, Affec-
caudate functional connectivity and striatal dopa- tive, & Behavioral Neuroscience, 14(2), 672–682.
mine transporter density predict memory change Sawaguchi, T., & Goldman-Rakic, P. S. (1991). D1
in normal aging. Proceedings of the National dopamine receptors in prefrontal cortex: Involve-
Academy of Sciences, 115(40), 10160–10165. ment in working memory. Science, 251,
Rieckmann, A., Karlsson, S., Fischer, H., & Backman, 947–950.
L. (2011a). Caudate dopamine D1 receptor density Schott, B. H., Minuzzi, L., Krebs, R. M., Elmenhorst,
is associated with individual differences in fron- D., Lang, M., Winz, O. H., … Bauer, A. (2008).
toparietal connectivity during working memory. Mesolimbic functional magnetic resonance imag-
Journal of Neuroscience, 31(40), 14284–14290. ing activations during reward anticipation corre-
Rieckmann, A., Karlsson, S., Karlsson, P., Brehmer, Y., late with reward-related ventral striatal dopamine
Fischer, H., Farde, L., Nyberg, L., & Bäckman, L. release. Journal of Neuroscience, 28(52),
(2011b). Dopamine D1 receptor associations 14311–14319.
within and between dopaminergic pathways in Schultz, W. (1998). Predictive reward signal of dopa-
younger and elderly adults: Links to cognitive per- mine neurons. Journal of Neurophysiology, 80(1),
formance. Cerebral Cortex, 21(9), 2023–2032. 1–27.
Rieckmann, A., & Nyberg, L. (2020). Cognitive aging: Schultz, W. (2007). Multiple dopamine functions at
The role of neurotransmitter systems. In A. Thomas different time courses. Annual Review of Neurosci-
& A. Gutchess (Eds.), The Cambridge handbook of ence, 30(1), 259–288.
cognitive aging: A life course perspective (pp. 82- Schultz, W. (2013). Updating dopamine reward sig-
100). Cambridge, UK: Cambridge University Press. nals. Current Opinion in Neurobiology, 23(2),
Rocchetti, J., Isingrini, E., Dal Bo, G., Sagheby, S., 229–238.
Menegaux, A., Tronche, F., … Giros, B. (2015). Schultz, W., Apicella, P., & Ljungberg, T. (1993).
Presynaptic D2 dopamine receptors control long- Responses of monkey dopamine neurons to
term depression expression and memory processes reward and conditioned stimuli during successive
in the temporal hippocampus. Biological Psychia- steps of learning a delayed response task. Journal
try, 77(6), 513–525. of Neuroscience, 13(3), 900–913.
Roffman, J. L., Tanner, A. S., Eryilmaz, H., Rodriguez- Schultz, W., Dayan, P., & Montague, P. R. (1997). A
Thompson, A., Silverstein, N. J., Ho, N. F., … neural substrate of prediction and reward.
Catana, C. (2016). Dopamine D1 signaling organ- Science, 275, 1593–1599.
izes network dynamics underlying working Seaman, K. L., Smith, C. T., Juarez, E. J., Dang, L. C.,
memory. Science Advances, 2(6). doi: 10.1126/ Castrellon, J. J., Burgess, L. L., … Samanez-Larkin,
sciadv.1501672 G. R. (2019). Differential regional decline in dopa-
Rossato, J. I., Bevilaqua, L. R. M., Izquierdo, I., mine receptor availability across adulthood: Linear
Medina, J. H., & Cammarota, M. (2009). Dopa- and nonlinear effects of age. Human Brain Map-
mine controls persistence of long-term memory ping, 40(10), 3125–3138.
storage. Science, 325, 1017–1020. Seamans, J. K., & Yang, C. R. (2004). The principal
Salami, A., Garrett, D. D., Wåhlin, A., Rieckmann, A., features and mechanisms of dopamine modula-
Papenberg, G., Karalija, N., … Nyberg, L. (2019). tion in the prefrontal cortex. Progress in Neurobi-
Dopamine D2/3 binding potential modulates ology, 74(1), 1–58.
neural signatures of working memory in a load- Seeman, P., Bzowej, N. H., Guan, H.-C., Bergeron, C.,
dependent fashion. Journal of Neuroscience, Becker, L. E., Reynolds, G. P., … Tourtellotte, W. W.
39(3), 537–547. (1987). Human brain dopamine receptors in chil-
Salami, A., Rieckmann, A., Karalija, N., Avelar- dren and aging adults. Synapse, 1(5), 399–404.
Pereira, B., Andersson, M., Wåhlin, A., … Nyberg, Shohamy, D., & Adcock, R. A. (2010). Dopamine and
L. (2018). Neurocognitive profiles of older adults adaptive memory. Trends in Cognitive Sciences,
with working-memory dysfunction. Cerebral 14(10), 464–472.
Cortex, 28(7), 2525–2539. Simon, J. R., Howard, J. H., & Howard, D. V. (2010).
Salamone, J. D., & Correa, M. (2012). The mysterious Adult age differences in learning from positive and
motivational functions of mesolimbic dopamine. negative probabilistic feedback. Neuropsychology,
Neuron, 76(3), 470–485. 24(4), 534–541.
Samanez-Larkin, G. R., & Knutson, B. (2015). Deci- Sojitra, R. B., Lerner, I., Petok, J. R., & Gluck, M. A.
sion making in the ageing brain: Changes in affec- (2018). Age affects reinforcement learning
tive and motivational circuits. Nature Reviews through dopamine-based learning imbalance and
Neuroscience, 16(5), 278–289. high decision noise: Not through Parkinsonian
Samanez-Larkin, G. R., Worthy, D. A., Mata, R., mechanisms. Neurobiology of Aging, 68,
McClure, S. M., & Knutson, B. (2014). Adult age 102–113.
132 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Squire, L. R. (1992). Memory and the hippocampus: Volkow, N. D., Gur, R. C., Wang, G. J., Fowler, J. S.,
A synthesis from findings with rats, monkeys, and Moberg, P. J., Ding, Y. S., Hitzemann, R., Smith,
humans. Psychological Review, 99(2), 195–231. G., & Logan, J. (1998a). Association between
Störmer, V. S., Passow, S., Biesenack, J., & Li, S.-C. decline in brain dopamine activity with age and
(2012). Dopaminergic and cholinergic modula- cognitive and motor impairment in healthy indi-
tions of visual-spatial attention and working viduals. American Journal of Psychiatry, 155(3),
memory: Insights from molecular genetic research 344–349.
and implications for adult cognitive development. Volkow, N. D., Wang, G. J., Fowler, J. S., Ding, Y. S.,
Developmental Psychology, 48(3), 875–889. Gur, R. C., Gatley, J., … Pappas, N. (1998b). Paral-
Surmeier, D. J., Ding, J., Day, M., Wang, Z., & Shen, lel loss of presynaptic and postsynaptic dopamine
W. (2007). D1 and D2 dopamine-receptor modu- markers in normal aging. Annals of Neurology,
lation of striatal glutamatergic signaling in striatal 44(1), 143–147.
medium spiny neurons. Trends in Neurosciences, Wang, M., Vijayraghavan, S., & Goldman-Rakic, P. S.
30(5), 228–235. (2004). Selective D2 receptor actions on the func-
ter Huurne, N., Fallon, S. J., van Schouwenburg, M., tional circuitry of working memory. Science, 303,
van der Schaaf, M., Buitelaar, J., Jensen, O., & 853–856.
Cools, R. (2015). Methylphenidate alters selective Wang, S.-H., Redondo, R. L., & Morris, R. G. M.
attention by amplifying salience. Psychopharma- (2010). Relevance of synaptic tagging and cap-
cology, 232(23), 4317–4323. ture to the persistence of long-term potentiation
Tompary, A., Duncan, K., & Davachi, L. (2015). Con- and everyday spatial memory. Proceedings of
solidation of associative and item memory is the National Academy of Sciences, 107(45),
related to post-encoding functional connectivity 19537–19542.
between the ventral tegmental area and different Westbrook, A., & Braver, T. S. (2016). Dopamine does
medial temporal lobe subregions during an unre- double duty in motivating cognitive effort. Neuron,
lated task. Journal of Neuroscience, 35(19), 89(4), 695–710.
7326–7331. Westbrook, A., Frank, M. J., & Cools, R. (2021). A
Treadway, M. T., Buckholtz, J. W., Cowan, R. L., mosaic of cost–benefit control over cortico-striatal
Woodward, N. D., Li, R., Ansari, M. S., … Zald, D. circuitry. Trends in Cognitive Sciences, 25(8),
H. (2012). Dopaminergic mechanisms of individual 710–721.
differences in human effort-based decision-mak- Westbrook, A., van den Bosch, R., Määttä, J. I., Hof-
ing. Journal of Neuroscience, 32(18), 6170–6176. mans, L., Papadopetraki, D., Cools, R., & Frank, M.
Tritsch, N. X., & Sabatini, B. L. (2012). Dopaminergic J. (2020). Dopamine promotes cognitive effort by
modulation of synaptic transmission in cortex and biasing the benefits versus costs of cognitive work.
striatum. Neuron, 76(1), 33–50. Science, 367, 1362–1366. doi: 10.1126/science.
Turner, M. P., Fischer, H., Sivakolundu, D. K., Hubbard, aaz5891
N. A., Zhao, Y., Rypma, B., & Bäckman, L. (2020). Williams, G. V., & Goldman-Rakic, P. S. (1995). Modu-
Age-differential relationships among dopamine D1 lation of memory fields by dopamine D1 receptors
binding potential, fusiform BOLD signal, and in prefrontal cortex. Nature, 376, 572–575.
face-recognition performance. NeuroImage, 206. Wittmann, B. C., Schott, B. H., Guderian, S., Frey, J.
doi: 10.1016/j.neuroimage.2019.116232 U., Heinze, H.-J., & Düzel, E. (2005). Reward-related
Vallone, D., Picetti, R., & Borrelli, E. (2000). Structure fMRI activation of dopaminergic midbrain is associ-
and function of dopamine receptors. Neurosci- ated with enhanced hippocampus-dependent long-
ence & Biobehavioral Reviews, 24(1), 125–132. term memory formation. Neuron, 45(3), 459–467.
Vijayraghavan, S., Wang, M., Birnbaum, S. G., Wil- Zald, D. H., Boileau, I., El-Dearedy, W., Gunn, R.,
liams, G. V, & Arnsten, A. F. T. (2007). Inverted-U McGlone, F., Dichter, G. S., & Dagher, A. (2004).
dopamine D1 receptor actions on prefrontal neu- Dopamine transmission in the human striatum
rons engaged in working memory. Nature Neuro- during monetary reward tasks. Journal of Neuro-
science, 10(3), 376–384. science, 24(17), 4105–4112.
9
Neurotransmission and
Neuromodulation of Recognition
Memory: Novelty vs. Familiarity
Daniel Osorio-Gómez, María-Isabel Miranda,
Federico Bermúdez-Rattoni, and
Kioko Guzmán-Ramos

INTRODUCTION and physiological changes through protein syn-


thesis to store information for extended periods,
Many organisms, including human beings, require even a lifetime (Dudai, 2004; Goelet et al., 1986;
modifying their behavior under variable situations McGaugh, 2000). Another classification of mem-
to promote and increase survival. Across evolutive ory involves the use of memory systems or neural
processes, memory has become an essential networks and is divided into explicit (declarative)
mechanism to cope with environmental modifica- and implicit (non-declarative) (Squire, 2004).
tions. In general, organisms need to remember Explicit memory refers to information that can be
adequate shelters, food sources, and dangerous recalled consciously and is related to facts (seman-
places or situations to avoid them. Usually, this tic) and events (episodic). Implicit memories are
knowledge is conserved through a process called related to information acquired through repetition,
memory, in which information is encoded, inte- such as habits or motor skills and conditioning; the
grated, and retrieved (Squire, 2009). Therefore, information is recalled without awareness. It has
memory promotes survival by shaping an organ- been reported that explicit memory requires the
ism’s behavior according to its acquired functional integrity of the medial temporal lobe.
experience. In contrast, implicit memory usually involves
Since the pioneering discovery in the 1950s by brain structures such as the cerebellum, amygdala,
Scoville and Milner of the loss of recent memory and basal ganglia among others (Nadel and Hardt,
following bilateral hippocampal lesions, consider- 2011; Squire, 2004). In this chapter we focus on
able evidence suggests that memory could be clas- recognition memory, an essential aspect of explicit
sified depending on the duration and participation memory in which organisms judge that a recent
of particular brain structures and circuits. In terms experience, including subject, location, or event,
of duration, memory can be divided into short-term has been previously experienced or encountered.
memory (STM) and long-term memory (LTM). Long-lasting memories undergo different
The neurobiological mechanisms involved in stages: encoding, consolidation, retrieval and
maintaining STM and LTM differ; STM modifies updating (Abel and Lattal, 2001; Dudai, 2004;
pre-existing brain networks by modulating exist- Rodriguez-Ortiz and Bermúdez-Rattoni, 2017;
ing proteins, while LTM demands morphological Sara, 2000). Memory encoding1 includes the
134 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

acquisition of information by learning events


during exposure to stimuli (McGaugh, 2000). Acquisition
As mentioned above, consolidation2 involves Information Encoding

changes in the structure of brain networks through


protein synthesis; in this process, memory is
stabilized into LTM suitable for future recalls
during memory retrieval (Bisaz et al., 2014;
McGaugh, 2000). Memory retrieval3 is defined
here as the process by which internal and external Consolidation
cues promote selection, reactivation, and assess-
ment of a behavioral outcome (Ben-Yakov et al.,
2015; Frankland et al., 2019). In addition, LTM
may go through a consolidation-like process after
memory retrieval. This process is known as recon-
solidation (Nader et al., 2000; Sara, 2000), and
it is when memory updating occurs (Lee et al., Retrieval
Reactivation Expression
2017; Rodriguez-Ortiz and Bermúdez-Rattoni,
2017; see Figure 9.1).
Various chemical substances carry information
between neurons at the synaptic level, underly-
ing neuronal communication within the brain.
Nevertheless, more than one hundred chemical
substances and even more receptors are involved
New information
in synaptic transmission (Holz and Fisher, 2012;
Hyman, 2005), promoting memory formation.
After the transduction4 of recently acquired infor-
mation, neurotransmitters are released into the
synaptic cleft, diffused across the synapse, and
bound to their respective receptors. According
Updating
to the neuroscience literature, there have been
attempts to categorize or group neurotransmit-
ters based on their chemical nature, function,
mechanisms, among other classifications. In this
regard, it has been proposed that neurotransmit-
ters are substances that produce rapid changes in
the membrane potential through the activation of
Figure 9.1 Long-term memory stages.
ligand-gated channels.5 In contrast, neuromodu-
lators are chemical substances that promote slow Exteroceptive and interoceptive informa-
biochemical changes in postsynaptic neurons, tion is encoded during acquisition and sub-
modulating presynaptic, and postsynaptic events. sequently stabilized into long-term memory
Unfortunately, this classification oversimplifies after structural changes within brain net-
the function and activity of neurotransmitters, works over memory consolidation. After the
since many neurochemicals activate ligand-gated integration of information, memories are
channels and metabotropic receptors6 (Hyman, recalled throughout retrieval, when they
2005). In this regard, we propose that neuro- are reactivated and expressed. Furthermore,
chemical substances can be identified as neuro- memory updating occurs only when new
transmitters and neuromodulators according to
information is integrated into the previ-
their effect on learning and memory processes.
Neurotransmitters are involved in signaling the ously formed memory traces.
functional properties of neural circuits required
for memory integration, whereas neuromodulators
modify the intrinsic characteristics of the neural
circuits regulating memory integration. Overall, required to promote and modulate cellular and
synaptic activity through neurotransmission and molecular mechanisms that subserve novelty
neuromodulation induces morphological changes detection and eventually transform novel stimuli
within brain networks for LTM establishment. into familiar stimuli necessary for recognition
Therefore, a series of neurochemical events are memory.
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 135

RECOGNITION MEMORY regarding the brain structures involved in familiar-


ity and recollection processes; evidence suggests
Memory is an effective strategy to cope with envi- that both processes depend on distinct neural sub-
ronmental changes; specifically, recognition strates. The hippocampal formation and prefrontal
memory identifies different types of information, cortex are pivotal in recovering the qualitative
including faces, places, sounds, objects, or changes information of experienced events, while familiar-
in the context to distinguish familiar events from ity relies on the parahippocampal (perirhinal,
novel ones. It may be appropriate to distinguish entorhinal, and postrhinal) cortex of the brain
between assessment of explicit memory through (Brown and Aggleton, 2001; Evans and Wilding,
recall or recognition and their differences in pro- 2012; Merkow et al., 2015; Yonelinas, 2002).
cedure and effort involved. Recognition memory Recognition memory has been studied using
plays a crucial role in episodic memory, which different strategies and stimuli. A widely used
comprises the acquisition, consolidation, retrieval, paradigm of taste recognition memory is the one
and updating of information (Balderas et al., 2015; regarding identifying a familiar food. The poten-
Morici et al., 2015) about episodes experienced by tial to ingest harmful foods is reduced when
organisms in a space-time frame (Squire and Zola, organisms restrict the consumption of novel tastes
1996; Tulving, 2002). Recognition memory (neophobia). With subsequent exposures, the
involves two differential processes: familiarity and taste becomes familiar and organisms increase
recollection (Brown and Aggleton, 2001; Merkow the ingestion of the familiar stimulus (attenuation
et al., 2015). Familiarity refers to the ability to of neophobia), since as pointed out by Michael
judge whether a particular stimulus or event has Domjan, this could be incorporated into their diet
already been experienced (Mandler, 1980). as a safe source of nutrition. Accordingly, the defi-
Recollection is an independent process related to nition of attenuation of neophobia indicates that
recovering the characteristics (qualitative) of a learning and memory events occur in organisms
stimulus or event (Evans and Wilding, 2012; see promoting the recognition of novel and familiar
Figure 9.2). There is a comprehensive discussion taste stimuli (Osorio-Gómez et al., 2018).

Figure 9.2 Recognition memory. Novelty/familiarity and recollection constitute recognition


memory. Familiarity is the ability to recognize if an experience, stimulus, or situation has
been encountered before, whereas recollection processes are associated with the contextual
integration of the characteristics of the event.
136 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Novel object recognition7 (NOR) and object they need to detect novel stimuli and discrimi-
location memory8 (OLM) tasks are regularly nate environmental information. Novelty pro-
employed to assess the neurobiological mecha- cessing is an essential cognitive function that
nisms of recognition memory, which involve modulates other cognitive processes. For exam-
periods of handling and habituation to an empty ple, novelty plays a crucial role in episodic
open field. During the sample phase, animals are memory, as novel objects, context, gustatory
exposed to one or two identical novel objects; stimuli, among others are remembered better
afterward, animals are reintroduced to the than less distinctive stimuli due to familiarity
open field during the test phase and recognition (Kishiyama and Yonelinas, 2003). Moreover, it
memory is evaluated either by presenting a dif- has been suggested that the novelty-familiarity
ferent novel object, in NOR tasks, or by displac- process modulates overall recognition memory
ing an object to a novel location, in OLM tasks performance (Parker et al., 1998).
(Ennaceur and Delacour, 1988; Moreno-Castilla Novelty can be defined as the quality of being
et al., 2018). Both tasks allude to the ability of new and unusual. It triggers attention, motiva-
animals to identify environmental changes by the tion, and memory processes (Bastin et al., 2019).
addition of an unknown object or a novel spatial Thus, novelty refers to an attribute of simple
configuration, where researchers can assess novel/ or complex stimuli that has never been experi-
familiar discrimination and the neurobiological enced; specifically, there are different types of
changes that subserve this process. Animal mod- novelty depending on the nature of their pre-
els are widely employed to evaluate learning and existing representations (Kafkas and Montaldi,
memory processes. Behavioral tasks evaluate 2018). Absolute novelty implies that a stimulus
the ability to recognize a previously experienced has never been encountered before, such as new
stimulus or situation and are useful tools to repre- objects or a novel taste. In contrast, contextual
sent human amnesia modeling. Although memory novelty detects an unexpected position/location
is often evaluated in humans through language of the components of an experienced stimulus–
(verbal and written), recognition memory is also context pairing (Bastin et al., 2019; Kafkas and
examined in humans through the visual-paired Montaldi, 2018; Ranganath and Rainer, 2003).
comparison task. In this task, two identical pic- Regardless of the type of stimulus, exposure
tures are presented side-by-side during a brief to a novel stimulus or context engages several
period; after a delay, one of the previous pictures brain regions, including the prefrontal cortex
is presented alongside a new picture. Individuals (Kishiyama and Yonelinas, 2003), insular cortex
will look more at the new picture than the familiar (Bermudez-Rattoni, 2014), amygdala (Blackford
one (Manns et al., 2000). As explained later, NOR, et al., 2010; Guzmán-Ramos et al., 2012b), and
OLM and visual-paired comparison tasks rely on medial temporal areas, including the hippocam-
brain structures involved in declarative memory. pus and perirhinal cortices (Ranganath and
Learning and memory require different neuro- Rainer, 2003; Tulving et al., 1996).
chemical substances to properly integrate infor- As indicated above, a widely used paradigm
mation into LTM. Recognition memory promotes to study recognition memory is the attenua-
identifying different stimuli through acquisition, tion of neophobia. Presentation of a novel taste
consolidation, retrieval, and updating, which are stimulus enhances c-Fos expression in the central
necessary for novel/familiar discrimination. In this (Yamamoto et al., 1997) and basolateral nuclei of
chapter, we review how neurotransmitters signal the amygdala (Lin et al., 2012), the perirhinal cor-
information about novel and familiar stimuli and tex (Gómez-Chacón et al., 2015), the nucleus of
how this signaling is regulated by neuromodula- the solitary tract (Houpt et al., 1994), the thala-
tors that enhance LTM formation. Many neu- mus and the insular cortex (Lin et al., 2012). In
rotransmitters and neuromodulators may have addition, it has been reported that these structures
convergent functions, but for clarity, these are pre- modify their electrophysiological responses after
sented separately in this chapter. the presentation of a novel taste stimulus (Adachi
et al., 1989; Carleton et al., 2010; Peng et al.,
2015; Verhagen et al., 2003). However, we will
focus on the neurochemicals involved in novel
taste recognition. Catecholaminergic denervation
NEUROTRANSMISSION RELATED TO by 6-hydroxydopamine9 (6-OHDA) administra-
NOVELTY SIGNALING tion within the olfactory bulb (Royet et al., 1983)
and dorsal bundle (Cole et al., 1988; Steketee
As indicated before, organisms search for shelter et al., 1989) reduces the neophobic response.
and food resources and constantly evaluate pos- Similar lesions in the lateral tegmental area pro-
sible dangers (Ahmadlou et al., 2021); therefore, mote a decrease in the consumption of novel
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 137

stimuli (Dunn and Everitt, 1987). On the other stimulus increases acetylcholine within the insular
hand, serotoninergic terminal denervation spares cortex (Miranda et al., 2000; Rodríguez-García
the detection of novel gustatory stimuli (Royet and Miranda, 2016). Also, the administration of
et al., 1983). Additionally, changes in several neu- scopolamine, a muscarinic receptor antagonist,
rotransmitters have been monitored during the spares the neophobic response when injected into
presentation of novel gustatory stimuli by using the insular cortex (Gutiérrez et al., 2003), perirhi-
in vivo microdialysis.10 Novel gustatory stimu- nal cortex (Gutiérrez et al., 2004), and nucleus
lation induces an elevation of dopamine within accumbens (Ramírez-Lugo et al., 2006). However,
the nucleus accumbens, medial prefrontal cortex administering a cholinergic agonist into the insu-
(Bassareo and Di Chiara, 1999; Bassareo et al., lar cortex generates a novelty signal that facili-
2002), amygdala (Guzmán-Ramos et al., 2012b) tates subsequent association leaning (Clark and
and insular cortex (Guzmán-Ramos et al., 2010; Bernstein, 2009). Moreover, systemic (Aguado
Moreno-Castilla et al., 2016). There is also an et al., 1994) or intra-amygdalar (Figueroa-Guzmán
augmentation of norepinephrine in the amygdala et al., 2006) administration of a non-competitive
(Guzmán-Ramos et al., 2012b) and insular cortex NMDA receptor antagonist does not affect the
(Guzmán-Ramos et al., 2010). Moreover, novel neophobic response. Contrarily, administering a
stimulus presentation is related to dopaminergic competitive NMDA antagonist within the baso-
activity within the insular cortex regardless of its lateral amygdala diminishes the rejection of novel
valence, since presenting either an innate aversive gustatory stimuli (Burns et al., 1994). Altogether,
gustatory stimulus (quinine) or an innate appeti- the presentation of novel gustatory stimuli elicits
tive gustatory stimulus promotes an elevation of an increase in acetylcholine, whereas glutamater-
dopamine (Osorio-Gómez et al., 2021). gic levels remain unaltered. Additionally, cholin-
Norepinephrine administration into the baso- ergic activation promotes taste novelty signaling.
lateral amygdala (Borsini and Rolls, 1984) or However, pharmacological manipulation of the
insular cortex (Rojas et al., 2015) enhances the cholinergic system does not alter the neophobic
neophobic response; in contrast, a blockade of response, indicating that acetylcholine and gluta-
beta-adrenergic receptors within the basolat- mate could act as an instructive signal required for
eral amygdala (Roozendaal and Cools, 1994) or a transition from novelty to familiarity, as will be
insular cortex (Rojas et al., 2015) diminishes the discussed later.
rejection of a novel taste stimulus. Therefore, As previously mentioned, besides recognizing
norepinephrine release and the subsequent acti- novel foods, animals must identify novel objects
vation of beta-adrenergic receptors within the or contexts. In this regard, NOR and OLM tasks
basolateral amygdala and insular cortex occur in have been used as behavioral models to study
novelty signaling. Similarly, a decrease in dopa- items and spatial recognition memory. Several
mine levels within the insular cortex, as a result studies have reported that OLM and NOR depend
of beta-amyloid accumulation in an Alzheimer’s on different brain structures; in general, OLM
disease model, reduces the neophobic response; requires the hippocampal formation, particularly
this detrimental effect is ameliorated by the inhibi- the dorsal CA1 (Assini et al., 2009; Barrett et al.,
tion of dopamine reuptake (Moreno-Castilla et al., 2011) and CA3 (Wagatsuma et al., 2018) por-
2016). The dopaminergic system is also involved tions, to acquire, consolidate, and retrieve spatial
in neophobic response through D2 receptors, since information (Mumby et al., 2002). Conversely,
systemic administration of a D2 receptor agonist NOR involves the insular cortex (Balderas et al.,
reduces the rejection to novel stimuli (Muscat 2008; Bermudez-Rattoni et al., 2005), perirhinal
et al., 1992). Hence, catecholaminergic activity is cortex (Balderas et al., 2008; Winters et al., 2004;
required to modulate the novel response because Winters and Bussey, 2005), and ventromedial cor-
norepinephrine and dopamine levels increase after tex (Akirav and Maroun, 2006). Studies have also
presenting a novel gustatory stimulus. Therefore, reported the involvement of the hippocampus in
the activation of beta-adrenergic and D2 receptors NOR (Broadbent et al., 2010; Cohen et al., 2013;
modulates novelty detection and the neophobic Furini et al., 2010, 2014, 2015; Mello-Carpes
response. et al., 2016; Myskiw et al., 2008; Reed and Squire,
Concerning the cholinergic system and amino 1997). However, some controversy arises for its
acids, the presentation of a novel taste stimulus does participation in short but not long-term mem-
not promote any changes in the extracellular levels ory (Balderas et al., 2008; Haettig et al., 2011;
of glutamate within the amygdala (Guzmán-Ramos Mumby, 2001).
et al., 2012b; Miranda et al., 2002; Tucci et al., Microdialysis experiments have shown that
1998) and insular cortex (Guzmán-Ramos presenting a novel context does not promote either
et al., 2010; Miranda et al., 2002; Osorio-Gómez glutamate or GABA elevation in the hippocam-
et al., 2021). On the contrary, exposure to a novel pus or prefrontal cortex (Giovannini et al., 2001).
138 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

However, exposure to novel objects induces Therefore, norepinephrine enhances behavioral


an elevation of acetylcholine in the hippocam- performance inducing exploration by modulat-
pus (Aloisi et al., 1997; Ceccarelli et al., 1999; ing other effects of excitatory or inhibitory neu-
Degroot et al., 2005; Ihalainen et al., 2010; Inglis rotransmitters. Information about serotonin and
et al., 1994; Stanley et al., 2012) and frontal cor- novelty detection is scarce, as the presentation
tex (Giovannini et al., 1998, 2001). Furthermore, of a novel environment does not induce changes
exposure to novel objects causes contradictory in serotonin levels within the ventral midbrain,
results since elevated glutamatergic levels in the basal ganglia, nucleus accumbens, or prefrontal
hippocampus have been reported (Stanley et al., cortex (Jones et al., 1996). However, it seems that
2012). In contrast, we reported no changes in serotoninergic denervation in a transgenic mouse
glutamate after the presentation of novel objects model for Down syndrome promotes an increased
(Guzmán-Ramos et al., 2012a). At the same time, response to novelty (Bell et al., 2003), suggesting
GABA levels remain similar to basal levels within its involvement in novelty response modulation or
the hippocampus and insular cortex after present- unspecific modifications in other neurotransmit-
ing novel stimuli (Guzmán-Ramos et al., 2012a; ters, such as dopamine (Bortolozzi et al., 2005;
Stanley et al., 2012). Díaz-Mataix et al., 2005).
The catecholaminergic system is widely Similarly, dopamine modulates learning signals
involved in NOR and OLM performance; dopa- that facilitate the prediction of events (Montague
mine is released within the insular cortex during et al., 2004). Traditionally, dopamine has been
the presentation of novel objects in the sample associated with the prediction and coding of
phase, whereas hippocampal dopamine remains rewarding events (Schultz, 1998; Schultz et al.,
unaltered (Guzmán-Ramos et al., 2012a). On 1993). Nevertheless, the dopaminergic response
the other hand, dopamine and noradrenaline could be a regulator of the behavioral response
levels increase within the dorsal hippocampus and a modulator of learning signals that strengthen
(Hernández-Ramírez et al., 2021; Mello-Carpes associative events between novel predictor-stimuli
et al., 2016; Moreno-Castilla et al., 2017), ven- and their possible consequences.
tral hippocampus (Titulaer et al., 2021), striatum
(Ihalainen et al., 1999), and prefrontal cortex
(Feenstra et al., 2000; Ihalainen et al., 1999) while
exploring a novel contextual configuration. In NEUROCHEMISTRY INVOLVED IN THE
addition, novel stimulus presentation elicits the
activation of the ventral tegmental area (Düzel
CONSOLIDATION OF RECOGNITION
et al., 2010; Schultz, 1998) and the subsequent MEMORY
release of dopamine in the nucleus accumbens
(Legault and Wise, 2001; Leonibus et al., 2006) After novelty detection, neurotransmitter release is
and prefrontal cortex (Feenstra and Botterblom, modified in response to a familiarity-consolidation
1996; Feenstra et al., 2000). The locus coeruleus process. The catecholaminergic response induced
also increases its firing response after presenting by the presentation of novel gustatory stimuli is
novel objects or contexts (Pudovkina et al., 2001; altered because of repetitive exposure to the same
Sara et al., 1994; Vankov et al., 1995; Wagatsuma stimulus. In this regard, once the taste stimulus is
et al., 2018). familiar, the initial dopaminergic release is
The presentation of novel stimuli, indepen- reduced within the nucleus accumbens (De Luca,
dently of the sensory modality, evokes several 2014), amygdala (Osorio-Gómez et al., 2016), and
neurotransmitters release, including acetylcho- insular cortex (Osorio-Gómez et al., 2017); like-
line, dopamine, and norepinephrine. Glutamate wise, noradrenaline release remains unaltered
and GABA activity in novelty detection during within the amygdala and insular cortex after the
NOR and OLM tasks is not apparent due to con- presentation of a familiar stimulus (Osorio-Gómez
tradictory results. This elevation induces a cascade et al., 2016, 2017). Thus, the dopaminergic and
of learning and memory events (Hasselmo, 2006) noradrenergic response is no longer engaged due
that could contribute to familiarity consolidation. to the recognition of familiar gustatory stimuli.
Thus, cholinergic signaling is involved in nov- Additionally, denervation of the dorsal noradren-
elty that instructs the integration of recognition ergic bundle accelerates attenuation of neophobia
memory. In addition, changes in norepinephrine (Sahakian et al., 1983), while the blockade of D1
levels could be associated with detecting salient dopaminergic receptors in the insular cortex
stimuli; the release of norepinephrine regulates before the presentation of a novel stimulus impairs
cortical function and mediates arousal, improving attenuation of neophobia (David et al., 2014). This
subsequent cognitive functions such as attention evidence shows that catecholaminergic activity
and motivation (Aston-Jones and Cohen, 2005). diminishes after repeated presentation of the
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 139

gustatory stimulus, suggesting that dopamine dorsal hippocampus (Hunsaker et al., 2007) or the
(Duszkiewicz et al., 2019) and noradrenaline perirhinal cortex (Tang et al., 1997; Tinsley et al.,
(McIntyre et al., 2012) enhance the consolidation 2011; Warburton et al., 2003) hinders NOR forma-
of gustatory recognition memory during the initial tion (STM and LTM) when administered before
presentations of the stimulus. the sample phase. Besides, the inhibition of acetyl-
Likewise, the extracellular levels of acetylcho- cholinesterase within the hippocampus maintains
line within the insular cortex decrease after the acetylcholine activity, enhancing NOR (Hunsaker
consecutive presentation of a gustatory stimulus. et al., 2007). The administration of a muscarinic
Decrements observed in acetylcholine are related antagonist into the insular or perirhinal cortices
to the increased consumption of the familiar gusta- immediately after the sample phase impairs NOR
tory stimulus (Miranda et al., 2000). Concerning consolidation (Balderas et al., 2008; Bermudez-
acetylcholine and consolidation of taste recogni- Rattoni et al., 2005) and object-in-context memory
tion memory, the blockade of muscarinic recep- recognition when administered in the hippocam-
tors in the nucleus accumbens (Ramírez-Lugo pus (Balderas et al., 2008). Therefore, cholinergic
et al., 2006), insular cortex (Gutiérrez et al., activity within the hippocampus and perirhinal
2003), or perirhinal cortex (Gutiérrez et al., 2004) and insular cortices is responsible for learning and
has no effect on the neophobic response. Still, it consolidation processes associated with recogni-
impairs consolidation of familiarity, observed tion memory, promoting the transition from nov-
as a reduction in the attenuation of neophobia. elty to familiarity. For many years acetylcholine
Therefore, the blockade of muscarinic receptors has been described as an essential modulator of
prevents the novel stimulus from becoming famil- learning/consolidation events through changes in
iar. Significantly, cholinergic response within the synaptic plasticity efficiency (Hasselmo, 2006).
insular cortex is modulated by the nucleus basalis However, there is evidence that activating mus-
magnocellularis, the main nucleus that provides carinic (Dennis et al., 2016) or nicotinic receptors
acetylcholine to several brain regions. The inac- (Matsuyama et al., 2000) within the hippocampus
tivation of the nucleus basalis magnocellularis is sufficient to induce long-term potentiation, sug-
disrupts familiar memory formation, preventing gesting that cholinergic transmission modulates
acetylcholine release within the insular cortex synaptic plasticity and makes a significant contri-
(Rodríguez-García and Miranda, 2016). Thereby, bution to memory-related events. Thus, it is prob-
cholinergic activity is widely involved in establish- able that acetylcholine release induced by novelty
ing taste familiarity, suggesting that acetylcholine initiates learning signals essential for recognition
encodes taste novelty triggering consolidation sig- memory regulated by neuromodulators such as
nals that promote gustatory recognition memory. catecholamines.
Concerning glutamate, we have previously As mentioned before, exposure to novel objects
mentioned that the presentation of a novel or or contexts induces an elevation of norepineph-
familiar gustatory stimulus does not induce an rine and dopamine within several brain regions.
elevation of glutamate within the amygdala or Regarding dopamine, systemic administration
the insular cortex (Osorio-Gómez et al., 2016, of a D1/D5 receptor agonist enhances NOR per-
2017; Tucci et al., 1998). Nevertheless, it has been sistence (de Lima et al., 2011) and facilitates the
observed that systemic (Aguado et al., 1994), retrieval of recognition memory (Hotte et al.,
intra-amygdalar (Figueroa-Guzmán and Reilly, 2005). Conversely, the pharmacological blockade
2008), or intra-cortical (Figueroa-Guzmán et al., of D1/D5 receptors within the perirhinal cortex
2006) administration of a non-competitive NMDA before the sample phase spares the acquisition
receptor antagonist impairs attenuation of neo- (intact STM) but impedes NOR consolidation
phobia. The administration of a specific mGluR1 (hindered LTM). However, the administration of
antagonist accelerated the attenuation of neopho- a D1/D5 receptor agonist promotes NOR consoli-
bia (Ramírez-Lugo et al., 2015). Therefore, even dation (Balderas et al., 2013a). Moreover, novelty
though changes in glutamatergic levels are not enhances memory persistence by activating hip-
observed during exposure to novel or familiar pocampal D1/D5 receptors (Takeuchi et al., 2016).
taste stimuli, it seems that glutamatergic receptors These results suggest that dopaminergic modula-
are involved in the consolidation of gustatory rec- tion is responsible for strengthening recognition
ognition memory, demonstrating that basal gluta- memories. Initially, novelty induces dopamine
matergic signaling is needed to sustain the taste release that improves memory persistence through
memory trace. the synthesis, tagging and capture of proteins
Regarding object-location recognition memory, associated with synaptic plasticity (Frey and
the reduction of acetylcholine impairs NOR con- Morris, 1998) generated by learning signals. In
solidation (De Jaeger et al., 2013). Additionally, the absence of a neuromodulator, synapses modi-
administration of a muscarinic antagonist into the fied during learning could return to the baseline
140 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

level without establishing memories (Duszkiewicz judged as familiar; this could be interpreted as
et al., 2019; Takeuchi et al., 2016). opposing signals that compete for the behavioral
Noradrenergic modulation of memories has outcome. However, evidence indicates that novel
been widely described (McGaugh, 2000, 2015); and familiar stimuli induce the activation of sev-
particularly, pharmacological manipulation of eral brain regions in a graded manner (Kafkas and
the noradrenergic system enhances the LTM Montaldi, 2014). Concerning neurotransmission,
of recognition tasks (Dornelles et al., 2007). novelty promotes an elevation in acetylcholine,
Norepinephrine administered into the hippocam- dopamine, and noradrenaline that gradually
pus immediately after the sample session pro- diminishes after the consecutive exposure to the
motes NOR persistence (Mello-Carpes et al., stimulus (Miranda et al., 2000; Osorio-Gómez
2016). Likewise, noradrenergic activation of the et al., 2016, 2017; Rodríguez-García and Miranda,
basolateral amygdala enhances the consolida- 2016). Thus, novelty elicits a maximum response,
tion of object-in-context recognition memory but a progressive response reduction accompanies
(Barsegyan et al., 2014), object recognition mem- the following presentations. This neurotransmitter
ory (Roozendaal et al., 2007), and object location modulation promotes the transition from novelty
(Song et al., 2021). Basolateral amygdala activity, to familiarity (Kafkas and Montaldi, 2018; see
through modulation of the noradrenergic system, Figure 9.3).
facilitates the consolidation of recognition mem- After learning, information is stabilized into
ory because of the regulation of arousal effects on LTM through protein synthesis. New proteins
the dorsal hippocampus and insular and prelimbic promote changes in the structure and physiology
cortices (Barsegyan et al., 2019). We have previ- of synaptic connections that strengthen memory
ously described that the locus coeruleus responds over time (McGaugh, 2000, 2013). Nevertheless,
to novelty stimuli. Although this brain structure consolidated memories can undergo a reconsoli-
has been classically associated with the produc- dation process11 (Nader et al., 2000) where memo-
tion of norepinephrine, recent evidence suggests ries are destabilized and restabilized after memory
that hippocampal terminal activation coming from retrieval (Lee et al., 2017; Rodriguez-Ortiz and
the locus coeruleus increases spatial memory Bermúdez-Rattoni, 2017; Sara, 2000). Memories
consolidation through D1/D5 receptor activity require updating to incorporate relevant and recent
(Takeuchi et al., 2016). Surprisingly, the locus information about the environment into the previ-
coeruleus induces noradrenergic and dopaminer- ously integrated memory. Memory retrieval trig-
gic release in the dorsal hippocampus (Kempadoo gers a period of lability where new information
et al., 2016). Hence, novelty enhances recogni- is incorporated, inducing memory updating. The
tion memory consolidation by the noradrenergic evidence suggests that reconsolidation only initi-
modulation of the amygdala and the hippocampal ates after new information arrives and cannot be
corelease of norepinephrine and dopamine via the induced without it (Garcia-Delatorre et al., 2014;
locus coeruleus. Lee, 2008; Lee et al., 2017; Rodriguez-Ortiz
and Bermúdez-Rattoni, 2017; Rodriguez-Ortiz
et al., 2005, 2008; Sevenster et al., 2014; Winters
et al., 2009). Therefore, the transition from nov-
elty to familiarity in recognition memory could
NEUROCHEMISTRY DURING be explained as progressive information updating
MEMORY UPDATING: LEARNING through reconsolidating processes until complete
AND RE-LEARNING familiarization is accomplished.
It has been documented that recognition memory
Recognition memory is achieved through two undergoes reconsolidation (Balderas et al., 2015;
aspects: recollection and familiarity. Recollection Kelly et al., 2003; Kwapis et al., 2020; Rodriguez-
requires the maintenance of information related to Ortiz et al., 2005; Santoyo-Zedillo et al., 2014).
the specific characteristics of the context or stimu- Thus, in taste recognition memory, each time a
lus, and familiarity involves remembering if a sit- taste stimulus is presented to the organism during
uation, stimulus, context, or event has been retrieval, memory is destabilized, and new infor-
previously experienced. The transition from nov- mation (familiarity) is integrated into the memory
elty to familiarity is not a single event; novelty trace. The inhibition of protein synthesis after the
promotes new information learning (Henson and first sessions of attenuation of neophobia impairs
Gagnepain, 2010) and plasticity changes (Lisman taste memory reconsolidation, and the familiar
et al., 2011) that are required to shift gradually taste is recognized as a novel again. Nevertheless,
toward familiarity. In this regard, it has been sug- when the stimulus has become very familiar, the
gested that novelty and familiarity are two sides of inhibition of protein synthesis spares taste rec-
the same coin since something novel cannot be ognition memory (Rodriguez-Ortiz et al., 2005).
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 141

Novel Familiar

Memory updating

Unstable
memory

Novel Novel Novel


Familiar
Familiar Familiar Familiar
Stable
memory
information

information
n

n
information
n

io

io

io
io

at

at

at
at

New

New
lid

lid

lid
New
lid

so

so

so
so

on

on

on
on

ec

ec

ec
C

R
Novel Novel Novel
Novel
Familiar Familiar Familiar

Figure 9.3 The transition from novelty to familiarity. Novelty promotes learning and memory
consolidation. Memory is destabilized and subsequently updated over reconsolidation when
new information is presented. The most significant amount of information is learned during
the first trials. Information updating during the last trials is diminished after the first sessions.

Interestingly, the blockade of muscarinic receptors since animals do not recognize the novel object.
within the insular cortex before the second famil- However, during the next test session, animals can
iarization session increases the neophobic response discriminate a novel object from a familiar one,
and delays attenuation of neophobia (Gutiérrez indicating memory updating. Nevertheless, sup-
et al., 2003), suggesting that novelty-familiarity pose the inactivation of the perirhinal cortex is fol-
updating is hindered. However, more studies are lowed by the administration of a protein synthesis
required to elucidate the role of other neurotrans- inhibitor during retrieval. In that case, animals do
mitters in taste memory recognition updating. not recognize the novel object during test sessions,
Object recognition memory (Akirav and suggesting a failure in memory updating (Balderas
Maroun, 2006; Balderas et al., 2013b, 2015; et al., 2013b, 2015). Similarly, the blockade of
Kelly et al., 2003; Rossato et al., 2007; Santoyo- AMPA receptors in the perirhinal cortex dis-
Zedillo et al., 2014) and object location memory rupts memory expression during retrieval, while
(Kwapis et al., 2020; Villain et al., 2016; Wright the blockade of NMDA receptors impairs NOR
et al., 2020) can also go through reconsolidation reconsolidation (Balderas et al., 2015; Santoyo-
processes. Inhibition of protein synthesis in the Zedillo et al., 2014). Therefore, evidence suggests
perirhinal cortex in the hippocampus hinders the that NOR reconsolidation occurs in the absence of
information previously integrated when updating the behavioral output during retrieval when new
with new information (Balderas et al., 2013b) or information is presented. NOR reconsolidation
the context (Winters et al., 2011) during the recon- requires NMDA receptor activation within the per-
solidation process. Moreover, systemic blockade irhinal cortex and subsequent protein synthesis.
of NMDA receptors impairs the reconsolidation The evidence related to catecholaminergic
of NOR (Winters et al., 2009) depending on glu- activity during NOR reconsolidation is scarce. D1/
tamatergic activity. Conversely, after contextual D5 receptors within the hippocampus are involved
updating, the systemic administration of nicotine in the destabilization process during reconsoli-
enhances NOR reconsolidation when done imme- dation since the inactivation of D1/D5 receptors
diately after retrieval (Tian et al., 2015). impedes the amnesic effect of a protein synthesis
Like other memories, NOR updating could inhibitor (Rossato et al., 2015). However, more
occur in the absence of the behavioral outcome data is necessary to comprehend the involvement
during retrieval. Inactivation of the perirhinal cor- of dopamine and noradrenaline on the transition
tex during retrieval impairs behavioral response of novelty to familiarity in recognition memory.
142 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

SUMMARY AND CONCLUSIONS processes associated with recognition memory


formation, promoting the transition from novelty
Learning and memory are critical cognitive pro- to familiarity. Moreover, cholinergic signaling
cesses that enable organisms to store relevant is modulated by catecholamines and released by
information obtained from the environment. the ventral tegmental area and locus coeruleus,
Learning and memory promote the acquisition, regulating the behavioral response associated with
storage and recall of data collected by sensory novelty and also enhancing consolidation signals
organs. During these processes, information is that strengthen recognition. In addition, we have
encoded and consolidated into LTM for subse- discussed how the ventral tegmental area and the
quent use during retrieval. Memory is a dynamic locus coeruleus midbrain structures modulate nov-
system in which stored information can be updated elty detection and promote recognition memory
after memory reactivation and reconsolidation consolidation through their catecholaminergic
when new information is added. Importantly, the projections. As well, we have addressed gluta-
consolidation, reconsolidation and updating pro- mate, GABA, and acetylcholine’s functional role
cesses require complex interaction and modula- in acquiring, consolidating, updating, and retriev-
tion between neurotransmitters, which promote ing recognition memory. Also, we have discussed
cellular and molecular mechanisms necessary for the main findings relating to neurotransmission
morphological and physiological modifications as related to novelty signaling, neurochemistry
involved with information maintenance. involved in the consolidation of recognition mem-
In this chapter, we have focused on one crucial ory, and neurochemistry during memory updating.
declarative memory – recognition memory – the Acknowledgments: We thank Gabriela Vera-
ability to discriminate or identify that something Rivera and Luis Rodriguez-Duran for technical
has been previously experienced. Recognition assistance, and we gratefully acknowledge Jessica
memory consists of two components: a collection G. Norris for her generous help in proofread-
of contextual information, which makes the recol- ing and editing the manuscript. DGAPA-PAPIIT
lection of the episode, where, and when the stimu- IN201420 and IN209122 to IM. Departamento de
lus was experienced, and the familiarity judgment, Ciencias de la Salud to KG-R. DGAPA-PAPIIT
which provides information on whether a stimulus IN212919 and IN213123 to FBR, and DGAPA-
had prior exposure. Recognition memory requires PAPIIT IA201420 and IA202922 to DOG.
the functional integrity of the medial temporal
lobe’s brain structures such as the perirhinal, para-
hippocampal, entorhinal, insular cortices, and the
hippocampus. Recognition memory allows organ- Notes
isms to remember specific characteristics (recol- 1  Memory encoding: Attention-dependent stage
lection) and recognize the familiarity of an event. where information from sensory inputs is incor-
Familiarity provides information on whether a porated through mechanisms that preserve it.
stimulus has been experienced previously with- 2  Consolidation: Time-dependent process of sta-
out the recollection component. The transition bilization of synaptic connectivity in local circuits
from novelty to familiarity in recognition memory and gradual reorganization of information at the
might be explained as a piece of progressive infor- systems level.
mation updated through reconsolidation processes 3  Memory retrieval: Process by which internal and
until complete familiarization is accomplished. external cues promote selection, reactivation and
Every time a new stimulus is presented to the assessment of a behavioral outcome.
organism during retrieval, memory is destabilized 4  Transduction: Conversion of a sensory stimulus
somewhat, and new information and familiar- into an electrical impulse.
ity information is integrated into the updating 5  Ligand-gated channels: Integral membrane pro-
memory trace. Recognition memory depends on teins forming a pore allowing passage of ions
cholinergic, glutamatergic, and dopaminergic sys- upon activation of a specific molecule.
tems that incorporate updated information into the 6  Metabotropic receptors: Transmembrane proteins
memory trace of “familiarity.” that activate coupled G-proteins, initiating several
Following the presentation of novel stimuli, signaling cascades that modulate cell activity.
acetylcholine, noradrenaline, and dopamine 7  Novel object recognition: Behavioral essay that
increase within multiple brain structures, includ- relies on rodent’s innate exploratory behaviour
ing the hippocampus and brain cortices (includ- in the absence of externally applied rules or
ing the perirhinal, entorhinal and insular cortices). reinforcement. In this task, animals discriminate
Acetylcholine encodes novelty and is also respon- between a previously seen object and a novel
sible for triggering learning and consolidation one.
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 143

8  Object location memory: Hippocampus-­ Balderas, I., Moreno-Castilla, P., & Bermudez-Rattoni,
ependent task where animals identify a familiar F. (2013a). Dopamine D1 receptor activity modu-
object in a novel contextual distribution. It has three lates object recognition memory consolidation in
components: (1) object processing, (2) spatial-loca- the perirhinal cortex but not in the hippocampus.
tion processing, and (3) object to location binding. Hippocampus, 23(10), 873–878.
9  6-Hydroxydopamine: A neurotoxin that leads to Balderas, I., Rodriguez-Ortiz, C. J., & Bermudez-Rat-
catecholaminergic cell death. toni, F. (2013b). Retrieval and reconsolidation of
10  In vivo microdialysis: Sampling and collecting object recognition memory are independent pro-
of small molecular weight substances from the cesses in the perirhinal cortex. Neuroscience, 253,
interstitial space. It allows the measurement of 398–405.
neurotransmitters in neuronal extracellular fluid Balderas, I., Rodriguez-Ortiz, C. J., & Bermudez-Rat-
in specific brain structures. toni, F. (2015). Consolidation and reconsolidation
11  Reconsolidation: Stabilization process by which of object recognition memory. Behavioural Brain
active (or reactivated) memories are converted Research, 285, 213–222.
into a lasting and stable long-term memory. Balderas, I., Rodriguez-Ortiz, C. J., Salgado-Tonda, P.,
Chavez-Hurtado, J., McGaugh, J. L., & Bermudez-
Rattoni, F. (2008). The consolidation of object and
context recognition memory involve different
regions of the temporal lobe. Learning & Memory,
REFERENCES 15(9), 618–624.
Barrett, R. M., Malvaez, M., Kramar, E., Matheos, D. P.,
Abel, T., & Lattal, K. M. (2001). Molecular mecha- Arrizon, A., Cabrera, S. M., Lynch, G., Greene, R. W.,
nisms of memory acquisition, consolidation and & Wood, M. A. (2011). Hippocampal focal knockout
retrieval. Current Opinion in Neurobiology, 11(2), of CBP affects specific histone modifications, long-
180–187. term potentiation, and long-term memory. Neu-
Adachi, M., Ohshima, T., Yamada, S., & Satoh, T. ropsychopharmacology, 36(8), 1545–1556.
(1989). Cross-correlation analysis of taste neuron Barsegyan, A., McGaugh, J. L., & Roozendaal, B.
pairs in rat solitary tract nucleus. Journal of Neuro- (2014). Noradrenergic activation of the basolateral
physiology, 62(2), 501–509. amygdala modulates the consolidation of object-
Aguado, L., San Antonio, A., Pérez, L., del Valle, R., in-context recognition memory. Frontiers in
& Gómez, J. (1994). Effects of the NMDA receptor Behavioral Neuroscience, 8. doi: 10.3389/fnbeh.
antagonist ketamine on flavor memory: Condi- 2014.00160
tioned aversion, latent inhibition, and habituation Barsegyan, A., Mirone, G., Ronzoni, G., Guo, C.,
of neophobia. Behavioral and Neural Biology, Song, Q., van Kuppeveld, D., … Roozendaal, B.
61(3), 271–281. (2019). Glucocorticoid enhancement of recogni-
Ahmadlou, M., Houba, J. H. W., Vierbergen, J. F. M. tion memory via basolateral amygdala-driven facil-
van, Giannouli, M., Gimenez, G.-A., Weeghel, C. itation of prelimbic cortex interactions. Proceedings
van, … Heimel, J. A. (2021). A cell type-specific of the National Academy of Sciences of the United
cortico-subcortical brain circuit for investigatory States of America, 116(14), 7077–7082.
and novelty-seeking behavior. Science, 372. doi: Bassareo, V., De Luca, M. A., & Di Chiara, G. (2002).
10.1126/science.abe9681 Differential expression of motivational stimulus
Akirav, I., & Maroun, M. (2006). Ventromedial pre- properties by dopamine in nucleus accumbens
frontal cortex is obligatory for consolidation and shell versus core and prefrontal cortex. Journal of
reconsolidation of object recognition memory. Neuroscience, 22(11), 4709–4719.
Cerebral Cortex, 16(12), 1759–1765. Bassareo, V., & Di Chiara, G. (1999). Differential
Aloisi, A. M., Casamenti, F., Scali, C., Pepeu, G., & responsiveness of dopamine transmission to food-
Carli, G. (1997). Effects of novelty, pain and stress stimuli in nucleus accumbens shell/core compart-
on hippocampal extracellular acetylcholine levels ments. Neuroscience, 89(3), 637–641.
in male rats. Brain Research, 748(1-2), 219–226. Bastin, C., Delhaye, E., Moulin, C., & Barbeau, E. J.
Assini, F. L., Duzzioni, M., & Takahashi, R. N. (2009). (2019). Novelty processing and memory impair-
Object location memory in mice: Pharmacological ment in Alzheimer’s disease: A review. Neurosci-
validation and further evidence of hippocampal ence and Biobehavioral Reviews, 100, 237–249.
CA1 participation. Behavioural Brain Research, Bell, K., Shokrian, D., Potenzieri, C., & Whitaker-
204(1), 206–211. Azmitia, P. M. (2003). Harm avoidance, anxiety,
Aston-Jones, G., & Cohen, J. D. (2005). An integra- and response to novelty in the adolescent S-100 β
tive theory of locus coeruleus-norepinephrine transgenic mouse: Role of serotonin and relevance
function: Adaptive gain and optimal performance. to Down syndrome. Neuropsychopharmacology,
Annual Review of Neuroscience, 28, 403–450. 28(10), 1810–1816.
144 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Ben-Yakov, A., Dudai, Y., & Mayford, M. R. (2015). rodent hippocampus is essential for nonspatial
Memory retrieval in mice and men. Cold Spring object memory. Current Biology, 23(17),
Harbor Perspectives in Biology, 7(12). doi: 1685–1690.
10.1101/cshperspect.a021790 Cole, B. J., Robbins, T. W., & Everitt, B. J. (1988).
Bermudez-Rattoni, F. (2014). The forgotten insular Lesions of the dorsal noradrenergic bundle simul-
cortex: Its role on recognition memory formation. taneously enhance and reduce responsivity to
Neurobiology of Learning and Memory, 109, novelty in a food preference test. Brain Research,
207–216. 472(4), 325–349.
Bermudez-Rattoni, F., Okuda, S., Roozendaal, B., & David, O., Barrera, I., Chinnakkaruppan, A., Kaphzan,
McGaugh, J. L. (2005). Insular cortex is involved in H., Nakazawa, T., Yamamoto, T., & Rosenblum, K.
consolidation of object recognition memory. (2014). Dopamine-induced tyrosine phosphoryla-
Learning & Memory, 12(5), 447–449. tion of NR2B (Tyr1472) is essential for ERK1/2
Bisaz, R., Travaglia, A., & Alberini, C. M. (2014). The activation and processing of novel taste informa-
neurobiological bases of memory formation: From tion. Frontiers in Molecular Neuroscience, 7. doi:
physiological conditions to psychopathology. Psy- 10.3389/fnmol.2014.00066
chopathology, 47(6), 347–356. De Jaeger, X., Cammarota, M., Prado, M. A. M.,
Blackford, J. U., Buckholtz, J. W., Avery, S. N., & Zald, Izquierdo, I., Prado, V. F., & Pereira, G. S. (2013).
D. H. (2010). A unique role for the human amyg- Decreased acetylcholine release delays the consoli-
dala in novelty detection. NeuroImage, 50(3), dation of object recognition memory. Behavioural
1188–1193. Brain Research, 238, 62–68.
Borsini, F., & Rolls, E. T. (1984). Role of noradrenaline de Lima, M. N. M., Presti-Torres, J., Dornelles, A.,
and serotonin in the basolateral region of the Scalco, F. S., Roesler, R., Garcia, V. A., & Schröder,
amygdala in food preferences and learned taste N. (2011). Modulatory influence of dopamine
aversions in the rat. Physiology & Behavior, 33(1), receptors on consolidation of object recognition
37–43. memory. Neurobiology of Learning and Memory,
Bortolozzi, A., Díaz-Mataix, L., Scorza, M. C., Celada, 95(3), 305–310.
P., & Artigas, F. (2005). The activation of 5-HT De Luca, M. A. (2014). Habituation of the responsive-
receptors in prefrontal cortex enhances dopamin- ness of mesolimbic and mesocortical dopamine
ergic activity. Journal of Neurochemistry, 95(6), transmission to taste stimuli. Frontiers in Integra-
1597–1607. tive Neuroscience, 8. doi: 10.3389/
Broadbent, N. J., Gaskin, S., Squire, L. R., & Clark, R. fnint.2014.00021
E. (2010). Object recognition memory and the Degroot, A., Wolff, M. C., & Nomikos, G. G. (2005).
rodent hippocampus. Learning & Memory, 17(1), Acute exposure to a novel object during consolida-
5–11. tion enhances cognition. Neuroreport, 16(1),
Brown, M. W., & Aggleton, J. P. (2001). Recognition 63–67.
memory: What are the roles of the perirhinal Dennis, S. H., Pasqui, F., Colvin, E. M., Sanger, H.,
cortex and hippocampus? Nature Reviews Neuro- Mogg, A. J., Felder, C. C., … Mellor, J. R. (2016).
science, 2(1), 51–61. Activation of muscarinic M1 acetylcholine recep-
Burns, L. H., Everitt, B. J., & Robbins, T. W. (1994). tors induces long-term potentiation in the hip-
Intra-amygdala infusion of the N-methyl-D-aspar- pocampus. Cerebral Cortex, 26(1), 414–426.
tate receptor antagonist AP5 impairs acquisition Díaz-Mataix, L., Scorza, M. C., Bortolozzi, A., Toth,
but not performance of discriminated approach to M., Celada, P., & Artigas, F. (2005). Involvement of
an appetitive CS. Behavioral and Neural Biology, 5-HT1A receptors in prefrontal cortex in the mod-
61(3), 242–250. ulation of dopaminergic activity: Role in atypical
Carleton, A., Accolla, R., & Simon, S. A. (2010). antipsychotic action. Journal of Neuroscience,
Coding in the mammalian gustatory system. 25(47), 10831–10843.
Trends in Neurosciences, 33(7), 326–334. Dornelles, A., de Lima, M. N. M., Grazziotin, M.,
Ceccarelli, I., Casamenti, F., Massafra, C., Pepeu, G., Presti-Torres, J., Garcia, V. A., Scalco, F. S., Roesler,
Scali, C., & Aloisi, A. M. (1999). Effects of novelty R., & Schröder, N. (2007). Adrenergic enhance-
and pain on behavior and hippocampal extracel- ment of consolidation of object recognition
lular ACh levels in male and female rats. Brain memory. Neurobiology of Learning and Memory,
Research, 815(2), 169–176. 88(1), 137–142.
Clark, E. W., & Bernstein, I. L. (2009). Boosting cho- Dudai, Y. (2004). The neurobiology of consolidations,
linergic activity in gustatory cortex enhances the or, how stable is the engram? Annual Review of
salience of a familiar CS in taste aversion learning. Psychology, 55, 51–86.
Behavioral Neuroscience, 123(4), 764–771. Dunn, L. T., & Everitt, B. J. (1987). The effects of
Cohen, S. J., Munchow, A. H., Rios, L. M., Zhang, G., lesions to noradrenergic projections from the locus
Asgeirsdóttir, H. N., & Stackman, R. W. (2013). The coeruleus and lateral tegmental cell groups on
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 145

conditioned taste aversion in the rat. Behavioral Furini, C. R., Rossato, J. I., Bitencourt, L. L., Medina,
Neuroscience, 101(3), 409–422. J. H., Izquierdo, I., & Cammarota, M. (2010). Beta-
Duszkiewicz, A. J., McNamara, C. G., Takeuchi, T., & adrenergic receptors link NO/sGC/PKG signaling to
Genzel, L. (2019). Novelty and dopaminergic BDNF expression during the consolidation of
modulation of memory persistence: A tale of two object recognition long-term memory. Hippocam-
systems. Trends in Neurosciences, 42(2), pus, 20(5), 672–683.
102–114. Garcia-Delatorre, P., Pérez-Sánchez, C., Guzmán-
Düzel, E., Bunzeck, N., Guitart-Masip, M., & Düzel, S. Ramos, K., & Bermúdez-Rattoni, F. (2014). Role of
(2010). Novelty-related motivation of anticipation glutamate receptors of central and basolateral
and exploration by dopamine (NOMAD): Implica- amygdala nuclei on retrieval and reconsolidation
tions for healthy aging. Neuroscience and Biobe- of taste aversive memory. Neurobiology of Learn-
havioral Reviews, 34(5), 660–669. ing and Memory, 111, 35–40.
Ennaceur, A., & Delacour, J. (1988). A new one-trial Giovannini, M. G., Bartolini, L., Kopf, S. R., & Pepeu,
test for neurobiological studies of memory in rats. G. (1998). Acetylcholine release from the frontal
1: Behavioral data. Behavioural Brain Research, cortex during exploratory activity. Brain Research,
31(1), 47–59. 784(1-2), 218–227.
Evans, L. H., & Wilding, E. L. (2012). Recollection and Giovannini, M. G., Rakovska, A., Benton, R. S., Paz-
familiarity make independent contributions to zagli, M., Bianchi, L., & Pepeu, G. (2001). Effects
memory judgments. Journal of Neuroscience, of novelty and habituation on acetylcholine,
32(21), 7253–7257. GABA, and glutamate release from the frontal
Feenstra, M. G. P., & Botterblom, M. H. A. (1996). cortex and hippocampus of freely moving rats.
Rapid sampling of extracellular dopamine in the Neuroscience, 106(1), 43–53.
rat prefrontal cortex during food consumption, Goelet, P., Castellucci, V. F., Schacher, S., & Kandel, E.
handling and exposure to novelty. Brain Research, R. (1986). The long and the short of long–term
742(1), 17–24. memory: A molecular framework. Nature, 322,
Feenstra, M. G. P., Botterblom, M. H. A., & Masten- 419–422.
broek, S. (2000). Dopamine and noradrenaline Gómez-Chacón, B., Morillas, E., & Gallo, M. (2015).
efflux in the prefrontal cortex in the light and dark Altered perirhinal cortex activity patterns during
period: Effects of novelty and handling and com- taste neophobia and their habituation in aged
parison to the nucleus accumbens. Neuroscience, rats. Behavioural Brain Research, 281,
100(4), 741–748. 245–249.
Figueroa-Guzmán, Y., Kuo, J. S., & Reilly, S. (2006). Gutiérrez, R., De la Cruz, V., Rodriguez-Ortiz, C. J., &
NMDA receptor antagonist MK-801 infused into Bermudez-Rattoni, F. (2004). Perirhinal cortex
the insular cortex prevents the attenuation of gus- muscarinic receptor blockade impairs taste recog-
tatory neophobia in rats. Brain Research, 1114(1), nition memory formation. Learning & Memory,
183–186. 11(1), 95–101.
Figueroa-Guzmán, Y., & Reilly, S. (2008). NMDA Gutiérrez, R., Téllez, L. A., & Bermúdez-Rattoni, F.
receptors in the basolateral amygdala and gusta- (2003). Blockade of cortical muscarinic but not
tory neophobia. Brain Research, 1210, 200–203. NMDA receptors prevents a novel taste from
Frankland, P. W., Josselyn, S. A., & Köhler, S. (2019). becoming familiar. European Journal of Neurosci-
The neurobiological foundation of memory ence, 17(8), 1556–1562.
retrieval. Nature Neuroscience, 22(10), Guzmán-Ramos, K., Moreno-Castilla, P., Castro-Cruz,
1576–1585. M., McGaugh, J. L., Martínez-Coria, H., LaFerla, F.
Frey, U., & Morris, R. G. (1998). Synaptic tagging: M., & Bermúdez-Rattoni, F. (2012a). Restoration of
Implications for late maintenance of hippocampal dopamine release deficits during object recogni-
long-term potentiation. Trends in Neurosciences, tion memory acquisition attenuates cognitive
21(5), 181–188. impairment in a triple transgenic mice model of
Furini, C. R. G., Myskiw, J. C., Schmidt, B. E., Mar- Alzheimer’s disease. Learning & Memory, 19(10),
condes, L. A., & Izquierdo, I. (2014). D1 and D5 453–460.
dopamine receptors participate on the consolida- Guzmán-Ramos, K., Osorio-Gómez, D., Moreno-
tion of two different memories. Behavioural Brain Castilla, P., & Bermúdez-Rattoni, F. (2010). Off-line
Research, 271, 212–217. concomitant release of dopamine and glutamate
Furini, C. R. G., Myskiw, J. de C., Schmidt, B. E., Zinn, involvement in taste memory consolidation. Jour-
C. G., Peixoto, P. B., Pereira, L. D., & Izquierdo, I. nal of Neurochemistry, 114(1), 226–236.
(2015). The relationship between protein synthesis Guzmán-Ramos, K., Osorio-Gómez, D., Moreno-
and protein degradation in object recognition Castilla, P., & Bermúdez-Rattoni, F. (2012b). Post-
memory. Behavioural Brain Research, 294, acquisition release of glutamate and
17–24. norepinephrine in the amygdala is involved in
146 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

taste-aversion memory consolidation. Learning & consumption of a palatable meal. Neuroscience,


Memory, 19(6), 231–238. 62(4), 1049–1056.
Haettig, J., Stefanko, D. P., Multani, M. L., Figueroa, Jones, B. C., Hou, X., & Cook, M. N. (1996). Effect of
D. X., McQuown, S. C., & Wood, M. A. (2011). exposure to novelty on brain monoamines in
HDAC inhibition modulates hippocampus- C57BL/6 and DBA/2 mice. Physiology & Behavior,
dependent long-term memory for object location 59(2), 361–367.
in a CBP-dependent manner. Learning & Memory, Kafkas, A., & Montaldi, D. (2014). Two separate, but
18(2), 71–79. interacting, neural systems for familiarity and nov-
Hasselmo, M. E. (2006). The role of acetylcholine in elty detection: A dual-route mechanism. Hip-
learning and memory. Current Opinion in Neuro- pocampus, 24(5), 516–527.
biology, 16(6), 710–715. Kafkas, A., & Montaldi, D. (2018). How do memory
Henson, R. N., & Gagnepain, P. (2010). Predictive, systems detect and respond to novelty? Neurosci-
interactive multiple memory systems. Hippocam- ence Letters, 680, 60–68.
pus, 20(11), 1315–1326. Kelly, A., Laroche, S., & Davis, S. (2003). Activation of
Hernández-Ramírez, S., Osorio-Gómez, D., Escobar, mitogen-activated protein kinase/extracellular sig-
M. L., Rodríguez-Durán, L., Velasco, M., Bermú- nal-regulated kinase in hippocampal circuitry is
dez-Rattoni, F., Hiriart, M., & Guzmán-Ramos, K. required for consolidation and reconsolidation of
R. (2021). Catecholaminergic stimulation restores recognition memory. Journal of Neuroscience,
high-sucrose diet-induced hippocampal dysfunc- 23(12), 5354–5360.
tion. Psychoneuroendocrinology, 127. doi: Kempadoo, K. A., Mosharov, E. V., Choi, S. J., Sulzer,
10.1016/j.psyneuen.2021.105178 D., & Kandel, E. R. (2016). Dopamine release from
Holz, R. W., & Fisher, S. K. (2012). Synaptic Transmis- the locus coeruleus to the dorsal hippocampus
sion and Cellular Signaling: An Overview. In S. T. promotes spatial learning and memory. Proceed-
Brady, G. J. Siegel, R. W. Albers, & D. L. Price ings of the National Academy of Sciences of the
(Eds.), Basic neurochemistry (8th ed.): Principles of United States of America, 113(51),
molecular, cellular, and medical neurobiology (pp. 14835–14840.
235–257). Amsterdam: Academic. Kishiyama, M. M., & Yonelinas, A. P. (2003). Novelty
Hotte, M., Naudon, L., & Jay, T. M. (2005). Modula- effects on recollection and familiarity in recogni-
tion of recognition and temporal order memory tion memory. Memory & Cognition, 31(7),
retrieval by dopamine D1 receptor in rats. Neuro- 1045–1051.
biology of Learning and Memory, 84(2), 85–92. Kwapis, J. L., Alaghband, Y., Keiser, A. A., Dong, T.
Houpt, T. A., Philopena, J. M., Wessel, T. C., Joh, T. N., Michael, C. M., Rhee, D., … Wood, M. A.
H., & Smith, G. P. (1994). Increased c-Fos expres- (2020). Aging mice show impaired memory updat-
sion in nucleus of the solitary tract correlated with ing in the novel OUL updating paradigm. Neu-
conditioned taste aversion to sucrose in rats. Neu- ropsychopharmacology, 45(2), 337–346.
roscience Letters, 172(1-2), 1–5. Lee, J. L. C. (2008). Memory reconsolidation medi-
Hunsaker, M. R., Rogers, J. L., & Kesner, R. P. (2007). ates the strengthening of memories by additional
Behavioral characterization of a transection of learning. Nature Neuroscience, 11(11),
dorsal CA3 subcortical efferents: Comparison with 1264–1266.
scopolamine and physostigmine infusions into Lee, J. L. C., Nader, K., & Schiller, D. (2017). An
dorsal CA3. Neurobiology of Learning and update on memory reconsolidation updating.
Memory, 88(1), 127–136. Trends in Cognitive Sciences, 21(7), 531–545.
Hyman, S. E. (2005). Neurotransmitters. Current Biol- Legault, M., & Wise, R. A. (2001). Novelty-evoked
ogy, 15(5), R154–R158. elevations of nucleus accumbens dopamine:
Ihalainen, J. A., Riekkinen, P., & Feenstra, M. G. Dependence on impulse flow from the ventral
(1999). Comparison of dopamine and noradrena- subiculum and glutamatergic neurotransmission in
line release in mouse prefrontal cortex, striatum the ventral tegmental area. European Journal of
and hippocampus using microdialysis. Neurosci- Neuroscience, 13(4), 819–828.
ence Letters, 277(2), 71–74. Leonibus, E. D., Verheij, M. M. M., Mele, A., & Cools,
Ihalainen, J., Sarajärvi, T., Kemppainen, S., Keski- A. (2006). Distinct kinds of novelty processing dif-
Rahkonen, P., Lehtonen, M., & Tanila, H. (2010). A ferentially increase extracellular dopamine in dif-
novel delayed non-match to sample object recog- ferent brain regions. European Journal of
nition task that allows simultaneous in vivo micro- Neuroscience, 23(5), 1332–1340.
dialysis. Journal of Neuroscience Methods, 189(2), Lin, J.-Y., Roman, C., Arthurs, J., & Reilly, S. (2012).
210–215. Taste neophobia and c-Fos expression in the rat
Inglis, F. M., Day, J. C., & Fibiger, H. C. (1994). brain. Brain Research, 1448, 82–88.
Enhanced acetylcholine release in hippocampus Lisman, J., Grace, A. A., & Duzel, E. (2011). A neo-
and cortex during the anticipation and Hebbian framework for episodic memory; role of
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 147

dopamine-dependent late LTP. Trends in Neuro- neuroscience (Vol. 27, pp. 403-413). London:
sciences, 34(10), 536–547. Elsevier/Academic.
Mandler, G. (1980). Recognizing: The judgment of Moreno-Castilla, P., Pérez-Ortega, R., Violante-Soria,
previous occurrence. Psychological Review, 87(3), V., Balderas, I., & Bermúdez-Rattoni, F. (2017). Hip-
252–271. pocampal release of dopamine and norepineph-
Manns, J. R., Stark, C. E. L., & Squire, L. R. (2000). rine encodes novel contextual information.
The visual paired-comparison task as a measure of Hippocampus, 27(5), 547–557.
declarative memory. Proceedings of the National Moreno-Castilla, P., Rodriguez-Duran, L. F., Guzman-
Academy of Sciences of the United States of Ramos, K., Barcenas-Femat, A., Escobar, M. L., &
America, 97(22), 12375–12379. Bermudez-Rattoni, F. (2016). Dopaminergic neuro-
Matsuyama, S., Matsumoto, A., Enomoto, T., & Nishi- transmission dysfunction induced by amyloid-β
zaki, T. (2000). Activation of nicotinic acetylcholine transforms cortical long-term potentiation into
receptors induces long-term potentiation in vivo in long-term depression and produces memory
the intact mouse dentate gyrus. European Journal impairment. Neurobiology of Aging, 41, 187–199.
of Neuroscience, 12(10), 3741–3747. Morici, J. F., Bekinschtein, P., & Weisstaub, N. V.
McGaugh, J. L. (2000). Memory: A century of con- (2015). Medial prefrontal cortex role in recognition
solidation. Science, 287, 248–251. memory in rodents. Behavioural Brain Research,
McGaugh, J. L. (2013). Making lasting memories: 292, 241–251.
Remembering the significant. Proceedings of the Mumby, D. G. (2001). Perspectives on object-recog-
National Academy of Sciences of the United States nition memory following hippocampal damage:
of America, 110(S2), 10402–10407. Lessons from studies in rats. Behavioural Brain
McGaugh, J. L. (2015). Consolidating memories. Research, 127(1-2), 159–181.
Annual Review of Psychology, 66, 1–24. Mumby, D. G., Gaskin, S., Glenn, M. J., Schramek, T.
McIntyre, C. K., McGaugh, J. L., & Williams, C. L. E., & Lehmann, H. (2002). Hippocampal damage
(2012). Interacting brain systems modulate and exploratory preferences in rats: Memory for
memory consolidation. Neuroscience and Biobe- objects, places, and contexts. Learning & Memory,
havioral Reviews, 36(7), 1750–1762. 9(2), 49–57.
Mello-Carpes, P. B., da Silva de Vargas, L., Gayer, M. Muscat, R., Papp, M., & Willner, P. (1992). Antide-
C., Roehrs, R., & Izquierdo, I. (2016). Hippocampal pressant-like effects of dopamine agonists in an
noradrenergic activation is necessary for object animal model of depression. Biological Psychiatry,
recognition memory consolidation and can pro- 31(9), 937–946.
mote BDNF increase and memory persistence. Myskiw, J. C., Rossato, J. I., Bevilaqua, L. R. M.,
Neurobiology of Learning and Memory, 127, Medina, J. H., Izquierdo, I., & Cammarota, M.
84–92. (2008). On the participation of mTOR in recogni-
Merkow, M. B., Burke, J. F., & Kahana, M. J. (2015). tion memory. Neurobiology of Learning and
The human hippocampus contributes to both the Memory, 89(3), 338–351.
recollection and familiarity components of recog- Nadel, L., & Hardt, O. (2011). Update on memory
nition memory. Proceedings of the National Acad- systems and processes. Neuropsychopharmacol-
emy of Sciences of the United States of America, ogy, 36(1), 251–273.
112(46), 14378–14383. Nader, K., Schafe, G. E., & Le Doux, J. E. (2000). Fear
Miranda, M. I., Ferreira, G., Ramirez-Lugo, L., & memories require protein synthesis in the amyg-
Bermudez-Rattoni, F. (2002). Glutamatergic activ- dala for reconsolidation after retrieval. Nature,
ity in the amygdala signals visceral input during 406, 722–726.
taste memory formation. Proceedings of the Osorio-Gómez, D., Bermúdez-Rattoni, F., & Guzmán-
National Academy of Sciences of the United States Ramos, K. R. (2021). Cortical neurochemical sign-
of America, 99(17), 11417–11422. aling of gustatory stimuli and their visceral
Miranda, M. I., Ramírez-Lugo, L., & Bermúdez- consequences during the acquisition and consoli-
Rattoni, F. (2000). Cortical cholinergic activity is dation of taste aversion memory. Neurobiology of
related to the novelty of the stimulus. Brain Learning and Memory, 181. doi: 10.1016/j.
Research, 882(1-2), 230–235. nlm.2021.107437
Montague, P. R., Hyman, S. E., & Cohen, J. D. (2004). Osorio-Gómez, D., Guzmán-Ramos, K., & Bermúdez-
Computational roles for dopamine in behavioural Rattoni, F. (2016). Differential involvement of glu-
control. Nature, 431, 760–767. tamatergic and catecholaminergic activity within
Moreno-Castilla, P., Guzman-Ramos, K., & Bermu- the amygdala during taste aversion retrieval on
dez-Rattoni, F. (2018). Object recognition and memory expression and updating. Behavioural
object location recognition memory: The role of Brain Research, 307, 120–125.
dopamine and noradrenaline. In A. Ennaceur & M. Osorio-Gómez, D., Guzmán-Ramos, K., & Bermúdez-
A. de Souza Silva (Eds.), Handbook of behavioral Rattoni, F. (2017). Memory trace reactivation and
148 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

behavioral response during retrieval are differen- Rodriguez-Ortiz, C. J., Garcia-DeLaTorre, P., Benavi-
tially modulated by amygdalar glutamate recep- dez, E., Ballesteros, M. A., & Bermudez-Rattoni, F.
tors activity: Interaction between amygdala and (2008). Intrahippocampal anisomycin infusions
insular cortex. Learning & Memory, 24(1), 14–23. disrupt previously consolidated spatial memory
Osorio-Gómez, D., Guzmán-Ramos, K., & Bermúdez- only when memory is updated. Neurobiology of
Rattoni, F. (2018). Neurobiology of neophobia and Learning and Memory, 89(3), 352–359.
its attenuation. In S. Reilly (Ed.), Food neophobia: Rojas, S., Diaz-Galarce, R., Jerez-Baraona, J. M.,
Behavioral and biological influences (pp. 111-128). Quintana-Donoso, D., Moraga-Amaro, R., &
Duxford, UK: Elsevier/Woodhead. Stehberg, J. (2015). The insula modulates arousal-
Parker, A., Wilding, E., & Akerman, C. (1998). The induced reluctance to try novel tastes through
Von Restorff effect in visual object recognition adrenergic transmission in the rat. Frontiers in
memory in humans and monkeys: The role of Behavioral Neuroscience, 9. doi: 10.3389/
frontal/perirhinal interaction. Journal of Cognitive fnbeh.2015.00164
Neuroscience, 10(6), 691–703. Roozendaal, B., Barsegyan, A., & Lee, S. (2007).
Peng, Y., Gillis-Smith, S., Jin, H., Tränkner, D., Ryba, Adrenal stress hormones, amygdala activation,
N. J. P., & Zuker, C. S. (2015). Sweet and bitter and memory for emotionally arousing experiences.
taste in the brain of awake behaving animals. In E. R. De Kloet, M. S. Oitzl & E. Vermetten (Eds.),
Nature, 527, 512–515. Progress in brain research: Stress hormones and
Pudovkina, O. L., Kawahara, Y., de Vries, J., & West- post traumatic stress disorder basic studies and
erink, B. H. C. (2001). The release of noradrenaline clinical perspectives (Vol. 167, pp. 79-97). Amster-
in the locus coeruleus and prefrontal cortex stud- dam: Elsevier.
ied with dual-probe microdialysis. Brain Research, Roozendaal, B., & Cools, A. R. (1994). Influence of
906(1), 38–45. the noradrenergic state of the nucleus accumbens
Ramírez-Lugo, L., Zavala-Vega, S., & Bermúdez-Rat- in basolateral amygdala mediated changes in neo-
toni, F. (2006). NMDA and muscarinic receptors of phobia of rats. Behavioral Neuroscience, 108(6),
the nucleus accumbens have differential effects on 1107–1118.
taste memory formation. Learning & Memory, Rossato, J. I., Bevilaqua, L. R. M., Myskiw, J. C.,
13(1), 45–51. Medina, J. H., Izquierdo, I., & Cammarota, M.
Ramírez-Lugo, L., Zavala-Vega, S., Pedroza-Llinas, R., (2007). On the role of hippocampal protein syn-
Núñez-Jaramillo, L., & Bermúdez-Rattoni, F. thesis in the consolidation and reconsolidation of
(2015). Effects of glutamate and its metabotropic object recognition memory. Learning & Memory,
receptors class 1 antagonist in appetitive taste 14(1), 36–46.
memory formation. Behavioural Brain Research, Rossato, J. I., Köhler, C. A., Radiske, A., Lima, R. H.,
284, 213–217. Bevilaqua, L. R. M., & Cammarota, M. (2015).
Ranganath, C., & Rainer, G. (2003). Neural mecha- State-dependent effect of dopamine D1/D5 recep-
nisms for detecting and remembering novel tors inactivation on memory destabilization and
events. Nature Reviews Neuroscience, 4(3), reconsolidation. Behavioural Brain Research, 285,
193–202. 194–199.
Reed, J. M., & Squire, L. R. (1997). Impaired recogni- Royet, J. P., Gervais, R., & Araneda, S. (1983). Effect
tion memory in patients with lesions limited to the of local 6-OHDA and 5,6-DHT injections into the
hippocampal formation. Behavioral Neuroscience, rat olfactory bulb on neophobia and learned aver-
111(4), 667–675. sion to a novel food. Behavioural Brain Research,
Rodríguez-García, G., & Miranda, M. I. (2016). 10(2-3), 297–309.
Opposing roles of cholinergic and GABAergic Sahakian, B. J., Winn, P., Robbins, T. W., Deeley, R. J.,
activity in the insular cortex and nucleus basalis Everitt, B. J., Dunn, L. T., Wallace, M., & James, W.
magnocellularis during novel recognition and P. T. (1983). Changes in body weight and food-
familiar taste memory retrieval. Journal of Neuro- related behaviour induced by destruction of the
science, 36(6), 1879–1889. ventral or dorsal noradrenergic bundle in the rat.
Rodriguez-Ortiz, C. J., & Bermúdez-Rattoni, F. (2017). Neuroscience, 10(4), 1405–1420.
Determinants to trigger memory reconsolidation: Santoyo-Zedillo, M., Rodriguez-Ortiz, C. J., Chavez-
The role of retrieval and updating information. Marchetta, G., Bermudez-Rattoni, F., & Balderas, I.
Neurobiology of Learning and Memory, 142(A), (2014). Retrieval is not necessary to trigger recon-
4–12. solidation of object recognition memory in the
Rodriguez-Ortiz, C. J., De la Cruz, V., Gutiérrez, R., & perirhinal cortex. Learning & Memory, 21(9),
Bermudez-Rattoni, F. (2005). Protein synthesis 452–456.
underlies post-retrieval memory consolidation to a Sara, S. J. (2000). Retrieval and reconsolidation:
restricted degree only when updated information Toward a neurobiology of remembering. Learning
is obtained. Learning & Memory, 12(5), 533–537. & Memory, 7(2), 73–84.
NEUROTRANSMISSION AND NEUROMODULATION OF RECOGNITION MEMORY 149

Sara, S. J., Vankov, A., & Hervé, A. (1994). Locus produced by nicotinic and muscarinic cholinergic
coeruleus-evoked responses in behaving rats: A antagonism in perirhinal cortex. Learning &
clue to the role of noradrenaline in memory. Brain Memory, 18(7), 484–492.
Research Bulletin, 35(5-6), 457–465. Titulaer, J., Björkholm, C., Feltmann, K., Malmlöf, T.,
Schultz, W. (1998). Predictive reward signal of dopa- Mishra, D., … & Konradsson-Geuken, Å. (2021).
mine neurons. Journal of Neurophysiology, 80(1). The importance of ventral hippocampal dopamine
doi: 10.1152/jn.1998.80.1.1 and norepinephrine in recognition memory. Fron-
Schultz, W., Apicella, P., & Ljungberg, T. (1993). tiers in Behavioral Neuroscience, 15. doi: 10.3389/
Responses of monkey dopamine neurons to fnbeh.2021.667244
reward and conditioned stimuli during successive Tucci, S., Rada, P., & Hernandez, L. (1998). Role of
steps of learning a delayed response task. Journal glutamate in the amygdala and lateral hypothala-
of Neuroscience, 13(3), 900–913. mus in conditioned taste aversion. Brain Research,
Sevenster, D., Beckers, T., & Kindt, M. (2014). Predic- 813(1), 44–49.
tion error demarcates the transition from retrieval, Tulving, E. (2002). Episodic memory: From mind to
to reconsolidation, to new learning. Learning & brain. Annual Review of Psychology, 53(1), 1–25.
Memory, 21(11), 580–584. Tulving, E., Markowitsch, H. J., Craik, F. I. M., Habib,
Song, Q., Bolsius, Y. G., Ronzoni, G., Henckens, M. R., & Houle, S. (1996). Novelty and familiarity acti-
J. A. G., & Roozendaal, B. (2021). Noradrenergic vations in PET studies of memory encoding and
enhancement of object recognition and object retrieval. Cerebral Cortex, 6(1), 71–79.
location memory in mice. Stress, 24(2), Vankov, A., Hervé-Minvielle, A., & Sara, S. J. (1995).
181–188. Response to novelty and its rapid habituation in locus
Squire, L. R. (2004). Memory systems of the brain: A coeruleus neurons of the freely exploring rat. Euro-
brief history and current perspective. Neurobiol- pean Journal of Neuroscience, 7(6), 1180–1187.
ogy of Learning and Memory, 82(3), 171–177. Verhagen, J. V., Giza, B. K., & Scott, T. R. (2003).
Squire, L. R. (2009). Memory and brain systems: Responses to taste stimulation in the ventropo-
1969–2009. Journal of Neuroscience, 29(41), steromedial nucleus of the thalamus in rats. Jour-
12711–12716. nal of Neurophysiology, 89(1), 265–275.
Squire, L. R., & Zola, S. M. (1996). Structure and Villain, H., Benkahoul, A., Drougard, A., Lafragette,
function of declarative and nondeclarative memory M., Muzotte, E., Pech, S., … Roullet, P. (2016).
systems. Proceedings of the National Academy of Effects of propranolol, a β-noradrenergic antago-
Sciences of the United States of America, 93(24), nist, on memory consolidation and reconsolidation
13515–13522. in mice. Frontiers in Behavioral Neuroscience, 10.
Stanley, E. M., Wilson, M. A., & Fadel, J. R. (2012). doi: 10.3389/fnbeh.2016.00049
Hippocampal neurotransmitter efflux during one- Wagatsuma, A., Okuyama, T., Sun, C., Smith, L. M.,
trial novel object recognition in rats. Neuroscience Abe, K., & Tonegawa, S. (2018). Locus coeruleus
Letters, 511(1), 38–42. input to hippocampal CA3 drives single-trial learn-
Steketee, J. D., Silverman, P. B., & Swann, A. C. ing of a novel context. Proceedings of the National
(1989). Forebrain norepinephrine involvement in Academy of Sciences of the United States of
selective attention and neophobia. Physiology & America, 115(2), E310–E316.
Behavior, 46(4), 577–583. Warburton, E. C., Koder, T., Cho, K., Massey, P. V.,
Takeuchi, T., Duszkiewicz, A. J., Sonneborn, A., Duguid, G., Barker, G. R. I., … Brown, M. W.
Spooner, P. A., Yamasaki, M., Watanabe, M., … (2003). Cholinergic neurotransmission is essential
Morris, R. G. M. (2016). Locus coeruleus and for perirhinal cortical plasticity and recognition
dopaminergic consolidation of everyday memory. memory. Neuron, 38(6), 987–996.
Nature, 537, 357–362. Winters, B. D., & Bussey, T. J. (2005). Removal of
Tang, Y., Mishkin, M., & Aigner, T. G. (1997). Effects cholinergic input to perirhinal cortex disrupts
of muscarinic blockade in perirhinal cortex during object recognition but not spatial working memory
visual recognition. Proceedings of the National in the rat. European Journal of Neuroscience,
Academy of Sciences of the United States of 21(8), 2263–2270.
America, 94(23), 12667–12669. Winters, B. D., Forwood, S. E., Cowell, R. A., Saksida,
Tian, S., Pan, S., & You, Y. (2015). Nicotine enhances L. M., & Bussey, T. J. (2004). Double dissociation
the reconsolidation of novel object recognition between the effects of peri-postrhinal cortex and
memory in rats. Pharmacology Biochemistry and hippocampal lesions on tests of object recognition
Behavior, 129, 14–18. and spatial memory: Heterogeneity of function
Tinsley, C. J., Fontaine-Palmer, N. S., Vincent, M., within the temporal lobe. Journal of Neuroscience,
Endean, E. P. E., Aggleton, J. P., Brown, M. W., & 24(26), 5901–5908.
Warburton, E. C. (2011). Differing time dependen- Winters, B. D., Tucci, M. C., & DaCosta-Furtado, M.
cies of object recognition memory impairments (2009). Older and stronger object memories are
150 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

selectively destabilized by reactivation in the pres- Current Protocols in Neuroscience, 91(1). doi:
ence of new information. Learning & Memory, 10.1002/cpns.87
16(9), 545–553. Yamamoto, T., Sako, N., Sakai, N., & Iwafune, A.
Winters, B. D., Tucci, M. C., Jacklin, D. L., Reid, J. M., (1997). Gustatory and visceral inputs to the amyg-
& Newsome, J. (2011). On the dynamic nature of dala of the rat: Conditioned taste aversion and
the engram: Evidence for circuit-level reorganiza- induction of c-Fos-like immunoreactivity. Neurosci-
tion of object memory traces following reactivation. ence Letters, 226(2), 127–130.
Journal of Neuroscience, 31(48), 17719–17728. Yonelinas, A. P. (2002). The nature of recollection
Wright, D. S., Bodinayake, K. K., & Kwapis, J. L. and familiarity: A review of 30 years of research.
(2020). Investigating memory updating in mice Journal of Memory and Language, 46(3),
using the objects in updated locations task. 441–517.
10
Neurotransmission and
Neuromodulation of Memory:
Intensity and Valence
María-Isabel Miranda, Kioko Guzmán-Ramos,
Daniel Osorio-Gómez, and
Federico Bermúdez-Rattoni

INTRODUCTION et al., 2014; LeDoux, 2012; Luchicchi et al., 2014;


Picciotto et al., 2012).
Learning is a fundamental function of survival
that induces behavior modifications. It requires
instructive brain circuits that trigger changes in
neural connectivity and adaptive changes in CHOLINERGIC ACTIVITY
behavior in response to environmental/internal
challenges. Numerous studies about aversive ACh activity has long been implicated in attend-
associative learning have used different condition- ing to and remembering emotional experiences.
ing models to shed light on the neural mechanisms The role of ACh signaling in the mnemonic pro-
of the aversive predictive and instructive signaling cessing of highly salient events has been exten-
required for learning and memory. Recently, it has sively described (Dani and Bertrand, 2007;
been proposed that the predictability of the aver- Hasselmo and Sarter, 2011; Luchicchi et al., 2014;
sive experience tightly regulates parallel aversive Sarter et al., 2014; also see Chapter 13 this
signaling in multiple circuits during behavioral volume). ACh release is related to different states
learning (Ozawa and Johansen, 2018). of arousal and associative conditioning (Acquas
Furthermore, during learning, neural circuits et al., 1996; Miranda and Bermudez-Rattoni,
transduce salient experiences (e.g., punishments, 1999; Rasmusson and Szerb, 1976; Sarter and
aversive stimuli, motor errors, among others) into Bruno, 1994). Accordingly, significant increments
instructive neural signals integrated into the of ACh release occur during appetitive condition-
memory circuitries (Schultz, 2015). Therefore, ing,1 acquisition of discrimination performance2
learning and recalling highly aversive or negative (Butt, 1997), and operational tasks3 (Orsetti et al.,
experiences engages distributed brain networks. 1996), among other examples.
This brain network activity is tuned by several In both humans and experimental rodent
neurotransmitters, including acetylcholine (ACh), models, the amygdala and several cortical areas
glutamate, gamma-amino butyric acid (GABA), receive a particularly significant cholinergic pro-
dopamine (DA), and noradrenaline/norepineph- jection arising primarily from the nucleus basalis
rine (NA) (Hasselmo and Sarter, 2011; Hermans magnocellularis (NBM) (Woolf, 1991). In this
152 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

regard, seminal findings indicate that M1 and M2 amygdala (Quirarte et al., 1998). This NA surge
muscarinic receptor activation is necessary for promotes aversive associative learning and
the modulation of memory consolidation induced memory through β-adrenergic receptor activation
by cholinergic influences within the amygdala (Uematsu et al., 2017). Evidence of β-adrenoceptors
(Power et al., 2003). Jiang et al. (2016) corrobo- located on vagal afferents to the nucleus of the
rated by optogenetics4 that endogenous choliner- solitary tract (NTS) (Schreurs et al., 1986) sug-
gic signaling modulates the excitability of putative gests that systemic epinephrine may modulate
amygdala principal neurons via interaction with neural processes through actions mediated by the
both muscarinic and nicotinic ACh receptors NTS. In this regard, epinephrine’s effects on
(Jiang et al., 2016). Thus, ACh modulates cortical– memory consolidation are initiated by peripheral
amygdala synaptic plasticity and influences the β-adrenoceptor activation (McInytire et al., 2006).
acquisition of fear-related behaviors (Jiang et al., Extensive research suggests that the release
2016). These results suggest that the NBM–­ of peripheral adrenal hormones in response to a
cortical neuromodulatory pathway functions in stressor influences memory consolidation by acti-
concert with ACh inputs to the basal amygdala, vating central adrenoceptors (McGaugh, 2000).
which enhances the persistence of fear and aver- NA activity and its interaction with circulating
sive memories, supporting the notion that ACh corticosterone are required during memory con-
modulation plays a crucial role in amygdala-based solidation (Roozendaal et al., 2006). Furthermore,
learning (McGaugh et al., 2002). corticosteroids activate locus coeruleus (LC) pro-
Furthermore, cholinergic activity is required jections to the amygdala, releasing noradrenaline
for encoding spatial and aversive cues during and enhancing anxiety (McCall et al., 2017). This
memory extinction5 or memory updating6 (Knox input may thus be specific to aversive learning. It
and Keller, 2016), indicating that cholinergic basal has also been demonstrated that pharmacological
forebrain inputs also shape responses to aversive stimulation of the NTS enhances aversive mem-
cues, cue context, and extinguished cues. ACh ory consolidation and NA levels in the amygdala
enhances cue detection across multiple regions (Garcia-Medina and Miranda, 2013; Likhtik and
through inhibitory and excitatory currents that Johansen, 2019b).
amplify signal-to-noise ratios. In other words, dis- Anatomical studies indicate that the pon-
tinct cholinergic inputs can convey salient infor- tine tegmentum and NTS could activate the LC;
mation to the amygdala and promote associative moreover, noradrenergic terminals from the LC
biophysical changes that underlie emotional mem- innervate the amygdala (Robertson et al., 2013;
ories (Aitta-Aho et al., 2018; Pidoplichko et al., Uematsu et al., 2017; Usunoff et al., 2006). These
2013; Unal et al., 2015). Since aversive learning7 findings corroborate a linear contribution of the
occurs during a heightened state of arousal, shift- NTS–LC–amygdala NA pathway in aversive
ing cortical sensory processing alters sensory- learning. In general, NA enhances excitatory sign-
perceptual thresholds and amplifies stimulus aling and neuronal plasticity,8 modulating aversive
generalization (Likhtik and Johansen, 2019b). memory acquisition and consolidation mecha-
ACh intensifies cue encoding during increased nisms (Garcia-Medina et al., 2015; McIntyre
attention at cortical sensory sites that communi- et al., 2003). Considerable evidence also indicates
cate with the cortex and at cortical afferents to the that amygdala activation influences memory by
amygdala (Letzkus et al., 2011; Weinberger et al., regulating consolidation in other brain regions;
2006), suggesting a cortical cholinergic impact thus, projections from the NTS to the amygdala
during aversive discrimination learning. However, are critically involved in mediating adrenergic
the role of ACh in memory cue/context discrimi- influences on memory consolidation (Clayton and
nation requires further research. In summary, Williams, 2000; Miyashita and Williams, 2002;
associations during aversive experiences engage Williams et al., 1998). Interestingly, LC projec-
distributed cholinergic activity. ACh intensifies tion neurons participate in memory processes in
cue encoding during increased attention at corti- different brain areas. For example, LC neurons
cal sensory sites, enhancing the persistence of fear enhance sensory cue responsiveness following
and aversive memories. fear conditioning (Rasmussen and Jacobs, 1986)
since significant increases in NA are evident in the
amygdala after fear conditioning (Quirarte et al.,
1998). Additionally, pharmacological findings
demonstrated that endogenous NA released in the
NORADRENERGIC ACTIVITY amygdala during aversive and emotional arousal
training improves memory consolidation (Ellis
It is well known that aversive experiences produce and Kesner, 1983; Hatfield and McGaugh, 1999;
a surge of NA levels in the brain, including the Liang et al., 1986, 1990).
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 153

According to recent studies, the LC projects 2015; Watabe-Uchida et al., 2017). However,
strongly to the hippocampus, where it can release some DA neurons in the VTA respond to aversive
both NA and DA, which are essential for contex- stimuli or when encoding negative reinforcer
tual and spatial forms of learning (Heath et al., saliency for aversive outcomes (de Jong et al.,
2015; Takeuchi et al., 2016). As mentioned in 2019; Matsumoto and Hikosaka, 2009; Menegas
Chapter 13, presentation of relevant stimuli elicits et al., 2018).
NA release from the LC (Robertson et al., 2013), These findings suggest that midbrain DA
within the insular cortex (IC) (Guzman-Ramos regulates fear learning through projections to the
et al., 2010; Osorio-Gomez et al., 2021). This NA amygdala. The amygdala receives DA projections
activity could be associated with anxiety-related from the VTA, increasing DA release during emo-
responses (Galvez et al., 1996; Quirarte et al., tional memories (Stevenson and Gratton, 2003).
1998). As reviewed, anatomical studies indicate Therefore, DA can regulate aversive learning
that NA neurons innervate several brain regions function by modulating the local synaptic, cellu-
from the LC, which in turn could be activated by lar, and microcircuit mechanisms of the amygdala.
the pontine tegmentum and the NTS. Interestingly, In addition, during emotional/aversive learning,
systemic epinephrine might modulate memory complex neurotransmission interaction is apparent
consolidation by NTS activation. Epinephrine because the amygdala sends a direct glutamater-
exerts its effects on memory consolidation upon gic projection to the nucleus accumbens (NAc)
peripheral β-adrenoceptor activation followed by (Ricardo and Koh, 1978). Moreover, amygdala
NTS stimulation. Furthermore, epinephrine pro- modulation induces a long-lasting increase in DA
motes an increase in NA in brain regions includ- efflux in the NAc, a brain structure that regulates
ing the amygdala, the hippocampus, and the IC, both appetitive and aversively motivated behav-
enhancing aversive memory consolidation. These iors. These behaviors modulate cortical ACh activ-
data confirm the peripheral influence of epi- ity (Howland et al., 2002; Louilot et al., 1985) by
nephrine on memory consolidation through the the activation of DA receptor activity in the NAc
activation of central NA activity. Therefore, stress- (Moore et al., 1999). Besides, during emotional/
induced activation of the NA system is pivotal in aversive learning, complex neurotransmission
enhancing memory consolidation. system interaction is evident given that the amyg-
It has been suggested that LC modulates rhyth- dala contributes to nociceptive sensory informa-
mic activity, promoting neural coding involved in tion by processing negative emotional value, thus
cognitive processes (Swift et al., 2018; Walling supporting the association between an aversive
et al., 2011). This modulation could explain, at response and the environment in which it occurs
least in part, the distributed oscillatory coupling (Ansah et al., 2010; Gao et al., 2007; Kissiwaa
between LC, amygdala, and hippocampus dur- and Bagley, 2018; Tanimoto et al., 2003). The
ing emotional/aversive discrimination learning. amygdala sends a direct glutamatergic projection
Further, the rhythmic activity modulated by NA to the NAc (Ricardo and Koh, 1978), which sug-
has been linked to sleep dysregulation in post- gests that amygdala–NAc interactions may lead
traumatic stress disorder (PTSD), which affects to changes in dopaminergic and glutamatergic
emotional memory (Hendrickson and Raskind, activity related to emotional component process-
2016). Additional studies are needed to under- ing during aversive memory formation (Howland
stand the relationship between sleep modulation et al., 2002; Ramirez-Lugo et al., 2006; Stevenson
and aversive memory formation. and Gratton, 2003, Yamamoto, 2006).
Furthermore, it seems that independent dopa-
minergic populations of VTA inputs to the mPFC
regulate aversive learning (de Jong et al., 2019;
Lammel et al., 2012; Likhtik and Johansen, 2019a;
DOPAMINERGIC ACTIVITY Stubbendorff et al., 2019). Similarly, another set
of dopaminergic neurons respond to unexpected
Different populations of dopaminergic neurons aversive events and the cues that predict them.
project to individual target sites (Likhtik and These VTA DA neurons participate in aversive
Johansen, 2019b). For instance, dopamine (DA) processes including those projecting to the ventro-
neuron populations in the ventral tegmental area medial shell of the NAc, substantia nigra and stria-
(VTA) project to the medial prefrontal cortex tum (de Jong et al., 2019; Menegas et al., 2018).
(mPFC), striatum and amygdala (Oades and As mentioned, recent work suggests that the LC
Halliday, 1987). Some of these neurons integrate releases DA in the hippocampus to modulate some
reward prediction error,9 responding primordi- forms of contextual learning since VTA projec-
ally to unexpected rewards as an instructive tions to the hippocampus are sparse (Broussard
signal for appetitive learning (Keiflin and Janak, et al., 2016; Takeuchi et al., 2016).
154 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

DA is involved in reward prediction error, unex- stimuli associated with the environment. Evidence
pected rewards, and instructive signals for appeti- indicates that the anterior cingulate cortex pro-
tive memory formation, as well as in processing jection to the amygdala is involved in descend-
negative emotional value and nociceptive sensory ing pain modulation (Fuchs et al., 2014) and the
information. Thus, DA contributes to the associa- cognitive-affective component of pain (Johansen
tion between an aversive response and the envi- et al., 2001).
ronment in which it occurs. It should also be noted As we will describe, the IC plays a crucial role
that amygdala and NAc interactions may lead to in the processing of interoceptive information such
changes in dopaminergic and glutamatergic activ- as noxious stimulation intensity. Interestingly,
ity related to emotional/aversive processing during using conditioned taste aversion11 as an aversively
memory formation. motivated learning model, studies have dem-
onstrated that novel taste stimuli fail to induce
changes in glutamate levels. However, glutamate
release is observed in the IC and amygdala with
gastric malaise induction (i.e., LiCl but not NaCl)
GLUTAMATERGIC ACTIVITY (Guzman-Ramos et al., 2010, 2012; Miranda et al.,
2002). Conversely, NaCl and LiCl administration
Nociception10 in all species is an essential defense elicits cortical NA release (Guzman-Ramos et al.,
against dangers in the environment. Accordingly, 2010; Osorio-Gomez et al., 2017). However, in
the experience of pain consists of somatosensory experiments modifying the LiCl and NaCl concen-
elements of intensity and location, negative emo- trations, researchers observed that the changes in
tional/aversive “feelings,” and subsequent nega- NA release were related to the intensity of the peri-
tive behavior because of a learned association toneal irritation. In contrast, glutamate was related
between specific events and pain (i.e., association to the gastric irritation induced by LiCl regardless
of aversive and nociceptive components). This of its concentration. Therefore, these results sug-
associative learning integrates polymodal sensory gest that glutamate could be associated with the
information and nociceptive information processing of gastric malaise by gastric irritation,
(Mareket al., 2013; Moga et al., 1995; Neugebauer, and NA could be related to the signaling of the
2015; Pape and Pare, 2010; Sah et al., 2003; noxious salt intensity (Osorio-Gomez et al., 2017).
Vertes and Hoover, 2008).
Evidence indicates that the central amygdala
promotes aversive association. It receives noci-
ceptive information via the spino-parabrachial-
amygdaloid pathway and polymodal sensory
PEPTIDES AND ENDOCANNABINOID
information via cortical and thalamic inputs SYSTEMS
(Bernard et al., 1992, 1993; Bester et al., 1997).
Nevertheless, the final amygdala circuitry could Regarding pain modulation and learning, a recent
be required for pain-induced associative learn- study suggests that the endocannabinoid system in
ing (Han et al., 2015; Sato et al., 2015; Watabe the mPFC plays a key role in mediating the
et al., 2013). Both nociceptive stimulus and previ- expression of fear-conditioned analgesia and con-
ously repeated subthreshold stimuli trigger gluta- ditioned fear in the presence of nociceptive tone
mate release from the parabrachial terminals. In (Rea et al., 2019). The results of that study are
addition, nociceptive amygdala neurons increase consistent with evidence that cannabinoid (CB1)
responsiveness, enhancing the aversive response receptors are essential in mediating the expression
and the association between nociception and con- of endogenous analgesia (e.g., delineating the
text (Baliki et al., 2006; Bornhovd et al., 2002; array of actions that the central nervous system
Cardinal et al., 2002; LeDoux, 2000) can use to reduce or, at times, augment pain) to
The mechanism of nociception association is unconditioned aversive stimuli (Freitas et al.,
not well described; however, some studies suggest 2013). However, the potential function of the can-
that it may be related to an increase in postsynap- nabinoid system during the association of aversive
tic AMPA receptors (Cheng et al., 2011) that could and nociceptive components has not been com-
induce plasticity reeling upon the synthesis of new pletely elucidated.
proteins. These postsynaptic changes are similar The endogenous opioid system is involved in
to those observed during stress when corticoster- fear learning in rodents. It has been suggested that
one produces an increment of AMPA receptors an opioidergic transmitter signal, engaged through
(Groc et al., 2008; Martin et al., 2009). However, conditioning, acts inhibitory on unconditioned
it is still unclear whether additional neurochemical stimulus input (Eippert et al., 2008). Blockade of
changes in the amygdala participate in nociceptive endogenous opioid neurotransmission enhances
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 155

the acquisition of conditioned fear in humans anticipation, and defined by some as a “state of
(Eippert et al., 2008). Opioid receptors and pep- readiness for a reward.” Moreover, during feed-
tides are expressed to varying degrees throughout back based on rewards and learning, a proper
the amygdala (Poulin et al., 2008). Therefore, opi- sequence of events is required for balanced inte-
oidergic neurotransmission impacts not only on gration between the expected value of a given
aversive stimulus processing (Calcagnetti et al., stimulus and the predicted effort to obtain a
1987; Fanselow et al., 1988) but also on appe- reward, thereby emitting an adequate motivation
titive conditioned associations (e.g., acquisition) to initiate goal-directed behavior. The consumma-
(Davis, 1979; McNally et al., 2004; Westbrook tory phase of reward processing occurs when the
et al., 1991; Young and Fanselow, 1992). Based goal is achieved, leading to the hedonic response
on neurophysiological simulations, Padlubnaya (Kring and Barch, 2014; Rizvi et al., 2016).
and coworkers (2006) suggested that opioids can-
not exert their function alone, but need to work in
concert with a faster-acting inhibitory neurotrans-
mitter such as GABA (Padlubnaya et al., 2006).
Moreover, human studies suggest that the endog- SYSTEMS THAT REGULATE REWARD
enous opioid system has an inhibitory role in fear
acquisition and is differentially involved in aver- Several systems that regulate reward and affective
sive learning. The effect of endogenous opioids processing also interact during associative learn-
may attenuate aversive/nociceptive impact via ing and memory retrieval. Nevertheless, signifi-
conditioned hypoalgesia. They might also directly cantly less is known about the role of reward and
regulate prediction errors during aversive learning hedonic processing in learning and memory.
(e.g., conditioned fear) (Eippert et al., 2008). So Manipulation of numerous neurotransmitter path-
far, we can conclude that the opioidergic transmit- ways can simultaneously impact food intake and
ter signal could act as an inhibitory input of the reward. The main systems controlling homeostatic
unconditioned stimulus. Thus, the blockade of and hedonic feeding are often treated as independ-
endogenous opioid neurotransmission enhances ent because homeostatic feeding is necessary for
the acquisition of conditioned fear memory. basic metabolic processes and survival. In com-
parison, hedonic feeding is driven by sensory
perception or pleasure/hedonic stimulation.
Nonetheless, functional and anatomical aspects
of homeostatic and hedonic feeding overlap,
VALENCE ASSOCIATION OF REWARDING which indicates a significant interaction that has
HEDONIC COMPONENTS: EMOTIONAL been greatly overlooked in learning research. Non-
human animal research points to opioid modula-
BALANCE tion of cognitive and decision-making processes
related to hedonic components. This emerging
The motivation to pursue happiness or pleasure evidence provides insight into how acute modula-
impacts greatly on our daily life behaviors. tion of opioids can influence cognitive function.
Integral hedonic capacity requires undisturbed Opioid receptors are distributed throughout the
interplay between several brain regions and neuro- brain, including the more “cognitive” regions in
transmitter systems, including those underlying the frontal and parietal lobes (van Steenbergen
associative learning (Hoflich et al., 2019). It et al., 2019). Reports also highlight the impor-
should be noted that there is growing evidence on tance of the opioid system in regulating not just
the difference between understanding “pleasure” aversive experiences but also motivation and the
in humans and the hedonic response in animals sense of hedonic impact (e.g., “liking”12) (Baldo,
during rewarding events (Berridge and 2016; Pecina and Smith, 2010). In this regard, sev-
Kringelbach, 2008). However, reward, hedonic eral neural circuits thought to orchestrate feeding
response, and pleasure are complex terms that behavior overlap with the reward circuitry (Rossi
involve various subcomponents such as incentive, and Stuber, 2018).
motivation, and learning processing to obtain/
perceive wellness in response to a stimulus or
event and subsequently to remember it.
Accordingly, reward processing is a multi-step
phenomenon initiated by forming an association AVERSIVE LEARNING TASKS
between a given stimulus and a related reward.
After learning, it is followed by the development In the presence of aversive stimuli, blocking
or increase of attention, motivation and/or opioid signaling can enhance conditioned
156 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 10.1 A hypothetical scheme summarizes the interactions in a cortical synapse, where
stimuli with different valence and intensity can be integrated during memory formation and/
or complex retrieval associations.

responses in relation to learning tasks (Eippert Furthermore, “hedonic spots” involved in sweet
et al., 2008; Haaker et al., 2017). Reports agree taste “liking” responses also have been described
with the idea that opioid peptide neurotransmis- in the rat insula and prefrontal cortices (Castro
sion causes a shift in the valuation of “hedonic and Berridge, 2017). In this regard, rodent research
gradient,” ranging from displeasure to pleasure, has argued convincingly for opioid involvement in
which is not limited to the liking of stimuli. For choice behaviors beyond the regulation of rein-
example, micro-stimulation with opioid peptides forcement and aversion (Laurent et al., 2015).
has been shown to increase motivation for differ- However, the role of opioid in associative hedonic
ent reward types in rodents (Mahler and Berridge, component processes and emotional memory
2012; Wassum et al., 2009). However, Laurent retrieval has not been clarified.
et al. (2015) also show opioid modulation of Several reports have suggested that the opioid
learning regardless of “liking” responses, suggest- system profoundly affects reward-based decisions
ing a dual role of this peptidergic system during by altering motivational processing and learning
associative learning and hedonic integration (Jacobson et al., 2018). Thus, it is conceivable
(Laurent et al., 2015). Overall, these findings that the opioid system regulates aversive arousal
partly explain that learning and motivation typi- (e.g., during aversive associations), influencing
cally increase together with reward valuation cognitive control that tunes goal-directed behav-
(Berridge et al., 2009). ior (Dreisbach and Fischer, 2015; Valentino and
Van Bockstaele, 2015; van Steenbergen et al.,
2019). Evidence indicates that the endogenous
opioid system is involved in value-based decision-­
making, since blocking opioid receptors may
REWARD LEARNING TASKS reduce motivational value when learning about
high-value stimuli (Berridge and Kringelbach,
In rodents, reward learning can be modulated by 2015; Eikemo et al., 2017; Laurent et al., 2015;
opioid receptor manipulation in several brain Lutz and Kieffer, 2013; van Steenbergen et al.,
regions related to memory processing like the stria- 2019; Weber et al., 2016).
tum and amygdala, among others (Berridge and Opioid activation and inhibition also affect
Kringelbach, 2015; Wassum et al., 2009a,b, 2011). other neurotransmitter systems important for
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 157

cognition, such as DA and NA (Chaijale et al., component processing (Cota et al., 2006; Friemel
2013; Fields and Margolis, 2015; Valentino and et al., 2014; Jarrett et al., 2007; Kirkham, 2009;
Van Bockstaele, 2015). These systems are the Mahler et al., 2007; Solinas et al., 2008), probably
core of reward learning circuits and code reward by close interaction with the endogenous opioid
prediction error (Tian et al., 2016). Nonetheless, system (Cota et al., 2006). Detailed information
mesolimbic and neostriatal DA systems do not about neurocircuits and mechanisms involved in
mediate the hedonic impact of rewarding stimuli the mediation of pleasure during learning is cru-
or hedonic reward learning and associative pre- cial to gain a better a better understanding of
diction (Berridge and Robinson, 1998; Schultz, hedonic processing during emotional behaviors
2006). Conversely, the DA system may mediate (Berridge and Kringelbach, 2008; Esch and
the incentive salience13 of rewards by modulating Stefano, 2004; Kringelbach and Berridge, 2009).
their motivational value separately from hedonic Future research will help to elucidate the emo-
and reward learning. tional modulation that occurs during the associa-
According to recent studies, DA is neither nec- tion of delightful experiences.
essary nor sufficient to mediate changes in hedonic The systems that regulate reward and affective
“liking” for sensory “pleasures” (Berridge, 2000; processing interact during associative learning and
Pecina and Berridge, 2005). Further, DA is not memory retrieval. Thus, current evidence indicates
needed for new learning and is insufficient to that the endogenous opioid system is implicated
mediate learning by directly causing teaching or in determining the motivational value of stimuli
prediction signals. By contrast, growing evidence during learning. In contrast, the DA system may
indicates that DA contributes causally to incen- mediate the incentive salience of rewards, modu-
tive salience (Koob and Volkow, 2016; Lynch and lating their motivational value in a manner separa-
Ryan, 2020). Regardless, the debate continues ble from hedonic and reward learning.
over the precise causal contribution of mesolim-
bic DA systems to reward (Berridge, 2007; Lin
et al., 2021). In this regard, the term “reward”
and its interpretation have various connotations
that are mainly linked to hedonic value, reward CONTEXT COMPLEXITY: EXTEROCEPTIVE
motivation, learning and extinction processes, and AND INTEROCEPTIVE MEMORY
anticipation or expectation of rewarding stimuli
(Barbano and Cador, 2007; Salamone et al., 2007; MODULATION
Schneider et al., 2010). These connotations make
it even more challenging to understand the neu- According to recent studies, contextual changes
rochemical processes involved in the complex could significantly affect learning, retrieval and
association of hedonic “pleasurable” components memory updating (see Chapter 13 this volume),
during learning. suggesting a more complex scenario in any moti-
vational task during stimulus associations that
include opposite or different valences (e.g., a posi-
tive/appetitive or negative/aversive context).
Indeed, everyday learning entails a series of
HEDONIC AND/OR REWARD PROCESSING changes in the context, temporality of experiences
and type of stimuli to be associated. In this regard,
The endocannabinoid system has recently interference paradigms15 in which stimuli are
emerged as another critical mediator for hedonic associated with different stages of learning and
and/or reward processing (Elphick, 2012). It is retrieval can modulate and update the strength of
well known that cannabinoid signaling has a regu- memory performance at any point in the learning
latory influence over several neurotransmitter process associated with contextual cues. For
systems that mediate reward-related behaviors example, testing memory in the same context in
(e.g., DA, glutamate, and endogenous opioids). which the memory was encoded leads to improved
Apparent involvement of cannabinoid signaling in retention relative to testing memory in a new con-
learning processes, neuroplasticity, motivational text (Bouton, 2019). Accordingly, context seems
behavior (e.g., operant conditioning), and reward to play a significant role in interference paradigms
“wanting”14 has been reported (Ameri, 1999; such as memory extinction, countercondition-
Fattore et al., 2010; Solinas et al., 2008). However, ing16, discrimination reversal,17 and latent inhibi-
just recently, studies have suggested that the tion18 (Pearce, 1994; Pearce and Hall, 1980;
endocannabinoid system is central in hedonic Wagner, 2003).
aspects of emotionally related learned behaviors, Context refers to the immediate environment
specifically for the mediation of hedonic in which a memory is encoded (Roberts, 2019).
158 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Therefore, context is commonly defined as the set behavioral evidence demonstrates that the hip-
of background stimuli that comprise the environ- pocampus and glutamatergic activity are crucial
ment during a behavior. Similar stimuli can also for context-based memory (Kim and Fanselow,
become foreground conditioned stimuli, depend- 1992). However, few studies have explored their
ing on the task (De la Casa et al., 2018; Nadel and involvement in context complexity. In this regard,
Willner, 1980). Nevertheless, a suitable general some reports have indicated a role for hippocam-
conceptualization of context should include vari- pal function in the complex taste learning pro-
ous exteroceptive and interoceptive stimuli that cess, such as blocking (Gallo and Cándido, 1995;
are thought to be available in the environment and Moron et al., 2002), and in taste learning tasks
play a key role. Theoretical approaches to inter- that critically depend on contextual information
ference have emphasized the possible influence of (Gallo et al., 1999). The dorsal hippocampus has
many kinds of contextual stimuli (Bouton, 1993, been implicated in both learned taste aversions
2019) that suggest the importance of context dur- to the physical context and the blocking of the
ing learning and memory. context in taste aversion learning (Aguado et al.,
1998) and extinction (Garcia-de la Torre et al.,
2010). Furthermore, dorsal hippocampal integ-
rity is required during the context dependency of
taste aversions and latent inhibition of taste aver-
EXTEROCEPTIVE INFLUENCES ON sion (Baldi and Bucherelli, 2015; Manrique et al.,
CONTEXT 2009a,b; Molero et al., 2005). DA activity has also
been reported in the consolidation of contextual
Standard exteroceptive context includes the room memories in the hippocampus. These reports sug-
and apparatus used during Pavlovian fear learn- gest that DA activity is modulatory in response
ing and context switching in different phases, to environmental novelty within the hippocampal
including extinction (Baldi and Bucherelli, neurons responsible for initial consolidation and
2015). As indicated above, the basal forebrain shifts after environmental novelty, when reward
cholinergic system provides dense neuromodula- expectancy or magnitude also changes.
tory inputs to the basolateral amygdala complex, In reward expectancy,19 DA is required for
targeting pyramidal neurons and interneurons in executing learned actions until they become habit-
this area (Lee and Kim, 2019; Muller et al., ual in a similar context (Kempadoo et al., 2016;
2011). The basolateral amygdala complex is Takeuchi et al., 2016; Yamasaki and Takeuchi,
implicated in numerous cognitive functions, such 2017). Moreover, systemic administration of the
as associative context learning and extinction DA receptor antagonist disrupts context-­dependent
processes. Particularly, cholinergic regulation attenuation of taste neophobia (De la Casa and
has been demonstrated in various studies using Diaz, 2013; Grau-Perales et al., 2019). Despite this
systemic and region-specific pharmacological evidence, the neurotransmission systems involved
manipulations during associative fear learning in these processes have not been clearly described.
and extinction paradigms (Gold, 2003; Gould Various studies suggest that the histaminergic
and Leach, 2014; Knox, 2016; Power et al., 2003; system could play a significant role in regulating
Robinson et al., 2011; Tinsley et al., 2004; contextual learning. Histamine, as a neurotrans-
Wilson and Fadel, 2017). mitter, is synthesized by neurons in the tubero-
Some plastic mechanisms underlying the origi- mammillary nucleus and projects to the whole
nal learning of the contextual or cue-conditioned brain. H1 and H2 potentiate excitatory inputs of
response suggest that extinction learning involves the four histamine receptors, while H3 receptors
disparate neuronal populations and signaling pro- down-regulate histamine synthesis and release
cesses specifically within prefrontal-amygdala other neurotransmitters (Haas and Panula, 2003;
circuits (Baldi and Bucherelli, 2015; Herry et al., Panula et al., 1989). Histamine-induced cholin-
2008; Orsini and Maren, 2012; Rozeske et al., ergic, GABAergic and glutamatergic release, for
2015; Tronson et al., 2009). Studies on the optoge- example, modulates several structures including
netic stimulation of cholinergic inputs and both the supramammillary nucleus, NBM, diagonal
muscarinic and nicotinic cholinergic receptors band nuclei, cerebral cortex, amygdala, and hip-
demonstrate that these circuits exert a significant pocampus (Bacciottini et al., 2002; Baldo, 2016;
influence on cued fear extinction (Aitta-Aho et al., Inagaki et al., 1988; Panula et al., 1989). Thus, his-
2018; Jiang et al., 2016; Unal et al, 2015). tamine controls fundamental aspects of learning,
Moreover, during acquisition, cholinergic ranging from the sleep-wake cycle to nociception
stimulation in the amygdala disrupts the extinc- (Passani et al., 2007). Histamine also modulates
tion, but not the acquisition, of cued fear memo- hippocampal activity during aversive memory
ries (Jiang et al., 2016). On the other hand, robust processing (Brown et al., 2001). In this regard,
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 159

hippocampal histamine receptors are involved Accordingly, Schepers and Bouton (2017)
in fear memory consolidation (Da Silva et al., reported that interoceptive states produced by
2006; Zarrindast et al. 2002). In addition, increas- hunger and satiety could control memory extinc-
ing ACh release in the BLA using histaminergic tion. They observed that the renewed response in
agonists improves the expression of fear memo- the context of satiety went against traditional
ries (Cangioli et al., 2002). Evidence corroborates notions about how behavior is motivated by
that intra-BLA administration of H3 antagonists hunger and satiety. Since the hunger response was
reduces both the freezing time of contextual fear- extinguished during several sessions, the partici-
conditioned rats and local ACh release (Passani pants, which were evaluated individually in both
et al., 2001). Furthermore, histamine decreases satiety and hunger states, exhibited a renewed
cortical ACh release, which has a differential response when retrieval was tested in satiety
impact on taste aversive memory formation and (Schepers and Bouton, 2017).
extinction. For example, an H3 agonist released Furthermore, hunger and satiety states can pro-
into the IC impaired conditioned taste aversive vide discriminative cues for Pavlovian-conditioned
memory and simultaneously decreased the local responses20 (Davidson, 1993; Davidson et al., 2005;
release of ACh (Puron-Sierra and Miranda, 2014). Kanoski et al., 2007; Sample et al., 2016). Similarly,
Conversely, CA1 infusion of the H2 receptor ago- interoceptive cues provided by drug states could
nist dimaprit facilitated the extinction of step- modify extinction performance (Bouton et al.,
down inhibitory avoidance (Bonini et al., 2011), 1990; Cunningham, 1979; Lattal, 2007). These
sustaining the hypothesis that histaminergic activ- findings indicate that associative learning involving
ity is required during context memory extinction. interoceptive stimuli could induce physiological,
Recent data illustrates that the central histamin- emotional, and perceptual changes during retrieval
ergic system is a necessary intermediary modula- and memory updating (Van Diest, 2019).
tor for arranging peripheral homeostatic signals The IC has been implicated in a wide range of
during emotionally salient cognitive processes. conditions and behaviors. It seems that this cor-
It exerts its actions through the recruitment of tex translates and integrates external cues into
emotional cortico-amygdala circuits to generate interoceptive states that regulate physiological
adequate behavioral responses (Provensi et al., and cognitive processes (Craig and Craig, 2009).
2017). Accordingly, the cortico-amygdala circuit Consequently, the IC functions as an integrative
is essential for appetitive behavior related to feed- hub due to the vast reciprocal connections that
ing (Valdés et al., 2010) and other goal-directed exist between it and an extensive network of cor-
behaviors, like sexual behavior, drug seeking or tical and subcortical structures (Benarroch, 2019;
drinking (Contreras et al., 2016). Altogether, sev- Craig and Craig, 2009; Nguyen et al., 2016; Saper,
eral studies suggest that histaminergic neurons 1982). As the IC area is responsible for the intero-
detect aversive and rewarding signals in various ceptive processing of multisensory information,
emotionally arousing contexts and relevant stimuli this region could be required for the processing
that could help to regulate their impact on moti- of internal states involved in memory formation
vated memory consolidation (Miklos and Kovacs, (Gu et al., 2013). The established role of the IC in
2003; Passani et al., 2007; Taylor and Snyder, the processing of pain (Lu et al., 2016; Starr et al.,
1971; Torrealba et al., 2012). More studies are 2009) and negative affective states like anxiety
needed to show that the effects of histaminergic (Paulus and Stein, 2006) supports this hypothesis.
activity may depend on interactions with brain Moreover, the IC is also a necessary compo-
structures that modulate the expression of learned nent in reward and aversion learning (Bermúdez-
behavior in complex contexts and its mediation Rattoni et al., 2004; Casanova Castillo et al., 2016;
with interoceptive signaling (Provens et al., 2020). Guzman-Ramos et al., 2012; Toyoda, 2019; Sun
et al., 2020). It has been implicated in the media-
tion of several processes related to craving and
drug seeking (Contreras et al., 2007; Moschak
et al., 2018; Naqvi and Bechara, 2009). Several
INTEROCEPTIVE INFLUENCES ON findings present a potential mechanism whereby
CONTEXT upregulation of the IC by opioid signaling can lead
to altered subcortical function and downstream
Interoception is defined as “a process by which activity (Pina et al., 2020). Also, recent data indi-
the nervous system senses, interprets, and inte- cate that combined effects of endocannabinoid
grates signals originating from within the body, signaling can modulate long-term plasticity at
providing a moment-by-moment mapping of the GABAergic synapses in the IC. This GABAergic
body’s internal landscape across conscious and synaptic plasticity may be a crucial synaptic
unconscious levels” (Khalsa et al., 2018). mechanism in memory formation (Toyoda, 2020).
160 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

SUMMARY AND CONCLUSIONS the sleep-wake cycle for the regulation of nocic-
eption, modulating the rhythmical activity of the
Identifying a stimulus with punishment or reward hippocampus during aversive learning processes.
is vital for survival throughout life. This associa- Histamine is an excellent candidate for peripheral
tive process ultimately drives targeted actions to homeostatic signals enabling cognition of salient
prevent danger, such as avoidance behaviors or stimuli related to contextual environments.
appetitive responses essential for survival. Acknowledgments: We thank Gabriela Vera-
Consequently, the previous association of stimuli Rivera and Luis Rodriguez-Duran for techni-
alters future learning, allowing rapid response and cal assistance, and we gratefully acknowledge
crucial adaptation to different situations. Several Jessica G. Norris for her generous help in proof-
studies indicate that the integration of nociceptive reading and editing the manuscript. DGAPA-
stimuli requires a complex interaction of neuro- PAPIITIN209122, IN201420 to IM. Departamento
transmission in brain areas related to the sensory- de Ciencias de la Salud to KG-R. DGAPA-PAPIIT
discriminative components of pain and those that IN212919 and IN213123 to FBR, and DGAPA-
contribute to the behavior associated with affec- PAPIIT IA201420 and IA202922 to DOG.
tive responses. In this chapter, we have discussed
the role of different neurotransmitters in several
brain areas which are crucial for cognitive func-
tions related to maintaining a balance between Notes
reward and aversive learning.
Recent evidence shows that contextual changes 1  Appetitive conditioning: A type of classical condi-
could affect learning, retrieval, and updating of tioning in which the unconditioned stimulus is a
memory, suggesting a more complex scenario positive reinforcer.
during stimuli associations, including opposite 2  Discrimination performance: Sensory decision-
or different valences, relating to any motivational making that focuses on relevant stimuli and ade-
task. Current knowledge points to the critical role quate signal/noise ratio and recognizes different
of integrating hubs, like the insular cortex, to attributes in them.
translate external signals into interoceptive states 3  Operational tasks: Involves behavior that “oper-
that elicit complex physiological, affective, and ates” on the environment according to the con-
behavioral responses. This chapter has provided sequences of the response output.
an overview of some of the most important neu- 4  Optogenetics: A combination of genetic and
rotransmitter interactions that mediate aversive optical methods to excite or inhibit neuronal
and appetitive association in different brain areas. function.
Particularly important are those areas that play an 5  Memory extinction: Reduction of a response that
essential role during the formation of association occurs because the conditioned stimulus is pre-
that require mediation of the affective/aversive sented without the unconditioned stimulus and
emotional balance according to their complexity, no longer predicts it.
resulting in the immense variation of valence and 6  Memory updating: Strengthening the current
intensity of memory. memory trace incorporating new information.
In addition, these findings support the hypoth- 7  Aversive learning: Development of dislike or
esis that the IC is an integrative hub combining unwillingness to display a behavior by pairing it
the opioid and cannabinoid systems during the with an unpleasant stimulus.
integration of associations of complex contexts. 8  Neuronal plasticity: Morphofunctional changes in
Thus, the IC could be a substantial area to under- the cell that modify synaptic efficiency.
stand exteroceptive and interoceptive memory 9  Reward prediction error: The difference between
modulation with complex physiological, affective, received and expected rewards.
and behavioral responses. However, more studies 10  Nociception: Encoding and processing of noxious
are needed to unveil the IC´s precise circuit and stimuli.
signaling mechanisms during context complex- 11  Conditioned taste aversion: Learned rejection to
ity learning to weigh the intensity and valence of a particular taste after its association with visceral
experiences. illness.
Evidently, the conceptualization of context 12  Liking: The pleasurable/hedonic impact or vari-
should include a variety of exteroceptive and ous expressions of subjective pleasure induced by
interoceptive stimuli thought to be available in the rewarded appetitive experience.
environment and which play a key role in mem- 13  Incentive salience: Motivation for rewards that is
ory formation. In this regard, it has been shown driven by both the physiological state and previ-
that histamine controls fundamental aspects of ously learned associations about a reward cue.
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 161

14  Wanting: Rewarding behavior that triggers atten- Bacciottini, L., Passani, M. B., Giovannelli, L., Cangi-
tion-grabbing and attraction processes. oli, I., Mannaioni, P. F., Schunack, W., & Blandina,
15  Interference paradigms: Tasks that activate non- P. (2002). Endogenous histamine in the medial
target memories, influencing learning or retrieval. septum-diagonal band complex increases the
16  Counterconditioning: Behavioral modification release of acetylcholine from the hippocampus: A
involving a new association with a stimulus of dual-probe microdialysis study in the freely moving
opposite valence. rat. European Journal of Neuroscience, 15(10),
17  Discrimination reversal: Learning about contin- 1669–1680.
gencies through the sensory properties of cues Baldi, E., & Bucherelli, C. (2015). Brain sites involved
that predict stimulus availability and the actions in fear memory reconsolidation and extinction of
required to experience the consequence provid- rodents. Neuroscience & Biobehavioral Reviews,
ing discriminated behavior. 53, 160–190.
18  Latent inhibition: Process in which stimulus pre- Baldo, B. A. (2016). Prefrontal cortical opioids and
exposure attenuates the ability of that stimulus dysregulated motivation: A network hypothesis.
to be associated with positive or negative conse- Trends Neuroscience, 39(6), 366–377.
quences. Baliki, M. N., Chialvo, D. R., Geha, P. Y., Levy, R. M.,
19  Reward expectancy: When animals display a Harden, R. N., Parrish, T. B., & Apkarian, A. V.
behavior motivated by a particular positive out- (2006). Chronic pain and the emotional brain:
come. Specific brain activity associated with spontaneous
20  Pavlovian conditioned responses: Association of a fluctuations of intensity of chronic back pain. Jour-
conditioned stimulus with a biologically relevant nal of Neuroscience, 26(47), 12165–12173.
unconditioned stimulus where the subject learns Barbano, M. F., & Cador, M. (2007). Opioids for
the predictive value of the conditioned stimulus. hedonic experience and dopamine to get ready for
it. Psychopharmacology, 191(3), 497–506.
Benarroch, E. E. (2019). Insular cortex: Functional
complexity and clinical correlations. Neurology,
93(21), 932–938.
REFERENCES Bermúdez-Rattoni, F., Ramírez-Lugo, L., Gutiérrez, R.,
& Miranda, M. I. (2004). Molecular signals into the
Acquas, E., Wilson, C., & Fibiger, H. C. (1996). Con- insular cortex and amygdala during aversive gusta-
ditioned and unconditioned stimuli increase fron- tory memory formation. Cellular and Molecular
tal cortical and hippocampal acetylcholine release: Neurobiology, 24(1), 25–36.
Effects of novelty, habituation, and fear. Journal of Bernard, J. F., Alden, M., & Besson, J. M. (1993). The
Neuroscience, 16(9), 3089–3096. organization of the efferent projections from the
Aguado, L., Hall, G., Harrington, N., & Symonds, M. pontine parabrachial area to the amygdaloid com-
(1998). Illness-induced context aversion learning in plex: A phaseolus vulgaris leucoagglutinin (PHA-L)
rats with lesions of the dorsal hippocampus. study in the rat. Journal of Comparative Neurol-
Behavioral Neuroscience, 112(5), 1142–1151. ogy, 329(2), 201–229.
Aitta-Aho, T., Hay, Y. A., Phillips, B. U., Saksida, L. M., Bernard, J. F., Huang, G. F., & Besson, J. M. (1992).
Bussey, T. J., Paulsen, O., & Apergis-Schoute, J. Nucleus centralis of the amygdala and the globus
(2018). Basal forebrain and brainstem cholinergic pallidus ventralis: Electrophysiological evidence for
neurons differentially impact amygdala circuits and an involvement in pain processes. Journal of Neu-
learning-related behavior. Current Biology, 28(16), rophysiology, 68(2), 551–569.
2557–2569. Berridge, K. C. (2000). Measuring hedonic impact in
Aitta-Aho, T., Hay, Y. A., Phillips, B. U., Saksida, L. M., animals and infants: Microstructure of affective
Bussey, T. J., Paulsen, O., & Apergis-Schoute, J. taste reactivity patterns. Neuroscience & Biobehav-
(2018). Basal forebrain and brainstem cholinergic ioral Reviews, 24(2), 173–198.
neurons differentially impact amygdala circuits and Berridge, K. C. (2007). The debate over dopamine’s
learning-related behavior. Current Biology, 28(16), role in reward: the case for incentive salience.
2557-2569. Psychopharmacology, 191(3), 391–431.
Ameri, A. (1999). The effects of cannabinoids on the Berridge, K. C., & Kringelbach, M. L. (2008). Affec-
brain. Progress in Neurobiology, 58(4), 315-348. tive neuroscience of pleasure: Reward in humans
Ansah, O. B., Bourbia, N., Goncalves, L., Almeida, and animals. Psychopharmacology, 199(3),
A., & Pertovaara, A. (2010). Influence of amygda- 457–480.
loid glutamatergic receptors on sensory and emo- Berridge, K. C., & Kringelbach, M. L. (2015). Pleasure
tional pain-related behavior in the neuropathic systems in the brain. Neuron, 86(3), 646–664.
rat. Behavioural Brain Research, 209(1), Berridge, K. C., & Robinson, T. E. (1998). What is the
174–178. role of dopamine in reward: Hedonic impact,
162 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

reward learning, or incentive salience? Brain A role for the interoceptive insular cortex in the
Research Reviews, 28(3), 309–369. consolidation of learned fear. Behavioural Brain
Berridge, K. C., Robinson, T. E., & Aldridge, J. W. Research, 296, 70–77.
(2009). Dissecting components of reward: Castro, D. C., & Berridge, K. C. (2017). Opioid and
“Liking”, “wanting”, and learning. Current Opin- orexin hedonic hotspots in rat orbitofrontal cortex
ion in Pharmacology, 9(1), 65–73. and insula. Proceedings of the National Academy
Bester, H., Matsumoto, N., Besson, J. M., & Bernard, of Sciences of the United States of America,
J. F. (1997). Further evidence for the involvement 114(43), E9125–E9134.
of the spinoparabrachial pathway in nociceptive Chaijale, N. N., Curtis, A. L., Wood, S. K., Zhang, X.-Y.,
processes: A c-Fos study in the rat. Journal of Bhatnagar, S., Reyes, B. A., & Valentino, R. J. (2013).
Comparative Neurology, 383(4), 439–458. Social stress engages opioid regulation of locus
Bonini, J. S., Da Silva, W. C., Da Silveira, C. K., Kohler, coeruleus norepinephrine neurons and induces a
C. A., Izquierdo, I., & Cammarota, M. (2011). state of cellular and physical opiate dependence.
Histamine facilitates consolidation of fear extinc- Neuropsychopharmacology, 38(10), 1833–1843.
tion. International Journal of Neuropsychophar- Cheng, S. J., Chen, C. C., Yang, H. W., Chang, Y. T.,
macology, 14(9), 1209–1217. Bai, S. W., Chen, C. C., & Min, M. Y. (2011). Role
Bornhovd, K., Quante, M., Glauche, V., Bromm, B., of extracellular signal-regulated kinase in synaptic
Weiller, C., & Buchel, C. (2002). Painful stimuli transmission and plasticity of a nociceptive input
evoke different stimulus-response functions in the on capsular central amygdaloid neurons in normal
amygdala, prefrontal, insula and somatosensory and acid-induced muscle pain mice. Journal of
cortex: A single-trial fMRI study. Brain, 125(6), Neuroscience, 31(6), 2258–2270.
1326–1336. Clayton, E. C., & Williams, C. L. (2000). Adrenergic
Bouton, M. E. (1993). Context, time, and memory activation of the nucleus tractus solitarius potenti-
retrieval in the interference paradigms of Pavlovian ates amygdala norepinephrine release and
learning. Psychological Bulletin, 114(1), 80–99. enhances retention performance in emotionally
Bouton, M. E. (2019). Extinction of instrumental arousing and spatial memory tasks. Behavioural
(operant) learning: interference, varieties of con- Brain Research, 112(1–2), 151–158.
text, and mechanisms of contextual control. Psy- Contreras, M., Ceric, F., & Torrealba, F. (2007). Inacti-
chopharmacology, 236(1), 7–19. vation of the interoceptive insula disrupts drug
Bouton, M. E., Kenney, F. A., & Rosengard, C. (1990). craving and malaise induced by lithium. Science,
State-dependent fear extinction with two benzo- 318, 655–658.
diazepine tranquilizers. Behavioral Neuroscience, Contreras, M., Riveros, M. E., Quispe, M., Sanchez,
104(1), 44–55. C., Perdomo, G., Torrealba, F., & Valdés, J. L.
Broussard, J. I., Yang, K., Levine, A. T., Tsetsenis, T., (2016). The histaminergic tuberomamillary nucleus
Jenson, D., Cao, F., & Dani, J. A. (2016). Dopamine is involved in appetite for sex, water and ampheta-
regulates aversive contextual learning and associ- mine. PLoS One, 11(2). doi: 10.1371/journal.
ated in vivo synaptic plasticity in the hippocampus. pone.0148484
Cell Reports, 14(8), 1930–1939. Cota, D., Tschöp, M. H., Horvath, T. L., & Levine, A.
Brown, R. E., Stevens, D. R., & Haas, H. L. (2001). The S. (2006). Cannabinoids, opioids and eating
physiology of brain histamine. Progress in Neuro- behavior: the molecular face of hedonism? Brain
biology, 63(6), 637–672. Research Reviews, 51(1), 85–107.
Calcagnetti, D. J., Helmstetter, F. J., & Fanselow, M. S. Craig, A. D., & Craig, A. (2009). How do you feel–
(1987). Quaternary naltrexone reveals the central now? The anterior insula and human awareness.
mediation of conditional opioid analgesia. Pharma- Nature Reviews Neuroscience, 10(1), 59–70.
cology Biochemistry & Behavior, 27(3), 529–531. Cunningham, C. L. (1979). Alcohol as a cue for
Cangioli, I., Baldi, E., Mannaioni, P. F., Bucherelli, C., extinction: State dependency produced by condi-
Blandina, P., & Passani, M. B. (2002). Activation of tioned inhibition. Animal Learning & Behavior,
histaminergic H3 receptors in the rat basolateral 7(1), 45–52.
amygdala improves expression of fear memory Dani, J. A., & Bertrand, D. (2007). Nicotinic acetyl-
and enhances acetylcholine release. European choline receptors and nicotinic cholinergic mecha-
Journal of Neuroscience, 16(3), 521–528. nisms of the central nervous system. Annual
Cardinal, R. N., Parkinson, J. A., Hall, J., & Everitt, B. Review of Pharmacology Toxicology, 47,
J. (2002). Emotion and motivation: The role of the 699–729.
amygdala, ventral striatum, and prefrontal cortex. da Silva WC, Bonini JS, Bevilaqua LR, Izquierdo I, &
Neuroscience & Biobehavioral Reviews, 26(3), Cammarota M. (2006). Histamine enhances inhibi-
321–352. tory avoidance memory consolidation through a
Casanova Castillo, J. P., Madrid, C., Contreras, M., H2 receptor-dependent mechanism. Neurobiology
Rodríguez, M., Vásquez, M., & Torrealba, F. (2016). of Learning & Memory, 86(1), 100–106.
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 163

Davidson, T. L. (1993). The nature and function of Fields, H. L., & Margolis, E. B. (2015). Understanding
interoceptive signals to feed: Toward integration opioid reward. Trends in Neurosciences, 38(4),
of physiological and learning perspectives. Psycho- 217–225.
logical Review, 100(4), 640–657. Freitas, R. L., Salgado-Rohner, C. J., Hallak, J. E.,
Davidson, T. L., Kanoski, S. E., Tracy, A. L., Walls, E. Crippa, J. A., & Coimbra, N. C. (2013). Involve-
K., Clegg, D., & Benoit, S. C. (2005). The intero- ment of prelimbic medial prefrontal cortex in
ceptive cue properties of ghrelin generalize to cues panic-like elaborated defensive behaviour and
produced by food deprivation. Peptides, 26(9), innate fear-induced antinociception elicited by
1602–1610. GABAA receptor blockade in the dorsomedial and
Davis, M. (1979). Morphine and naloxone: Effects on ventromedial hypothalamic nuclei: Role of the
conditioned fear as measured with the potenti- endocannabinoid CB1 receptor. International Jour-
ated startle paradigm. European Journal of Phar- nal of Neuropsychopharmacology, 16(8),
macology, 54(4), 341–347. 1781–1798.
de Jong, J. W., Afjei, S. A., Pollak Dorocic, I., Peck, J. Friemel, C. M., Zimmer, A., & Schneider, M. (2014).
R., Liu, C., Kim, C. K., & Lammel, S. (2019). A The CB1 receptor as an important mediator of
neural circuit mechanism for encoding aversive hedonic reward processing. Neuropsychopharma-
stimuli in the mesolimbic dopamine system. cology, 39(10), 2387–2396.
Neuron, 101(1), 133–151. Fuchs, P. N., Peng, Y. B., Boyette-Davis, J. A., & Uhel-
De la Casa, L. G., Cárcel, L., Ruiz-Salas, J. C., Vicente, ski, M. L. (2014). The anterior cingulate cortex and
L., & Mena, A. (2018). Conditioned increase of pain processing. Frontiers in Integrative Neurosci-
locomotor activity induced by haloperidol. PLoS ence, 8. doi: 10.3389/fnint.2014.00035
One, 13(10). doi: 10.1371/journal.pone.0200178 Gallo, M., & Cándido, A. (1995). Reversible inactiva-
De la Casa, L. G., & Diaz, E. (2013). Contextual con- tion of dorsal hippocampus by tetrodotoxin
trol of flavor neophobia. Physiology and Behavior, impairs blocking of taste aversion selectively
118, 45–51. during the acquisition but not the retrieval in rats.
Dreisbach, G., & Fischer, R. (2015). Conflicts as aver- Neuroscience Letters, 186(1), 1–4.
sive signals for control adaptation. Current Direc- Gallo, M., Marquez, S., Ballesteros, M., & Mal-
tions in Psychological Science, 24(4), 255–260. donado, A. (1999). Functional blockade of the
Eikemo, M., Biele, G., Willoch, F., Thomsen, L., & parabrachial area by tetrodotoxin disrupts the
Leknes, S. (2017). Opioid modulation of value- acquisition of conditioned taste aversion induced
based decision-making in healthy humans. Neu- by motion-sickness in rats. Neuroscience Letters,
ropsychopharmacology, 42(9), 1833–1840. 265(1), 57–60.
Eippert, F., Bingel, U., Schoell, E., Yacubian, J., & Galvez, R., Mesches, M. H., & McGaugh, J. L. (1996).
Buchel, C. (2008). Blockade of endogenous opioid Norepinephrine release in the amygdala in
neurotransmission enhances acquisition of condi- response to footshock stimulation. Neurobiology
tioned fear in humans. Journal of Neuroscience, of Learning and Memory, 66(3), 253–257.
28(21), 5465–5472. Gao, Y.-J., Ren, W.-H., Zhang, Y.-Q., & Zhao, Z.-Q.
Ellis, M. E., & Kesner, R. P. (1983). The noradrenergic (2004). Contributions of the anterior cingulate
system of the amygdala and aversive information cortex and amygdala to pain-and fear-conditioned
processing. Behavioral Neuroscience, 97(3), place avoidance in rats. Pain, 110(1–2), 343–353.
399–415. Garcia-Medina, N. E., & Miranda, M. I. (2013).
Elphick, M. R. (2012). The evolution and comparative Nucleus of the solitary tract chemical stimulation
neurobiology of endocannabinoid signalling. Phil- induces extracellular norepinephrine release in the
osophical Transactions of the Royal Society B: lateral and basolateral amygdala. Brain Stimula-
Biological Sciences, 367, 3201–3215. tion, 6(2), 198–201.
Esch, T., & Stefano, G. B. (2004). The neurobiology of Garcia-Medina, N. E., Vera, G., & Miranda, M. I.
pleasure, reward processes, addiction and their (2015). Chemical stimulation or glutamate injec-
health implications. Neuroendocrinology Letters, tions in the nucleus of solitary tract enhance con-
25(4), 235–251. ditioned taste aversion. Behavioral Brain Research,
Fanselow, M. S., Calcagnetti, D. J., & Helmstetter, F. 278, 202–209.
J. (1988). Peripheral versus intracerebroventricular Garcia-de la Torre, P., Rodríguez-Ortiz, C. J., Balderas,
administration of quaternary naltrexone and the I., & Bermúdez-Rattoni, F. (2010). Differential par-
enhancement of Pavlovian conditioning. Brain ticipation of temporal structures in the consolida-
Research, 444(1), 147–152. tion and reconsolidation of taste aversion
Fattore, L., Melis, M., Fadda, P., Pistis, M., & Fratta, extinction. European Journal of Neuroscience,
W. (2010). The endocannabinoid system and non- 32(6), 1018–1023.
drug rewarding behaviours. Experimental Neurol- Gold, P. E. (2003). Acetylcholine modulation of
ogy, 224(1), 23–36. neural systems involved in learning and memory.
164 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Neurobiology of Learning and Memory, 80(3), PTSD. Experimental Neurology, 284(B),


194–210. 181–195.
Gould, T. J., & Leach, P. T. (2014). Cellular, molecular, Hermans, E. J., Henckens, M. J., Joels, M., & Fernan-
and genetic substrates underlying the impact of dez, G. (2014). Dynamic adaptation of large-scale
nicotine on learning. Neurobiology of Learning brain networks in response to acute stressors.
and Memory, 107, 108–132. Trends in Neuroscience, 37(6), 304–314.
Grau-Perales, A., Levy, E., Fenton, A., & Gallo, M. Herry, C., Ciocchi, S., Senn, V., Demmou, L., Müller,
(2019). Dorsal hippocampal damage disrupts the C., & Lüthi, A. (2008). Switching on and off fear
auditory context-dependent attenuation of taste by distinct neuronal circuits. Nature, 454,
neophobia in mice. Neurobiology of Learning and 600–606.
Memory, 157, 121–127. Hoflich, A., Michenthaler, P., Kasper, S., & Lanzen-
Groc, L., Choquet, D., & Chaouloff, F. (2008). The berger, R. (2019). Circuit mechanisms of reward,
stress hormone corticosterone conditions AMPAR anhedonia, and depression. International Journal
surface trafficking and synaptic potentiation. of Neuropsychopharmacology, 22(2), 105–118.
Nature of Neuroscience, 11(8), 868–870. Howland, J. G., Taepavarapruk, P., & Phillips, A. G.
Gu, X., Hof, P. R., Friston, K. J., & Fan, J. (2013). Ante- (2002). Glutamate receptor-dependent modula-
rior insular cortex and emotional awareness. Journal tion of dopamine efflux in the nucleus accumbens
of Comparative Neurology, 521(15), 3371–3388. by basolateral, but not central, nucleus of the
Guzman-Ramos, K., Osorio-Gomez, D., Moreno- amygdala in rats. Journal of Neuroscience, 22(3),
Castilla, P., & Bermudez-Rattoni, F. (2010). Off-line 1137–1145.
concomitant release of dopamine and glutamate Inagaki, N., Yamatodani, A., Ando-Yamamoto, M.,
involvement in taste memory consolidation. Jour- Tohyama, M., Watanabe, T., & Wada, H. (1988).
nal of Neurochemistry, 114(1), 226–236. Organization of histaminergic fibers in the rat
Guzman-Ramos, K., Osorio-Gomez, D., Moreno- brain. Journal of Comparative Neurology, 273(3),
Castilla, P., & Bermudez-Rattoni, F. (2012). Post- 283–300.
acquisition release of glutamate and Jacobson, M. L., Wulf, H. A., Browne, C. A., & Lucki,
norepinephrine in the amygdala is involved in I. (2018). Opioid modulation of cognitive impair-
taste-aversion memory consolidation. Learning ment in depression. Progress in Brain Research,
and Memory, 19(6), 231–238. 239, 1–48.
Haaker, J., Yi, J., Petrovic, P., & Olsson, A. (2017). Jarrett, M. M., Scantlebury, J., & Parker, L. A. (2007).
Endogenous opioids regulate social threat learning Effect of Δ9-tetrahydrocannabinol on quinine pal-
in humans. Nature Communications, 8. doi: atability and AM251 on sucrose and quinine palat-
10.1038/ncomms15495 ability using the taste reactivity test. Physiology
Haas, H., & Panula, P. (2003). The role of histamine and Behavior, 90(2–3), 425–430.
and the tuberomamillary nucleus in the nervous Jiang, L., Kundu, S., Lederman, J. D., Lopez-Hernan-
system. Nature Reviews Neuroscience, 4(2), dez, G. Y., Ballinger, E. C., Wang, S., & Role, L. W.
121–130. (2016). Cholinergic signaling controls conditioned
Han, S., Soleiman, M. T., Soden, M. E., Zweifel, L. S., fear behaviors and enhances plasticity of cortical-
& Palmiter, R. D. (2015). Elucidating an affective amygdala circuits. Neuron, 90(5), 1057–1070.
pain circuit that creates a threat memory. Cell, Johansen, J. P., Fields, H. L., & Manning, B. H. (2001).
162(2), 363–374. The affective component of pain in rodents: Direct
Hasselmo, M. E., & Sarter, M. (2011). Modes and evidence for a contribution of the anterior cingu-
models of forebrain cholinergic neuromodulation late cortex. Proceedings of the National Academy
of cognition. Neuropsychopharmacology, 36(1), of Sciences of the United States of America,
52–73. 98(14), 8077–8082.
Hatfield, T., & McGaugh, J. L. (1999). Norepinephrine Kanoski, S. E., Walls, E. K., & Davidson, T. (2007).
infused into the basolateral amygdala posttraining Interoceptive “satiety” signals produced by leptin
enhances retention in a spatial water maze task. and CCK. Peptides, 28(5), 988–1002.
Neurobiology of Learning and Memory, 71(2), Keiflin, R., & Janak, P. H. (2015). Dopamine predic-
232–239. tion errors in reward learning and addiction: From
Heath, F. C., Jurkus, R., Bast, T., Pezze, M. A., Lee, J. theory to neural circuitry. Neuron, 88(2),
L., Voigt, J. P., & Stevenson, C. W. (2015). Dopa- 247–263.
mine D1-like receptor signalling in the hippocam- Kempadoo, K. A., Mosharov, E. V., Choi, S. J., Sulzer,
pus and amygdala modulates the acquisition of D., & Kandel, E. R. (2016). Dopamine release from
contextual fear conditioning. Psychopharmacol- the locus coeruleus to the dorsal hippocampus
ogy, 232(14), 2619–2629. promotes spatial learning and memory. Proceed-
Hendrickson, R. C., & Raskind, M. A. (2016). Noradr- ings of the National Academy of Sciences of the
energic dysregulation in the pathophysiology of United States of America, 113(51), 14835–14840.
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 165

Khalsa, S. S., Adolphs, R., Cameron, O. G., Critchley, Letzkus, J. J., Wolff, S. B., Meyer, E. M., Tovote, P.,
H. D., Davenport, P. W., Feinstein, J. S., & Mehling, Courtin, J., Herry, C., & Luthi, A. (2011). A disin-
W. E. (2018). Interoception and mental health: a hibitory microcircuit for associative fear learning in
roadmap. Biological Psychiatry: Cognitive Neuro- the auditory cortex. Nature, 480, 331–335.
science and Neuroimaging, 3(6), 501–513. Liang, K. C., Juler, R. G., & McGaugh, J. L. (1986).
Kim, J. J., & Fanselow, M. S. (1992). Modality-specific Modulating effects of posttraining epinephrine on
retrograde amnesia of fear. Science, 256, memory: Involvement of the amygdala noradren-
675–677. ergic system. Brain Research, 368(1), 125–133.
Kirkham, T. C. (2009). Cannabinoids and appetite: Liang, K. C., McGaugh, J. L., & Yao, H. Y. (1990).
Food craving and food pleasure. International Involvement of amygdala pathways in the influ-
Review of Psychiatry, 21(2), 163–171. ence of post-training intra-amygdala norepineph-
Kissiwaa, S. A., & Bagley, E. E. (2018). Central sensi- rine and peripheral epinephrine on memory
tization of the spino-parabrachial-amygdala path- storage. Brain Research, 508(2), 225–233.
way that outlasts a brief nociceptive stimulus. Likhtik, E., & Johansen, J. P. (2019a). Neuromodula-
Journal of Physiology, 596(18). doi: 10.1113/ tion in circuits of aversive emotional learning.
JP273976 Nature Neuroscience, 22(10), 1586–1597.
Knox, D. (2016). The role of basal forebrain choliner- Likhtik, E., & Johansen, J. P. (2019b). Publisher
gic neurons in fear and extinction memory. Neuro- Correction: Neuromodulation in circuits of aversive
biology of Learning and Memory, 133, 39–52. emotional learning. Nature Neuroscience,
Knox, D., & Keller, S. M. (2016). Cholinergic neu- 22(11), 1946.
ronal lesions in the medial septum and vertical Lin R, Liang J, & Luo M. (2021) The Raphe dopamine
limb of the diagonal bands of Broca induce con- system: Roles in salience encoding, memory
textual fear memory generalization and impair expression, and addiction. Trends in Neuroscience,
acquisition of fear extinction. Hippocampus, 44(5), 366–377.
26(6), 718–726. Louilot, A., Simon, H., Taghzouti, K., & Le Moal, M.
Koob, G. F., & Volkow, N. D. (2016). Neurobiology of (1985). Modulation of dopaminergic activity in the
addiction: a neurocircuitry analysis. Lancet Psychia- nucleus accumbens following facilitation or block-
try, 3(8), 760–773. ade of the dopaminergic transmission in the
Kring, A. M., & Barch, D. M. (2014). The motivation amygdala: A study by in vivo differential pulse
and pleasure dimension of negative symptoms: voltammetry. Brain Research, 346(1), 141–145.
Neural substrates and behavioral outputs. Euro- Lu, C., Yang, T., Zhao, H., Zhang, M., Meng, F., Fu,
pean Neuropsychopharmacology, 24(5), H., & Xu, H. (2016). Insular cortex is critical for the
725–736. perception, modulation, and chronification of
Kringelbach, M. L., & Berridge, K. C. (2009). Towards pain. Neuroscience Bulletin, 32(2), 191–201.
a functional neuroanatomy of pleasure and happi- Luchicchi, A., Bloem, B., Viana, J. N., Mansvelder, H.
ness. Trends in Cognitive Sciences, 13(11), D., & Role, L. W. (2014). Illuminating the role of
479–487. cholinergic signaling in circuits of attention and
Lammel, S., Lim, B. K., Ran, C., Huang, K. W., Betley, emotionally salient behaviors. Frontiers in Synaptic
M. J., Tye, K. M., & Malenka, R. C. (2012). Input- Neuroscience, 6. doi: 10.3389/fnsyn.2014.00024
specific control of reward and aversion in the Lutz, P.-E., & Kieffer, B. L. (2013). The multiple facets
ventral tegmental area. Nature, 491, 212–217. of opioid receptor function: Implications for addic-
Lattal, K. M. (2007). Effects of ethanol on encoding, tion. Current Opinion in Neurobiology, 23(4),
consolidation, and expression of extinction follow- 473–479.
ing contextual fear conditioning. Behavioral Neu- Lynch, K. S., & Ryan, M. J. (2020). Understanding the
roscience, 121(6), 1280–1292. role of incentive salience in sexual decision-mak-
Laurent, V., Morse, A. K., & Balleine, B. W. (2015). ing. Integrative and Comparative Biology, 60(3),
The role of opioid processes in reward and deci- 712–721.
sion-making. British Journal of Pharmacology, Mahler, S. V., & Berridge, K. C. (2012). What and
172(2), 449–459. when to “want”? Amygdala-based focusing of
LeDoux, J. E. (2000). Emotion circuits in the brain. incentive salience upon sugar and sex. Psychop-
Annual Review of Neuroscience, 23, 155–184. harmacology, 221(3), 407–426.
LeDoux, J. E. (2012). Evolution of human emotion: a Mahler, S. V., Smith, K. S., & Berridge, K. C. (2007).
view through fear. Progress in Brain Research, Endocannabinoid hedonic hotspot for sensory
195, 431–442. pleasure: Anandamide in nucleus accumbens shell
Lee, S., & Kim, J.-H. (2019). Basal forebrain choliner- enhances “liking” of a sweet reward. Neuropsy-
gic-induced activation of cholecystokinin inhibi- chopharmacology, 32(11), 2267–2278.
tory neurons in the basolateral amygdala. Manrique, T., Gamiz, F., Moron, I., Ballesteros, M. A.,
Experimental Neurobiology, 28(3), 320–328. & Gallo, M. (2009a). Peculiar modulation of taste
166 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

aversion learning by the time of day in developing acetylcholine release and acquisition, but not
rats. Journal of the International Society for Devel- retrieval, of aversive memories. Proceedings of the
opmental Psychobiology, 51(2), 147–157. National Academy of Sciences of the United States
Manrique, T., Morón, I., Ballesteros, M. A., Guerrero, of America, 96(11), 6478–6482.
R. M., Fenton, A. A., & Gallo, M. (2009b). Hip- Miranda, M. I., Ferreira, G., Ramirez-Lugo, L., & Ber-
pocampus, aging, and segregating memories. mudez-Rattoni, F. (2002). Glutamatergic activity in
Hippocampus, 19(1), 57–65. the amygdala signals visceral input during taste
Marek, R., Strobel, C., Bredy, T. W., & Sah, P. (2013). memory formation. Proceedings of the National
The amygdala and medial prefrontal cortex: Part- Academy of Sciences of the United States of
ners in the fear circuit. Journal of Physiology, America, 99(17), 11417–11422.
591(10), 2381–2391. Miyashita, T., & Williams, C. L. (2002). Glutamatergic
Martin, S., Henley, J. M., Holman, D., Zhou, M., transmission in the nucleus of the solitary tract
Wiegert, O., van Spronsen, M., … Krugers, H. J. modulates memory through influences on amyg-
(2009). Corticosterone alters AMPAR mobility and dala noradrenergic systems. Behavoural Neurosci-
facilitates bidirectional synaptic plasticity. PLoS ence, 116(1), 13–21.
One, 4(3). doi: 10.1371/journal.pone.0004714 Moga, M. M., Weis, R. P., & Moore, R. Y. (1995).
Matsumoto, M., & Hikosaka, O. (2009). Two types of Efferent projections of the paraventricular tha-
dopamine neuron distinctly convey positive and lamic nucleus in the rat. Journal of Comparative
negative motivational signals. Nature, 459, Neurology, 359(2), 221–238.
837–841. Molero, A., Moron, I., Angeles Ballesteros, M., Man-
McCall, J. G., Siuda, E. R., Bhatti, D. L., Lawson, L. A., rique, T., Fenton, A., & Gallo, M. (2005). Hip-
McElligott, Z. A., Stuber, G. D., & Bruchas, M. R. pocampus, temporal context and taste memories.
(2017). Locus coeruleus to basolateral amygdala Chemical Senses, 30(S1), 160–161.
noradrenergic projections promote anxiety-like Moore, H., Fadel, J., Sarter, M., & Bruno, J. P. (1999).
behavior. Elife, 6. doi: 10.7554/eLife.18247 Role of accumbens and cortical dopamine recep-
McGaugh, J. L. (2000). Memory: A century of con- tors in the regulation of cortical acetylcholine
solidation. Science, 287, 248–251. release. Neuroscience, 88(3), 811–822.
McGaugh, J. L., McIntyre, C. K., & Power, A. E. Moron, I., Ballesteros, M., Candido, A., & Gallo, M.
(2002). Amygdala modulation of memory consoli- (2002). Taste aversion learning and aging: a com-
dation: interaction with other brain systems. Neu- parison with the effect of dorsal hippocampal
robiology of Learning and Memory, 78(3), lesions in rats. Physiological Research, 51, S21–S28.
539–552. Moschak, T. M., Wang, X., & Carelli, R. M. (2018). A
McGaugh, J. L. (2013). Making lasting memories: neuronal ensemble in the rostral agranular insula
Remembering the significant. Proceedings of the tracks cocaine-induced devaluation of natural
National Academy of Sciences of the United States reward and predicts cocaine seeking. Journal of
of America, 110, 10402–10407. Neuroscience, 38(39), 8463–8472.
McIntyre, C. K., Power, A. E., Roozendaal, B., & Muller, J. F., Mascagni, F., & McDonald, A. J. (2011).
McGaugh, J. L. (2003). Role of the basolateral Cholinergic innervation of pyramidal cells and
amygdala in memory consolidation. Annals of the parvalbumin-immunoreactive interneurons in the
New York Academy Sciences, 985, 273–293. rat basolateral amygdala. Journal of Comparative
McNally, G. P., Pigg, M., & Weidemann, G. (2004). Neurology, 519(4), 790–805.
Opioid receptors in the midbrain periaqueductal Nadel, L., & Willner, J. (1980). Context and condition-
gray regulate extinction of Pavlovian fear condi- ing: A place for space. Physiological Psychology,
tioning. Journal of Neuroscience, 24(31), 8(2), 218–228.
6912–6919. Naqvi, N. H., & Bechara, A. (2009). The hidden island
Menegas, W., Akiti, K., Amo, R., Uchida, N., & of addiction: the insula. Trends in Neurosciences,
Watabe-Uchida, M. (2018). Dopamine neurons 32(1), 56–67.
projecting to the posterior striatum reinforce Neugebauer, V. (2015). Amygdala pain mechanisms.
avoidance of threatening stimuli. Nature Neurosci- Handbook Experimental Pharmacology, 227,
ence, 21(10), 1421–1430. 261–284.
Miklos, I., & Kovacs, K. (2003). Functional heteroge- Nguyen, V. T., Breakspear, M., Hu, X., & Guo, C. C.
neity of the responses of histaminergic neuron (2016). The integration of the internal and exter-
subpopulations to various stress challenges. Euro- nal milieu in the insula during dynamic emotional
pean Journal of Neuroscience, 18(11), experiences. NeuroImage, 124(A), 455–463.
3069–3079. Oades, R. D., & Halliday, G. M. (1987). Ventral teg-
Miranda, M. I., & Bermudez-Rattoni, F. (1999). mental (A10) system: Neurobiology. 1. Anatomy
Reversible inactivation of the nucleus basalis mag- and connectivity. Brain Research, 434(2),
nocellularis induces disruption of cortical 117–165.
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 167

Orsetti, M., Casamenti, F., & Pepeu, G. (1996). Pecina, S., & Smith, K. S. (2010). Hedonic and moti-
Enhanced acetylcholine release in the hippocam- vational roles of opioids in food reward: Implica-
pus and cortex during acquisition of an operant tions for overeating disorders. Pharmacology
behavior. Brain Research, 724(1), 89–96. Biochemistry and Behavior, 97(1), 34–46.
Orsini, C. A., & Maren, S. (2012). Neural and cellular Pedersen, L. H., Scheel-Krüger, J., & Blackburn-
mechanisms of fear and extinction memory forma- Munro, G. (2007). Amygdala GABA-A receptor
tion. Neuroscience & Biobehavioral Reviews, 36(7), involvement in mediating sensory-discriminative
1773–1802. and affective-motivational pain responses in a rat
Osorio-Gomez, D., Bermudez-Rattoni, F., & Guzman- model of peripheral nerve injury. Pain, 127(1-2),
Ramos, K. R. (2021). Cortical neurochemical sign- 17–26.
aling of gustatory stimuli and their visceral Picciotto, M. R., Higley, M. J., & Mineur, Y. S. (2012).
consequences during the acquisition and consoli- Acetylcholine as a neuromodulator: Cholinergic
dation of taste aversion memory. Neurobiology of signaling shapes nervous system function and
Learning and Memory, 181, 107437. behavior. Neuron, 76(1), 116–129.
Osorio-Gomez, D., Guzman-Ramos, K., & Bermudez- Pidoplichko, V. I., Prager, E. M., Aroniadou-Ander-
Rattoni, F. (2017). Memory trace reactivation and jaska, V., & Braga, M. F. (2013). alpha7-Containing
behavioral response during retrieval are differen- nicotinic acetylcholine receptors on interneurons
tially modulated by amygdalar glutamate receptors of the basolateral amygdala and their role in the
activity: Interaction between amygdala and insular regulation of the network excitability. Journal of
cortex. Learning and Memory, 24(1), 14–23. Neurophysiology, 110(10), 2358–2369.
Ozawa, T., & Johansen, J. P. (2018). Learning rules for Pina, M. M., Pati, D., Hwa, L. S., Wu, S. Y., Mahoney,
aversive associative memory formation. Current A. A., Omenyi, C. G., & Kash, T. L. (2020). The
Opinion in Neurobiology, 49, 148–157. kappa opioid receptor modulates GABA neuron
Padlubnaya, D. B., Parekh, N. H., & Brown, T. H. excitability and synaptic transmission in midbrain
(2006). Neurophysiological theory of Kamin block- projections from the insular cortex. Neuropharma-
ing in fear conditioning. Behavioral Neuroscience, cology, 165. doi: 10.1016/j.neuropharm.2019.
120(2), 337–352. 107831
Panula, P., Pirvola, U., Auvinen, S., & Airaksinen, M. Poulin, J. F., Castonguay-Lebel, Z., Laforest, S., &
(1989). Histamine-immunoreactive nerve fibers in Drolet, G. (2008). Enkephalin co-expression with
the rat brain. Neuroscience, 28(3), 585–610. classic neurotransmitters in the amygdaloid com-
Pape, H. C., & Pare, D. (2010). Plastic synaptic net- plex of the rat. Journal of Comparative Neurology,
works of the amygdala for the acquisition, expres- 506(6), 943–959.
sion, and extinction of conditioned fear. Power, A. E., McIntyre, C. K., Litmanovich, A., &
Physiological Reviews, 90(2), 419–463. McGaugh, J. L. (2003). Cholinergic modulation of
Passani, M. B., Cangioli, I., Baldi, E., Bucherelli, C., memory in the basolateral amygdala involves acti-
Mannaioni, P. F., & Blandina, P. (2001). Histamine vation of both m1 and m2 receptors. Behavioral
H3 receptor-mediated impairment of contextual Pharmacology, 14(3), 207–213.
fear conditioning and in-vivo inhibition of cholin- Power, A. E., Vazdarjanova, A., & McGaugh, J. L.
ergic transmission in the rat basolateral amygdala. (2003). Muscarinic cholinergic influences in
European Journal of Neuroscience, 14(9), memory consolidation. Neurobiology of Learning
1522–1532. and Memory, 80(3), 178–193.
Passani, M. B., Giannoni, P., Bucherelli, C., Baldi, E., Provensi, G., Fabbri, R., Munari, L., Costa, A., Baldi,
& Blandina, P. (2007). Histamine in the brain: E., Bucherelli, C., & Passani, MB. (2017). Histamin-
Beyond sleep and memory. Biochemical Pharma- ergic neurotransmission as a gateway for the
cology, 73(8), 1113–1122. cognitive effect of oleoylethanolamide in contex-
Paulus, M. P., & Stein, M. B. (2006). An insular view tual fear conditioning. International Journal of
of anxiety. Biological Psychiatry, 60(4), 383–387. Neuropsychopharmacology, 20(5), 392–399.
Pearce, J. M. (1994). Similarity and discrimination: A Provensi, G., Passani, M. B., Costa, A., Izquierdo, I.,
selective review and a connectionist model. Psy- & Blandina, P. (2020). Neuronal histamine and the
chological Review, 101(4), 587–607. memory of emotionally salient events. British Jour-
Pearce, J. M., & Hall, G. (1980). A model for Pavlo- nal of Pharmacology, 177(3), 557–569.
vian learning: Variations in the effectiveness of Purón-Sierra, L., & Miranda, M. I. (2014). Histaminer-
conditioned but not of unconditioned stimuli. gic modulation of cholinergic release from the
Psychological Review, 87(6), 532–552. nucleus basalis magnocellularis into insular cortex
Pecina, S., & Berridge, K. C. (2005). Hedonic hot spot during taste aversive memory formation. PLoS
in nucleus accumbens shell: Where do μ-opioids One, 9(3). doi: 10.1371/journal.pone.0091120
cause increased hedonic impact of sweetness? Quirarte, G. L., Galvez, R., Roozendaal, B., &
Journal of Neuroscience, 25(50), 11777–11786. McGaugh, J. L. (1998). Norepinephrine release in
168 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

the amygdala in response to footshock and opioid fear conditioning. Genes, Brain and Behavior,
peptidergic drugs. Brain Research, 808(2), 14(1), 22–36.
134–140. Sah, P., Faber, E. S., Lopez De Armentia, M., & Power,
Ramirez-Lugo, L., Zavala-Vega, S., & Bermudez-Rat- J. (2003). The amygdaloid complex: Anatomy and
toni, F. (2006). NMDA and muscarinic receptors of physiology. Physiological Reviews, 83(3),
the nucleus accumbens have differential effects on 803–834.
taste memory formation. Learning and Memory, Salamone, J. D., Correa, M., Farrar, A., & Mingote,
13(1), 45–51. S. M. (2007). Effort-related functions of nucleus
Rasmussen, K., & Jacobs, B. L. (1986). Single unit accumbens dopamine and associated forebrain
activity of locus coeruleus neurons in the freely circuits. Psychopharmacology, 191(3),
moving cat. II. Conditioning and pharmacologic 461–482.
studies. Brain Research, 371(2), 335–344. Sample, C. H., Jones, S., Hargrave, S. L., Jarrard, L. E.,
Rasmusson, D., & Szerb, J. C. (1976). Acetylcholine & Davidson, T. L. (2016). Western diet and the
release from visual and sensorimotor cortices of weakening of the interoceptive stimulus control of
conditioned rabbits: the effects of sensory cuing appetitive behavior. Behavioural Brain Research,
and patterns of responding. Brain Research, 312, 219–230.
104(2), 243–259. Saper, C. B. (1982). Convergence of autonomic and
Rea, K., McGowan, F., Corcoran, L., Roche, M., & limbic connections in the insular cortex of the rat.
Finn, D. P. (2019). The prefrontal cortical endocan- Journal of Comparative Neurology, 210(2),
nabinoid system modulates fear-pain interactions 163–173.
in a subregion-specific manner. British Journal of Sarter, M., Lustig, C., Howe, W. M., Gritton, H., &
Pharmacology, 176(10), 1492–1505. Berry, A. S. (2014). Deterministic functions of cor-
Recorla, R. A., & Wagner, A. R. (1972). A theory of tical acetylcholine. European Journal of Neurosci-
Pavlovian conditioning: Variations in the effective- ence, 39(11), 1912–1920.
ness of reinforcement and nonreinforcement. In A. Sarter, M. F., & Bruno, J. P. (1994). Cognitive func-
H. Black & W. F. Prokasy (Eds.), Classical condition- tions of cortical ACh: Lessons from studies on
ing II: Current research and theory (pp. 64–99). trans-synaptic modulation of activated efflux.
New York: Appleton- Century-Crofts. Trends of Neuroscience, 17(6), 217–221.
Ricardo, J. A., & Koh, E. T. (1978). Anatomical evi- Sato, M., Ito, M., Nagase, M., Sugimura, Y. K., Taka-
dence of direct projections from the nucleus of the hashi, Y., Watabe, A. M., & Kato, F. (2015). The
solitary tract to the hypothalamus, amygdala, and lateral parabrachial nucleus is actively involved in
other forebrain structures in the rat. Brain the acquisition of fear memory in mice. Molecular
Research, 153(1), 1–26. Brain, 8. doi: 10.1186/s13041-015-0108-z
Rizvi, S. J., Pizzagalli, D. A., Sproule, B. A., & Ken- Schepers, S. T., & Bouton, M. E. (2017). Hunger as a
nedy, S. H. (2016). Assessing anhedonia in depres- context: Food seeking that is inhibited during
sion: Potentials and pitfalls. Neuroscience and hunger can renew in the context of satiety. Psy-
Biobehavioral Reviews, 65, 21–35. chological Science, 28(11), 1640–1648.
Roberts, W. A. (2019). The role of context in animal Schneider, M., Heise, V., & Spanagel, R. (2010). Dif-
memory. Learning and Behavior, 47(2), 117–130. ferential involvement of the opioid receptor antag-
Robertson, S. D., Plummer, N. W., de Marchena, J., & onist naloxone in motivational and hedonic
Jensen, P. (2013). Developmental origins of central aspects of reward. Behavioral Brain Research,
norepinephrine neuron diversity. Nature Neurosci- 208(2), 466–472.
ence, 16(8), 1016–1023. Schreurs, J., Seelig, T., & Schulman, H. (1986).
Robinson, L., Platt, B., & Riedel, G. (2011). Involve- β2-Adrenergic receptors on peripheral nerves.
ment of the cholinergic system in conditioning Journal of Neurochemistry, 46(1), 294–296.
and perceptual memory. Behavioral Brain Schultz, W. (2006). Behavioral theories and the neu-
Research, 221(2), 443–465. rophysiology of reward. Annual Review of Psychol-
Roozendaal, B., Hui, G. K., Hui, I. R., Berlau, D. J., ogy, 57, 87–115.
McGaugh, J. L., & Weinberger, N. M. (2006). Baso- Schultz, W. (2015). Neuronal reward and decision
lateral amygdala noradrenergic activity mediates signals: From theories to data. Physiology Review,
corticosterone-induced enhancement of auditory 95(3), 853–951.
fear conditioning. Neurobiology of Learning and Solinas, M., Goldberg, S. R., & Piomelli, D. (2008).
Memory, 86(3), 249–255. The endocannabinoid system in brain reward pro-
Rossi, M. A., & Stuber, G. D. (2018). Overlapping cesses. British Journal of Pharmacology, 154(2),
Brain Circuits for Homeostatic and Hedonic Feed- 369–383.
ing. Cell Metabolism, 27(1), 42–56. Starr, C. J., Sawaki, L., Wittenberg, G. F., Burdette, J.
Rozeske, R., Valerio, S., Chaudun, F., & Herry, C. H., Oshiro, Y., Quevedo, A. S., & Coghill, R. C.
(2015). Prefrontal neuronal circuits of contextual (2009). Roles of the insular cortex in the
NEUROTRANSMISSION AND NEUROMODULATION OF MEMORY 169

modulation of pain: insights from brain lesions. mouse insular cortex. Scientific Reports, 10. doi:
Journal of Neuroscience, 29(9), 2684–2694. 10.1038/s41598-020-64236-5
Stevenson, C. W., & Gratton, A. (2003). Basolateral Tronson, N. C., Schrick, C., Guzman, Y. F., Huh, K. H.,
amygdala modulation of the nucleus accumbens Srivastava, D. P., Penzes, P., & Radulovic, J. (2009).
dopamine response to stress: Role of the medial Segregated populations of hippocampal principal
prefrontal cortex. European Journal of Neurosci- CA1 neurons mediating conditioning and extinc-
ence, 17(6), 1287–1295. tion of contextual fear. Journal of Neuroscience,
Stubbendorff, C., Hale, E., Cassaday, H. J., Bast, T., & 29(11), 3387–3394.
Stevenson, C. W. (2019). Dopamine D1-like recep- Uematsu, A., Tan, B. Z., Ycu, E. A., Cuevas, J. S.,
tors in the dorsomedial prefrontal cortex regulate Koivumaa, J., Junyent, F., … Johansen, J. P. (2017).
contextual fear conditioning. Psychopharmacol- Modular organization of the brainstem noradrena-
ogy, 236(6), 1771–1782. line system coordinates opposing learning states.
Sun, K., Xiao, L., Wu, Y., Zuo, D., Zhang, C., Liu, S., Nature Neuroscience, 20(11), 1602–1611.
& Sun, T. (2020). GABAergic neurons in the insular Unal, C. T., Pare, D., & Zaborszky, L. (2015). Impact
cortex play an important role in cue-morphine of basal forebrain cholinergic inputs on basolateral
reward memory reconsolidation. Life Science, 254. amygdala neurons. Journal of Neuroscience, 35(2),
doi: 10.1016/j.lfs.2020.117655 853–863.
Swift, K. M., Gross, B. A., Frazer, M. A., Bauer, D. S., Unal, C. T., Pare, D., & Zaborszky, L. (2015). Impact
Clark, K. J. D., Vazey, E. M., & Poe, G. R. (2018). of basal forebrain cholinergic inputs on basolateral
Abnormal locus coeruleus sleep activity alters amygdala neurons. Journal of Neuroscience, 35(2),
sleep signatures of memory consolidation and 853–863.
impairs place cell stability and spatial memory. Usunoff, K. G., Itzev, D. E., Rolfs, A., Schmitt, O., &
Current Biology, 28(22), 3599–3609. Wree, A. (2006). Brain stem afferent connections
Takeuchi, T., Duszkiewicz, A. J., Sonneborn, A., of the amygdala in the rat with special references
Spooner, P. A., Yamasaki, M., Watanabe, M., to a projection from the parabigeminal nucleus: A
…Morris, R. G. (2016). Locus coeruleus and dopa- fluorescent retrograde tracing study. Anatomy and
minergic consolidation of everyday memory. Embryology, 211(5), 475–496.
Nature, 537, 357–362. Valdés, J. L., Sánchez, C., Riveros, M. E., Blandina, P.,
Tanimoto, S., Nakagawa, T., Yamauchi, Y., Minami, Contreras, M., Farías, P., & Torrealba, F. (2010). The
M., & Satoh, M. (2003). Differential contributions histaminergic tuberomammillary nucleus is critical
of the basolateral and central nuclei of the amyg- for motivated arousal. European Journal of Neuro-
dala in the negative affective component of science, 31, 2073–2085.
chemical somatic and visceral pains in rats. Euro- Valentino, R. J., & Van Bockstaele, E. (2015). Endog-
pean Journal of Neuroscience, 18(8), enous opioids: The downside of opposing stress.
2343–2350. Neurobiology of Stress, 1, 23–32
Taylor, K. M., & Snyder, S. H. (1971). Brain histamine: Van Diest, I. (2019). Interoception, conditioning, and
Rapid apparent turnover altered by restraint and fear: The panic threesome. Psychophysiology,
cold stress. Science, 172, 1037–1039. 56(8). doi: 10.1111/psyp.13421
Tian, J., Huang, R., Cohen, J. Y., Osakada, F., Kobak, Van Steenbergen, H., Eikemo, M., & Leknes, S.
D., Machens, C. K., & Watabe-Uchida, M. (2016). (2019). The role of the opioid system in decision
Distributed and mixed information in monosynap- making and cognitive control: A review. Cognitive,
tic inputs to dopamine neurons. Neuron, 91(6), Affective and Behavioral Neuroscience, 19(3),
1374–1389. 435–458.
Tinsley, M. R., Quinn, J. J., & Fanselow, M. S. (2004). Vertes, R. P., & Hoover, W. B. (2008). Projections of
The role of muscarinic and nicotinic cholinergic the paraventricular and paratenial nuclei of the
neurotransmission in aversive conditioning: Com- dorsal midline thalamus in the rat. Journal of Com-
paring Pavlovian fear conditioning and inhibitory parative Neurology, 508(2), 212–237.
avoidance. Learning and Memory, 11(1), 35–42. Wagner, A. R. (2003). Context-sensitive elemental
Torrealba, F., Riveros, M. E., Contreras, M., & Valdes, theory. Quarterly Journal of Experimental Psychol-
J. L. (2012). Histamine and motivation. Frontiers in ogy: Section B, 56(1), 7–29.
Systems Neuroscience, 6. doi: 10.3389/ Walling, S. G., Brown, R. A., Milway, J. S., Earle, A.
fnsys.2012.00051 G., & Harley, C. W. (2011). Selective tuning of hip-
Toyoda, H. (2019). Interaction of nicotinic acetylcho- pocampal oscillations by phasic locus coeruleus
line receptors with dopamine receptors in synaptic activation in awake male rats. Hippocampus,
plasticity of the mouse insular cortex. Synapse, 21(11), 1250–1262.
73(7). doi: 10.1002/syn.22094 Wassum, K. M., Cely, I. C., Balleine, B. W., & Maid-
Toyoda, H. (2020). CB1 cannabinoid receptor-medi- ment, N. T. (2011). Micro-opioid receptor activa-
ated plasticity of GABAergic synapses in the tion in the basolateral amygdala mediates the
170 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

learning of increases but not decreases in the Westbrook, R. F., Greeley, J. D., Nabke, C. P., & Swin-
incentive value of a food reward. Journal of Neu- bourne, A. L. (1991). Aversive conditioning in the
roscience, 31(5), 1591–1599. rat: Effects of a benzodiazepine and of an opioid
Wassum, K. M., Cely, I. C., Maidment, N. T., & Bal- agonist and antagonist on conditioned hypoalge-
leine, B. W. (2009a). Disruption of endogenous sia and fear. Journal Experimental Psychology:
opioid activity during instrumental learning Animal Behavior Processes, 17(3), 219–230.
enhances habit acquisition. Neuroscience, 163(3), Williams, C. L., Men, D., Clayton, E. C., & Gold, P. E.
770–780. (1998). Norepinephrine release in the amygdala
Wassum, K. M., Ostlund, S. B., Maidment, N. T., & after systemic injection of epinephrine or escapa-
Balleine, B. W. (2009b). Distinct opioid circuits ble footshock: Contribution of the nucleus of the
determine the palatability and the desirability of solitary tract. Behavior Neuroscience, 112(6),
rewarding events. Proceedings of the National 1414–1422.
Academy of Sciences of the United States of Wilson, M. A., & Fadel, J. R. (2017). Cholinergic regu-
America, 106(30), 12512–12517. lation of fear learning and extinction. Journal of
Watabe-Uchida, M., Eshel, N., & Uchida, N. (2017). Neuroscience Research, 95(3), 836–852.
Neural circuitry of reward prediction error. Annual Woolf, N. J. (1991). Cholinergic systems in mamma-
Review of Neuroscience, 40, 373–394. lian brain and spinal cord. Progress in Neurobiol-
Watabe, A. M., Ochiai, T., Nagase, M., Takahashi, Y., ogy, 37(6), 475–524.
Sato, M., & Kato, F. (2013). Synaptic potentiation Yamasaki, M., & Takeuchi, T. (2017). Locus coeruleus
in the nociceptive amygdala following fear learn- and dopamine-dependent memory consolidation.
ing in mice. Molecular Brain, 6. doi: Neural Plasticity, 2017. doi: 10.1155/2017/
10.1186/1756-6606-6-11 8602690
Weber, S. C., Beck-Schimmer, B., Kajdi, M. E., Müller, Young, S. L., & Fanselow, M. S. (1992). Associative
D., Tobler, P. N., & Quednow, B. B. (2016). Dopa- regulation of Pavlovian fear conditioning: uncon-
mine D2/3-and μ-opioid receptor antagonists ditional stimulus intensity, incentive shifts, and
reduce cue-induced responding and reward impul- latent inhibition. Journal of Experimental Psychol-
sivity in humans. Translational Psychiatry, 6(7). doi: ogy: Animal Behavior Processes, 18(4),
10.1038/tp.2016.113 400–413.
Weinberger, N. M., Miasnikov, A. A., & Chen, J. C. Zarrindast M.R., Eidi M., Eidi A., & Oryan S. (2002)
(2006). The level of cholinergic nucleus basalis Effects of histamine and opioid systems on
activation controls the specificity of auditory asso- memory retention of passive avoidance learning in
ciative memory. Neurobiology of Learning and rats. European Journal of Pharmacology, 452(2),
Memory, 86(3), 270–285. 193–197.
11
Neuromodulator Adenosine A2A
Receptor Control of Cognition
J i a n g - F a n C h e n a n d Ya n L i

INTRODUCTION A2A receptors to the striatopallidal neurons, the


molecular interaction with other neurotransmit-
Adenosine is a ubiquitous nucleotide throughout ters such as dopamine and glutamate signaling
the body that is directly involved in the key intra- systems and their ability to modulate (striatal)
cellular processes such as the energy metabolism synaptic plasticity led us that the A2AR represents
and biosynthesis of DNA and RNA. With this a novel target for improving cognition in normal
intrinsic link to cellular metabolism, adenosine is and disordered brain. With the recent approval of
also widely used as an extracellular signal by the A2AR antagonist istradefylline for treatment of
acting at four G-protein coupled receptors, namely OFF-episode Parkinson’s disease (PD) by US FDA
adenosine A1, A2A, A2B, and A3 receptors (Borea (Chen and Cunha, 2020), we proposed to translate
et al., 2018; Fredholm et al., 2011). In the brain the cognitive enhancement of A2AR antagonists in
where adenosine receptors are located more abun- rodents and non-human primates, and the safety
dantly than in other mammalian organs, adenosine profile of A2AR in clinical phase IIB-III trials and
functions primarily as a neuromodulator. In par- to reevaluate and repurpose the ability of the A2AR
ticular, adenosine is traditionally considered an antagonist istadefylline to control cognitive dete-
inhibitory neuromodulator to decrease in the activ- rioration in Alzheimer’s disease (AD) and PD.
ity of excitatory synapses via feedback mechanism
(Cunha, 2016; Dunwiddie and Masino, 2001).
A2A receptors (A2AR) are expressed at high
levels in the striatum (dorsal striatum and nucleus STRIATOPALLIDAL A2A RECEPTORS
accumbens) (Svenningsson et al., 1999) and olfac- CONTROL THE NO-GO PATHWAY TO
tory tubercle and at low levels in neurons outside
the striatum and in glial cells (Christofi et al.,
EXERT “BRAKE” CONTROL OVER A WIDE
2001). Adenosine A2A receptors are also one of the RANGE OF COGNITIVE FUNCTIONS
best characterized GPCRs (Carpenter and Lebon,
2017; Goldfeld et al., 2011; Hino et al., 2012), A2ARs are abundantly expressed in the striato-
among the first to be crystalized and with 3D struc- pallidal medium spiny neurons (MSNs;
turally analyzed at nm resolution. As discussed in Svenningsson et al., 1999), 20-folds higher than
detail below, the selective cellular localization of other cells and tissues. A2ARs are located
172 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

predominantly on the axon terminal and cell of the at corticostriatal terminals, which is attenuated
striatopallidal neurons which constitute the output by the D2R but enhanced by A2AR activation
projection pathway (i.e., striatopallidal pathway) (Higley and Sabatini, 2010). Thus, the striato-
from the striatum to the external segment of the pallidal pathway activity is regulated by multiple
globus pallidus (GPe). This selective expression A2AR-mediated actions via cAMP/PKA signaling
pattern is key to understanding the potential of mechanisms (Mori, 2020): (a) generation of mIP-
A2AR antagonism in the control of motor as well SCs and (b) counting D2R inhibition of calcium
as cognition. Striatal GABAergic projection neu- entry of striatopallidal neurons, (c) GABA release
rons are divided into the striatonigral pathway in striatal collaterals and GPe output projections.
(expressing dopamine D1 receptor) that receives A2AR antagonists and adenyly cyclase or PKA
preferential innervation from the sensory cortical inhibitors blocked these effects, resulting in over-
and limbic structures and projects directly to sub- all reduction of excitability of striatopallidal neu-
stantia nigra pars reticulate (SNr), and the striato- rons with increased GABA release in GPe. Thus,
pallidal pathway (expressing dopamine D 2 the striatopallidal A2ARs are uniquely positioned
receptor, D2R and A2AR) that receives preferential to selectively modulate the activity of the ‘NoGo’
innervation from motor cortex and projects to the pathway (Mori, 2020).
external globus pallidus (GPe) and indirectly to In functional terms, A2AR antagonism relieves
SNr via subthalamic nucleus(Gerfen et al., 1990; the ‘NoGo’ signal (a ‘brake’) and facilitating the
Wall et al., 2013). Striatopallidal projection neu- D2R effects. In contrast to most of neuropharmaco-
rons are characterized electrophysiologically by logical manipulations which often result in disrup-
the strengthened and more excitable neural firing tion of normal behaviors, this “brake” mechanism
patterns (Yin et al., 2009) and by specific form of by striatopallidal neurons enable A2AR antago-
synaptic plasticity, long-term depression (LTD) nists to facilitate motor and a range of cognitive
(W. Shen et al., 2008). At behavioral level, the behaviors, including working memory (Li et al.,
striatonigral pathway facilitates (i.e., “Go” pathway) 2018), reversal learning (Amodeo et al., 2018),
and the striatopallidal pathway inhibits (i.e., Pavlovian fear conditioning (Simoes et al., 2016),
“No-Go” pathway) motor and motivational behav- set-­shifting (Zhou et al., 2019), and goal-directed
iors (Kreitzer and Malenka, 2008). Accordingly, behavior (Li et al., 2016, 2018). This feature also
pharmacogenetic (Ferguson et al., 2011) and makes the A2AR antagonist a novel therapeutic
optogenetic (Kravitz et al., 2012) activation of strategy for improving cognitive behaviors under
striatopallidal pathways can suppress psycho- various neuropsychiatric conditions.
stimulant sensitization and motivation-related It should be noted that A2ARs in brain areas
behaviors, including motor skill learning (Durieux outside the striatum may differently control cog-
et al., 2012), the selection accuracy of learned nitions through distinct modulatory mechanisms
motor action (Nishizawa et al., 2012), and depending on different neural networks. Thus,
decision-making in a probabilistic switching task A2ARs in forebrain have been shown to exert oppo-
(Tai et al., 2012), sequence-learning initiation and site effects on working memory (Li et al., 2018),
execution (Tecuapetla et al., 2016), and the fear conditioning (Simoes et al., 2016; Wei et al.,
response initiation of instrumental behaviors. 2014) and psychomotor activity (H.Y. Shen et al.,
Striatopallidal A2ARs are positively coupled to 2008, 2013) than the striatal A2ARs. This differen-
Gs protein to activation adenylyl cyclase (Chen tial effects by striatal versus extra-striatal A2ARs
et al., 2014), resulting in the increase in the confers the A2AR an unique ability to balance each
3’,5’-cyclic adenosine monophosphate (cAMP)/ of cognitive behavioral elements from becoming
PKA level. A2AR agonist (CGS21680) or an extreme and rigidity control.
adenyly cyclase activator forskolin significantly
increased cAMP accumulation, and activate
the miniature inhibitory postsynaptic currents
(mIPSCs) frequency of striatopallidal neurons,
resulting inhibition of GABA release in the GPe STRIATOPALLIDAL A2A RECEPTORS
(Fredholm et al., 2007; Morales-Figueroa et al., INTEGRATE CORTICAL GLUTAMATE
2019; Shindou et al., 2002). However, A2AR AND NIGRAL DOPAMINE SIGNALS
activation also inhibit GABA release from intra-
striatal collaterals, leading to excitation of the
TO CONTROL STRIATAL SYNAPTIC
striatopallidal neurons (Mori et al., 1996). The PLASTICITY
study with combined optogenetics, two-photon
microscopy, and glutamate uncaging shows that Synaptic plasticity, including long-term potentia-
this PKA-dependent mechanism result in Ca2 + tion (LTP) and long-term depression (LTD), is
entry through NMDA-type glutamate receptors considered the neurophysiological basis of
NEUROMODULATOR ADENOSINE 173

memory (Martin et al., 2000; Neves et al., 2008). A2ARs impair LTP at corticoaccumbal synapses
A2ARs are expressed at the cortico-striatal synapse (d’Alcantara et al., 2001) and spike-timing-
(i.e., post-synaptic sites of the striatopallidal neu- dependent LTP at glutamatergic synapses onto
rons) and in synapses throughout the rest of the striatopallidal neurons (H.Y. Shen et al., 2008).
brain including excitatory (glutamatergic) syn- In the setting of FGFR coactivation, A2AR activa-
apses (Rebola et al., 2005; Tetzlaff et al., 1987), tion also promotes LTP at the corticostriatopalli-
GABAergic (Cunha and Ribeiro, 2000; Rombo dal synapses through a PKA-dependent pathway
et al., 2015; Shindou et al., 2002), and probably (Flajolet et al., 2008).
other synapses. At the postsynaptic sites, striato- LTD: LTD is the predominant and best-
pallidal A2ARs interact postsynaptically with D2Rs characterized form of long-term synaptic plastic-
(Canals et al., 2003; Hillion et al., 2002) in an ity that is restricted to striatopallidal MSNs where
antagonistic manner as A2AR activation inhibits A2ARs are highly concentrated in the striatum.
D2R binding in the striatum, D2R-mediated neuro- In particular, endocannabinoid (eCB)-dependent
transmitter release, and immediate early gene LTD is found to be restricted to striatopallidal
expression (Ferre et al., 1991, 1997). Functional MSNs and is dependent on endocannabinoid
A2AR-D2R antagonistic interaction is further sup- release and D2R activation (Kreitzer and Malenka,
ported by the demonstration of A2AR-D2R heter- 2007). A2ARs interact with both D2Rs and CB1Rs,
odimers and A2AR-mGluR5 heterodimers using likely contribute to the modulation of striatal LTD.
FRET and coimmunoprecipitation analyses Indeed, we showed that A2AR activity preferen-
(Canals et al., 2003) and proximity ligation assay tially modulates LTD in striatal slices from R6/2
(PLA) analysis. There is also evidence for the pos- mice, a genetic model of Huntington’s disease
sible postsynaptic A2AR-NMDAR crosstalk since (Li et al., 2015; Villar-Menendez et al., 2013).
A2ARs can control expression, recruitment, and This impact of postsynaptic striatal A2AR activ-
the rate of desensitization, phosphorylation of ity on striatal LTD is key to our understanding of
NMDAR and NMDA current (Franco et al., 2020; A2AR control of cognition.
Kouvaros and Papatheodoropoulos, 2016; LTD-LTP shift: Interestingly, concomitant
Temido-Ferreira et al., 2020). In additional, post- stimulation of A2AR and D2R changes the stri-
synaptic A2ARs in the striatopallidal neurons syn- atopallidal MSN plasticity from LTD to LTP
ergistically interaction with metabotropic (W. Shen et al., 2008) and modulate a form of
glutamate 5 receptors (Coccurello et al., 2004; striatal eCB-LTD that is dependent on endocan-
Ferre et al., 2002; Kachroo et al., 2005) and can- nabinoid release and D2R activation (Kreitzer and
nabinoid CB1 receptors (Lerner et al., 2010) in the Malenka, 2007). Notably, overactivation of the
modulation of NMDAR-mediated effects. At the A2AR under condition of aging and stress can led to
presynaptic sites, A2ARs are also present at cor- the LTD-to-LTP shift and increased NMDA recep-
tico-striatal projection terminals (Rebola et al., tor gating in hippocampus (Temido-Ferreira et al.,
2005; Rosin et al., 2003) to modulate glutamate 2020). Accordingly, A2AR blockade reverse this
release (Ciruela et al., 2006) and at striatopallidal plasticity shift and accompanied cognitive impair-
output projection terminals as well as striatal col- ment (Temido-Ferreira et al., 2019). It is sug-
laterals to modulate GABA release (Gomez- gested that mGluR5 may act as a switch between
Castro et al., 2021; Morales-Figueroa et al., 2019). the A2AR and NMDAR by sensing glutamate
As NMDAR activity is required for long-term and translating it into NMDAR overactivation as
potentiation (LTP and LTD) and D2R activation mGluR5 blockade prevents the LTD-to LTP shift
is critical to induction of LTD, the postsynaptic and the NMDAR overactivation (Temido-Ferreira
interaction between A2AR and D2R and mGluR5/ et al., 2020).
NMDAR, possibly through A2AR-D2R heterodi-
mer formation, provide a strong anatomical and
molecular bases for A2AR control of striatal syn-
aptic plasticity and cognition. Through these post- A2A RECEPTOR ACTIVITY IS NECESSARY
synaptic and presynaptic mechanisms, adenosine
has been shown to provide homeostatic modula-
AND SUFFICIENT TO TRIGGER MEMORY
tion of synapse by providing the permissive condi- IMPAIRMENT
tion to set the stage for Hebbian forms of plasticity
(Dias et al., 2013). The ability of the striatopallidal A2AR to integrate
LTP: Striatal A2AR activity did not affect short- dopamine and glutamate signaling to control syn-
term plasticity (such as the paired-pulse facilita- aptic plasticity and to exert the “brake” control of
tion index of presynaptic function) but plays an cognition uniquely position the A2AR as important
important role in modulating striatal LTP since therapeutic target for modulating cognitive behav-
pharmacological blockade or gene deletion of iors in normal and neuropsychiatric disorders.
174 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

A growing body of evidence indicates that A2AR animals. Given the A2AR antagonists relieves
activation inhibits various cognitive behaviors in the ‘NoGo’ in the striatum and increase the stri-
normal mice (Cunha et al., 2008; Ferre et al., atopallidal neuron activity, this “brake” effect of
2008; Schiffmann et al., 2007; H.Y. Shen et al., striatal A2AR activation collaborates with the find-
2008). For example, A2AR activation in the cingu- ing that the activity of the striatopallidal pathway
late cortex impairs memory retrieval (Pereira provides inhibitory control for amphetamine sen-
et al., 2005) and social recognition memory, sitization (Bateup et al., 2010), instrumental learn-
which is prevented by A2AR antagonists (Prediger ing (Lobo et al., 2007; Yu et al., 2009), addiction
et al., 2005; Prediger and Takahashi, 2005). Over- (Durieux et al., 2009; Lobo et al., 2010), and goal-
expression of A2ARs in the cortex impairs spatial oriented behavior (Yu et al., 2009). These findings
working memory without affecting spatial refer- not only herald the notion that the suppression of
ence memory (Gimenez-Llort et al., 2007). A2AR activity is pro-cognitive, but also support
Optogenetic activation of A2AR signaling in hip- the possibility that in contrast to targeting hip-
pocampus is sufficient to trigger CREB phospho- pocampus-cortex by cholinesterase inhibitors, the
rylation and cognitive impairments (Li et al., selective localization of A2AR in the striatopallidal
2015). Optogenetic activation of A2AR signaling pathway would provide a novel and promising tar-
in the striatopallidal neurons suppress value sensi- get for selectively alleviating cognitive deficits in
tivity and inhibit goal-directed behavior as dem- AD and PD.
onstrated by increased habit formation (Li et al., However, conditional striatum-specific A2AR
2016). Similarly, the pharmacological activation knockout did not affect latent inhibition (LI)
of A2AR in the hippocampus (Pagnussat et al., and prepulse inhibition (PPI), two common
2015) or cingulate cortex (Pereira et al., 2005) can cross-species translational behavior tests for the
trigger memory impairment. assessment of selective attention and sensori-
Conversely, using three different A2AR KO motor gating deficits reported in schizophrenia
models (global, forebrain-specific and striatum- patients (Singer et al., 2013). It should be noted
specific A2AR KO), we recently showed that inac- that A2ARs in the brain regions outside striatum or
tivation of striatal A2ARs is sufficient to enhance in glial cells have been shown to produce oppo-
working memory (Wei et al., 2011; Zhou et al., site effects on cognitions (H.Y. Shen et al., 2008,
2009), reversal learning (Wei et al., 2011), goal- 2013). For example, selective deletion of the
directed behavior (Yu et al., 2009), and Pavlovian astrocytic A2AR gene trigger working memory
fear conditioning (Wei et al., 2014), Pavlovian- deficits and the exacerbated responses to psy-
to-Instrumental Transfer (Li et al., 2020), with- chosis-inducing drugs such as MK-801 (Matos
out affecting spatial reference memory, motor et al., 2015). Thus, astroglial and neuronal A2AR
function, and anxiety-like behaviors. The pro- interplay critically control glutamate homeosta-
cognitive effects of A2AR inactivation were dem- sis and provide a mechanistic key link between
onstrated using multiple behavioral tests: Eight astrocytic dysfunction and abnormal psychomotor
arm radial maze (working memory and reference and cognitive behavior. This indicates that rather
memory), the Morris water maze (reversal learn- than through the postulated antagonistic interac-
ing, working memory), instrumental learning tion between A2AR and D2Rs in neurons (Boison
(goal-directed behavior) and Pavlovian fear condi- et al., 2012; Rimondini et al., 1997; Shen et al.,
tioning and motor sequence learning. These cogni- 2012), astroglial A2AR may directly affect gluta-
tive enhancement in WM, goal-directed behavior mate transport activity as a primary target to drive
and motor sequence learning can be achieved an astrocyte-to-neuron communication, leading
similarly by pharmacological blockade of the ultimately to the deregulated glutamate neuro-
A2AR (He et al., 2021; Li et al., 2018). Notably, transmission and cognitive impairments.
this precognitive effect of the A2AR can be isolated
to the striatopallidal A2ARs since selective dele-
tion of striatal A2AR (st-A2AR-KO) was sufficient
to bolster WM performance (Wei et al., 2011), MULTIPLE BEHAVIORAL MECHANISMS
goal-directed behavior and fear conditioning
(Wei et al., 2014).
MAY CONTRIBUTE TO STRIATAL A2A
Thus, A2AR in striatopallidal neurons may RECEPTOR CONTROL OF COGNITION
serve as an inhibitory control system for a range
of cognitive behaviors, including working mem- How does the striatal A2AR bring changes in cog-
ory (Li et al., 2018), reversal learning (Amodeo nitions by modifying behavioral elements? Recent
et al., 2018), Pavlovian fear conditioning (Simoes studies coupled well-controlled, sophisticated
et al., 2016), Set-shifting (Zhou et al., 2019), goal- behavioral paradigms with pharmacological,
directed behavior (Li et al., 2016, 2018), and motor genetic, and optogenetics approaches have uncov-
sequence learning (He et al., 2021) in normal ered multiple distinct behavioral elements/
NEUROMODULATOR ADENOSINE 175

mechanisms underlying striatal A2AR control of enhancing Pavlovian-to-instrumental transfer and


cognitive processes. thereby promoted reward approaching behaviors,
that is invigorating the performance of instrumen-
1 Value sensitivity versus action-outcome contin- tal actions to gaining rewards (Li et al., 2020). This
gency: One important finding is that striatal A2AR finding collaborates with recent study that the NAc
affect neither acquisition of instrumental behav- played a critical role in the motivational aspect of
iors during the random interval training and out- instrumental behavior by a Pavlovian-instrumental
come-contingency as evident by the omission test transfer (Yin et al., 2008). Thus, accumbal A2AR
(Li et al., 2016, 2018), nor the encoding of working activity also plays an important role in linking
memory information during the sample phase value information with the sensory features of the
(Li et al., 2018). Rather, striatal A2AR activity modi- reward or reward-related cues and A2AR control
fied the animal’s sensitivity to the value of the of goal-directed behavior by modulating animal’s
reward as evident by devaluation test. These results adaptive utility of cue-motivated behavior.
are in line with the findings that A2AR antagonism 4 Behavioral inhibition: Down regulation of A2ARs
and genetic deletion can improve effort-based expression by shA2AR in the medial prefrontal
decision making (Lopez-Cruz et al., 2018; Pardo cortex which innervates NAc impaired the per-
et al., 2012). The ability to control the animal’s formance of rat in a cost-benefit decision-making
sensitivity to the value of the outcome may have task by controlling impulsive behavior (Leffa
therapeutic implication since the deficits in this et al., 2018). On the other hand, selective dele-
NAc incentive processes can produce a range of tion of the striatal A2ARs apparently improved
motivational dysfunctions (Salamone et al., 2016). the control of impulsive behavior using the
2 Motivation: As the striatum (nucleus accumbens omission test (Yu et al., 2009). Furthermore,
in particular) is the critical locus for motivational nucleus accumbal A2AR knockdown apparently
control, inhibition of the striatopallidal pathway modulated the reversal learning with the reduced
in either the NAc or dorsomedial striatum (DMS) regressive and perseverative errors, indicating
leads to enhanced motivation (Carvalho Poyraz the enhanced control of impulsive behavior
et al., 2016), The view that NAc A2ARs may affect accumbal A2AR (Zhou et al., 2019). Thus, the
goal-directed behavior by modulating the motiva- A2ARs in the medial prefrontal cortex and stria-
tion is supported by the finding that pharmaco- tum may exert different effects on impulsive
logical blockade of A2AR increases the breakpoints behavior for control of goal-directed behavior.
and total lever presses in the progressive ratio 5 Mood-related behaviors: Chronic stress shifted
test (Li et al., 2020; Zhou et al., 2019), indicating behavioral control from goal-directed toward
increased effort-related motivation that mice were habitual strategy (Radenbach et al., 2015),
willing to expend to earn a reward. In the progres- mainly through corticostriatal pathway involving
sive-hold down test, the A2AR antagonist KW6002 the dorsomedial striatum and nucleus accumbal
produces a greater number of responses, with the circuits (Dias-Ferreira et al., 2009; Taylor et al.,
main responding increases in the short durations 2014). Importantly, caffeine and A2AR specific
(<2 s). This increased general motivational effect antagonists can prevent mood and memory dys-
(with increased vigor) may explain an enhanced function triggered by chronic stress (Kaster et al.,
sensitivity to reinforcement and rapid initiation of 2015). Thus, A2AR signaling in NAc might modu-
lever pressing. Thu, A2AR antagonist may modulate late the goal-directed behavior and the decision-
goal-directed behavior by modifying motivation making processes triggered by mood-related
with increased vigor/responding rate and by invig- behavior, which need to be further investigated.
orating the performance of instrumental actions
to gain rewards.
3 Cue-motivated behavior: Cue-(evoked) Pavlovian
incentive learning mediates the ability of a stimulus ABERRANT INCREASE OF A2A
previously paired with a reward to acquire incen- RECEPTOR SIGNALING IN AGED AND
tive motivational properties and thereby enhances NEURODEGENERATIVE BRAINS
reward approaching behaviors, as well as invig-
orates the performance of actions to gaining Increasing evidence have documented that patho-
rewards (Aitken et al., 2016). Using a Pavlovian- logical brain conditions associated with memory
to-instrumental transfer test, Li et al. found that impairment (such as AD, stress, and inflamma-
NAc A2ARs controlled goal-directed behavior by tion) are characterized by a local increase of the
176 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

extracellular levels of adenosine and up-regulation synaptotoxicity after intracerebroventricular Aβ


and aberrant signaling of brain A2ARs (Chen et al., injection (Canas et al., 2009; Cunha et al., 2008;
2013; Cunha and Agostinho, 2010). A2ARs is up- Dall’Igna et al., 2007). Interestingly, long-term
regulated in the brain of AD patients, detectable (between early 4-9 months) administration of a
by radioligand binding assay at early Braak stages 1.5 mg daily dose of caffeine (equivalent to 500 mg
(Albasanz et al., 2008). A recent study demon- in human) to AD transgenic (APPsw, Swedish
strated that human with AD had increased level of mutation) mice reduced brain A levels and protect
A2ARs selectively in astrocytes which may con- against certain cognitive deficits (Arendash et al.,
tribute to memory impairments in AD (Orr et al., 2006, 2009; Cao et al., 2009). A similar protective
2015). Aging also induced upregulation of A2ARs effect of the A2AR antagonist SCH58261 was also
in hippocampal neurons of aged humans, a pheno- observed in AD transgenic (3xTgAD and APP/
type further exacerbated in AD patients PS1) mouse models (Silva et al., 2018; Viana da
(Temido-Ferreira et al., 2020). Furthermore, the Silva et al., 2016). Furthermore, conditional
density of hippocampal A2AR increases in aged genetic deletion of astrocytic A2AR enhanced
animals (Canas et al., 2009; Cunha et al., 1995; long-term memory in young and aging mice.
Lopes et al., 1999; Rebola et al., 2003) as well as Conversely, chemogenetic activation of astrocytic
in transgenic mice displaying memory impair- Gs-coupled signaling reduced long-term memory
ments (Cognato et al., 2010; Cunha et al., 2006; in mice without affecting learning (Orr et al.,
Espinosa et al., 2013). 2015). A recent study further demonstrates that
Similarly, there is aberrant increase of A2AR genetic deletion of or oral therapy of A2AR antago-
signaling in postmortem brains of PD models nists protect THY-Tau22 mice from Tau
and human PD patients. For example, dopa- pathology-induced deficits in spatial memory and
mine depletion by mitochondrial neurotoxins hippocampal plasticity, along with reduced Tau
such as 6-hydroxydopamine (6-OHDA) and hyperphosphorylation (Laurent et al., 2016).
MPTP increased ATP release and upregulated Thus, A2AR blockade and caffeine can reverses
ecto-5’-nucleotidase (CD73) and A2ARs in stri- memory impairments in AD models.
atal synaptosomes (Carmo et al., 2019) and in In animal models of PD, dopamine depletion in
the striatum of MPTP-treated mice (Meng et al., the prefrontal cortex produces cognitive deficits
2019). A2AR upregulation in neurons and glial in executive dysfunction (Brozoski et al., 1979;
cells of hippocampus is also induced by intra- Clinton et al., 2006), visuo-spatial and non-spatial
hippocampal injection of A53T α-Syn fibrils working memory that resemble those seen in PD
(Hu et al., 2016). Furthermore, A2AR upregulation (Decamp and Schneider, 2004). The A2AR antago-
has been documented in putamen of early stage nist istradefylline reverses these cognitive defi-
of PD (Braak stage 1-2) (Villar-Menendez et al., cits, with increased level of dopamine content in
2014) and in PD with dyskinesia (compared to PD prefrontal cortex (Kadowaki Horita, et al., 2013).
patients without dyskinesia) by positron emission A2AR antagonists or A2AR KO reverse working
tomography (PET) studies (Mishina et al., 2011; memory deficits induced by intracerebral injec-
Ramlackhansingh et al., 2011). tion of alpha-synuclein (Ko et al., 2016). rescue
Therefore, increased adenosine-A2AR signal- impairment of LTP associated with α-Syn expo-
ing represents brain response to insults and thus a sure through an NMDA receptor-dependent mech-
novel therapeutic target for pharmacological inter- anism (Durante et al., 2019).
vention to reverse cognitive impairment in AD In animal models of Huntington’s disease,
and PD. A2AR blockade reverse working memory defi-
cits with selective reversal of LTD (but not LTP)
( Li et al., 2015). In animals chronically exposed
to cannabinoid (WIN 55,212-2), A2AR antago-
A2A RECEPTOR ANTAGONISTS nists also revert memory deficits (as evident on
REVERSE COGNITIVE IMPAIRMENTS administration of the Novel Object Recognition
Test) and partially rescue impaired LTP of syn-
IN ALZHEIMER’S AND PARKINSON’S
aptic potentials recorded from the CA1 area of
DISEASE MODELS the hippocampus (Mouro et al., 2019). In cortical
controlled impact model of traumatic brain injury,
A2AR antagonists can not only enhance cognition A2AR antagonists improved working memory
in normal animals but also reverse cognitive deficits with reduced neuroinflammation (Ning
impairments in AD and PD models. The selective et al., 2013, 2019). Interestingly, a recent study
A2AR antagonist SCH58261 and non-specific show that A2AR overexpression triggers the HPA-
adenosine receptor antagonist caffeine prevent axis dysfunction, including loss of plasmatic cor-
Aβ-induced cognitive impairments and ticosterone circadian oscillation, and promotes
NEUROMODULATOR ADENOSINE 177

reduction of GR hippocampal levels (Batalha drinking was associated with a reduced severity of
et al., 2016) and memory impairments. This the mood/cognition domain of NMSS (p = 0.003)
memory and HPA-axis dysfunction was mecha- (Cho et al., 2018). Curiously, caffeine is report-
nistically accompanied by a hippocampal LTD- edly to enhance the consolidation of long-term
to-LTP shift and increased NMDA receptor gating memories in humans, 24 hours after a one-dose
(Temido-Ferreira et al., 2020). Accordingly, A2AR (200mg) administration (Borota et al., 2014).
blockade reverse this plasticity shift and dysfunc- Despite the considerable strength of the corre-
tion in HPA-axis (Batalha et al., 2016), suggesting lation in these large, longitudinal studies, epidemi-
that the pro-cognitive effects of A2AR antagonists ological investigation cannot definitively isolate
on age-related cognitive impairments may be caffeine intake from other lifestyle choices that
associated with its ability to modulate HPA-axis. potentially affect cognition. In fact, a recent clini-
cal trial with caffeine in PD has failed to confirm
a procognitive effect of caffeine in PD patients
(Postuma et al., 2017). Thus, it is importance to
HUMAN STUDIES SUPPORT POSSIBLE further investigate the effect of chronic treatment
PRO-COGNITIVE EFFECTS OF CAFFEINE of caffeine on cognition in AD and PD.
Lastly, a human gene-based association analy-
IN AGING AND NEURODEGENERATIVE sis in cognitively normal and impaired partici-
pants reported that a minor allele rs9608282 of
Caffeine is doubtless the most widely consumed the ADORA2A gene is associated with larger hip-
psychoactive substance by more than 50% of the pocampal volumes and better memory in mild AD
world’s adult population, largely for its psycho- patients (Horgusluoglu-Moloch et al., 2017).
stimulant (and presumably cognitive enhance-
ment) effect. Importantly, the cognitive beneficial
effects of caffeine attainable by regular human
consumption (around 200–300mg caffeine or 3–5
cups of coffee/day) are largely attributed to the SUMMARY AND CONCLUSIONS
antagonism of the brain A2AR (Cunha and
Agostinho, 2010; Ferre, 2008; Fredholm et al., In summary, there is a striking convergence of
2005). At least six longitudinal studies support an epidemiological evidence from multiple longitu-
inverse relationship between caffeine consump- dinal studies strongly establishing an inverse
tion and decreased memory decline associated relationship between chronic caffeine consump-
with aging as well as a reduced risk of developing tion in humans and cognitive decline in aging, AD
AD, including the Masssatricht Aging Study and PD, with the animal studies demonstrating
(Hameleers et al., 2000; van Boxtel et al., 2003), that selective A2AR antagonists and caffeine have
the Canadian Study of Health and Aging (CSHA) the strong pro-cognitive impact in normal as well
study (Lindsay et al., 2002), the Finland, Italy, as aging and AD and PD models. This led us
Netherlands Elderly (FINE) study (van Gelder to postulate that propose that in addition to
et al., 2007), the French Three Cities study hippocampal-cortex circuits, it may be possible to
(Ritchie et al., 2007), the Cardiovascular risk fac- selectively target the striatopallidal pathway of the
tors, Aging and Dementia (CAIDE) study striatum to improve memory impairment in aging
(Eskelinen et al., 2009), and the Honolulu-Asia and AD. The validity of this target is supported by
Aging study (Gelber et al., 2011). Human studies the up-regulation of A2ARs in the brain of AD and
show that plasma levels of caffeine in individuals PD patients (including astrocytes) and by the abil-
with mild cognitive impairment (MCI) who later ity of A2AR antagonists to enhance conditioning in
progress to dementia are lower than in those normal and disease models including AD and PD.
whose MCI remains stable (Solfrizzi et al., 2015). The confirmation of this hypothesis will constitute
For example, the Honolulu-Asia Aging Study a novel view of the control of cognition including
involved 3494 men found that the men in the working memory and goal-directed behaviors by
highest quartile of caffeine intake were less likely the targeting striatal A2ARs.
than men in the lowest quartile to have any neuro- Over at least the last 15 years, at least six
pathologic lesions at death in the 226 men with double-blind, placebo controlled clinical trials of
dementia and the 347 men with cognitive impair- istradefylline (KW-6002), altogether involving
ment who underwent brain autopsy (Gelber a total of >2500 advanced PD patients and one
et al., 2011). phase IIB trial with preladenant (SCH420814)
Furthermore, among patients with PD, a involving 253 PD patients, have been reported to
cross-sectional study involving 196 early-stage, demonstrate motor benefits in PD (Hauser et al.,
treatment-naïve PD patients reveals that coffee 2011). These clinical IIB-III trials have ultimately
178 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

led to the approval of istradefylline for treatment Batalha, V. L., Ferreira, D. G., Coelho, J. E., Valadas,
of OFF-PD patients in both the United States J. S., Gomes, R., Temido-Ferreira, M., … Lopes, L. V.
(2019) and in Japan (2013). Particularly relevant (2016). The caffeine-binding adenosine A2A
to AD drug discovery, these clinical IIB-III trials receptor induces age-like HPA-axis dysfunction by
with the A2AR antagonists have shown a consist- targeting glucocorticoid receptor function. Scien-
ent safety profile in more than 3000 advanced PD tific Reports, 6. doi: 10.1038/srep31493
patients (Hauser et al., 2011; Jenner et al., 2009). Bateup, H. S., Santini, E., Shen, W., Birnbaum, S.,
This safety profile of A2AR antagonists is entirely Valjent, E., Surmeier, D. J., … Greengard, P.
consistently with the widespread use of caffeine (2010). Distinct subclasses of medium spiny neu-
among 70% of the human population. The effect rons differentially regulate striatal motor behav-
of A2AR antagonists on cognition deficits in PD iors. Proceedings of the National Academy of
has not been, however, specifically assessed in Sciences of the United States of America, 107(33),
these large phase IIB-III clinical trials in PD for 14845–14850.
motor effects (Aarsland et al., 2010; Chase et al., Boison, D., Singer, P., Shen, H. Y., Feldon, J., & Yee,
2003). With the approval of istradefylline (Chen B. K. (2012). Adenosine hypothesis of schizophre-
and Cunha, 2020), it would be critically important nia: Opportunities for pharmacotherapy. Neurop-
within clinical trials to reevaluate the capability of harmacology, 62(3), 1527–1543.
A2AR antagonists to reverse cognitive deficits in Borea, P. A., Gessi, S., Merighi, S., Vincenzi, F., &
PD patients. Varani, K. (2018). Pharmacology of adenosine
receptors: The state of the art. Physiological
Reviews, 98(3), 1591–1625.
Borota, D., Murray, E., Keceli, G., Chang, A., Watabe,
J. M., Ly, M., … Yassa, M. A. (2014). Post-study
REFERENCES caffeine administration enhances memory consoli-
dation in humans. Nature Neuroscience, 17(2),
Aarsland, D., Bronnick, K., Williams-Gray, C., Wein- 201–203.
traub, D., Marder, K., Kulisevsky, J., … Emre, M. Brozoski, T. J., Brown, R. M., Rosvold, H. E., &
(2010). Mild cognitive impairment in Parkinson Goldman, P. S. (1979). Cognitive deficit caused by
disease: A multicenter pooled analysis. Neurology, regional depletion of dopamine in prefrontal
75(12), 1062–1069. cortex of rhesus monkey. Science, 205, 929–932.
Aitken, T. J., Greenfield, V. Y., & Wassum, K. M. Canals, M., Marcellino, D., Fanelli, F., Ciruela, F., de
(2016). Nucleus accumbens core dopamine signal- Benedetti, P., Goldberg, S. R. … Franco, R. (2003).
ing tracks the need-based motivational value of Adenosine A2A-dopamine D2 receptor-receptor
food-paired cues. Journal of Neurochemistry, heteromerization: qualitative and quantitative
136(5), 1026–1036. assessment by fluorescence and bioluminescence
Albasanz, J. L., Perez, S., Barrachina, M., Ferrer, I., & energy transfer. Journal of Biological Chemistry,
Martin, M. (2008). Up-regulation of adenosine 278(47), 46741–46749.
receptors in the frontal cortex in Alzheimer’s Canas, P. M., Duarte, J. M., Rodrigues, R. J., Kofalvi,
disease. Brain Pathology, 18(2), 211–219. A., & Cunha, R. A. (2009). Modification upon
Amodeo, D. A., Cuevas, L., Dunn, J. T., Sweeney, J. A., aging of the density of presynaptic modulation
& Ragozzino, M. E. (2018). The adenosine A2A systems in the hippocampus. Neurobiology of
receptor agonist, CGS 21680, attenuates a proba- Aging, 30(11), 1877–1884.
bilistic reversal learning deficit and elevated Canas, P. M., Porciuncula, L. O., Cunha, G. M., Silva,
grooming behavior in BTBR mice. Autism Research, C. G., Machado, N. J., Oliveira, J. M., … Cunha, R. A.
11(2), 223–233. (2009). Adenosine A2A receptor blockade pre-
Arendash, G. W., Mori, T., Cao, C., Mamcarz, M., vents synaptotoxicity and memory dysfunction
Runfeldt, M., Dickson, A., … Potter, H. (2009). caused by beta-amyloid peptides via p38 mitogen-
Caffeine reverses cognitive impairment and activated protein kinase pathway. Journal of Neu-
decreases brain amyloid-beta levels in aged Alzhei- roscience, 29(47), 14741–14751.
mer’s disease mice. Journal of Alzheimers Disease, Cao, C., Cirrito, J. R., Lin, X., Wang, L., Verges, D. K.,
17(3), 661–680. Dickson, A., … Potter, H. (2009). Caffeine sup-
Arendash, G. W., Schleif, W., Rezai-Zadeh, K., Jackson, presses amyloid-beta levels in plasma and brain of
E. K., Zacharia, L. C., Cracchiolo, J. R., … Tan, J. Alzheimer’s disease transgenic mice. Journal of
(2006). Caffeine protects Alzheimer’s mice against Alzheimers Disease, 17(3), 681–697.
cognitive impairment and reduces brain Carmo, M., Goncalves, F. Q., Canas, P. M., Oses, J. P.,
beta-amyloid production. Neuroscience, 142(4), Fernandes, F. D., Duarte, F. V., … Cunha, R. A.
941–952. (2019). Enhanced ATP release and CD73-mediated
NEUROMODULATOR ADENOSINE 179

adenosine formation sustain adenosine A2A Caffeine and an adenosine A(2A) receptor antago-
receptor over-activation in a rat model of Parkin- nist prevent memory impairment and synaptotox-
son’s disease. British Journal of Pharmacology, icity in adult rats triggered by a convulsive episode
176(18), 3666–3680. in early life. Journal of Neurochemistry, 112(2),
Carpenter, B., & Lebon, G. (2017). Human adenosine 453–462.
A2A receptor: Molecular mechanism of ligand Cunha, G. M., Canas, P. M., Oliveira, C. R., & Cunha,
binding and activation. Frontiers in Pharmacology, R. A. (2006). Increased density and synapto-pro-
8. doi: 10.3389/fphar.2017.00898 tective effect of adenosine A2A receptors upon
Carvalho Poyraz, F., Holzner, E., Bailey, M. R., Mesza- sub-chronic restraint stress. Neuroscience, 141(4),
ros, J., Kenney, L., Kheirbek, M. A., … Kellendonk, 1775–1781.
C. (2016). Decreasing striatopallidal pathway Cunha, R. A. (2016). How does adenosine control
function enhances motivation by energizing the neuronal dysfunction and neurodegeneration?
initiation of goal-directed action. Journal of Journal of Neurochemistry, 139(6), 1019–1055.
Neuroscience, 36(22), 5988–6001. Cunha, R. A., & Agostinho, P. M. (2010). Chronic
Chase, T. N., Bibbiani, F., Bara-Jimenez, W., Dimitrova, caffeine consumption prevents memory distur-
T., & Oh-Lee, J. D. (2003). Translating A2A antago- bance in different animal models of memory
nist KW6002 from animal models to parkinsonian decline. Journal of Alzheimers Disease, 20(S1),
patients. Neurology, 61(11,S6), S107–S111. S95–S116.
Chen, J. F., & Cunha, R. A. (2020). The belated US Cunha, R. A., Constantino, M. C., Sebastiao, A. M.,
FDA approval of the adenosine A2A receptor & Ribeiro, J. A. (1995). Modification of A1 and
antagonist istradefylline for treatment of Parkin- A2a adenosine receptor binding in aged striatum,
son’s disease. Purinergic Signal, 16(2), 167–174. hippocampus and cortex of the rat. Neuroreport,
Chen, J. F., Eltzschig, H. K., & Fredholm, B. B. (2013). 6(11), 1583–1588.
Adenosine receptors as drug targets: What are the Cunha, R. A., Ferre, S., Vaugeois, J. M., & Chen, J. F.
challenges? Nature Reviews: Drug Discovery, (2008). Potential therapeutic interest of adenosine
12(4), 265–286. A2A receptors in psychiatric disorders. Current
Chen, J. F., Lee, C. F., & Chern, Y. (2014). Adenosine Pharmaceutical Design, 14(15), 1512–1524.
receptor neurobiology: Overview. International Cunha, R. A., & Ribeiro, J. A. (2000). Purinergic
Review of Neurobiology, 119, 1–49. modulation of [(3)H]GABA release from rat hip-
Cho, B. H., Choi, S. M., Kim, J. T., & Kim, B. C. pocampal nerve terminals. Neuropharmacology,
(2018). Association of coffee consumption and 39(7), 1156–1167.
non-motor symptoms in drug-naive, early-stage d’Alcantara, P., Ledent, C., Swillens, S., & Schiff-
Parkinson’s disease. Parkinsonism & Related mann, S. N. (2001). Inactivation of adenosine A2A
Disorders, 50, 42–47. receptor impairs long term potentiation in the
Christofi, F. L., Zhang, H., Yu, J. G., Guzman, J., Xue, accumbens nucleus without altering basal synaptic
J., Kim, M., … Cooke, H. J. (2001). Differential transmission. Neuroscience, 107(3), 455–464.
gene expression of adenosine A1, A2a, A2b, and Dall’Igna, O. P., Fett, P., Gomes, M. W., Souza, D. O.,
A3 receptors in the human enteric nervous system. Cunha, R. A., & Lara, D. R. (2007). Caffeine and
Journal of Comparative Neurology, 439(1), adenosine A(2a) receptor antagonists prevent
46–64. beta-amyloid (25–35)-induced cognitive deficits in
Ciruela, F., Casado, V., Rodrigues, R. J., Lujan, R., mice. Experimental Neurology, 203(1), 241–245.
Burgueno, J., Canals, M., … Franco, R. (2006). Decamp, E., & Schneider, J. S. (2004). Attention and
Presynaptic control of striatal glutamatergic neuro- executive function deficits in chronic low-dose
transmission by adenosine A1-A2A receptor heter- MPTP-treated non-human primates. European
omers. Journal of Neuroscience, 26(7), Journal of Neuroscience 20(5), 1371–1378.
2080–2087. Dias-Ferreira, E., Sousa, J. C., Melo, I., Morgado, P.,
Clinton, S. M., Sucharski, I. L., & Finlay, J. M. (2006). Mesquita, A. R., Cerqueira, J. J., … Sousa, N.
Desipramine attenuates working memory impair- (2009). Chronic stress causes frontostriatal reor-
ments induced by partial loss of catecholamines in ganization and affects decision-making. Science,
the rat medial prefrontal cortex. Psychopharmacol- 325, 621–625.
ogy, 183(4), 404–412. Dias, R. B., Rombo, D. M., Ribeiro, J. A., Henley, J. M.,
Coccurello, R., Breysse, N., & Amalric, M. (2004). Simul- & Sebastiao, A. M. (2013). Adenosine: Setting the
taneous blockade of adenosine A2A and metabo- stage for plasticity. Trends in Neurosciences, 36(4),
tropic glutamate mGlu5 receptors increase their 248–257.
efficacy in reversing Parkinsonian deficits in rats. Dunwiddie, T. V., & Masino, S. A. (2001). The role
Neuropsychopharmacology, 29(8), 1451–1461. and regulation of adenosine in the central nervous
Cognato, G. P., Agostinho, P. M., Hockemeyer, J., system. Annual Review of Neuroscience, 24,
Muller, C. E., Souza, D. O., & Cunha, R. A. (2010). 31–55.
180 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Durante, V., de Iure, A., Loffredo, V., Vaikath, N., De of the United States of America, 88(16),
Risi, M., Paciotti, S., … Calabresi, P. (2019). Alpha- 7238–7241.
synuclein targets GluN2A NMDA receptor subunit Flajolet, M., Wang, Z., Futter, M., Shen, W., Nuang-
causing striatal synaptic dysfunction and visuospa- chamnong, N., Bendor, J., … Greengard, P. (2008).
tial memory alteration. Brain, 142(5), 1365–1385. FGF acts as a co-transmitter through adenosine
Durieux, P. F., Bearzatto, B., Guiducci, S., Buch, T., A(2A) receptor to regulate synaptic plasticity.
Waisman, A., Zoli, M., … de Kerchove d’Exaerde, Nature Neuroscience, 11(12), 1402–1409.
A. (2009). D2R striatopallidal neurons inhibit both Franco, R., Rivas-Santisteban, R., Casanovas, M.,
locomotor and drug reward processes. Nature Lillo, A., Saura, C. A., & Navarro, G. (2020).
Neuroscience, 12(4), 393–395. Adenosine A2A receptor antagonists affect NMDA
Durieux, P. F., Schiffmann, S. N., & de Kerchove glutamate receptor function potential to address
d’Exaerde, A. (2012). Differential regulation of neurodegeneration in Alzheimer’s disease. Cells,
motor control and response to dopaminergic 9(5). doi: 10.3390/cells9051075
drugs by D1R and D2R neurons in distinct dorsal Fredholm, B. B., AP, I. J., Jacobson, K. A., Linden, J.,
striatum subregions. EMBO Journal, 31(3), & Müller, C. E. (2011). International union of basic
640–653. and clinical pharmacology. LXXXI. Nomenclature
Eskelinen, M. H., Ngandu, T., Tuomilehto, J., Soininen, and classification of adenosine receptors: An
H., & Kivipelto, M. (2009). Midlife coffee and tea update. Pharmacological Reviews, 63(1), 1–34.
drinking and the risk of late-life dementia: A pop- Fredholm, B. B., Chen, J. F., Masino, S. A., &
ulation-based CAIDE study. Journal of Alzheimers Vaugeois, J. M. (2005). Actions of adenosine at its
Disease, 16(1), 85–91. receptors in the CNS: Insights from knockouts and
Espinosa, J., Rocha, A., Nunes, F., Costa, M. S., drugs. Annual Review of Pharmacology and
Schein, V., Kazlauckas, V., … Porciuncula, L. O. Toxicology, 45, 385–412.
(2013). Caffeine consumption prevents memory Fredholm, B. B., Chern, Y., Franco, R., & Sitkovsky,
impairment, neuronal damage, and adenosine M. (2007). Aspects of the general biology of
A2A receptors upregulation in the hippocampus adenosine A2A signaling. Progress in Neurobiology,
of a rat model of sporadic dementia. Journal of 83(5), 263–276.
Alzheimers Disease, 34(2), 509–518. Gelber, R. P., Petrovitch, H., Masaki, K. H., Ross, G.
Ferguson, S. M., Eskenazi, D., Ishikawa, M., Wanat, W., & White, L. R. (2011). Coffee intake in midlife
M. J., Phillips, P. E., Dong, Y., … Neumaier, J. F. and risk of dementia and its neuropathologic cor-
(2011). Transient neuronal inhibition reveals relates. Journal of Alzheimer’s Disease, 23(4),
opposing roles of indirect and direct pathways in 607–615.
sensitization. Nature Neuroscience, 14(1), 22–24. Gerfen, C. R., Engber, T. M., Mahan, L. C., Susel, Z.,
Ferre, S. (2008). An update on the mechanisms of Chase, T. N., Monsma, F. J., Jr., & Sibley, D. R.
the psychostimulant effects of caffeine. Journal of (1990). D1 and D2 dopamine receptor-regulated
Neurochemistry, 105(4), 1067–1079. gene expression of striatonigral and striatopallidal
Ferre, S., Fredholm, B. B., Morelli, M., Popoli, P., & neurons. Science, 250, 1429–1432.
Fuxe, K. (1997). Adenosine-dopamine receptor- Gimenez-Llort, L., Schiffmann, S. N., Shmidt, T.,
receptor interactions as an integrative mechanism Canela, L., Camon, L., Wassholm, M., … Bader,
in the basal ganglia. Trends in Neurosciences, M. (2007). Working memory deficits in transgenic
20(10), 482–487. rats overexpressing human adenosine A2A recep-
Ferre, S., Karcz-Kubicha, M., Hope, B. T., Popoli, P., tors in the brain. Neurobiology of Learning and
Burgueno, J., Gutierrez, M. A., … Ciruela, F. Memory, 87(1), 42–56.
(2002). Synergistic interaction between adenosine Goldfeld, D. A., Zhu, K., Beuming, T., & Friesner, R. A.
A2A and glutamate mGlu5 receptors: implications (2011). Successful prediction of the intra- and
for striatal neuronal function. Proceedings of the extracellular loops of four G-protein-coupled
National Academy of Sciences of the United States receptors. Proceedings of the National Academy
of America, 99(18), 11940–11945. of Sciences of the United States of America,
Ferre, S., Quiroz, C., Woods, A. S., Cunha, R., Popoli, 108(20), 8275–8280.
P., Ciruela, F., … Schiffmann, S. N. (2008). An Gomez-Castro, F., Zappettini, S., Pressey, J. C., Silva,
update on adenosine A2A-dopamine D2 receptor C. G., Russeau, M., Gervasi, N., … Levi, S. (2021).
interactions: Implications for the function of G Convergence of adenosine and GABA signaling
protein-coupled receptors. Current Pharmaceutical for synapse stabilization during development. Sci-
Design, 14(15), 1468–1474. ence, 374. doi: 10.1126/science.abk2055.
Ferre, S., von Euler, G., Johansson, B., Fredholm, B. B., Hameleers, P. A., Van Boxtel, M. P., Hogervorst, E.,
& Fuxe, K. (1991). Stimulation of high-affinity Riedel, W. J., Houx, P. J., Buntinx, F., & Jolles, J.
adenosine A2 receptors decreases the affinity of (2000). Habitual caffeine consumption and its
dopamine D2 receptors in rat striatal membranes. relation to memory, attention, planning capacity
Proceedings of the National Academy of Sciences and psychomotor performance across multiple
NEUROMODULATOR ADENOSINE 181

age groups. Human Psychopharmacology-Clinical dysfunction triggered by chronic stress. Proceedings


and Experimental, 15(8), 573–581. of the National Academy of Sciences of the United
Hauser, R. A., Cantillon, M., Pourcher, E., Micheli, F., States of America, 112(25), 7833–7838.
Mok, V., Onofrj, M., … Wolski, K. (2011). Prelad- Ko, W. K. D., Camus, S. M., Li, Q., Yang, J., McGuire,
enant in patients with Parkinson’s disease and S., Pioli, E. Y., & Bezard, E. (2016). An evaluation
motor fluctuations: A phase 2, double-blind, ran- of istradefylline treatment on Parkinsonian motor
domised trial. Lancet Neurology, 10(3), 221–229. and cognitive deficits in 1-methyl-4-phenyl-
He, Y., Huang, L., Wang, K., Pan, X., Cai, Q., Zhang, 1,2,3,6-tetrahydropyridine (MPTP)-treated
F., … Chen, J. F. (2021). alpha-Synuclein selectively macaque models. Neuropharmacology, 110(A),
impairs motor sequence learning and value sensi- 48–58.
tivity: Reversal by the adenosine A2A receptor Kouvaros, S., & Papatheodoropoulos, C. (2016).
antagonists. Cerebral Cortex. 32(4), 808–823. Major dorsoventral differences in the modulation
Higley, M. J., & Sabatini, B. L. (2010). Competitive of the local CA1 hippocampal network by NMDA,
regulation of synaptic Ca2+ influx by D2 dopa- mGlu5, adenosine A2A and cannabinoid CB1
mine and A2A adenosine receptors. Nature receptors. Neuroscience, 317, 47–64.
Neuroscience, 13(8), 958–966. Kravitz, A. V., Tye, L. D., & Kreitzer, A. C. (2012).
Hillion, J., Canals, M., Torvinen, M., Casado, V., Scott, Distinct roles for direct and indirect pathway stri-
R., Terasmaa, A., … Fuxe, K. (2002). Coaggrega- atal neurons in reinforcement. Nature Neurosci-
tion, cointernalization, and codesensitization of ence, 15(6), 816–818.
adenosine A2A receptors and dopamine D2 recep- Kreitzer, A. C., & Malenka, R. C. (2007). Endocan-
tors. Journal of Biochemistry and Molecular nabinoid-mediated rescue of striatal LTD and
Biology, 277(20), 18091–18097. motor deficits in Parkinson’s disease models.
Hino, T., Arakawa, T., Iwanari, H., Yurugi-Kobayashi, Nature, 445, 643–647.
T., Ikeda-Suno, C., Nakada-Nakura, Y., … Murata, Kreitzer, A. C., & Malenka, R. C. (2008). Striatal plas-
T. (2012). G-protein-coupled receptor inactivation ticity and basal ganglia circuit function. Neuron,
by an allosteric inverse-agonist antibody. Nature, 60(4), 543–554.
482, 237–240. Laurent, C., Burnouf, S., Ferry, B., Batalha, V. L.,
Horgusluoglu-Moloch, E., Nho, K., Risacher, S. L., Coelho, J. E., Baqi, Y., … Blum, D. (2016). A2A
Kim, S., Foroud, T., Shaw, L. M., … Alzheimer’s adenosine receptor deletion is protective in a
disease neuroimaging, I. (2017). Targeted neuro- mouse model of Tauopathy. Molecular Psychiatry,
genesis pathway-based gene analysis identifies 21(1), 97–107.
ADORA2A associated with hippocampal volume in Leffa, D. T., Pandolfo, P., Goncalves, N., Machado, N.
mild cognitive impairment and Alzheimer’s J., de Souza, C. M., Real, J. I., … Ferreira, S. G.
disease. Neurobiology of Aging, 60, 92–103. (2018). Adenosine A2A receptors in the rat prelim-
Hu, Q., Ren, X., Liu, Y., Li, Z., Zhang, L., Chen, X., … bic medial prefrontal cortex control delay-based
Chen, J. F. (2016). Aberrant adenosine A2A recep- cost-benefit decision making. Frontiers in Molecu-
tor signaling contributes to neurodegeneration lar Neuroscience, 11. doi: 10.3389/
and cognitive impairments in a mouse model of fnmol.2018.00475
synucleinopathy. Experimental Neurology, 283(A), Lerner, T. N., Horne, E. A., Stella, N., & Kreitzer, A. C.
213–223. (2010). Endocannabinoid signaling mediates psy-
Jenner, P., Mori, A., Hauser, R., Morelli, M., Fredholm, chomotor activation by adenosine A2A antago-
B. B., & Chen, J. F. (2009). Adenosine, adenosine nists. Journal of Neuroscience, 30(6), 2160–2164.
A2A antagonists, and Parkinson’s disease. Parkin- Li, P., Rial, D., Canas, P. M., Yoo, J. H., Li, W., Zhou,
sonism & Related Disorders, 15(6), 406–413. X., … Chen, J. F. (2015). Optogenetic activation of
Kachroo, A., Orlando, L. R., Grandy, D. K., Chen, J. intracellular adenosine A2A receptor signaling in
F., Young, A. B., & Schwarzschild, M. A. (2005). the hippocampus is sufficient to trigger CREB
Interactions between metabotropic glutamate 5 phosphorylation and impair memory. Molecular
and adenosine A2A receptors in normal and par- Psychiatry, 20(11). doi: 10.1038/mp.2015.43
kinsonian mice. Journal of Neuroscience, 25(45), Li, W., Silva, H. B., Real, J., Wang, Y. M., Rial, D., Li,
10414–10419. P., … Chen, J. F. (2015). Inactivation of adenosine
Kadowaki Horita, T., Kobayashi, M., Mori, A., Jenner, A2A receptors reverses working memory deficits
P., & Kanda, T. (2013). Effects of the adenosine at early stages of Huntington’s disease models.
A2A antagonist istradefylline on cognitive perfor- Neurobiology Disease, 79, 70–80.
mance in rats with a 6-OHDA lesion in prefrontal Li, Y., He, Y., Chen, M., Pu, Z., Chen, L., Li, P., …
cortex. Psychopharmacology, 230(3), 345–352. Chen, J. F. (2016). Optogenetic activation of
Kaster, M. P., Machado, N. J., Silva, H. B., Nunes, A., adenosine A2A receptor signaling in the dorsome-
Ardais, A. P., Santana, M., … Cunha, R. A. (2015). dial striatopallidal neurons suppresses goal-
Caffeine acts through neuronal adenosine A2A directed behavior. Neuropsychopharmacology,
receptors to prevent mood and memory 41(4), 1003–1013.
182 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Li, Y., Pan, X., He, Y., Ruan, Y., Huang, L., Zhou, Y., Mishina, M., Ishiwata, K., Naganawa, M., Kimura, Y.,
… Chen, J. F. (2018). Pharmacological blockade of Kitamura, S., Suzuki, M., … Ishii, K. (2011).
adenosine A2A but Not A1 receptors enhances Adenosine A(2A) receptors measured with [C]
goal-directed valuation in satiety-based instrumen- TMSX PET in the striata of Parkinson’s disease
tal behavior. Frontiers in Pharmacology, 9. doi: patients. PLoS One, 6(2). doi: 10.1371/journal.
10.3389/fphar.2018.00393 pone.0017338.
Li, Y., Ruan, Y., He, Y., Cai, Q., Pan, X., Zhang, Y., … Morales-Figueroa, G. E., Rivera-Ramirez, N., Gonza-
Chen, J. F. (2020). Striatopallidal adenosine A2A lez-Pantoja, R., Escamilla-Sanchez, J., Garcia-Her-
receptors in the nucleus accumbens confer moti- nandez, U., Galvan, E. J., & Arias-Montano, J. A.
vational control of goal-directed behavior. Neurop- (2019). Adenosine A2A and histamine H3 recep-
harmacology, 168. doi: 10.1016/j. tors interact at the cAMP/PKA pathway to modu-
neuropharm.2020.108010 late depolarization-evoked [(3)H]-GABA release
Li, Z., Chen, X., Wang, T., Gao, Y., Li, F., Chen, L., … from rat striato-pallidal terminals. Purinergic
Chen, J. F. (2018). The corticostriatal adenosine Signal, 15(1), 85–93.
A2A receptor controls maintenance and retrieval Mori, A. (2020). How do adenosine A2A receptors
of spatial working memory. Biological Psychiatry, regulate motor function? Parkinsonism & Related
83(6), 530–541. Disorders, 80(S1), S13–S20.
Lindsay, J., Laurin, D., Verreault, R., Hebert, R., Helli- Mori, A., Shindou, T., Ichimura, M., Nonaka, H., &
well, B., Hill, G. B., & McDowell, I. (2002). Risk Kase, H. (1996). The role of adenosine A2a recep-
factors for Alzheimer’s disease: a prospective tors in regulating GABAergic synaptic transmission
analysis from the Canadian Study of Health and in striatal medium spiny neurons. Journal of Neu-
Aging. American Journal of Epidemiology, 156(5), roscience, 16(2), 605–611.
445–453. Mouro, F. M., Kofalvi, A., Andre, L. A., Baqi, Y.,
Lobo, M. K., Covington, H. E., 3rd, Chaudhury, D., Muller, C. E., Ribeiro, J. A., & Sebastiao, A. M.
Friedman, A. K., Sun, H., Damez-Werno, D., … (2019). Memory deficits induced by chronic can-
Nestler, E. J. (2010). Cell type-specific loss of BDNF nabinoid exposure are prevented by adenosine
signaling mimics optogenetic control of cocaine A2AR receptor antagonism. Neuropharmacology,
reward. Science, 330, 385–390. 155, 10–21.
Lobo, M. K., Cui, Y., Ostlund, S. B., Balleine, B. W., & Neves, G., Cooke, S. F., & Bliss, T. V. (2008). Synaptic
Yang, X. W. (2007). Genetic control of instrumen- plasticity, memory and the hippocampus: a neural
tal conditioning by striatopallidal neuron-specific network approach to causality. Nature Reviews
S1P receptor Gpr6. Nature Neuroscience, 10(11), Neuroscience, 9(1), 65–75.
1395–1397. Ning, Y. L., Yang, N., Chen, X., Tian, H. K., Zhao, Z.
Lopes, L. V., Cunha, R. A., & Ribeiro, J. A. (1999). A., Zhang, X. Z., … Zhou, Y. G. (2019). Caffeine
Increase in the number, G protein coupling, and attenuates brain injury but increases mortality
efficiency of facilitatory adenosine A2A receptors induced by high-intensity blast wave exposure.
in the limbic cortex, but not striatum, of aged rats. Toxicology Letters, 301, 90–97.
Journal of Neurochemistry, 73(4), 1733–1738. Ning, Y. L., Yang, N., Chen, X., Xiong, R. P., Zhang,
Lopez-Cruz, L., Salamone, J. D., & Correa, M. (2018). X. Z., Li, P., … Zhou, Y. G. (2013). Adenosine A2A
Caffeine and selective adenosine receptor antago- receptor deficiency alleviates blast-induced cogni-
nists as new therapeutic tools for the motivational tive dysfunction. Journal of Cerebral Blood Flow
symptoms of depression. Frontiers in Pharmacol- and Metabolism, 33(11), 1789–1798.
ogy, 9. doi: 10.3389/fphar.2018.00526 Nishizawa, K., Fukabori, R., Okada, K., Kai, N., Uchi-
Martin, S. J., Grimwood, P. D., & Morris, R. G. (2000). gashima, M., Watanabe, M., …Kobayashi, K.
Synaptic plasticity and memory: an evaluation of (2012). Striatal indirect pathway contributes to
the hypothesis. Annual Review of Neuroscience, selection accuracy of learned motor actions.
23, 649–711. Journal of Neuroscience, 32(39), 13421–13432.
Matos, M., Shen, H. Y., Augusto, E., Wang, Y., Wei, Orr, A. G., Hsiao, E. C., Wang, M. M., Ho, K., Kim, D. H.,
C. J., Wang, Y. T., … Chen, J. F. (2015). Deletion Wang, X., … Mucke, L. (2015). Astrocytic adenosine
of adenosine A2A receptors from astrocytes dis- receptor A2A and Gs-coupled signaling regulate
rupts glutamate homeostasis leading to psycho- memory. Nature Neuroscience, 18(3), 423–434.
motor and cognitive impairment: relevance to Pagnussat, N., Almeida, A. S., Marques, D. M.,
schizophrenia. Biological Psychiatry, 78(11), Nunes, F., Chenet, G. C., Botton, P. H., … Porciun-
763–774. cula, L. O. (2015). Adenosine A(2A) receptors are
Meng, F., Guo, Z., Hu, Y., Mai, W., Zhang, Z., Zhang, necessary and sufficient to trigger memory impair-
B., … Gao, Z. (2019). CD73-derived adenosine ment in adult mice. British Journal of Pharmacol-
controls inflammation and neurodegeneration by ogy, 172(15), 3831–3845.
modulating dopamine signalling. Brain, 142(3), Pardo, M., Lopez-Cruz, L., Valverde, O., Ledent, C.,
700–718. Baqi, Y., Muller, C. E., … Correa, M. (2012).
NEUROMODULATOR ADENOSINE 183

Adenosine A2A receptor antagonism and genetic prospective population study (the Three City
deletion attenuate the effects of dopamine D2 Study). Neurology, 69(6), 536–545.
antagonism on effort-based decision making in Rombo, D. M., Newton, K., Nissen, W., Badurek, S.,
mice. Neuropharmacology, 62(5–6), 2068–2077. Horn, J. M., Minichiello, L., … Lamsa, K. P. (2015).
Pereira, G. S., Rossato, J. I., Sarkis, J. J., Cammarota, Synaptic mechanisms of adenosine A2A receptor-
M., Bonan, C. D., & Izquierdo, I. (2005). Activation mediated hyperexcitability in the hippocampus.
of adenosine receptors in the posterior cingulate Hippocampus, 25(5), 566–580.
cortex impairs memory retrieval in the rat. Neuro- Rosin, D. L., Hettinger, B. D., Lee, A., & Linden, J.
biology of Learning and Memory, 83(3), (2003). Anatomy of adenosine A2A receptors in
217–223. brain: morphological substrates for integration of
Postuma, R. B., Anang, J., Pelletier, A., Joseph, L., striatal function. Neurology, 61(11,S6), S12–S18.
Moscovich, M., Grimes, D., … Lang, A. E. (2017). Salamone, J. D., Yohn, S. E., Lopez-Cruz, L., San
Caffeine as symptomatic treatment for Parkinson Miguel, N., & Correa, M. (2016). Activational and
disease (Cafe-PD): A randomized trial. Neurology, effort-related aspects of motivation: Neural mech-
89(17), 1795–1803. anisms and implications for psychopathology.
Prediger, R. D., Pamplona, F. A., Fernandes, D., & Brain, 139(5), 1325–1347.
Takahashi, R. N. (2005). Caffeine improves spatial Schiffmann, S. N., Fisone, G., Moresco, R., Cunha, R. A.,
learning deficits in an animal model of attention & Ferre, S. (2007). Adenosine A2A receptors and
deficit hyperactivity disorder (ADHD): The sponta- basal ganglia physiology. Progress in Neurobiol-
neously hypertensive rat (SHR). International Jour- ogy, 83(5), 277–292.
nal of Neuropsychopharmacology, 8(4), 583–594. Shen, H. Y., Canas, P. M., Garcia-Sanz, P., Lan, J. Q.,
Prediger, R. D., & Takahashi, R. N. (2005). Modulation Boison, D., Moratalla, R., … Chen, J. F. (2013).
of short-term social memory in rats by adenosine Adenosine A(2)A receptors in striatal glutamater-
A1 and A(2A) receptors. Neuroscience Letters, gic terminals and GABAergic neurons oppositely
376(3), 160–165. modulate psychostimulant action and DARPP-32
Radenbach, C., Reiter, A. M., Engert, V., Sjoerds, Z., phosphorylation. PLoS One, 8(11). doi: 10.1371/
Villringer, A., Heinze, H. J., … Schlagenhauf, F. journal.pone.0080902
(2015). The interaction of acute and chronic stress Shen, H. Y., Coelho, J. E., Ohtsuka, N., Canas, P. M.,
impairs model-based behavioral control. Psycho- Day, Y. J., Huang, Q. Y., … Chen, J. F. (2008). A
neuroendocrinology, 53, 268–280. critical role of the adenosine A2A receptor in
Ramlackhansingh, A. F., Bose, S. K., Ahmed, I., Tur- extrastriatal neurons in modulating psychomotor
kheimer, F. E., Pavese, N., & Brooks, D. J. (2011). activity as revealed by opposite phenotypes of
Adenosine 2A receptor availability in dyskinetic striatum and forebrain A2A receptor knock-outs.
and nondyskinetic patients with Parkinson disease. Journal of Neuroscience, 28(12), 2970–2975.
Neurology, 76(21), 1811–1816. Shen, H. Y., Singer, P., Lytle, N., Wei, C. J., Lan, J. Q.,
Rebola, N., Canas, P. M., Oliveira, C. R., & Cunha, R. A. Williams-Karnesky, R. L., … Boison, D. (2012).
(2005). Different synaptic and subsynaptic locali- Adenosine augmentation ameliorates psychotic
zation of adenosine A2A receptors in the hip- and cognitive endophenotypes of schizophrenia.
pocampus and striatum of the rat. Neuroscience, Journal of Clinical Investigation, 122(7),
132(4), 893–903. 2567–2577.
Rebola, N., Rodrigues, R. J., Lopes, L. V., Richardson, Shen, W., Flajolet, M., Greengard, P., & Surmeier, D. J.
P. J., Oliveira, C. R., & Cunha, R. A. (2005). Adeno- (2008). Dichotomous dopaminergic control of
sine A1 and A2A receptors are co-expressed in striatal synaptic plasticity. Science, 321, 848-851.
pyramidal neurons and co-localized in glutamater- Shindou, T., Nonaka, H., Richardson, P. J., Mori, A.,
gic nerve terminals of the rat hippocampus. Neu- Kase, H., & Ichimura, M. (2002). Presynaptic
roscience, 133(1), 79–83. adenosine A2A receptors enhance GABAergic
Rebola, N., Sebastiao, A. M., de Mendonca, A., synaptic transmission via a cyclic AMP dependent
Oliveira, C. R., Ribeiro, J. A., & Cunha, R. A. mechanism in the rat globus pallidus. British
(2003). Enhanced adenosine A2A receptor facilita- Journal of Pharmacology, 136(2), 296–302.
tion of synaptic transmission in the hippocampus Silva, A. C., Lemos, C., Goncalves, F. Q., Pliassova, A.
of aged rats. Journal of Neurophysiology, 90(2), V., Machado, N. J., Silva, H. B., … Agostinho, P.
1295–1303. (2018). Blockade of adenosine A2A receptors
Rimondini, R., Ferre, S., Ogren, S. O., & Fuxe, K. recovers early deficits of memory and plasticity in
(1997). Adenosine A2A agonists: a potential new the triple transgenic mouse model of Alzheimer’s
type of atypical antipsychotic. Neuropsychophar- disease. Neurobiology of Disease, 117, 72–81.
macology, 17(2), 82–91. Simoes, A. P., Machado, N. J., Goncalves, N., Kaster,
Ritchie, K., Carriere, I., de Mendonca, A., Portet, F., M. P., Simoes, A. T., Nunes, A., … Cunha, R. A.
Dartigues, J. F., Rouaud, O., … Ancelin, M. L. (2016). Adenosine A2A receptors in the amygdala
(2007). The neuroprotective effects of caffeine: A control synaptic plasticity and contextual fear
184 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

memory. Neuropsychopharmacology, 41(12), Viana da Silva, S., Haberl, M. G., Zhang, P., Bethge,
2862–2871. P., Lemos, C., Goncalves, N., … Mulle, C. (2016).
Singer, P., Wei, C. J., Chen, J. F., Boison, D., & Yee, B. Early synaptic deficits in the APP/PS1 mouse model
K. (2013). Deletion of striatal adenosine A(2A) of Alzheimer’s disease involve neuronal adenosine
receptor spares latent inhibition and prepulse inhi- A2A receptors. Nature Communications, 7. doi:
bition but impairs active avoidance learning. 10.1038/ncomms11915
Behavioural Brain Research, 242, 54–61. Villar-Menendez, I., Blanch, M., Tyebji, S., Pereira-
Solfrizzi, V., Panza, F., Imbimbo, B. P., D’Introno, A., Veiga, T., Albasanz, J. L., Martin, M., … Barra-
Galluzzo, L., Gandin, C. (2015). Coffee con- china, M. (2013). Increased 5-methylcytosine and
sumption habits and the risk of mild cognitive decreased 5-hydroxymethylcytosine levels are
impairment: The Italian longitudinal study on associated with reduced striatal A2AR levels in
aging. Journal of Alzheimer’s Disease, 47(4), Huntington’s disease. Neuromolecular Medicine,
889–899. 15(2), 295–309.
Svenningsson, P., Le Moine, C., Fisone, G., & Fred- Villar-Menendez, I., Porta, S., Buira, S. P., Pereira-Veiga,
holm, B. B. (1999). Distribution, biochemistry and T., Diaz-Sanchez, S., Albasanz, J. L., … Barrachina,
function of striatal adenosine A2A receptors. Pro- M. (2014). Increased striatal adenosine A2A recep-
gress in Neurobiology, 59(4), 355–396. tor levels is an early event in Parkinson’s disease-
Tai, L. H., Lee, A. M., Benavidez, N., Bonci, A., & related pathology and it is potentially regulated by
Wilbrecht, L. (2012). Transient stimulation of dis- miR-34b. Neurobiology of Disease, 69, 206–214.
tinct subpopulations of striatal neurons mimics Wall, N. R., De La Parra, M., Callaway, E. M., & Kre-
changes in action value. Nature Neuroscience, itzer, A. C. (2013). Differential innervation of
15(9), 1281–1289. direct- and indirect-pathway striatal projection
Taylor, S. B., Anglin, J. M., Paode, P. R., Riggert, A. G., neurons. Neuron, 79(2), 347–360.
Olive, M. F., & Conrad, C. D. (2014). Chronic stress Wei, C. J., Augusto, E., Gomes, C. A., Singer, P.,
may facilitate the recruitment of habit- and addic- Wang, Y., Boison, D., … Chen, J. F. (2014). Regula-
tion-related neurocircuitries through neuronal tion of fear responses by striatal and extrastriatal
restructuring of the striatum. Neuroscience, 280, adenosine A2A receptors in forebrain. Biological
231–242. Psychiatry, 75(11), 855–863.
Tecuapetla, F., Jin, X., Lima, S. Q., & Costa, R. M. Wei, C. J., Singer, P., Coelho, J., Boison, D., Feldon,
(2016). Complementary Contributions of striatal J., Yee, B. K., & Chen, J. F. (2011). Selective inacti-
projection pathways to action initiation and execu- vation of adenosine A(2A) receptors in striatal
tion. Cell, 166(3), 703–715. neurons enhances working memory and reversal
Temido-Ferreira, M., Coelho, J. E., Pousinha, P. A., & learning. Learning & Memory, 18(7), 459–474.
Lopes, L. V. (2019). Novel players in the aging Yin, H. H., Mulcare, S. P., Hilario, M. R., Clouse, E.,
synapse: Impact on cognition. Journal of Caffeine Holloway, T., Davis, M. I., … Costa, R. M. (2009).
and Adenosine Research, 9(3), 104–127. Dynamic reorganization of striatal circuits during
Temido-Ferreira, M., Ferreira, D. G., Batalha, V. L., the ac quisition and consolidation of a skill. Nature
Marques-Morgado, I., Coelho, J. E., Pereira, P., … Neuroscience, 12(3), 333–341.
Lopes, L. V. (2020). Age-related shift in LTD is Yin, H. H., Ostlund, S. B., & Balleine, B. W. (2008).
dependent on neuronal adenosine A2A receptors Reward-guided learning beyond dopamine in the
interplay with mGluR5 and NMDA receptors. nucleus accumbens: The integrative functions of
Molecular Psychiatry, 25(8), 1876–1900. cortico-basal ganglia networks. European Journal
Tetzlaff, W., Schubert, P., & Kreutzberg, G. W. (1987). of Neuroscience, 28(8), 1437–1448.
Synaptic and extrasynaptic localization of adeno- Yu, C., Gupta, J., Chen, J. F., & Yin, H. H. (2009).
sine binding sites in the rat hippocampus. Neuro- Genetic deletion of A2A adenosine receptors in
science, 21(3), 869–875. the striatum selectively impairs habit formation.
van Boxtel, M. P., Schmitt, J. A., Bosma, H., & Jolles, Journal of Neuroscience, 29(48), 15100–15103.
J. (2003). The effects of habitual caffeine use on Zhou, J., Wu, B., Lin, X., Dai, Y., Li, T., Zheng, W., …
cognitive change: A longitudinal perspective. Chen, J. F. (2019). Accumbal adenosine A2A
Pharmacology, Biochemistry, and Behavior, 75(4), receptors enhance cognitive flexibility by facilitat-
921–927. ing strategy shifting. Frontiers in Cellular Neurosci-
van Gelder, B. M., Buijsse, B., Tijhuis, M., Kalmijn, S., ence, 13. doi: 10.3389/fncel.2019.00130
Giampaoli, S., Nissinen, A., & Kromhout, D. Zhou, S. J., Zhu, M. E., Shu, D., Du, X. P., Song, X. H.,
(2007). Coffee consumption is inversely associated Wang, X. T., … He, J. C. (2009). Preferential
with cognitive decline in elderly European men: enhancement of working memory in mice lacking
The FINE study. European Journal of Clinical Nutri- adenosine A(2A) receptors. Brain Research, 1303,
tion, 61(2), 226–232. 74–83.
12
Hormonal Influences on Cognition
Linda Becker and Nicolas Rohleder

INTRODUCTION are composed of a small number of amino acids.


Relevant peptide hormones are adrenocortico-
Hormones are messenger molecules that are tropic hormone (ACTH), epinephrine (EPI), nor-
released by endocrine glands within the body and epinephrine (NE), and oxytocin. As opposed to
also by specialized neurons within the brain. Free steroid hormones, peptide hormones cannot eas-
circulating hormones in the bloodstream or in the ily cross cell membranes. Therefore, they cannot
interstitial fluid interact with the brain through pass the BBB either and can (in most cases) not
positive and negative feedback loops, which can alter cognitive functions directly. However, mech-
be related with changes in cognitive functioning. anisms to circumvent this restriction have been
In general, there are two different classes of hor- described (e.g., through activating nerves such as
mones, mainly distinguished by their biochemical the vagus nerve in the periphery that can then alter
characteristics, which have direct effects on the brain function through this neural pathway; cf. van
way they gain access to, and signal to target cells. Bochove et al., 2018).
The two main classes are steroid hormones and In addition to the direct and indirect effects of
peptide hormones. actual hormones, this chapter cannot provide a com-
One class of hormones are steroid hormones. prehensive overview without addressing neurotrans-
They are synthesized from the precursor molecule mitters that are part of intra-CNS pathways. These
cholesterol. Important steroid hormones are glu- pathways involve for example the locus coeruleus
cocorticoids (e.g., the stress hormone cortisol), (LC), which is the origin of noradrenergic projections
mineralocorticoids (e.g., aldosterone), and sex to the prefrontal cortex (PFC), as well as dopamin-
hormones (e.g., testosterone, estradiol, and pro- ergic projections to the prefrontal cortex originating
gesterone). Steroid hormones are highly stable and in the ventral tegmental area (VTA) (Chandler et al.,
typically bound to transport proteins in the blood. 2014). This pathway involves the norepinephrine as
In their free form (i.e., when they are not bound to a neurotransmitter, and thereby affects for example
proteins), steroid hormones can pass through cell executive functions located in the PFC. The LC also
membranes. Importantly, they can pass the blood- projects to the amygdala, also with norepinephrine as
brain barrier (BBB), and can therefore, affect neu- a neurotransmitter, providing regulation of memory
ral and brain processing directly. processes by arousal, which interacts closely with
A further important class of hormones are pep- stress hormones such as cortisol from the periphery
tide hormones, short protein molecules, which (McGaugh and Roozendaal, 2009).
186 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Brain Structures Involved in Or it requires the circulating substance to exert its


Cognitive Functioning effects on protected tissues (such as CNS neurons)
via mediating pathways, such as activating the
The main brain structures, which are relevant for vagus nerve in the periphery, which can then
cognitive functioning in general are the prefrontal transmit a message into the brain. Any such effect
cortex (PFC), the hippocampus, and the amyg- on cognitive function not only depends on the
dala, of which the former is particularly relevant ability of the hormone to enter the brain in
for executive functioning and the others for general, but more specifically, to reach an area of
memory processes. the brain that is involved in performing the
The prefrontal cortex (PFC) is part of the specific cognitive function.
cerebral cortex. The PFC receives projections It is therefore necessary to differentiate
from the mediodorsal nucleus of the thalamus. between (1) hormone effects in the classical way
Different parts of the PFC can be distinguished. (i.e., hormones being secreted into the periphery),
One distinction is the differentiation between a and finding their way into the brain; (2) hormone
medial and a lateral part, of which the former can effects through alternate mediating pathways
be subdivided into a dorsal (dorsomedial PFC; (i.e., hormones activate peripheral nerve endings
dmPFC) and a ventral one (ventromedial PFC; and signal to the CNS that way); and (3) intra-
vmPFC), and the latter a dorsolateral (dlPFC) and CNS pathways as described above (e.g., the LC
a ventrolateral part (vlPFC; Carlén, 2017). The noradrenergic system that can be activated by
PFC is involved in several functions, of which stress or arousal, and projects to target areas in the
cognitive functioning can be seen as the most CNS relaying the arousal signal).
important ones. Besides it is involved in emo- To study effects of hormones on cognitive func-
tional, motivational, and social processes. The tions such as memory and EF, different methodo-
PFC plays a major role for executive functioning logical approaches have been used. The type of
(e.g., attention, working memory and -updating, method also depends on the structure and ability
inhibition, and cognitive flexibility). of the hormone(s) of interest. Steroid hormones
The hippocampus is a seahorse shaped structure can simply be administered to research partici-
in the temporal part of the cerebral cortex, located pants via typical pharmacological techniques such
along the lateral ventricles. According to Lopes da as pills, injections, or skin patches (Schlosser
Silva and Arnolds (1978), it includes several ana- et al., 2013). These methods are not feasible for
tomical components – Ammon’s horn, the dentate peptide hormones, because they would not be able
gyrus, and the subiculum (most inferior compo- to penetrate the BBB and therefore will not reach
nent of the hippocampal formation). The main the brain. One alternative is the pharmacological
function of the hippocampus is its involvement in activation of the central noradrenergic system for
memory and learning. example by peripheral application of the alpha-2
The amygdala is an almond-shaped brain receptor antagonist yohimbine (Roozendaal et al.,
structure, located in the medial temporal cortex 2006). Another method is the application of recep-
(Sah et al., 2003). It includes several nuclei, the tor blockers, such as the beta-adrenergic antago-
basolateral amygdala (BLA), as well as surround- nist propranolol (Roozendaal et al., 2006). In
ing lateral nuclei. The amygdala is involved in addition to pharmacological approaches, hormone
bottom-up processing of (e.g., emotional) salient levels can also be experimentally manipulated by
stimuli. It is highly relevant for emotional memory behavioral paradigms in the laboratory, that for
foundation. Other structures involve for exam- example induce psychosocial stress, or negative
ple the anterior cingulate cortex (ACC) and the emotional arousal (Canli et al., 2000; Wolf et al.,
inferior frontal gyrus (IFG). 2004). Finally, natural fluctuations of hormones
during biological rhythms or hormone changes
over the lifespan can be investigated in observa-
tional studies (Pretscher et al., 2021).
HORMONE INTERACTIONS WITH SPECIFIC
COGNITIVE DOMAINS
Memory
Effects of any substance circulating in the blood,
including hormones, on any target tissue, depend One of the most important cognitive functions is
on the ability to reach the respective target tissue. memory. Memory is important for remembering
In the case of the brain, this either requires being and learning. Different types of memory can
able to pass the BBB, as for example, steroid hor- be distinguished. One of these is the differentia-
mones can do, while peptide hormones cannot. tion between explicit and implicit memory
Hormonal Influences on Cognition 187

(or declarative vs. non-declarative), of which the found in the morning, when endogenous cortisol
former is happening consciously and the latter is higher, but stimulating cortisol effects were
unconsciously (Squire and Zola-Morgan, 1988). found in the afternoon, when endogenous cortisol
Another neuro-biological distinction is whether the is lower. The importance of timing relative to
hippocampus is involved in the learning process or memory phase was also the result of a review of
not (so-called hippocampus-dependent vs. non- findings from animal studies by (Roozendaal,
hippocampus-dependent memory (Henke, 2010). 2002).
In the case of memory, a well-known exam- Glucocorticoid effects on memory retrieval
ple is the effect of the glucocorticoid cortisol on were traced to high glucocorticoid levels directly
declarative memory function (Wingenfeld and affecting glucocorticoid receptors in the hip-
Wolf, 2014). Cortisol, as a steroid hormone, is pocampus. Memory consolidation, in contrast,
able to pass the BBB and reach the hippocampus, was found to depend on more complicated inter-
an area responsible for declarative memory forma- actions with other brain regions, for example
tion. The effect of endogenous cortisol responses, the amygdala. In general, memory consolidation
or glucocorticoid application before learning, on appears to be enhanced by glucocorticoid hor-
the ability to later remember newly learned declar- mones. The key finding that cortisol, or gluco-
ative information is a robust finding (Wingenfeld corticoids, enhance memory consolidation, but
and Wolf, 2014). Furthermore, increased cortisol impair memory retrieval, in healthy humans has
levels (e.g., after acute stress exposure) are asso- been confirmed (Wingenfeld and Wolf, 2014). In
ciated with decreased declarative memory perfor- this review, it was further elaborated that imme-
mance (Becker and Rohleder, 2019; Wolf et al., diate short-term effects of cortisol on the hip-
2004). This is different for hormones that cannot pocampus, which might be stimulating, need to be
pass the BBB, which therefore can only indirectly distinguished from longer-term, inhibitory effects
affect cognitive function, for example for EF such on memory. One disadvantage of these pharma-
as working memory (Becker and Rohleder, 2019). cological studies is that artificial hormone effects
The situation becomes more complicated when are observed, for example because hormone con-
different classes of hormones are involved. This is centrations achieved through pharmacological
the case for emotional memory. A robust finding application tend to be supraphysiological, and
here is that information with negative emotional that interactions with other systems that occur in
content is more easily remembered than neu- real-life situations of for example stress, cannot be
tral or positive information. This effect depends investigated.
on an interaction of two different systems, the
(intra-CNS) noradrenergic system, with the glu-
cocorticoid system, in the amygdala: Here results Experimental manipulation of
show that memory is only enhanced if noradr- hormone levels
energic activation is present at the same time as Studies using experimental manipulations of hor-
glucocorticoids in the amygdala (Canli et al., mones mainly target the stress response system,
2000; Roozendaal et al., 2006). This example which includes increases in peripheral glucocorti-
highlights not only the importance of co-signaling coid concentrations, and activation of the central
of two pathways (i.e., glucocorticoids and nor- LC-based noradrenergic system. The specific role
epinephrine), but also the importance to differ- of location and timing, as well as interactions with
entiate between central and peripheral effects of different hormone or neurotransmitter systems
norepinephrine. and brain structures comes into play when think-
ing about the role of stress and stress hormones in
Pharmacological hormone application formation and consolidation, as well as retrieval
Application of hormones allows for the most of emotional memories.
stringent experimental manipulation, used to As an overall summary it can be noted that expe-
study cortisol effects on memory. As summarized riences that elicit arousal and/or intense emotions,
in a meta-analysis by (Het et al., 2005), which in particular negative emotions, enhance memory
includes 16 studies with a total of more than 500 formation (i.e., consolidation; Canli et al., 2000).
participants, the effect of cortisol on declarative This is mediated in particular by the interaction of
memory depends on the timing of hormone appli- the intra-CNS pathways, specifically the noradren-
cation relative to memory phase. It was found that ergic system originating from the LC and project-
cortisol application before retrieval impaired ing to the amygdala, with glucocorticoid hormones
memory, while cortisol before learning had differ- from the periphery entering the brain (Roozendaal
ent effects depending on the time of day, which is et al., 2006). This expands the view presented above
related with the circadian rhythm of endogenous that the effects of hormones depend on the memory
cortisol. Impairment of memory formation was phase to also include the timing relative to an actual
188 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

experience. Through the interactions described Cognitive flexibility refers to switching between
above and in more detail by (Joëls et al., 2006), it tasks, operations, actions, or cognitions. Working
can be concluded that declarative memory forma- memory is a limited capacity cognitive system,
tion (consolidation) is enhanced by the interaction which is responsible for temporary storage and
of noradrenergic with glucocorticoid signaling, processing of information (Baddeley, 2001).
which leads to even stronger memory enhancement Working memory-updating refers to the monitor-
when stimuli or experiences are arousing, and / ing of incoming information in working memory.
or characterized by negative emotions. Memory Verbal fluency is associated with cognitive flex-
retrieval in contrast is impaired in the context of ibility. At least two forms can be distinguished,
negatively arousing experiences. semantic fluency and second phonemic fluency.
Impairments in verbal fluency are in many cases
Associations with Natural Hormone associated with frontal (phonemic and semantic
fluency) or temporal (semantic fluency only) brain
Fluctuations lesions (Henry and Crawford, 2004).
A further way to assess the effect of hormones on
Attention involves both bottom-up and top-
memory is to observe changes in memory in asso-
down attentional processes. The former refers to
ciation with natural changes of hormone levels, for
the automatic processing of salient (e.g., threat-
example as part of natural rhythms or in association
ening) stimuli, whereas the latter, which is also
with age-related changes in hormone levels.
called attentional control, is the ability to engage
Cortisol fluctuates significantly during the day,
and to sustain attention upon information that
with highest concentrations within the first hour
is relevant to the current tasks or to one’s goals
of waking up (Cortisol Awakening Response;
(Engle, 2018). The most important brain structure
CAR) and continuous declines during the day.
which is involved in EF is the PFC, which is in
During the CAR, memory consolidation appears
particular modulated by dopaminergic, noradr-
to be enhanced, and a missing or lower morning
energic, serotonergic, and cholinergic pathways
cortisol increase seems to have negative effects on
(Logue and Gould, 2014).
memory, highlighting once again the role of cor-
tisol in memory formation (Law and Clow, 2020).
Less cortisol during the entire diurnal cycle fur- Pharmacological Hormone Application
ther relates to lower emotional memory formation The effects of pharmacological cortisol adminis-
(Nagamine et al., 2017). tration have been investigated in a comprehensive
Results of longitudinal studies investigating meta-analysis (Shields et al., 2015). One key find-
associations of changes in cortisol levels with ing was that somewhat similar to cortisol effects
memory function revealed that older individuals on memory, timing and domain are important, in
with higher cortisol levels experienced steeper that different domains of EF are differentially
declines in declarative memory (Li et al., 2006; affected by early versus late after cortisol admin-
Segerstrom et al., 2016). This underscores poten- istration. Here, the effect of timing is based on
tial long-term effects of hormones on brain struc- different mechanisms through which glucocorti-
tures such as the hippocampus. coids affect target tissues, with rapid non-genomic
effects being different from the slower, genomic
effects (Joëls et al., 2006). Working memory was
impaired via rapid non-genomic cortisol effects,
Executive Function but enhanced by slower, genomic effects. In
contrast, inhibition was stimulated by rapid
Executive functioning (EF) is one of the most non-genomic effects, but impaired by slower,
important cognitive functions for successfully genomic effects. Set-shifting was not affected by
managing daily life. Executive functioning is a cortisol (Shields et al., 2016).
higher-order neurocognitive control system ena-
bling individuals to organize actions, to plan and
engage in goal- and future-directed behaviour, and Experimental Manipulation
to successfully adapt to changing environments of Hormone Levels
(Jurado and Rosselli, 2007). Executive function- This already complex picture again becomes more
ing can be conceptualized as a multidimensional complicated when investigating the effect of
construct, including several subdomains such as experimental manipulation of hormone levels on
inhibition, cognitive flexibility, working memory different EF domains. Stress, which activates not
and working memory-updating, verbal fluency, only cortisol but also affects the Autonomic
and attention (Jurado and Rosselli, 2007; Snyder Nervous System and the LC-noradrenergic
et al., 2015). Inhibition refers to the ability to system, has been found to negatively affect work-
inhibit automatic or inadequate responses. ing memory and set-shifting (Plessow et al., 2011;
Hormonal Influences on Cognition 189

Schoofs et al., 2008). When stress is induced with domains also depend on timing of hormone appli-
the cold pressor test, a higher cortisol response cation relative to testing. Here, fast, non-genomic
was associated with better updating flexibility and effects often have opposite effects from slow,
with lower switching between different tasks genomic effects. For both, memory and execu-
(Goldfarb et al., 2017). Targeting the central tive function it is relatively clear that age-related
noradrenergic system by vagus nerve stimulation changes, mainly in the form of cortisol increases
(VNS) showed that norepinephrine has positive and flattening of diurnal curves are consistently
effects on inhibition (van Bochove et al., 2018). associated with declines in cognitive function.

Associations with Natural Hormone


Fluctuations
Similar to memory, studies have investigated the
relationship between natural hormone fluctuations
ASSESSMENT OF HORMONE EFFECTS
and executive function. A higher CAR seems to ON COGNITIVE FUNCTIONS
have positive effects on EF, with better verbal
memory (Labad et al., 2020) and better task The studies summarized above highlight the dif-
switching performance (Law et al., 2020). Higher ficulties when aiming to assess the effects of hor-
diurnal total cortisol was found to be associated mones on the brain, specifically, cognitive
with lower inhibition (stroop performance) but not function, or the associations of hormones with
with cognitive flexibility (Pretscher et al., 2021). cognitive function.
Higher hair cortisol was further found to be Experimental designs in which hormones are
related with lower working memory in children either pharmacologically administered, or behav-
(Kim et al., 2022) and adults (van den Heuvel iorally manipulated allow establishing causality
et al., 2022). Similar associations were also found and are therefore most powerful. However, both
with morning serum cortisol (i.e., higher cortisol study designs have their strengths and limitations.
related with lower verbal memory and flexibility; Hormone application typically – at least in human
Echouffo-Tcheugui et al., 2018). studies – only allows altering the concentration
In older adults as well, higher diurnal cortisol, of one specific hormone, for example glucocorti-
flatter diurnal cortisol slopes and higher bedtime coids. While this is useful to establish the specific
cortisol have been found to be associated with effect of this specific hormone, it typically does
lesser function on all EF domains studied (Evans not reflect what happens in a real-life situation, in
et al., 2011; Franz et al., 2011; Lee et al., 2007; which a whole network of hormones may interact.
Montoliu et al., 2019). Here as well, a higher CAR Furthermore, hormone application often leads to
is predictive of better EF (Evans et al., 2012). Here supra physiological concentrations, which limits
as well, higher cortisol most likely relates to more the generalizability of the findings. The opposite
advanced brain aging in relevant structures. is true in experiments that alter hormone concen-
Taken together, the effect of hormones on trations through behavioral paradigms. Here, the
memory and executive function has been mainly entire network of hormones and brain pathways is
investigated for stress hormones. Memory effects altered in a natural way.
of stress hormones cortisol and norepinephrine However, because humans respond very dif-
are well-understood, in that timing and emo- ferently to laboratory situations, hormone con-
tional valence or arousal determine if memory centrations typically show large variations, which
is enhanced or suppressed. Nevertheless, while requires large sample sizes. Despite the higher
declarative memory is well-investigated, much external validity of behavioral paradigms, they still
less is known about other forms, such as proce- do not reflect a real-life situation, which is where
dural memory. More questions are open with observational studies can help. In these studies,
regard to executive function, which is, at least in natural fluctuations of hormone concentrations
part, caused by the fact that many different sub- can be measured and associations with different
domains need to be studied, and different tests cognitive domains can be tested. These designs do
need to be administered to test a specific domain. not allow establishing causal relationships.
Since many of these tests are complicated and To measure the associations between hormones
also require full attention of the participants, it and cognitive functions, both must be measured
is almost impossible to test all domains in one adequately. Because hormonal concentrations
experimental setup. in the brain cannot be measured non-invasively,
Despite these difficulties, research using hor- they must be measured in the periphery, e.g., in
mone administration or behavioral paradigms has blood samples. An alternative is the assessment
revealed that glucocorticoid effects on different through saliva samples, which is much easier and
190 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

relatively stress-free for participants than collec- the limitations of the TSST are the relatively high
tion of blood samples. Concentrations of steroid number of personnel required.
hormones assessed in saliva are in many cases Alternatives to the TSST are less personnel-
proportional to hormone levels in the blood. intensive, such as the socially evaluated cold
Therefore, salivary hormone assessment has pressor test (SECPT; Boyle et al., 2016) or the
often become the method of choice, especially for Montreal imaging stress task (MIST (Dedovic
the assessment of steroids. However, most ster- et al., 2005)). With regard to cortisol, both SECPT
oid hormones are bound to transport proteins in and MIST are limited by their lower effect on the
blood, and only the free fraction can be measured HPA axis, which sometimes requires eliminating
in saliva fraction (about 10% for example of cor- non-responder participants from the sample. Both,
tisol). In contrast, due to the fact that peptides do TSST and SECPT can be applied in a group set-
not cross the blood-brain barrier, peptide hormone ting, which somewhat reduces their limitations
concentrations measured in the periphery do not (Becker et al., 2019; von Dawans et al., 2011).
accurately reflect peptide hormone concentrations Similar to pharmacological studies, measurement
in the brain. Some ways of measuring peptide hor- of peripheral hormone concentrations is a neces-
mone levels indirectly through the assessment of sity, because of high inter-individual variability of
enzymes (that are related with hormones) from for example cortisol responses to stress (Kudielka
saliva have been suggested. An example is the et al., 2009). Also, here as well, timing of stressor
assessment of the enzyme alpha-amylase from application and careful timing of the respective
saliva samples (salivary alpha- amylase, sAA) cognitive test is of major importance. Cortisol for
as an indicator for sympathetic nervous system example peaks about 18 minutes after the end of
(SNS) activity, which is associated with the acute a TSST (Goodman et al., 2017), so that cognitive
stress response (Nater and Rohleder, 2009). testing of cortisol effects should be done in this
Similarly, the application of hormones in the time window.
periphery can only effectively be used for steroid
hormones that easily cross the blood brain barrier.
To test the effects of peptide hormones, such as
norepinephrine, peripheral administration would
not reach the desired brain structures. One worka- BIDIRECTIONALITY
round is to activate the desired target system in
the way through alternative pathways: The central One important fact to be aware of is that the asso-
noradrenergic system, for example, can be stimu- ciation between hormones and cognition is bidi-
lated by administration of the alpha-2 adrenergic rectional, i.e., while hormones can have a
receptor antagonist Yohimbine (van Stegeren et al., facilitating or deteriorating effect on cognitive
2010), which is safe to use in humans and reliably functioning as described above, cognitive abilities
activates the noradrenergic system, as well as the can also affect the release and thereby concentra-
peripheral sympathetic nervous system. Given the tion of hormones.
importance of timing for hormone effects on cog- In longitudinal studies, results seem to support
nitive function, a main issue in all pharmacologi- that cognitive function can predict hormone con-
cal studies is to mimic natural hormone changes as centrations much later in life. It was for example
adequately as possible with regard to concentra- shown that general cognitive ability measured
tion and temporal response patterns. Here as well, at the age of twenty was correlated with higher
assessment of peripheral hormone concentrations, overall diurnal cortisol 35 years later (Franz
as described above, is essential in ensuring proper et al., 2011). While in this study, no specific
hormone concentrations. domains were analyzed with regard to this long-
When designing behavioral experiments term relationship, results support the notion that
aimed at activating specific hormone systems, higher cognitive abilities might offer some level
a major issue is the selection of the experimen- of protection from long-term pathophysiological
tal paradigm, and appropriate controls. Using hormone effects. Results from the Midlife in the
the example of experimental stress induction, the US (MIDUS) study point to a similar direction by
research has the choice between a number of dif- showing that higher executive function predicted
ferent stress paradigms, which all have strengths steeper diurnal cortisol declines at a later time
and limitations. The most widely used stress point (Stawski et al., 2011). Since steeper diurnal
paradigm is the Trier Social Stress Test (TSST; slopes comprise a healthier pattern, here as well, a
(Kirschbaum et al., 1993), which reliably activates better cognitive function seems to predict healthier
HPA axis and sympathetic nervous system, and for hormone secretion patterns. In experimental stud-
which a non-stress control condition, the friendly ies, higher scores on specific domains of execu-
TSST is available (Wiemers et al., 2013). One of tive functioning were found to be associated with
Hormonal Influences on Cognition 191

lower acute stress cortisol responses. Specifically, old age, later in life. Conversely, better cognitive
individuals with better working memory func- abilities in general, most notably executive func-
tion showed a lesser stress response, pointing to tions, help to dampen acute stress responses and
a potentially healthier ability to adapt to stressors predict more adaptive cortisol concentrations in
(Grimm et al., 2021). later life. These facts highlight the importance
of understanding the bidirectional interactions of
hormones and cognitive functions, and the impor-
tant need to examine more deeply neuroanatomi-
SUMMARY AND CONCLUSIONS cal brain areas affected by different hormones.

The release of hormones can interact with the


brain through different direct and indirect path-
ways. This can result either in deterioration or
improvement of cognitive functioning. Of particu- REFERENCES
lar importance are steroid hormones (e.g., gluco-
corticoid and sex hormones), as well as peptide Baddeley, A. D. (2001). Is working memory still work-
hormones (e.g., norepinephrine). In this chapter, ing? American Psychologist, 56(11), 851–864.
we have described through which pathways these Becker, L., & Rohleder, N. (2019). Time course of the
interactions occur and how they can be assessed. physiological stress response to an acute stressor
We started with an overview of different hormone and its associations with the primacy and recency
classes and brain structures that are particularly effect of the serial position curve. PloS One, 14(5).
relevant for cognitive functioning as well as for doi: 10.1371/journal.pone.0213883
the interaction between hormones and cognitive Becker, L., Schade, U., & Rohleder, N. (2019). Evalua-
functioning in general. We then examined differ- tion of the socially evaluated cold-pressor group
ent cognitive domains and their associations with test (SECPT-G) in the general population. Peer
peripheral concentrations of different hormone Journal, 7. doi: 10.7717/peerj.7521
types: First, we have described the hormonal Boyle, N. B., Lawton, C., Arkbåge, K., West, S. G.,
influences on different types of memory and, Thorell, L., Hofman, D., … Dye, L. (2016). Stress
second, the effects on executive functions (e.g., responses to repeated exposure to a combined
inhibition, working memory, and cognitive flexi- physical and social evaluative laboratory stressor in
bility). We then outlined how the hormonal effects young healthy males. Psychoneuroendocrinology,
on cognitive functions can be assessed and what 63, 119–127.
should be considered in designing an empirical Canli, T., Zhao, Z., Brewer, J., Gabrieli, J. D., & Cahill,
study. Finally, we have briefly outlined the bidi- L. (2000). Event-related activation in the human
rectionality of these effects whereby cognitive amygdala associates with later memory for
functions can be affected by hormones on the one individual emotional experience. Journal of
hand, but cognitive functions can influence the Neuroscience, 20(19). doi: 10.1523/JNEUROSCI.
release of hormones on the other hand. 20-19-j0004.2000
In summary, hormones circulating in the Carlén, M. (2017). What constitutes the prefrontal
periphery, as well as hormones and neurotrans- cortex? Science, 358, 478–482.
mitter systems entering or being produced in the Chandler, D. J., Waterhouse, B. D., & Gao, W.-J.
brain have profound effects on cognitive function. (2014). New perspectives on catecholaminergic
This is best established for stress hormones and regulation of executive circuits: Evidence for inde-
declarative memory, but also for other memory pendent modulation of prefrontal functions by
domains and different executive functions. It midbrain dopaminergic and noradrenergic neu-
is important to note a high level of interaction rons. Frontiers in Neural Circuits, 8. doi: 10.3389/
between different hormone systems, and the role fncir.2014.00053
of timing of the ability to use a specific cogni- Dedovic, K., Renwick, R., Mahani, N. K., Engert, V.,
tive function relative to a situation that changes Lupien, S. J., & Pruessner, J. C. (2005). The Mon-
hormone levels. With regard to memory, it can treal Imaging Stress Task: Using functional imaging
be assumed that stress hormone effects modu- to investigate the effects of perceiving and pro-
late memory in an adaptive manner, by enabling cessing psychosocial stress in the human brain.
easier long-term storage of information that is Journal of Psychiatry & Neuroscience, 30(5),
important for survival, at the expense of informa- 319–325.
tion that is of less importance. With regard to the Echouffo-Tcheugui, J. B., Conner, S. C., Himali, J. J.,
long-term effects of hormones on cognitive func- Maillard, P., DeCarli, C. S., Beiser, A. S., Vasan, R. S.,
tion, higher levels of stress hormones have been & Seshadri, S. (2018). Circulating cortisol and cog-
shown to predict declines in cognitive function in nitive and structural brain measures: The
192 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Framingham heart study. Neurology, 91(21). doi: stress disorder symptoms via executive function
10.1212/WNL.0000000000006549 deficits among urban American Indian children.
Engle, R. W. (2018). Working memory and executive Stress. doi: 10.1080/10253890.2021.2024164
attention: A revisit. Perspectives on Psychological Kirschbaum, C., Pirke, K. M., & Hellhammer, D. H.
Science, 13(2), 190–193. (1993). The Trier Social Stress Test: A tool for investi-
Evans, P. D., Fredhoi, C., Loveday, C., Hucklebridge, gating psychobiological stress responses in a labora-
F., Aitchison, E., Forte, D., & Clow, A. (2011). The tory setting. Neuropsychobiology, 28(1-2), 76–81.
diurnal cortisol cycle and cognitive performance in Kudielka, B. M., Hellhammer, D. H., & Wüst, S.
the healthy old. International Journal of Psycho- (2009). Why do we respond so differently?
physiology, 79(3), 371–377. Reviewing determinants of human salivary cortisol
Evans, P., Hucklebridge, F., Loveday, C., & Clow, A. responses to challenge. Psychoneuroendocrinol-
(2012). The cortisol awakening response is related ogy, 34(1), 2–18.
to executive function in older age. International Labad, J., Salvat-Pujol, N., Armario, A., Cabezas, Á.,
Journal of Psychophysiology, 84(2), 201–204. Arriba-Arnau, A. de, Nadal, R., … Soria, V. (2020).
Franz, C. E., O’Brien, R. C., Hauger, R. L., Mendoza, The role of sleep quality, trait anxiety and hypotha-
S. P., Panizzon, M. S., Prom-Wormley, E., … lamic-pituitary-adrenal axis measures in cognitive
Kremen, W. S. (2011). Cross-sectional and 35-year abilities of healthy individuals. International Jour-
longitudinal assessment of salivary cortisol and nal of Environmental Research and Public Health,
cognitive functioning: The Vietnam era twin study 17(20). doi: 10.3390/ijerph17207600
of aging. Psychoneuroendocrinology, 36(7), Law, R., & Clow, A. (2020). Stress, the cortisol awak-
1040–1052. ening response and cognitive function. Interna-
Goldfarb, E. V., Froböse, M. I., Cools, R., & Phelps, E. tional Review of Neurobiology, 150, 187–217.
A. (2017). Stress and cognitive flexibility: Cortisol Law, R., Evans, P., Thorn, L., Hucklebridge, F., Love-
increases are associated with enhanced updating day, C., & Clow, A. (2020). The cortisol awakening
but impaired switching. Journal of Cognitive response predicts a same-day index of executive
Neuroscience, 29(1), 14–24. function in healthy young adults. International
Goodman, W. K., Janson, J., & Wolf, J. M. (2017). Journal of Psychophysiology, 158, 27–33.
Meta-analytical assessment of the effects of proto- Lee, B. K., Glass, T. A., McAtee, M. J., Wand, G. S.,
col variations on cortisol responses to the Trier Bandeen-Roche, K., Bolla, K. I., & Schwartz, B. S.
Social Stress Test. Psychoneuroendocrinology, 80, (2007). Associations of salivary cortisol with cogni-
26–35. tive function in the Baltimore memory study.
Grimm, E., Agrigoroaei, S., Rohleder, N., & Becker, L. Archives of General Psychiatry, 64(7), 810–818.
(2021). Executive functioning as a predictor of Li, G., Cherrier, M. M., Tsuang, D. W., Petrie, E. C.,
physiological and subjective acute stress responses Colasurdo, E. A., Craft, S., Schellenberg, G. D., …
in non-clinical adult populations: A systematic Wilkinson, C. W. (2006). Salivary cortisol and
literature review and meta-analysis. Neuroscience memory function in human aging. Neurobiology
and Biobehavioral Reviews, 131, 1096–1115. of Aging, 27(11), 1705–1714.
Henke, K. (2010). A model for memory systems based Logue, S. F., & Gould, T. J. (2014). The neural and
on processing modes rather than consciousness. genetic basis of executive function: Attention,
Nature Reviews, Neuroscience, 11(7), 523–532. cognitive flexibility, and response inhibition.
Henry, J. D., & Crawford, J. R. (2004). A meta- Pharmacology, Biochemistry, and Behavior, 123,
analytic review of verbal fluency performance fol- 45–54.
lowing focal cortical lesions. Neuropsychology, Lopes da Silva, F. H., & Arnolds, D. E. (1978). Physiol-
18(2), 284–295. ogy of the hippocampus and related structures.
Het, S., Ramlow, G., & Wolf, O. T. (2005). A meta- Annual Review of Physiology, 40, 185–216.
analytic review of the effects of acute cortisol McGaugh, J. L., & Roozendaal, B. (2009). Drug
administration on human memory. Psychoneu- enhancement of memory consolidation: Historical
roendocrinology, 30(8), 771–784. perspective and neurobiological implications.
Joëls, M., Pu, Z., Wiegert, O., Oitzl, M. S., & Krugers, Psychopharmacology, 202(1-3), 3–14.
H. J. (2006). Learning under stress: How does it Montoliu, T., Hidalgo, V., & Salvador, A. (2019). The
work? Trends in Cognitive Sciences, 10(4), relationship between loneliness and cognition in
152–158. healthy older men and women: The role of corti-
Jurado, M. B., & Rosselli, M. (2007). The elusive sol. Psychoneuroendocrinology, 107, 270–279.
nature of executive functions: A review of our cur- Nagamine, M., Noguchi, H., Takahashi, N., Kim, Y., &
rent understanding. Neuropsychology Review, Matsuoka, Y. (2017). Effect of cortisol diurnal
17(3), 213–233. rhythm on emotional memory in healthy young
Kim, J., Li, L., Korous, K. M., Valiente, C., & Tsethlikai, adults. Scientific Reports, 7(1). doi: 10.1038/
M. (2022). Chronic stress predicts post-traumatic s41598-017-10002-z
Hormonal Influences on Cognition 193

Nater, U. M., & Rohleder, N. (2009). Salivary alpha- with cortisol. Neuroscience and Biobehavioral
amylase as a non-invasive biomarker for the sym- Reviews. doi: 10.1016/j.neubiorev.2016.06.038
pathetic nervous system: Current state of research. Snyder, H. R., Miyake, A., & Hankin, B. L. (2015).
Psychoneuroendocrinology, 34(4), 486–496. Advancing understanding of executive function
Plessow, F., Fischer, R., Kirschbaum, C., & Goschke, T. impairments and psychopathology: Bridging the
(2011). Inflexibly focused under stress: Acute psy- gap between clinical and cognitive approaches.
chosocial stress increases shielding of action goals Frontiers in Psychology, 6. doi: 10.3389/fpsyg.
at the expense of reduced cognitive flexibility with 2015.00328
increasing time lag to the stressor. Journal of Cog- Squire, L. R., & Zola-Morgan, S. (1988). Memory:
nitive Neuroscience, 23(11), 3218–3227. Brain systems and behavior. Trends in Neuro-
Pretscher, A., Kauzner, S., Rohleder, N., & Becker, L. sciences, 11(4), 170–175.
(2021). Associations between social burden, per- Stawski, R. S., Almeida, D. M., Lachman, M. E., Tun,
ceived stress, and diurnal cortisol profiles in older P. A., Rosnick, C. B., & Seeman, T. (2011). Associa-
adults: Implications for cognitive aging. European tions between cognitive function and naturally
Journal of Ageing, 18(4), 575–590. occurring daily cortisol during middle adulthood:
Roozendaal, B. (2002). Stress and memory: opposing Timing is everything. Journals of Gerontology.
effects of glucocorticoids on memory consolida- Series B, Psychological Sciences and Social
tion and memory retrieval. Neurobiology of Learn- Sciences, 66(S1), i71–81.
ing and Memory, 78(3), 578–595. van Bochove, M. E., De Taeye, L., Raedt, R., Vonck,
Roozendaal, B., Okuda, S., Van der Zee, E. A., & K., Meurs, A., Boon, P., … Verguts, T. (2018).
McGaugh, J. L. (2006). Glucocorticoid enhancement Reduced distractor interference during vagus
of memory requires arousal-induced noradrenergic nerve stimulation. International Journal of Psycho-
activation in the basolateral amygdala. Proceedings physiology, 128, 93–99.
of the National Academy of Sciences of the United van den Heuvel, L. L., Suliman, S., Bröcker, E., Kilian,
States of America, 103(17), 6741–6746. S., Stalder, T., Kirschbaum, C., & Seedat, S. (2022).
Sah, P., Faber, E. S. L., Lopez De Armentia, M., & Power, The association between hair cortisol levels,
J. (2003). The amygdaloid complex: Anatomy and inflammation and cognitive functioning in females.
physiology. Physiological Reviews, 83(3), 803–834. Psychoneuroendocrinology, 136. doi: 10.1016/j.
Schlosser, N., Wolf, O. T., Fernando, S. C., Terfehr, K., psyneuen.2021.105619
Otte, C., Spitzer, C., … Wingenfeld, K. (2013). van Stegeren, A. H., Roozendaal, B., Kindt, M., Wolf,
Effects of acute cortisol administration on response O. T., & Joëls, M. (2010). Interacting noradrenergic
inhibition in patients with major depression and and corticosteroid systems shift human brain acti-
healthy controls. Psychiatry Research, 209(3), vation patterns during encoding. Neurobiology of
439–446. Learning and Memory, 93(1), 56–65.
Schoofs, D., Preuss, D., & Wolf, O. T. (2008). Psycho- von Dawans, B., Kirschbaum, C., & Heinrichs, M.
social stress induces working memory impairments (2011). The Trier Social Stress Test for Groups
in an n-back paradigm. Psychoneuroendocrinol- (TSST-G): A new research tool for controlled simul-
ogy, 33(5), 643–653. taneous social stress exposure in a group format.
Segerstrom, S. C., Geiger, P. J., Boggero, I. A., Psychoneuroendocrinology, 36(4), 514–522.
Schmitt, F. A., & Sephton, S. E. (2016). Endoge- Wiemers, U. S., Schoofs, D., & Wolf, O. T. (2013). A
nous cortisol exposure and declarative verbal friendly version of the trier social stress test does
memory: A longitudinal study of healthy older not activate the HPA axis in healthy men and
adults. Psychosomatic Medicine, 78(2), 182–191. women. Stress, 16(2), 254–260.
Shields, G. S., Bonner, J. C., & Moons, W. G. (2015). Wingenfeld, K., & Wolf, O. T. (2014). Stress, memory,
Does cortisol influence core executive functions? A and the hippocampus. Frontiers of Neurology and
meta-analysis of acute cortisol administration Neuroscience, 34, 109–120.
effects on working memory, inhibition, and set- Wolf, O. T., Kuhlmann, S., Buss, C., Hellhammer, D. H.,
shifting. Psychoneuroendocrinology, 58, 91–103. & Kirschbaum, C. (2004). Cortisol and memory
Shields, G. S., Sazma, M. A., & Yonelinas, A. P. retrieval in humans: influence of emotional
(2016). The effects of acute stress on core execu- valence. Annals of the New York Academy of
tive functions: A meta-analysis and comparison Sciences, 1032, 195–197.
This page intentionally left blank
PART III
Neuroanatomical
Brain Systems
This page intentionally left blank
13
Modeling of Basal Ganglia
Structure and Function
Anneke Alkemade, Bernadette C.M. van Wijk,
and Birte U. Forstmann

INTRODUCTION features, such as main infrastructure created by


humans. In order to understand how cities are con-
Brain function can be studied from various per- nected, one can start to map the traffic architec-
spectives, including computational modeling, ture. Mapping how the roads are connected and
which has the potential to deepen our insight into embedded in the landscape is crucial to model and
structure-function relationships, as evidenced by understand traffic behavior. The level of descrip-
the progress made in relation to the basal ganglia tion can be limited to the identification of the
(BG). In this chapter we provide an overview of main roads, but could also include information on
current knowledge regarding BG structure-func- framework characteristics that are required to
tion relationships, and highlight the role of com- allow the system to function, for instance the
putational modeling in advancing our availability and location of gas stations, or bridges
understanding. Deciphering the neural processes connecting different roads. This relatively high
that the BG perform, as well as the overall behav- level of description can help us to understand how
ioral functions covered by this system, continues the landscape architecture gives rise to function,
to be a valid focus of research, as is the role of the and can further be refined, for instance, through
individual BG nuclei in disease, and case reports the inclusion of information on speed limits.
of BG lesions. Imagine that information is available on the dis-
tance between point A and B, together with the
speed with which a car travels. This information
now allows us to calculate how much time is
needed to get from point A to point B. If this cal-
RELATION BETWEEN ANATOMICAL culation results in a predicted seven-day drive,
STRUCTURE AND FUNCTION while in reality the drive only takes three days,
this likely indicates that part of the architecture,
Imagine a satellite image of the world, which such as a bridge or tunnel has been omitted in the
provides detailed information on the items that created model. The mismatch between structure
together form the landscape. This image provides (traffic architecture) and function (drive) will then
a high level of detail without highlighting specific trigger the search for this bridge or tunnel on the
198 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

satellite image, and the description can be updated meaningful, reusable, instantly understandable
accordingly. However, if the time needed to travel mapping of the brain for application in various
from point A to point B in reality is only 30 min, fields of research.
this indicates that the model used to estimate the
time of travel does not capture the data adequately,
and warrants a more thorough revision.
This principle of describing structure and link-
ing it to function can be applied to the brain in a ANATOMY OF THE BASAL GANGLIA
comparable manner. Studying the brain through
the microscope in sections that are stained using We summarize a number of the major anatomical
histological techniques, or by applying structural characteristics, required for understanding and inter-
Magnetic Resonance Imaging (MRI) presents preting the underlying building blocks that are
information on the anatomical structures of the incorporated in leading BG models. The BG is a
brain. To understand how function rises from the group of closely connected structures of varying
underlying anatomical architecture, the func- developmental origin. The main structures are the
tional neuroanatomy needs to be uncovered. The striatum (STR), globus pallidus (GP), substantia
relative contribution of specific brain areas can nigra (SN), and the subthalamic nucleus (STN). The
for instance be addressed using functional (f) caudate (CAU) and putamen (PUT) together form
MRI or electroencephalography (EEG), applied the dorsal striatum. The ventral striatum contains the
in combination with behavioral or physiologi- nucleus accumbens (NACC). Additionally, the
cal outcome parameters. This research process is globus pallidus is divided in an external (GPe) and
ongoing, and may never be completed. It is pur- internal segment (GPi) separated by the medial
sued using a wealth of description levels, varying medullary lamina (mml), each segment with differ-
from expert opinions for the recognition of struc- ent in- and output connections and with distinct
tural features, which have already been linked to functions. In turn, the substantia nigra is divided into
specific functions, to techniques from the field of a compact (SNc) and a reticular (SNr) part (Yelnik
molecular biology to identify specific signaling et al., 1987). The STN is located on the dorsolateral
systems, or individual proteins in cell popula- aspect of the SN and has a characteristic almond
tions. Using a variety of functional anatomical shape (Alkemade et al., 2015; also Figure 13.1).
studies, it is possible to identify main brain struc- The structure–function relationships are cap-
tures and anatomical networks that provide a tured in today’s most prominent and successful
backbone for specific behavior. The basic ana- BG models describing direct, indirect and hyper-
tomical architecture identified can then be refined direct pathways in the regulation of motor, cogni-
based on studies providing information on brain tive, as well as limbic behavior (cf. Helie et al.;
activation. As a next step, the anatomo-functional also Figure 13.2). This model represents a high
models can be validated, adjusted, extended, or level of description, and can be used in various
rejected based on behavioral testing in combina- ways within various disciplines including com-
tion with formal computational modeling stud- putational modeling approaches. Researchers
ies. If the identified neural architecture does studying brain function can use this model as a
not accommodate the observed behavior, or the basis to structure their research, and to refine their
model is insufficiently able to predict behavior, understanding of the molecular underpinning of
this can steer the descriptive anatomical research BG function. Additionally, this box-and-arrow
toward the identification of the missing nodes diagram can form the basis for the design of for-
related to latent parameters identified through the mal cognitive models that can be applied for the
computational modeling, or it can trigger rigor- prediction of behavior.
ous paradigm shifts, thereby changing our under-
standing of brain processes.
Fully annotated anatomical descriptions cover-
ing all intricacies of the networks of the human
brain do not exist. If we compare anatomical INDIVIDUAL NUCLEI OF THE BASAL
descriptions of the human brain to different geo- GANGLIA
graphical maps, distinct types of maps (BG maps
vs. maps of endocrine feedback systems) have dif- In addition to the general architecture of the BG
ferent value for understanding function and inter- nodes and their connections, the individual BG
preting behavior. High resolution satellite images nuclei each have their own complex internal make
can form the basis for atlasing efforts to derive up. A basic understanding of this architecture is
a multitude of maps that can be reused for vari- required as it delivers an anatomical backbone, as
ous purposes. The challenge then is how to create well as biological plausibility for the formal
Modeling of Basal Ganglia Structure and Function 199

Figure 13.1 Post-mortem anatomical images of the human basal ganglia.


A and B show the striatum (STR) and the globus pallidus interna and externa (GPi and GPe)
at different levels of magnification. C and D show the subthalamic nucleus (STN) and sub-
stantia nigra (SN), which are located at a more caudal level.

computational models of BG function, which are underestimate the extent and number of the pro-
used to explain both normal and aberrant behavior jections (Alkemade, 2013). Immunohistochemical
resulting from BG-related disease. Information on results are often extrapolated from animal experi-
the number and the nature of neuronal connec- ments as well, which do not take potential inter-
tions between individual BG structures are com- species differences into account. Alternatively,
monly derived from neural tracing studies in immunohistochemical results are derived from
rodents and/or non-human primates. Neuronal post mortem human studies which have inherent
tracing studies are performed using molecules that limitations imposed by ante mortem illness, cause
are taken up by neuronal cell bodies at the site of of death, tissue quality and antibody sensitivity
injection, and which travel through the axons to (Alkemade et al., 2019). Although it is unlikely
their terminal processes (Saleeba et al., 2019). that these technical limitations will have resulted
Dependent on the type of tracer, they may travel in a misrepresentation of the underlying human
antero- and/or retrogradely over the synaptic con- neuroanatomy, it is important to realize that some
nection or can be monosynaptically restricted differences may be present.
(Saleeba et al., 2019).
Although many studies have targeted the
BG, it is important to realize that the techniques
underlying the observations that form the basis of Striatum
the available anatomical models of the BG have
technical limitations. Tracing results are usually The striatum (STR) is the largest structure of the
obtained in rodents, and to a lesser extent in non- BG. The dorsal STR is formed by the CAU and
human primates. These studies systematically PUT, which share their histological characteristics
200 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 13.2 Basal ganglia models describing a direct, indirect, and hyperdirect pathway in
the regulation of motor, cognitive, as well as limbic behavior.
A Various studies have identified different parts of the cortico-basal ganglia-thalamic net-
work (indicated with shades of grey) as potential generators of neural oscillations.
B Using dynamic causal modeling, Moran et al. (2011) pinpointed the emergence of patholog-
ical beta oscillations (left panels) to altered connection strengths of the hyperdirect pathway
and STN-GPe circuit (light blue). Similar studies with data from human patients identified
additional connections: ‘+’ for Marreiros et al. (2013) and ‘*’ for Van Wijk et al. (2018). Overall,
findings suggest an increased activation of the hyperdirect and indirect pathway in PD.
Source: Adapted from Moran et al., 2011.

and functional profile, although they can easily be At the same time, the matrix of the striatum
distinguished anatomically due to their separation receives input from the sensorimotor cortex, and
by the internal capsule (ic). The NACC is located provides output via the SNr. It is important to
ventral to the CAU and PUT, and its border to the note that although this is an attractive anatomical
dorsal striatum can be visualized using histo- model, other studies have not been able to confirm
chemical approaches (Meredith et al., 1996). The its validity (Voorn et al., 2004; Yin and Knowlton,
NACC is often referred to as the ventral STR. The 2006). More research is therefore needed to deter-
STR predominantly contains medium spiny pro- mine to what extent this compartmentalization
jection neurons (MSN) and a smaller proportion translates into functional outcomes, including spa-
of interneurons (Bishop et al., 1982; Matamales tial weighting and regulated signaling.
et al., 2009; Plenz and Wickens, 2016). The STR The anatomo-functional specificity in the stria-
represents the main input nucleus of the BG and tum is attributed to the distinct function and dis-
receives information from multiple cortical areas tribution patterns of the dopamine receptors. Both
as well as the thalamus (THA), the brain stem and D1 and D2 receptors are expressed in striato-nigral
GP (Aston-Jones and Cohen, 2005; Beckstead, and striato-pallidal MSN of the STR, respectively
1983; Calabresi et al., 1996; Sharpe and Tepper, (Clark et al., 2005). Constitutive activity of the BG
1998; Soghomonian et al., 1989). reflects a situation in which the indirect pathway
The microscopic appearance of the STR is is active, as a result of D2-receptor stimulation.
in-homogenous and patchy. The matrix is ana- Striatal GABAergic enkephalin MSN that are part
tomically distinct from the striosomes (patches), of the indirect pathway, subsequently affect other
forming areas that are functionally separate, with BG nuclei through polysynaptic connections to the
differing neurochemical and connectivity pro- GPi /SNr via the GPe and STN (Shink and Smith,
files (Herkenham et al., 1984). The striosomes 1995). These output nuclei subsequently project
have been identified as part of the limbic circuitry (indirectly) to the thalamus, the superior colli-
based on their frontal cortical input regions, and culus, brain stem, and other regions to modulate
their projections to the SNc (Smith et al., 2016). behavior (Bolam et al., 2000). Activation is shifted
Modeling of Basal Ganglia Structure and Function 201

in anticipation of movement when increased dopa- same population of GPi neurons, suggestive of a
mine availability in the STR shifts the balance tight regulatory system to balance the direct, indi-
towards the direct pathway (Clark et al., 2005). rect, and hyperdirect pathways of the BG (Nambu
The direct pathway is activated via binding and et al., 2000).
activation of the D1 receptors through dopamin-
ergic input originating from the SNc. Striatal
GABAergic substance P-positive MSN project
monosynaptically to the GPi and SNr (Shink et al.,
1996). Overall increases in striatal dopamine con- GLOBUS PALLIDUS
centrations shift the balance toward activation of
the direct pathway, which is in line with increased GPi and SNr are output nuclei of the basal gan-
overall motor activity that is observed in rodents. glia. The GPi plays a role in the direct, indirect as
Additionally, topographical zonation has been well as the hyperdirect pathway of the BG. The
described for the STR. Functional differences GPi receives extensive GABA-ergic input from
between dorsomedial and dorsolateral STR point both the STR and GPe. Interestingly, the GABA-
toward discrete roles in motor learning, habits, ergic GPi neurons have only very limited local
action evaluation, and decision-making (Balleine axon collaterals (Ilinsky et al., 1997; Nakanishi
et al., 2007; Bloem et al., 2017; Castañé et al., et al., 1991; Parent et al., 1999; Smith et al.,
2010; Wang et al., 2013). 1987). This specific organization of dendrites and
fibers has led researchers to attribute information
convergence to the GPi (Percheron et al., 1984;
Yelnik et al., 1984).
Single neurons of the GPe can project to all
SUBTHALAMIC NUCLEUS nuclei of the BG, and through their extensive
GABAergic axon terminals, provide an anatomi-
The subthalamic nucleus receives direct cortical cal framework allowing coherent network activ-
input, and is the input nucleus for the hyperdirect ity (Bevan, 2021; Bevan et al., 1994, 1996; Shink
pathway of the BG (Temel et al., 2005). At the et al., 1996). The GPe is innervated by GABA-
same time the STN is part of the indirect pathway ergic fibers from the STR, as well as glutamatergic
of the BG (Temel et al., 2005). It conveys excita- fibers originating from the STN and the thalamus,
tory signals from the cortex directly to the GPi SNc, and brainstem (Deschênes, 1996; Fink-
and SNr via tonic glutamatergic signaling, provid- Jensen and Mikkelsen, 1991; Hazrati et al., 1990;
ing a faster pathway than the direct and indirect Kita and Kitai, 1994; Parent and Hazrati, 1995).
pathways (Nambu et al., 2000; Nambu et al., GPe signaling can be modulated via the endocan-
2002; Robledo and Féger, 1990; Rosales et al., nabinoid system through the altering of GABA
1994; Smith and Parent, 1988). The STN has uptake (Di Marzo et al., 1998; Maneue et al.,
received strong research attention in view of its 1994; Venderova et al., 2005). In the GPe and STR
role as a target for deep brain stimulation in the the endocannabinoid signaling was reported to be
treatment of Parkinson’s disease (PD) (Temel reduced in animal models of PD, as evidenced by
et al., 2005). The tripartite hypothesis dividing the reduced CB1 receptor mRNA expression (Hurley
STN in a dorsolateral motor area, a ventromedial, et al., 2003). Additionally, elevated levels of endo-
cognitive area, and a limbic tip has provided ana- cannabinoid were reported, as well as positive
tomical guidance for deep brain stimulation effects of receptor activation on the motor pheno-
(DBS) surgery (Temel et al., 2005). Interestingly, type of primate models of PD (Ferrer et al., 2003;
the discussion on the location and sharpness of the Fox et al., 2002; Marzo et al., 2000).
anatomical borders of the putative subdivisions
has fueled anatomical research into the micro-
anatomy of this nucleus (Alkemade et al., 2019;
Alkemade and Forstmann, 2014; Lambert et al.,
2012, 2015). This has led to detailed accounts of SUBSTANTIA NIGRA
protein marker distribution patterns throughout
the human STN, although to date it remains chal- The substantia nigra (SN) is also an output
lenging to link function directly to individual nucleus of the BG. The reticular and compact
protein expression. STN neurons display axon parts of the SN receive largely comparable inputs,
collaterals that innervate both the internal and but differ substantially in their output which is
external segment of the GP. Communicating neu- exerted by chemically distinct neuronal types
rons both from the GPe and STN innervate the (Gerfen and Wilson, 1996). The SN transmits
202 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

information received from the STR, via the pars inhibitory connections within a pathway and infer
reticulata, to the thalamus, superior colliculus or the overall effect on cortical activation through
brainstem. Additionally, the SN provides feedback thalamocortical projections. As illustrated in
to modulate information flow through the BG via Figure 13.2, the direct BG pathway consists of an
the STR. This modulatory feedback is mediated excitatory corticostriatal connection, an inhibitory
by the dopaminergic nigrostriatal neurons that are striatopallidal connection, another inhibitory pal-
located in the pars compacta of the SN and that lidothalamic connection, and an excitatory projec-
has strong and diverse projections onto the STR tion from the thalamus to the cortex. Since the
(Bolam et al., 1991). resulting effect on the cortex is excitatory, this
pathway is often referred to as the ‘go’ pathway.
Conversely, the indirect pathway exerts an overall
inhibitory effect on cortical activity, and is there-
USING ANATOMICAL INFORMATION TO fore referred to as the ‘no go’ pathway. This basic
STUDY FUNCTION understanding of these pathways forms the under-
pinning of leading theories that offer an explana-
The level of description of the BG nuclei and their tion of hypo- and hyperkinetic movement
connections provided here is sufficient to under- disorders and our general understanding of BG
stand the major anatomical nodes incorporated in function (Albin et al., 1989; DeLong, 1990). This
leading models of BG function. We would like to broadly applicable, but very general model does
emphasize that the levels of description of the not capture all experimental data on neuronal
internal anatomy provided here do not reflect the activity patterns, and does not cover more specific
full complexity of the BG. They can be compared aspects of behavioral control such as action selec-
to the level of a single main road on a map telling tion and learning. In this section, we will illustrate
you in which direction you need to travel, but not how extension of available theories through
what you may encounter on the road. Additional formal computational models based on BG anat-
signaling pathways, which can be major regulatory omy could contribute to the elucidation of under-
signals or ones that serve a more modulatory func- lying brain processes.
tion are not included. Reasons for their omission A next level of detail is reached through cor-
are either that the additional anatomical informa- relation of measures of task performance or indi-
tion does not contribute to explaining behavior vidual disease characteristics with recorded brain
captured in a model, or that the function of the responses using functional (f)MRI or electro-
anatomical characteristics is currently unknown. physiological recordings. It is important to note
Isolated anatomical information including that the small size of a number of the individual
connectomic data will not explain how the brain BG nuclei, their close proximity to each other and
works. However, when combined with data on other structures, and their dispersed location deep
electrophysiological properties, synaptic strength, inside the brain render it difficult to reliably record
neurotransmitter content, receptor expression activity from all network nodes simultaneously
etc., anatomy provide insights into the general (Forstmann et al., 2017). Moreover, such correla-
mechanisms that underlie brain circuit function tions provide only limited insights into the contri-
(Rockland, 2015). Subsequently, the resulting bution of individual pathways as they might to a
functional neuroanatomy can be integrated in fur- large extent contain the same structures.
ther research to understand individual differences Computational models can provide an account
in brain and behavior. Creation of formal models of the generative mechanisms that underlie
integrating all of this information allows us to observed experimental data, motivated by current
improve our understanding of how the brain gives theories in cognitive and systems neuroscience.
rise to complex human behavior. We will explain These descriptions include exact mathematical
how anatomy can funnel into computational mod- equations, therefore leaving little room for seman-
eling of human behavior, and vice versa, how com- tic ambiguities and facilitating scientific replica-
putational models can shape anatomical research. bility. The parameters of such models generally
represent biological quantities or psychological
constructs of interest, such as synaptic connection
strengths or learning rates. One important objec-
tive for using computational models is to quantify
COMPUTATIONAL MODELS OF THE the contribution of these individual parameters
BASAL GANGLIA to aspects of behavior. This can be achieved by
identifying the most plausible values for these
A straightforward approach to model BG function parameters for different (groups of) individuals
is to quantify the number of excitatory and or experimental conditions, or by investigating
Modeling of Basal Ganglia Structure and Function 203

the impact of individual parameters on model direct and indirect pathway activation with an
predictions through simulations. Another major overall exacerbated activation of the indirect path-
objective is to formulate and compare alterna- way, leading to inhibitory effects on motor control
tive models to experimental data. Through this and in the presentation of clinical motor symp-
approach, theories can be tested and further devel- toms. This theory fits with the observed changes
oped and refined. in neuronal firing rates in GPe, STN, and GPi
Since computational models have diverse (Albin et al., 1989; DeLong, 1990), although it
applications in neuroscience, it is not surprising cannot readily explain altered firing patterns in
that models are available in a wide range of forms these structures. In the Parkinsonian state, neurons
and level of detail. We can roughly distinguish of the indirect pathway have been demonstrated to
between models that aim to explain observed exhibit an increased incidence of bursting and
behavior, models that explain observed neural oscillatory firing (Bergman et al., 1994;
measurements, and so-called ‘joint’ models that Steigerwald et al., 2008). In particular, the local
capture both behavior and neural measurements field potential amplitude of beta band (∼13- 35Hz)
(Turner et al., 2017). Behavioral models are oscillations in STN is positively correlated with
widely used in the field of mathematical psychol- severity of bradykinesia and rigidity (Neumann
ogy, in which estimates of cognitive parameters et al., 2016; van Wijk et al., 2016) and its reduc-
are often statistically linked to neural measure- tion in response to dopaminergic medication
ments (Forstmann et al., 2015). These models (Kühn et al., 2006; Kühn et al., 2009; Ray et al.,
provide handles to understand the type of com- 2008) or DBS (Kühn et al., 2008; Ray et al., 2008)
putations implemented in the brain that underlie predicts observed clinical improvement.
cognition and behavior. At the same time, neu- Identifying the synaptic circuits involved in beta
ral models are biophysical models that typically generation is important for understanding the
describe the time evolution of neuronal membrane pathophysiology of PD and the development of
potential or firing rates with differential equations. targeted treatments.
These are valuable for investigating how neuronal Dynamical systems theory indicates that oscil-
functions arise from biological properties such as lations in a neuronal network can emerge when
specific ionic currents or synaptic connectivity excitatory and inhibitory neurons are recurrently
patterns. We refer to (Dayan and Abbott, 2005; connected. Considering the cortico-basal ganglia-
Forstmann and Wagenmakers, 2015; Izhikevich, thalamus network, there are several ways in which
2005) for excellent introductions on these topics. oscillations could potentially arise in the system
Here, we will illustrate how the use of biophysical (Figure 13.2A). Many studies have focused on
models has provided more insights into the origin the STN-GPe circuit. Several research groups
of pathological neural oscillations in Parkinson’s (e.g., Gillies et al., 2002; Holgado et al., 2010;
disease (PD), and how biologically inspired mod- Humphries et al., 2006; Liu et al., 2016; Terman
els of behavior have served to elucidate the neural et al., 2002) have applied computational models
underpinnings of reinforcement learning. to understand how the membrane potential or fir-
ing rate of individual neurons or populations of
neurons in this circuit responds to synaptic input.
This work shows that oscillations emerge within
certain parameter ranges of synaptic connection
BIOPHYSICAL MODELING IN strengths and transmission time delays within
PARKINSON’S DISEASE the circuit. Notably, connections leading directly
to the STN-GPe circuit have a critical influence
PD is associated with dysfunction of the basal (Holgado et al., 2010). A mere increase in striatal
ganglia. Cardinal symptoms such as bradykinesia inhibitory input to GPe could trigger oscillations
(slowness of movement), rigidity, and tremor arise (Gillies et al., 2002; Kumar et al., 2011; Terman
as a result of the degeneration of dopaminergic et al., 2002). Alternatively, inhibitory interactions
neurons in the SNc. According to current view- between striatal MSNs (McCarthy et al., 2011) or
points (Surmeier et al., 2007), D1 and D2 receptor in combination with fast-spiking interneurons and
activation modulate the intrinsic excitability of the feedback loop with GPe (Corbit et al., 2016)
MSNs in distinct fashions: D1-receptor activation have been proposed as a possible source of beta
in MSNs that belong to the direct pathway have oscillations. Long feedback loops involving cor-
excitatory effects, whereas D2-receptor activation tex can also generate oscillatory activity (Leblois
in MSNs that belong to the indirect pathway give et al., 2006; Pavlides et al., 2015). Last, but not
rise to inhibitory effects on the excitability of the least it is possible that oscillations originate else-
MSNs. The reduced dopamine availability as where in cortex or the cortico-thalamic circuit and
observed in PD would cause a disbalance between propagate through the BG (Hahn and Mcintyre,
204 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

2010; Pavlides et al., 2015; van Albada and competing ones (Mink, 1996). Dopamine has
Robinson, 2009). Many of these studies motivate been implicated in learning to select the most
their parameter values or validate their results with rewarding action given a particular context of
findings from experimental studies. sensory input. Striatal neurons receive phasic
The question then rises which of these pos- dopaminergic input from the SNc in response to
sibilities most likely underlies the emergence of unexpected rewards to a conditioned stimulus
beta oscillations in PD. Moran et al. (2011) used (Schultz, 1998). Conversely, the omission of an
dynamic causal modelling (DCM) to address expected reward leads to a dip in dopaminergic
this question through computational analyses. signaling compared to tonic baseline innervation.
They constructed a generative model comprised Synaptic plasticity has been demonstrated in stud-
of coupled neural masses, one for each excita- ies where the stimulus-response combination
tory or inhibitory neural population included repeatedly leads to unexpected rewards. The
in a cortico-basal ganglia-thalamus network. release of dopamine promotes long-term potentia-
Parameters derived from this model included tion by activating MSN D1 receptors (Surmeier
synaptic coupling strengths between neural pop- et al., 2007), resulting in the activation of the
ulations. Their values were estimated through direct pathway and increasing the probability for
Bayesian model inversion by comparing model selecting the same response option next time
predictions of power- and cross-spectral densi- (Cohen and Frank, 2009). The same action repre-
ties with experimental data. These data were sentation in the indirect pathway weakens as
obtained from a group of 6-hydroxydopamine dopamine promotes long-term depression through
(6-OHDA)-lesioned rats (an established model the activation of MSN D2 receptors. A decrease in
of PD), and compared to an appropriate control dopamine release would result in an inverse effect
group. Electrocorticography and local field poten- through long-term potentiation of D2 MSN neu-
tials were recorded from four sites in the net- rons (Shen et al., 2008). This process creates a
work: cortex, GPe, STN, and the entopeduncular dynamic balance between facilitation and inhibi-
nucleus (EPN, the rodent orthologue of the GPi). tion of response actions and, although speculative,
6-OHDA-lesioned rats showed a marked spec- could underlie reinforcement learning.
tral peak in the beta frequency range that was not Frank et al. (2005, 2007) performed a series
observed in control animals (Figure 13.2B). The of computational modeling studies to investi-
difference between the two groups of animals was gate whether the BG neural architecture supports
best explained by a significantly increased input action selection learning behavior as observed in
from cortex to the STN and a reduced input from healthy individuals and can predict how behavior
the STN to GPe in the 6-OHDA-lesioned animals. relates to varying dopamine levels. Inspired both
Later DCM studies using local field potentials by BG anatomy, and the computational properties
recorded with DBS electrodes in human patients of neurons, a neural network model was designed
confirmed that the presence of beta oscillations is (Figure 13.3A) (Frank, 2005). The model learned
associated with altered connection strengths of the to select one of two behavioral responses for given
hyperdirect pathway and the STN-GPe circuit, as input stimuli. Each node in the network contained
well as other connections in the indirect pathway a column of neurons that encoded activity for one
and elsewhere (Figure 13.2B) (Marreiros et al., of the response options. Neurons were activated
2013; van Wijk et al., 2018). These results are in through input from the previous node, incorpo-
support of widespread effects of dopamine deple- rating the synaptic weight of the connection. The
tion on synaptic transmission in the cortico-basal balance in activity between the direct and indirect
ganglia-thalamus network. Multiple connections pathway ultimately determined response option
are likely to contribute to the onset of pathological selection at the output level of the network, the
beta oscillations in the indirect pathway. premotor cortex. Hebbian learning as well as error
learning was included to drive the updating of syn-
aptic weights after receiving feedback whether the
selected response was correct.
ROLE OF THE BASAL GANGLIA IN ACTION Simulations under normal conditions revealed
a good learning performance (Frank, 2005).
SELECTION AND REINFORCEMENT Interestingly, the model performed significantly
LEARNING worse if only the direct pathway was included
compared to the intact network, suggesting that
The classical view poses that the direct and indi- the enhanced contrast created by opposing modu-
rect pathways serve to gait action representations lations in the direct vs. indirect pathway improves
from frontal cortex by selectively facilitating one discriminability between response options (Cohen
of the action representations whilst inhibiting all and Frank, 2009). Furthermore, the model could
Modeling of Basal Ganglia Structure and Function 205

explain how an altered balance of tonic and phasic cortex via the hyperdirect pathway would serve as
dopamine leads to unequal learning effects from a rapid signal to withhold all responses in case of
positive vs. negative outcomes (Frank et al., 2004). conflicting sensory input. This mechanism avoids
The model correctly predicted that PD patients are premature responses by allowing extra decision-
better in learning from negative than from posi- making time. To test this, Frank et al. (2007)
tive outcomes when in a low dopaminergic state, a included the STN in their network and tested the
pattern that reverses after taking levodopa (Figure effect of STN-DBS on impulsive behavior in a
13.3B). This finding may help explain why dopa- decision-making task. DBS did not affect learn-
minergic medication improves performance for ing but speeded up reaction times (RTs) for high-
some cognitive tasks but yields opposite effects conflict (difficult) trials. The modeling confirmed
in others. Next, the role of the STN was consid- that interfering with STN activity increases the
ered as it is one of the primary targets for DBS probability of premature responses triggered by
treatment of PD. Projections from STN to GPi are early firing in the direct pathway. These examples
more diffuse than those from GPe to GPi (Mink, highlight how computational modeling can aid in
1996; Parent and Hazrati, 1995), which has fueled identifying the BG anatomical pathways and bio-
the hypothesis that the STN acts to globally sup- logical processes involved in controlling aspects
press responses. Its monosynaptic input from of behavior.

Figure 13.3 A biologically constrained computational model of action selection correctly


predicts learning behavior in Parkinson’s disease.
A Network architecture of the model with example activation (in different grey shades). Each
cell represents a processing unit whose activation dynamics is inspired from real neurons.
Each column encodes one of two response options. Direct (“go”) and indirect (“no go”) path-
ways originate in the striatum. Relative activation in these pathways for the two response
options determines the final behavioral response (“R1” vs. “R2”). Synaptic weights in the
network are adjusted according to Hebbian and error learning after outcome presentation.
B Model predictions are tested against performance in a probabilistic decision-making task.
Individuals implicitly learn which of two stimuli in a training-pair is associated with the
higher probability of positive outcome. Subsequently, they are tested during presentation
of novel test-pairs of stimuli, to infer whether they rely more on learning from positive
(“Choose”) or negative feedback (“Avoid”). PD patients took part before (“OFF”) and after
(“ON”) receiving dopaminergic medication. Observed performance during the task is shown
top right (cf. model predictions bottom right). Note the reversal in learning from negative vs.
positive outcomes in PD with medication is well-captured by the model.
Source: Adapted from Frank et al., 2004. Reprinted with permission from AAAS.
206 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

CONSIDERATIONS IN COMPUTATIONAL identify the missing part of the anatomical archi-


MODELING tecture. If we return to the comparison to the traffic
architecture, imagine the planning of a three-day
holiday trip, and a calculation of whether the traf-
By definition, computational models are simplifi- fic architecture allows the completion of such a
cations of the biological system they reflect. For trip. The hypothesis formulated could be that an
example, despite evidence for anatomical projec- overnight accommodation should be included
tions from GPe to STR (Mallet et al., 2012), and along the set route. Identification of accommoda-
from GPe to GPi (Smith et al., 1998), both Moran tion for overnight stays along the route would then
et al. (2011) and Marreiros et al. (2013) concluded represent the equivalent of anatomical research
through formal model comparisons that incorpo- inspired by the modeling results.
rating these connections into the basal BG circuit Computational modeling could lead to the
did not improve the model’s performance in description of the anatomical nodes beyond those
explaining beta oscillations beyond the cost of already in the model. The incorporation of addi-
added complexity. Similarly, initial models by tional structures into the model architecture may
Frank et al. (2005) did not include the STN, pre- not be needed to explain the main function of the
sumably because the indirect pathway connection model, but could for instance allow the refinement
between GPe and GPi was sufficient for obtaining of specific models. One could for instance specu-
qualitatively correct model predictions of action late that models based on dopamine signaling
selection. On the other hand, leaving out relevant would benefit from further refinement through the
components could significantly alter the behavior incorporation of modulating systems. Although
of the system. Rubin (2017) provides an in-depth speculative, the cannabinoid system is an interest-
discussion on the importance of biological details ing candidate.
in computational models of BG dysfunction. Conversely, hypothesis generation can be
Validation of computational models on the basis driven by anatomy. Questions raised from the field
of experimental data will remain crucial for reach- of anatomy include: What is the function of neu-
ing reliable and robust conclusions. A challenge ronal populations or sources of neuromodulation
for future research is to integrate neural and that are currently not included in circuit models?
behavioral models for finding even more direct For instance, what is the role of serotonergic mod-
links between behavior and underlying biophysi- ulation of STN function, and to what extent does
ological processes. DBS affect serotonin signaling? Additionally,
does potential modulation of serotonergic signal-
ing contribute to the desired clinical effects, or to
the unwanted side effects of DBS (Temel et al.,
SYNERGISTIC APPROACHES 2005)? Can we identify additional anatomical
components that regulate behavior other than that
Data from individual research fields will not described in available models, or do they provide
explain how the brain works. The field of descrip- additional modulatory components for the model?
tive anatomy provides an account of the character, Bringing together hypotheses from modeling and
form, size, and position of structures in the brain. anatomy is expected to contribute to the refinine-
The combining of maps that contain multiple ment of available models, the identification of
levels of description, can grow into sizable atlas- latent parameters, as well as the identification
ing efforts which provide valuable resources. of structures responsible for these parameters.
These resources can be used by researchers from Ultimately these efforts can lead to a deeper
other fields to retrieve the data relevant for their understanding of the BG nuclei and regulation of
theory formation, and provide anatomical validity their activity.
to their research findings. Through these integra-
tive efforts, descriptive anatomy is transferred to
the field of functional anatomy and computational
modeling, which in turn can indicate missing SUMMARY AND CONCLUSIONS
nodes in the network models, which should be
retrieved via anatomical studies. Understanding the role of the BG in human
The combined research data can subsequently behavior is the goal of many research groups
be used for hypothesis generation. If the anatomi- working in individual research disciplines world-
cal architecture and model behavior do not match, wide. Currently, we can integrate findings from
this can either lead to the adjustment of the model, different fields using an interdisciplinary
or to additional research in the field of anatomy to approach. Individual research disciplines can
Modeling of Basal Ganglia Structure and Function 207

benefit from other disciplines through the filling and non-human primate subthalamic nucleus.
of knowledge gaps that result from technical limi- Brain Structure and Function, 220(6), 3075–3086.
tations inherent to specific approaches, which are Aston-Jones, G., & Cohen, J. D. (2005). An integra-
present in every type of research, and which could tive theory of locus coeruleus-norepinephrine
potentially lead to unwanted biases within specific function: Adaptive gain and optimal performance.
research fields. Annual Review of Neuroscience, 28, 403–450.
Formal computational modeling is crucial in Balleine, B. W., Delgado, M. R., & Hikosaka, O.
this process, through providing a level of descrip- (2007). The role of the dorsal striatum in reward
tion that allows the translation of quantitative and decision-making. Journal of Neuroscience,
research parameters between behavioral and 27(31), 8161–8165.
anatomo-functional data. Additionally, formal Beckstead, R. M. (1983). A pallidostriatal projection
modeling can lead to the identification of latent in the cat and monkey. Brain Research Bulletin,
factors that are needed to accurately explain and 11(6), 629–632.
predict behavior and behavioral changes. For BG Bergman, H., Wichmann, T., Karmon, B., & DeLong,
modeling, these interdisciplinary approaches have M. R. (1994). The primate subthalamic nucleus. II.
already led to a deeper understanding of the brain Neuronal activity in the MPTP model of parkinson-
alterations that are observed in people with PD, as ism. Journal of Neurophysiology 72(2), 507–520.
well as more insight in the mechanisms underly- Bevan, M. D., Bolam, J. P., & Crossman, A. R. (1994).
ing reinforcement learning. Convergent synaptic input from the neostriatum
Crucial for the success of such efforts is the and the subthalamus onto identified nigrothalamic
availability of neural and behavioral data for com- neurons in the rat. European Journal of Neurosci-
putational modeling. With increasing importance ence, 6(3), 320–334.
of open science and open data, more research Bevan, M. D., Smith, A. D., & Bolam, J. P. (1996). The
groups are now sharing their data allowing reuse substantia nigra as a site of synaptic integration of
and reanalysis of brain and behavioral data- functionally diverse information arising from the
sets. The process of sharing data within the field ventral pallidum and the globus pallidus in the rat.
of cognitive neuroscience is further facilitated Neuroscience, 75(1), 5–12.
through initiatives that aim to create uniformity in Bevan, Mark D. (2021). Motor control: A basal gan-
data sharing structure that facilitates the algorith- glia feedback circuit for action suppression. Cur-
mic processing of individual datasets. An excel- rent Biology, 31(4), R191–R193.
lent example of such efforts is the introduction of Bishop, G. A., Chang, H. T., & Kitai, S. T. (1982). Mor-
the Brain Imaging Data Structure (BIDS) format, phological and physiological properties of neostri-
which provides an intuitive and consistent method atal neurons: An intracellular horseradish peroxidase
for the arrangement of anatomo-functional and study in the rat. Neuroscience, 7(1), 179–191.
behavioral data (Gorgolewski et al., 2016). Bloem, B., Huda, R., Sur, M., & Graybiel, A. M.
(2017). Two-photon imaging in mice shows stri-
osomes and matrix have overlapping but differen-
tial reinforcement-related responses. ELife, 6. doi:
10.7554/eLife.32353
REFERENCES Bogacz, R., & Gurney, K. (2007). The basal ganglia
and cortex implement optimal decision making
Albin, R. L., Young, A. B., & Penney, J. B. (1989). The between alternative actions. Neural Computation,
functional anatomy of basal ganglia disorders. 19(2), 442–477.
Trends in Neurosciences, 12(10), 366–375. Bolam, J. P., Francis, C. M., & Henderson, Z. (1991).
Alkemade, A. (2013). Subdivisions and anatomical Cholinergic input to dopaminergic neurons in the
boundaries of the subthalamic nucleus. Journal of substantia nigra: A double immunocytochemical
Neuroscience, 33(22), 9233–9244. study. Neuroscience, 41(2-3), 483–494.
Alkemade, A., de Hollander, G., Miletic, S., Keuken, Bolam, J. P., Hanley, J. J., Booth, P. A. C., & Bevan, M.
M. C., Balesar, R., de Boer, O. J., … Forstmann, B. D. (2000). Synaptic organisation of the basal gan-
U. (2019). The functional microscopic neuroanat- glia. Journal of Anatomy, 196(4), 527–542.
omy of the human subthalamic nucleus. Brain Calabresi, P., Pisani, A., Mercuri, N. B., & Bernardi, G.
Structure and Function, 224(9), 3213–-3227. (1996). The corticostriatal projection: From synap-
Alkemade, A., & Forstmann, B. U. (2014). Do we tic plasticity to dysfunctions of the basal ganglia.
need to revise the tripartite subdivision hypothesis Trends in Neurosciences, 19(1), 19–24.
of the human subthalamic nucleus (STN)? Neuro- Castañé, A., Theobald, D. E. H., & Robbins, T. W.
Image, 95, 326–329. (2010). Selective lesions of the dorsomedial stria-
Alkemade, A., Schnitzler, A., & Forstmann, B. U. tum impair serial spatial reversal learning in rats.
(2015). Topographic organization of the human Behavioural Brain Research, 210(1), 74–83.
208 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Clark, L., Roiser, J. P., Cools, R., Rubinsztein, D. C., the MPTP-lesioned nonhuman primate model of
Sahakian, B. J., & Robbins, T. W. (2005). Stop Parkinson’s disease. Movement Disorders, 17(6),
signal response inhibition is not modulated by 1180–1187.
tryptophan depletion or the serotonin transporter Frank, M. J. (2005). Dynamic dopamine modulation
polymorphism in healthy volunteers: Implications in the basal ganglia: A neurocomputational
for the 5-HT theory of impulsivity. Psychopharma- account of cognitive deficits in medicated and
cology, 182(4), 570–578. nonmedicated Parkinsonism. Journal of Cognitive
Cohen, M. X., & Frank, M. J. (2009). Neurocomputa- Neuroscience, 17(1), 51–72.
tional models of basal ganglia function in learning, Frank, M. J., Samanta, J., Moustafa, A. A., & Sher-
memory and choice. Behavioural Brain Research, man, S. J. (2007). Hold your horses: Impulsivity,
199(1), 141–156. deep brain stimulation, and medication in Parkin-
Corbit, V. L., Whalen, T. C., Zitelli, K. T., Crilly, S. Y., sonism. Science, 318, 1309–1312.
Rubin, J. E., & Gittis, A. H. (2016). Pallidostriatal Frank, M. J., Seeberger, L. C., & O’Reilly, R. C. (2004).
projections promote β oscillations in a dopamine- By carrot or by stick: Cognitive reinforcement learn-
depleted biophysical network model. Journal of ing in Parkinsonism. Science, 306, 1940–1943.
Neuroscience, 36(20), 5556–5571. Frank, M. J., Woroch, B. S., & Curran, T. (2005). Error-
Dayan, P., & Abbott, L. F. (2005). Theoretical neurosci- related negativity predicts reinforcement learning
ence: Computational and mathematical modeling and conflict biases. Neuron, 47(4), 495–501.
of neural systems (Computational Neuroscience Gerfen, C. R., & Wilson, C. J. (1996). Chapter II:The
Series). Cambridge, MA. MIT Press. basal ganglia. In L. W. Swanson, A. Björklund & T.
DeLong, M. R. (1990). Primate models of movement Hökfelt (Eds.), Handbook of chemical neuroanat-
disorders of basal ganglia origin. Trends in Neuro- omy (Vol 12, pp 371-468). Amsterdam: Elsevier.
sciences, 13(7), 281–285. Gillies, A., Willshaw, D., & Li, Z. (2002). Subthalamic-
Deschênes, M. (1996). A single-cell study of the pallidal interactions are critical in determining
axonal projections arising from the posterior intral- normal and abnormal functioning of the basal
aminar thalamic nuclei in the rat. European Jour- ganglia. Proceedings of the Royal Society of
nal of Neuroscience, 8(2), 329–343. London. Series B: Biological Sciences, 269(1491),
Di Marzo, V., Melck, D., Bisogno, T., & De Petrocellis, 545–551.
L. (1998). Endocannabinoids: Endogenous can- Hahn, P. J., & Mcintyre, C. C. (2010). Modeling shifts
nabinoid receptor ligands with neuromodulatory in the rate and pattern of subthalamopallidal net-
action. Trends in Neurosciences, 21(12), 521–528. work activity during deep brain stimulation. Journal
Ferrer, B., Asbrock, N., Kathuria, S., Piomelli, D., & Giuf- of Computational Neuroscience, 28(3), 425–441.
frida, A. (2003). Effects of levodopa on endocannabi- Hazrati, L. N., Parent, A., Mitchell, S., & Haber, S. N.
noid levels in rat basal ganglia: Implications for the (1990). Evidence for interconnections between the
treatment of levodopa-induced dyskinesias. Euro- two segments of the globus pallidus in primates:
pean Journal of Neuroscience, 18(6), 1607–1614. A PHA-L anterograde tracing study. Brain Research,
Fink-Jensen, A., & Mikkelsen, J. D. (1991). A direct 533(1), 171–175.
neuronal projection from the entopeduncular Helie, S., Chakravarthy, S., & Moustafa, A. A. (2013).
nucleus to the globus pallidus. A PHA-L antero- Exploring the cognitive and motor functions of the
grade tracing study in the rat. Brain Research, basal ganglia: An integrative review of computa-
542(1), 175–179. tional cognitive neuroscience models. Frontiers in
Forstmann, B. U., De Hollander, G., Van Maanen, L., Computational Neuroscience, 7. doi: 10.3389/
Alkemade, A., & Keuken, M. C. (2017). Towards a fncom.2013.00174
mechanistic understanding of the human subcor- Herkenham, M., Edley, S. M., & Stuart, J. (1984). Cell
tex. Nature Reviews Neuroscience, 18(1), 57–65. clusters in the nucleus accumbens of the rat, and
Forstmann, B. U., Ratcliff, R., & Wagenmakers, E.-J. the mosaic relationship of opiate receptors, acetyl-
(2016). Sequential sampling models in cognitive cholinesterase and subcortical afferent termina-
neuroscience: Advantages, applications, and exten- tions. Neuroscience, 11(3), 561–593.
sions. Annual Review of Psychology, 67, 641–666. Holgado, A. J., Terry, J. R., & Bogacz, R. (2010). Con-
Forstmann, B. U., & Wagenmakers, E.-J. (2015). ditions for the generation of beta oscillations in
Model-based cognitive neuroscience: A concep- the subthalamic nucleus-globus pallidus network.
tual introduction. In B. U. Forstmann & E. J. Journal of Neuroscience, 30(37), 12340–12352.
Wagenmakers (Eds.), An introduction to model- Humphries, M. D., Stewart, R. D., & Gurney, K. N.
based cognitive neuroscience (pp. 139–156). New (2006). A physiologically plausible model of action
York: Springer-Verlag. selection and oscillatory activity in the basal ganglia.
Fox, S. H., Henry, B., Hill, M., Crossman, A., & Journal of Neuroscience, 26(50), 12921–12942.
Brotchie, J. (2002). Stimulation of cannabinoid Hurley, M. J., Mash, D. C., & Jenner, P. (2003).
receptors reduces levodopa-induced dyskinesia in Expression of cannabinoid CB1 receptor mRNA in
Modeling of Basal Ganglia Structure and Function 209

basal ganglia of normal and parkinsonian human Liu, F., Wang, J., Liu, C., Li, H., Deng, B., Fietkiewicz,
brain. Journal of Neural Transmission, 110(11), C., & Loparo, K. A. (2016). A neural mass model
1279–1288. of basal ganglia nuclei simulates pathological beta
Ilinsky, I. A., Yi, H., & Kultas-Ilinsky, K. (1997). Mode rhythm in Parkinson’s disease. Chaos, 26(12). doi:
of termination of pallidal afferents to the thala- 10.1063/1.4972200
mus: A light and electron microscopic study with Mallet, N., Micklem, B. R., Henny, P., Brown, M. T.,
anterograde tracers and immunocytochemistry in Williams, C., Bolam, J. P., … Magill, P. J. (2012).
Macaca Mulatta. Journal of Comparative Neurol- Dichotomous organization of the external globus
ogy, 386(4), 601–612. pallidus. Neuron, 74(6), 1075–1086.
Izhikevich, E. M. (2010). Dynamical systems in neuro- Maneue, Y. P., Mitchell, I. J., Crossman, A. R., &
science: The geometry of excitability and bursting Brotchiel, J. M. (1994). On the role of enkephalin
(Computational Neuroscience Series). Cambridge, cotransmission in the GABAergic striatal efferents
MA. MIT Press. to the globus pallidus. Experimental Neurology,
Kita, H., & Kitai, S. T. (1994). The morphology of 125(1), 65–71.
globus pallidus projection neurons in the rat: An Marreiros, A. C., Cagnan, H., Moran, R. J., Friston, K.
intracellular staining study. Brain Research, 636(2), J., & Brown, P. (2013). Basal ganglia–cortical inter-
308–319. actions in Parkinsonian patients. NeuroImage, 66,
Kühn, A. A., Kempf, F., Brücke, C., Doyle, L. G., 301–310.
Martinez-Torres, I., Pogosyan, A., … Brown, P. Marzo, V. di, Hill, M. P., Bisogno, T., Crossman, A. R.,
(2008). High-frequency stimulation of the subtha- & Brotchie, J. M. (2000). Enhanced levels of
lamic nucleus suppresses oscillatory β activity in endogenous cannabinoids in the globus pallidus
patients with Parkinson’s disease in parallel with are associated with a reduction in movement in an
improvement in motor performance. Journal of animal model of Parkinson’s disease. FASEB Jour-
Neuroscience, 28(24), 6165–6173. nal, 14(10), 1432–1438.
Kühn, A. A., Kupsch, A., Schneider, G.-H., & Brown, Matamales, M., Bertran-Gonzalez, J., Salomon, L.,
P. (2006). Reduction in subthalamic 8-35 Hz oscil- Degos, B., Deniau, J. M., Valjent, E., Hervé, D., &
latory activity correlates with clinical improvement Girault, J. A. (2009). Striatal medium-sized spiny
in Parkinson’s disease. European Journal of Neuro- neurons: Identification by nuclear staining and study
science, 23(7), 1956–1960. of neuronal subpopulations in BAC transgenic mice.
Kühn, A. A., Tsui, A., Aziz, T., Ray, N., Brücke, C., PLoS One, 4(3). doi: 10.1371/journal.pone.0004770
Kupsch, A., … Brown, P. (2009). Pathological syn- McCarthy, M. M., Moore-Kochlacs, C., Gu, X.,
chronisation in the subthalamic nucleus of patients Boyden, E. S., Han, X., & Kopell, N. (2011). Striatal
with Parkinson’s disease relates to both bradykin- origin of the pathologic beta oscillations in Parkin-
esia and rigidity. Experimental Neurology, 215(2), son’s disease. Proceedings of the National Acad-
380–387. emy of Sciences of the United States of America,
Kumar, A., Cardanobile, S., Rotter, S., & Aertsen, A. 108(28), 11620–11625.
(2011). The role of inhibition in generating and Meredith, G. E., Pattiselanno, A., Groenewegen, H.
controlling Parkinson’s disease oscillations in the J., & Haber, S. N. (1996). Shell and core in monkey
basal ganglia. Frontiers in Systems Neuroscience, and human nucleus accumbens identified with
5. doi; 10.3389/fnsys.2011.00086 antibodies to calbindin-D28k. Journal of Compara-
Lambert, C., Zrinzo, L., Nagy, Z., Lutti, A., Hariz, M., tive Neurology, 365(4), 628–639.
Foltynie, T., … Frackowiak, R. (2012). Confirma- Middleton, F. A., & Strick, P. L. (2000). Basal ganglia
tion of functional zones within the human subtha- and cerebellar loops: Motor and cognitive circuits.
lamic nucleus: patterns of connectivity and Brain Research: Brain Research Reviews, 31(2–3),
sub-parcellation using diffusion weighted imag- 236–250.
ing. Neuroimage, 60(1), 83–94. Mink, J. W. (1996). The basal ganglia: Focused selec-
Lambert, C., Zrinzo, L., Nagy, Z., Lutti, A., Hariz, M., tion and inhibition of competing motor programs.
Foltynie, T., … Frackowiak, R. (2015). Do we need Progress in Neurobiology, 50(4), 381–425.
to revise the tripartite subdivision hypothesis of Moran, R. J., Mallet, N., Litvak, V., Dolan, R. J.,
the human subthalamic nucleus (STN)? Response Magill, P. J., Friston, K. J., & Brown, P. (2011).
to Alkemade and Forstmann. NeuroImage, 110(1- Alterations in brain connectivity underlying beta
2). doi: 10.1016/j.neuroimage.2015.01.038 oscillations in Parkinsonism. PLoS Computational
Leblois, A., Boraud, T., Meissner, W., Bergman, H., & Biology, 7(8). Doi: 10.1371/journal.pcbi.1002124
Hansel, D. (2006). Competition between feedback Nakanishi, H., Kita, H., & Kitai, S. T. (1991). Intracellular
loops underlies normal and pathological dynamics study of rat entopeduncular nucleus neurons in an
in the basal ganglia. Journal of Neuroscience, in vitro slice preparation: Response to subthalamic
26(13), 3567–3583. stimulation. Brain Research, 549(2), 285–291.
210 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Nambu, A, Tokuno, H., & Takada, M. (2002). Func- reticulata: A microdialysis study in the rat. Brain
tional significance of the cortico-subthalamo- Research, 645(1–2), 335–337.
pallidal “hyperdirect” pathway. Neuroscience Rubin, J. E. (2017). Computational models of basal
Research, 43(2), 111–117. ganglia dysfunction: The dynamics is in the details.
Nambu, Atsushi, Tokuno, H., Hamada, I., Kita, H., Current Opinion in Neurobiology, 46, 127–135.
Imanishi, M., Akazawa, T., … Hasegawa, N. Saleeba, C., Dempsey, B., Le, S., Goodchild, A., &
(2000). Excitatory conical inputs to pallidal neu- McMullan, S. (2019). A student’s guide to neural
rons via the subthalamic nucleus in the monkey. circuit tracing. Frontiers in Neuroscience, 13. doi:
Journal of Neurophysiology, 84(1), 289–300. 10.3389/fnins.2019.00897
Neumann, W.-J., Degen, K., Schneider, G.-H., Brücke, Schultz, W. (1998). Predictive reward signal of dopa-
C., Huebl, J., Brown, P., & Kühn, A. A. (2016). mine neurons. Journal of Neurophysiology, 80(1),
Subthalamic synchronized oscillatory activity cor- 1–27.
relates with motor impairment in patients with Sharpe, N. A., & Tepper, J. M. (1998). Postnatal devel-
Parkinson’s disease. Movement Disorders, 31(11), opment of excitatory synaptic input to the rat
1748–1751. neostriatum: An electron microscopic study. Neu-
Parent, A., & Hazrati, L. N. (1995). Functional anat- roscience, 84(4), 1163–1175.
omy of the basal ganglia. I. The cortico-basal Shen, W., Flajolet, M., Greengard, P., & Surmeier, D.
ganglia-thalamo-cortical loop. Brain Research: J. (2008). Dichotomous dopaminergic control of
Brain Research Reviews, 20(1), 91–127. striatal synaptic plasticity. Science, 321, 848–851.
Parent, M., Lévesque, M., & Parent, A. (1999). The Shink, E., Bevan, M. D., Bolam, J. P., & Smith, Y.
pallidofugal projection system in primates: evi- (1996). The subthalamic nucleus and the external
dence for neurons branching ipsilaterally and con- pallidum: two tightly interconnected structures
tralaterally to the thalamus and brainstem. Journal that control the output of the basal ganglia in the
of Chemical Neuroanatomy, 16(3), 153–165. monkey. Neuroscience, 73(2), 335–357.
Pavlides, A., Hogan, S. J., & Bogacz, R. (2015). Com- Shink, E., & Smith, Y. (1995). Differential synaptic
putational models describing possible mechanisms innervation of neurons in the internal and external
for generation of excessive beta oscillations in segments of the globus pallidus by the GABA- and
Parkinson’s disease. PLoS Computational Biology, glutamate-containing terminals in the squirrel
11(12), doi: 10.1371/journal.pcbi.1004609 monkey. Journal of Comparative Neurology,
Percheron, G., Yelnik, J., & François, C. (1984). A 358(1), 119–141.
Golgi analysis of the primate globus pallidus. III. Smith, J. B., Klug, J. R., Ross, D. L., Howard, C. D.,
Spatial organization of the striato-pallidal com- Hollon, N. G., Ko, V. I., … Jin, X. (2016). Genetic-
plex. Journal of Comparative Neurology, 227(2), based dissection unveils the inputs and outputs of
214–227. striatal patch and matrix compartments. Neuron,
Plenz, D., & Wickens, J. R. (2016). The striatal skele- 91(5), 1069–1084.
ton: Medium spiny projection neurons and their Smith, Y., & Parent, A. (1988). Neurons of the sub-
lateral connections. In H. Steiner & K. Y. Tseng thalamic nucleus in primates display glutamate but
(Eds.), Handbook of behavioral neuroscience: Part not GABA immunoreactivity. Brain Research,
B. Anatomy and physiology of the striatum (Vol. 453(1-2), 353–356.
24, pp. 121–136). London: Elsevier/Academic. Smith, Y., Parent, A., Seguela, P., & Descarries, L.
Ray, N. J., Jenkinson, N., Wang, S., Holland, P., Brit- (1987). Distribution of GABA-immunoreactive
tain, J. S., Joint, C., … Aziz, T. (2008). Local field neurons in the basal ganglia of the squirrel
potential beta activity in the subthalamic nucleus monkey (Saimiri sciureus). Journal of Comparative
of patients with Parkinson’s disease is associated Neurology, 259(1), 50–64.
with improvements in bradykinesia after dopa- Soghomonian, J. J., Descarries, L., & Watkins, K. C.
mine and deep brain stimulation. Experimental (1989). Serotonin innervation in adult rat neostria-
Neurology, 213(1), 108–113. tum. II. Ultrastructural features: A radioautographic
Robledo, P., & Féger, J. (1990). Excitatory influence of and immunocytochemical study. Brain Research,
rat subthalamic nucleus to substantia nigra pars 481(1), 67–86.
reticulata and the pallidal complex: electrophysio- Steigerwald, F., Pötter, M., Herzog, J., Pinsker, M.,
logical data. Brain Research, 518(1-2), 47–54. Kopper, F., Mehdorn, H., … Volkmann, J. (2008).
Rockland, K. S. (2015). About connections. Frontiers in Neuronal activity of the human subthalamic nucleus
Neuroanatomy, 9. doi: 10.3389/fnana.2015.00061 in the parkinsonian and nonparkinsonian state.
Rosales, M. G., Flores, G., Hernández, S., Martínez- Journal of Neurophysiology, 100(5), 2515–2524.
Fong, D., & Aceves, J. (1994). Activation of subtha- Surmeier, D. J., Ding, J., Day, M., Wang, Z., & Shen,
lamic neurons produces NMDA receptor-mediated W. (2007). D1 and D2 dopamine-receptor modu-
dendritic dopamine release in substantia nigra pars lation of striatal glutamatergic signaling in striatal
Modeling of Basal Ganglia Structure and Function 211

medium spiny neurons. Trends in Neurosciences, Venderova, K., Brown, T. M., & Brotchie, J. M. (2005).
30(5), 228–235. Differential effects of endocannabinoids on [3H]-
Temel, Y., Blokland, A., Steinbusch, H. W. M., & Visser- GABA uptake in the rat globus pallidus. Experi-
Vandewalle, V. (2005). The functional role of the mental Neurology, 194(1), 284–287.
subthalamic nucleus in cognitive and limbic circuits. Voorn, P., Vanderschuren, L. J., Groenewegen, H. J.,
Progress in Neurobiology, 76(6), 393–413. Robbins, T. W., & Pennartz, C. M. (2004). Putting
Terman, D., Rubin, J. E., Yew, A. C., & Wilson, C. J. a spin on the dorsal-ventral divide of the striatum.
(2002). Activity patterns in a model for the sub- Trends in Neurosciences, 27(8), 468–474.
thalamopallidal network of the basal ganglia. Wang, A. Y., Miura, K., & Uchida, N. (2013). The
Journal of Neuroscience, 22(7), 2963–2976. dorsomedial striatum encodes net expected
Turner, B. M., Forstmann, B. U., Love, B. C., Palmeri, return, critical for energizing performance vigor.
T. J., & Van Maanen, L. (2017). Approaches to Nature Neuroscience, 16(5), 639–647.
analysis in model-based cognitive neuroscience. Smith, Y., Bevan, M.D., Shink, E., & Bolam, J.P.
Journal of Mathematical Psychology, 76, 65–79. (1998). Microcircuitry of the direct and indirect
van Albada, S. J., & Robinson, P. A. (2009). Mean- pathways of the basal ganglia. Neuroscience,
field modeling of the basal ganglia-thalamocorti- 86(2), 353–387.
cal system. I: Firing rates in healthy and Yelnik, J., François, C., Percheron, G., & Heyner, S.
parkinsonian states. Journal of Theoretical Biology, (1987). Golgi study of the primate substantia
257(4), 642–663. nigra. I. Quantitative morphology and typology of
van Wijk, B. C. M., Beudel, M., Jha, A., Oswal, A., nigral neurons. Journal of Comparative Neurology,
Foltynie, T., Hariz, M. I., … Litvak, V. (2016). Sub- 265(4), 455–472.
thalamic nucleus phase–amplitude coupling cor- Yelnik, J., Percheron, G., & François, C. (1984). A
relates with motor impairment in Parkinson’s Golgi analysis of the primate globus pallidus. II.
disease. Clinical Neurophysiology, 127(4), Quantitative morphology and spatial orientation
2010–2019. of dendritic arborizations. Journal of Comparative
van Wijk, B. C. M., Cagnan, H., Litvak, V., Kühn, A. Neurology, 227(2), 200–213.
A., & Friston, K. J. (2018). Generic dynamic causal Yin, H. H., & Knowlton, B. J. (2006). The role of the
modelling: An illustrative application to Parkin- basal ganglia in habit formation. Nature Reviews
son’s disease. NeuroImage, 181, 818–830. Neuroscience, 7(6), 464–476.
14
Cerebellum: Advances in
Understanding of
Cerebellar Functions
Mario Manto

INTRODUCTION Cajal (1852–1934) reported the fine network struc-


ture of the cerebellar cortex, and subsequently,
Gordon Holmes (1876–1965) described motor
The cerebellum has intrigued scientists and phi-
deficits, underlining the clumsiness in patients
losophers from the Antiquity. During the eight-
presenting with a cerebellar injury/lesion (cf.
eenth century, Vincenzo Malacarne (1744–1816)
Coco and Perciavalle, 2015). During World War I,
provided the first detailed macroanatomical
hundreds of soldiers suffered injuries to the occip-
description of the human cerebellum (cf. Zanatta
ital region and cerebellum, because the issued hel-
et al., 2018). In particular, he suggested that there
mets did not cover these regions adequately. This
was a correlation between the number of cerebel- gave the opportunity to Gordon Holmes to observe
lar folia and the environment, which was the first and report the clinical deficits in soldiers suffering
pioneering idea of neuroplasticity related to exter- from acute cerebellar lesions (e.g., from gunshot
nal factors. He suggested that the number of folia wounds). Subsequently, Olof Larsell (1886–1964)
was linked with the degree of intelligence, provid- identified the 10 lobules (I–X) of the cerebel-
ing the first hypothesis of the role of the cerebel- lum (Larsell and Jansen, 1973). More recently,
lum in cognition. He also introduced the current toward the end of the twentieth century, Jeremy
terminology of vermis, tonsil, pyramid, and uvula. D. Schmahmann reported the cognitive and affec-
Subsequently, Luigi Rolando (1773–1831) tive deficits in cerebellar ataxias, thereby identi-
observed that cerebellar lesions can impair pos- fying the third cornerstone of clinical ataxiology
ture and voluntary movements (Schmahmann, (Schmahmann and Sherman, 1998; Manto and
2016). Luigi Luciani (1840–1919) reported three Marien, 2015).
elemental deficits ipsilateral to a cerebellar lesion
performed in primates and in dogs: atonia, asthe-
nia and astasia. In the early 1800s, Jean Pierre
Flourens (1794–1867) was the first to associ-
ate cerebellar lesions with lack of coordination. STRUCTURAL ANATOMY
Likewise, Jan Evangelista Purkyně first identified
Purkinje neurons in the early 1800s. At the begin- Cerebellum occupies a large portion of the poste-
ning of the twentieth century, Santiago Ramón y rior fossa, posteriorly to the brainstem. Although
Cerebellum: Advances in Understanding of Cerebellar Functions 213

the human cerebellum contains about 60% of all Whereas granule cells are excitatory, the other
neurons of the brain, it occupies only 10% of brain neurons are all inhibitory and use principally
volume. Structurally, the cerebellum is composed gamma-aminobutyric acid (GABA). Purkinje neu-
of four pairs of nuclei embedded in the white rons inhibit cerebellar nuclei, the sole output of
matter which is surrounded by the cerebellar the cerebellar circuitry. Purkinje neurons form a
cortex (Colin et al., 2002). By comparison with monolayer (ganglionic layer) between the exter-
the cerebral cortex, the cytoarchitecture of the nal molecular layer and the internal granular layer.
cerebellum is particularly uniform. Their number is around 15 million in the human
In terms of evolution, the cerebellum consists cerebellum (Colin et al., 2002). They target cer-
of a single folium in amphibians and most teleosts ebellar nuclei and vestibular nuclei (considered by
(Colin et al., 2002). The vermis is already visible some authors as belonging to cerebellar nuclei).
in reptiles and birds. The number of folia increases The single primary dendrite of the Purkinje neu-
markedly from birds to primates as a result of a ron is directed towards the molecular layer and is
rostro-caudal expansion. The medio-lateral expan- divided into a rich arborization (width of about
sion of the hemispheres follows the development 300 microns) perpendicular to the folium axis.
of the cerebral cortex. The phylogenetic expansion The numerous spines along the arbor make excita-
of the neocerebellum is associated with expanding tory synapses with parallel fibers emerging from
interactions with the cerebral cortex, impacting the parallel fibers arising from granule cells. The
on information coding and processing capac- climbing fiber embraces the smooth proximal
ity (Huang and Ricklefs, 2013). Both in birds portion of the dendrite, forming one of the most
and mammals, the expansion of the cerebellum is powerful synapses in the brain. There is a 1:1 rela-
nearly proportional to the telencephalon (Sultan, tionship between one ascending fiber and the den-
2002). The constancy of the cerebellar fraction in dritic tree of a Purkinje neuron.
birds and mammals (about 10–15% of the whole It can be considered that three types of fib-
brain) has been interpreted as an argument to ers enter in the cerebellum: The climbing fibers
consider that the cerebellum is a general-purpose arising from the contralateral inferior olive and
computational machine showing plasticity mecha- ascending in the inferior cerebellar peduncle (res-
nisms (Huang and Ricklefs, 2013; Herculano- tiform body), the mossy fibers having numerous
Houzel, 2010). origins (somesthetic, vestibular, visual, acoustic,
Anatomically, the cerebellum is highly con- and cortical) and the cholinergic/monoaminer-
served across mammals, with a division of the gic afferents (noradrenergic fibers, dopaminergic
cortex by two fissures: (1) the primary fissure sep- fibers, serotoninergic fibers). The inferior olive
arating the anterior lobe from the posterior lobe, nucleus is a complex of multi-lamella structure
and (2) posterolateral fissure separating the poste- packed with small-sized gabaergic neurons (Luo
rior lobe from the flocculonodular lobe. The criti- and Sugihara, 2016). The large majority of oli-
cal works of Jan Jansen, Olof Larsell, Robert Dow, vary neurons are projecting to the cerebellum and
and Jan Voogd have founded our current under- each olivocerebellar axon gives rise to collaterals
standing of the structural organization of the cer- targeting cerebellar nuclei and provide also thin
ebellum (cf. Larsell and Jansen, 1973; Voogd and collaterals terminating in the granular layer with a
Koehler, 2018; Colin et al., 2002). Jansen divided small number of swellings. The functional signifi-
the human cerebellum on the basis of the fissures cance of these collaterals remains undetermined
into an anterior lobe, a posterior lobe and the floc- (Luo and Sugihara, 2016). Whereas the multiple
culus/paraflocculus. Larsell subdivided the cer- climbing fibers originating from a single axon are
ebellum into 10 lobules (numbered I to X; Larsell distributed in a longitudinal band-shaped area, the
and Jansen, 1973; Colin et al., 2002). Dow used projection pattern of mossy fiber axons is trans-
the mossy fiber projections to the cerebellar cor- versal and wide (Luo and Sugihara, 2016).
tex to divide the cerebellum into three main areas: Whereas fish and amphibians have a single
the flocculonodular lobe (vestibulocerebellum), unique cerebellar nucleus, reptiles and birds have
the vermal anterior and posterior lobes receiving a medial and lateral nucleus. In lower mammals,
spinal projections (paleocerebellum) and a medio- three nuclei are identified: a medial nucleus,
lateral portion receiving mainly ponto-cerebellar an interpositus and a lateral nucleus. In higher
projections (neocerebellum). mammals, the interpositus nucleus is subdivided
The cerebellar cortex is arranged into three lay- into an anterior and posterior part. In human, the
ers: the molecular layer, the Purkinje cell layer and medial nucleus is the fastigial nucleus, the glo-
the granular layer. These three cortical layers con- bosus nucleus corresponds to the anterior inter-
tain six main neuronal populations (Purkinje, stel- positus, the emboliformis nucleus to the posterior
late, basket, Lugaro, Golgi and granule neurons). interpositus, and the dentate nucleus to the lateral
214 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

nucleus. The dentate nucleus contains the majority neuron by activation of voltage-gated calcium
of nuclear neurons. It appears convoluted with a channels densely distributed throughout the den-
medial hilus and contains mainly large multipolar drites. It is generally considered that complex
neurons. Nucleofugal fibers are excitatory, with spikes slow the simple spikes firing (Otis, 2016).
the exception of the nucleo-olivary tract which is The number of simple spikes is decreased after
inhibitory. Collaterals from climbing and mossy a complex spike (“post-complex spike pause”).
ascending tracts excite nuclear neurons. Rates of complex spikes and simple spikes are
Vestibular nuclei project on the flocculonodu- anticorrelated in response to sensory stimuli
lar lobe at the level of the flocculus-paraflocculus, (Barmack and Yakhnitsa, 2011).
the nodulus/ventral uvula, and the dorsal oculo-
motor vermis corresponding to lobules V to VII.
It can be considered that vestibular nuclei are at
a sensorimotor crossroad (Barmack, 2016) since Rebound Depolarization
they are influenced by visual information, pro-
prioceptive signals, cerebellar signals, and signals Cerebellar nuclei neurons receive collateral excit-
from the cerebrum. They target thalamic nuclei, atory projections from mossy and climbing fibers
brainstem nuclei, and also the spinal cord. The and a strong inhibitory projection from Purkinje
descending routes to the spinal cord run through neurons. Neurons in nuclei show a unique feature
the ventral funiculus (medial descending sys- of rebound in firing after hyperpolarization
tems including the uncrossed corticospinal tract, (Dykstra and Turner, 2016). There is a first
the tectospinal tract, the reticulospinal tract, and increase in the firing frequency of about 100 msec
the vestibulospinal tract) and the lateral funiculus (role of T-type calcium currents), followed by a
(lateral descending systems gathering the crossed late rebound lasting a few seconds (persistent
corticospinal tract and the rubrospinal tract). The sodium currents). The rebound firing likely con-
medial vestibulospinal tract terminates bilaterally tributes to the sculpting of nuclear commands
in the ventromedial aspects of the spinal gray mat- towards extra-cerebellar targets (Dykstra and
ter, until midthoracic levels (Ruigrok, 2016). The Turner, 2016).
lateral vestibulospinal tract targets the ventrome-
dial laminae of the spinal gray matter. The first
tract influences motoneurons innervating neck and
axial muscles, the second influences motoneu-
Plasticity Mechanisms
rons controlling extensor or anti-gravity muscles The cerebellum is endowed with multiple forms
(Ruigrok, 2016). of plasticity at various synaptic sites. Learning
and compensatory mechanisms involve plasticity
mechanisms both in the cerebellar cortex and in
cerebellar nuclei. The various forms of plasticity
PHYSIOLOGICAL ASPECTS are integrated in order to fit with the environmen-
tal requirements and for learning purposes.
Concomitant stimulation at a low frequency of the
Simple Spikes and Complex Spikes climbing fiber and a parallel fiber is associated
with a persistent attenuation of the parallel fiber-
Purkinje neurons fire two types of action poten- Purkinje cell synapse (Ito et al., 1982). This paral-
tials: conventional action potentials (simple lel fiber long-term depression (LTD) is associative,
spikes) and stereotyped bursts of decrementing saturable, and involves mGluR, clathrin-depend-
spikes (complex spikes; Otis, 2016). Simple ent AMPAR endocytosis and subsequent rise in
spikes are typical voltage-gated sodium channel- internal Ca++ (Shen, 2016). This LTD is consid-
dependent action potentials. Complex spikes are ered critical for motor learning.
sodium channel driven spikelets on a depolarized If the parallel fiber stimulation preceded the
plateau potential. climbing fiber-evoked complex spikes by 50–250
Purkinje neurons fire spontaneously at rest at a msec, a spike-timing dependent plasticity (STDP)
high frequency of about 40–80 Hz (simple spikes), occurs. The parallel fiber-Purkinje cell synapse
in a very regular fashion. This pace-making fea- is also the site a homosynaptic LTP depend-
ture is fundamental for the cerebellar cortex, as ing on the stimulus frequency which is applied
it allows the Purkinje neurons to inhibit tonically (1 Hz-post-synaptic LTP and 4–8 Hz pre-synaptic
the cerebellar nuclei and vestibular nuclei (Otis, LTP). Presumably, the 1 Hz LTP is used as a reset-
2016). Climbing fibers evoke a complex spike ting mechanism for motor learning purposes. LTP
(frequency about 1 Hz) at the level of the Purkinje occurs also between interneurons and Purkinje
Cerebellum: Advances in Understanding of Cerebellar Functions 215

neurons, presumably to modulate the spike firing neurons. The level of simple spikes in the cer-
pattern of Purkinje neurons (Hausser and Clark, ebellar cortex modulates the complex spike syn-
1997). Another form of LTP is observed at the chrony by acting remotely on the GABA release
level of the mossy fiber-granule cell synapse, at the olivary gap junctions (Marshall and Lang,
modulating the local coding in the granular layer. 2009). In other words, a reduction in the rate of
The Marr-Albus-Ito theory was based on com- simple spikes in the cerebellar cortex will disin-
putational principle of the cerebellum related to hibit cerebellar nuclei, thus reducing the complex
a blueprint of the cerebellar circuit (Yamazaki, spike synchrony via the nucleo-olivary tract. A
2021). The model was established following dis- decreased rate of simple spikes will thus result
covery of LTD and has been challenged over the in a reduction of the rate of complex spikes in a
years. Cerebellar plasticity is now considered as specific cortical region (Broersen et al., 2016).
distributed amongst the cerebellar cortex and Optogenetic manipulation confirm these observa-
nuclei (D’Angelo, 2016). The original idea that tions (Witter et al., 2013).
plasticity was restrained to the parallel fiber- Cerebellum is also organized into discrete
Purkinje neuron under olivocerebellar guidance microcomplexes. Each microcomplex is the
is not considered as valid nowadays (D’Angelo, recipient of a specific motor error signal from
2016). The current theory considers that cerebel- the inferior olive, this signal being used to teach
lar plasticity is distributed and dynamically trans- the cerebellar cortex. Climbing fibers arising from
ferred through the various synaptic sites during multiple subunits of the inferior olive target sev-
a specific behavior and according to the needs eral microcomplexes, all projecting on the same
to achieve a task successfully (D’Angelo, 2016). subunit of cerebellar nuclei. The microcomplex
During learning, numerous forms of plasticity can be seen as an operating unit from the compu-
occur in various sites of the circuitry, with plastic- tational standpoint.
ity being organized in specific temporal patterns
starting in the granular layer, extending to the
molecular layer and finally to the cerebellar nuclei
(Shen, 2016).
CEREBRO-CEREBELLAR CONNECTIVITY

Anatomical Cerebro-cerebellar Loops


CEREBELLAR MODULES Cerebro-cerebellar connectivity is organized into
multiple parallel loops arranged topographically.
The cerebellum is divided into a series of olivo- Their existence is supported in particular by trac-
cortico-nuclear modules arranged longitudinally ing studies in animals and the technique of diffu-
in the rostro-caudal plane. Each module is defined sion tensor imaging (DTI) which allows to identify
by its climbing fiber input from a specific subdi- white matter tracts in vivo. Cerebral cortex pro-
vision of the inferior olivary complex which tar- jects to the contralateral cerebellum via the pon-
gets one or more longitudinal zones of Purkinje tine nuclei (feed-forward loop through the middle
cells. Specific zones of Purkinje neurons send cerebellar peduncle: crossed pontocerebellar
inhibitory projections to defined clusters of neu- tract). In monkeys, the majority of the cortico-
rons in cerebellar nuclei/vestibular nuclei, which pontine fibers arise from the motor system,
themselves target the inferior olive neurons (via whereas they arise from the prefrontal areas in
inhibitory nucleo-olivary tract) which will project human (Ramnani, 2006; Ramnani et al., 2006).
to the Purkinje neuron (closed-loop triangulation: The cerebellum projects back to the cerebral
inferior olive – Purkinje neurons – nuclei; cortex via the cerebello-thalamo-cortical pathway
Ruigrok, 2011). One module may include multi- (feedback loop through the superior cerebellar
ple microzones which correspond to clusters of peduncle). Distinct territories of the dentate nuclei
neighboring Purkinje cells coherently active target distinct thalamic and cortical areas (motor,
during a given operation (Broersen et al., 2016). premotor, associative) (Strick et al., 2009). The
Each microzone is composed of a group of ventral part of the dentate nucleus projects to pre-
approximately 1000 Purkinje neurons having the frontal and pre-supplementary motor cortices
same somatotopic receptive field and arranged in whereas the dorsal part project to the motor cortex
a long narrow strip. The cellular interactions (Middleton and Strick 2001). Lobules III–VI and
within a microzone are much stronger than a VIII are connected to the motor cortex and Crus II
those between different microzones. is connected with area BA46 (Kelly and Strick,
The inhibitory nucleo-olivary tract regulates the 2003). Overall, the following closed loops run in
degree of gap junction coupling between olivary parallel (Habas, 2016):
216 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

• dorsal dentate nucleus, caudal ventro-lateral posterior vermis and paraflocculus are associated
thalamus, area BA4 with errors in saccades, irregular smooth pursuit,
• lateral dentate nucleus, X thalamus, area BA6 and impaired adaptation.
• caudal dentate nucleus, X thalamus, area BA8
• ventromedial dentate nucleus, caudal ventrolat-
eral and mediodorsal thalamus, area BA9 Functional Anatomy
• ventralateral dentate nucleus, caudal ventro-
lateral and mediodorsal thalamus, dorsolateral Important progress has been made these last 15
prefrontal cortex (BA46) years in the understanding of resting-state net-
• lateral dentate nucleus, caudal ventrolateral works and the mapping of motor versus non-
motor tasks in the cerebellum (Brissenden et al.,
thalamus, BA7
2016; Buckner et al., 2011; Guell et al., 2018a,b;
King et al., 2019; Marek et al., 2018). Functional
In addition, striato-cerebellar loops have been imaging studies have confirmed that the cerebellar
identified: indeed, the dentate nucleus projects to circuitry is involved in multiple functions, beyond
the pallidum via the thalamus and the subthalamic the pure motor control. Functional maps can even
nucleus projects to the cerebellar cortex (crus II, be extracted at a single-subject level (Marek et al.,
lobule VIIb) via pontine nuclei (Bostan et al., 2018). Globally, the meta-analysis of neuroimag-
2010). Thanks to Strick et al. (2009), it is clear ing studies confirms the neuroanatomical findings
that the cerebellum communicates with the basal explained above (Stoodley and Schmahmann,
ganglia via disynaptic loops. 2009). The concept of task-dependent engage-
The spinocerebellar tracts convey the informa- ment of specific cerebellar subregions is now
tion from the spinal cord to the cerebellum via widely accepted.
the inferior and superior cerebellar peduncles. The independent component analysis (ICA)-
Lobules I–V, part of the lobule VI and lobule VIII based functional connectivity has delineated major
receive direct spinal afferents and spinal cord pro- networks from resting-state fMRI data (Habas,
jects also to the inferior olive (spino-olivary tract). 2016), including: the sensorimotor network
Unlike these lobules which are directly connected involved in sensorimotor operations, motor and
with the spinal cord (hence the terminology of premotor cortex, lobules V–VI and VIII. Simple
spino-cerebellum), the remainder of lobule VI and movements or tactile tasks activate the sensori-
lobule VII (VIIa, Crus I, Crus II, VIIb) receive no motor homunculi ipsilaterally: The right and left
direct spinal projection (Brodal and Brodal, 1981) executive networks, dorsolateral prefrontal and
but are massively involved in cerebro-cerebellar parietal prefrontal cortices, Crus I/II – the lim-
loops (prefrontal cortex, posterior parietal cortex, bic “salience” network involved in interoception,
superior temporal cortex, cingulate gyrus, para- emotional, and autonomic regulation, frontal and
hippocampus) as reported above. A dichotomy of insular cortices, lobules VI/VII – the default-mode
cerebellar connections with the cerebral cortex, network involved in self-agency, memory, and
brainstem and spinal cord has thus emerged. mental imagery – dorsomedian prefrontal, poste-
Cerebellum receives numerous inputs from vis- rior cingulate, retrosplenial and parahippocampal
ual and eye movement-related regions via the pons cortices, precuneus, lobules VII and IX. Regarding
(Roth et al., 2016). The information is integrated language, clusters are predominantly right-lateral-
with proprioceptive and vestibular signals to adapt ized and involve lobules VI–VII. Verb generation
spatial orientation. The cerebro-pontine projec- typically engages right-lateralized lobules VI–VII
tions originate from the frontal eye fields, the sup- (Ashida et al., 2019; Stoodley, 2012).
plementary eye fields, lateral intra-parietal area, It has been proposed recently that three main
medial intra-parietal area, medial temporal area, networks actively drive cerebellar activity (Guell
and middle temporal area (Roth et al., 2016). The and Schmahmann 2018b): motor network: acti-
ponto-cerebellar projections terminate as mossy vation during movements; attentional/executive
fibers mainly in VI and VII of the posterior vermis network: stimulus-driven attention (ventral atten-
and the paraflocculus (Thier, 2011). The cerebel- tion network, salience network), goal-oriented
lum optimizes goal-directed movements not only attention (dorsal attention network), and related
by acting on neck/trunk/limb muscles, but also by executive functions (frontoparietal network);
controlling saccades, smooth pursuit as well as default-mode network: mind wandering, and deac-
ocular reflexes (Roth et al., 2016). Furthermore, tivation during attention-demanding processes.
the cerebellum contributes to the adaptation and Guell and Schmahmann (2018a,b) suggested
recalibration of eye movements (see internal mod- that cerebellar functional anatomy is founded on
els below). Damage to lobules VI and VII of the these three poles. There are two representations of
Cerebellum: Advances in Understanding of Cerebellar Functions 217

motor processing in the cerebellar cortex: lobule emotion (Thomasson and Péron, 2022). For
I–VI and lobule VIII. For the attentional/execu- instance, an integrated model of emotional pro-
tive network and the default-mode network, there cessing for prosody has been proposed and sug-
are three representations: lobules VI-Crus I, Crus gests that the stimuli are processed by the cerebral
II-lobule VIIB, lobules IX–X. Crus I-Crus II are cortex, basal ganglia, and cerebellum (Thomasson
at the intersection of a first and a second default- and Péron, 2022). Within each of the three struc-
mode representation. It is plausible that the pro- tures, areas related to sensorimotor, associative,
gression at a macroscale functional organization and limbic tasks would be activated, with a func-
evolves from (a) motor to (b) attentional/execu- tional gradient. Temporal processing would be
tive, and to (c) default-mode processing. divided into two functions: detection of global
Our appraisal of emotion processing (engage- similarity with previous responses by the basal
ment of cerebellar vermis), vestibular functions ganglia, and detection of irregularities in the tem-
(engagement of cerebellar vermis and lobules poral pattern (violations in fine variations) by the
IX–X), language processing (lateralization on the cerebellum. Emotions recruit lobules VI–VII,
right side), and social cognition (engagement of with activation of lobules II–VI/VIII in relation to
the default-mode network) still require further motor responses, rostral and caudal vermal activa-
research. Ongoing studies show that the social tions in case of autonomic regulation and associa-
mentalizing depends on CrusI/II (Van Overwalle tive learning in conjunction with amygdala
et al., 2020). (Habas, 2022).
The functional organization reported here is The anatomical connectivity of these networks
growingly recognized at the level of the cerebellar would allow the processing of emotions at a tem-
cortex, but the functional organization at the level poral level and allow also to contribute to the
of cerebellar nuclei still remains to be refined. multidimensional aspect of the emotional expe-
Recent data indicate that the human dentate riences of daily life (Habas, 2022). By using the
nucleus contains three distinct territories belong- widespread connectivity with the cerebral cortex
ing to the default-mode, salience-motor, and visual and basal ganglia, the cerebellum would modulate
networks (Guell et al., 2019). It is unclear whether patterns of events in the time domain not only by
and how the interpositus nucleus and the fastigial using predictions but also by taking advantage of
nucleus contribute to these networks. a reward-based error feedback, so that the cere-
Regional gray matter volumes in lobules IV– bellum would help in the selection of emotional
VI are correlated with motor skills, whereas gray responses which are the most rewarded in a given
matter in the posterior lobe is correlated with per- context (Pierce and Péron, 2022). Even in the
formances on cognitive operations (Bernard and emotional domain, prediction errors would be con-
Seidler, 2013; Moore et al., 2017). Older adults veyed by the olivo-cerebellar pathway. This would
have smaller cerebellar volume than young adults tune the sensitivity of the parallel fibers/Purkinje
and lobules in the anterior cerebellum are more neurons, resulting in reinforcement (or the reverse)
impacted by age. A disrupted cortico-cerebellar of projections towards the limbic system, the
resting state network connectivity has been identi- orbitofrontal cortex, and the amygdala (Pierce and
fied in older adults (Bernard et al., 2013). There is Péron, 2022). Furthermore, cerebellum contrib-
a disruption of cerebellar connectivity with both utes to both biological motion perception and dis-
the striatum and the medial temporal lobe in the crimination in body emotional expressions, which
elderly. Engagement with the default mode net- represent a powerful form of non-verbal com-
work and striatal pathways is associated with bet- munication (Ferrari et al., 2022). Understanding
ter performance for older adults. others’ emotional/mental states such as intentions
are particularly relevant (Adamaszek et al., 2017).
Although faces have been considered as a major
supply to transmit intentions and feelings, bogy
Assessment of Emotions language contributes greatly as cues. The reper-
toire of emotional body postures has appeared as
The example of emotions reflects very well the extremely rich (Ferrari et al., 2022). Examples are
importance of the numerous hubs of the brain and embarrassment, shame, pride, and anger.
the re-appraisal of their complex interactions When individuals are distant, the assessment
(Thomasson and Péron, 2022). The models of of body expressions becomes a major tool to infer
Papez and MacLean stressed the cortical struc- other’s intentions. In some cases, body signals are
tures in the processing of emotions, with the critical to clarify ambiguous facial expressions.
exception of the amygdala and the anterior nucleus The streams of information go through a ventral
of the thalamus. Research has moved towards an path (the extra-striate body area and the fusiform
integrated cerebello-basal ganglia-cortical view of body area), a visuomotor circuit involving the
218 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

posterior superior temporal sulcus, the inferior and Sherman, 1998). Dentate nuclei are anatomi-
parietal sulcus, and the premotor areas (Ferrari cally connected with the prefrontal, temporo-
et al., 2022). The visuomotor circuit interacts parietal and limbic areas (Schmahmann, 2010).
with the amygdala to assess body movements and Each portion of the cerebellar cortex has specific
trigger adaptative behaviors. The posterior supe- feedforward and feedback connections to the cer-
rior temporal sulcus is a key-structure to process ebral cortex (Pisotta and Molinari, 2014). The
motion and integrates the signals with three net- connectivity can be understood as a large set of
works: the mentalizing, the action-observation, loops running in parallel (see above).
and the mirror networks. Furthermore, body Using transcranial electrical stimulation and
posture elicits reflex-like responses at the level transcranial magnetic stimulation (TMS), Ugawa
of the amygdala, the superior colliculus, and the has demonstrated that stimulation over the human
striatum. The amygdala can be envisioned as an cerebellum modulates the responses to a second
interface between the cerebral cortex and the sub- stimulus delivered over the contralateral primary
cortical structures. Interestingly, the insula, the motor cortex (M1). Delivering a TMS pulse (con-
somatosensory cortices, and the anterior cingulum ditioning stimulus, CS) 5–7 msecs over the con-
are closely connected with the amygdala, likely to tralateral cerebellar hemisphere before a second
take into account visceral and attentional aspects stimulus over M1 (test stimulus, TS) inhibits the
related to emotional processing (Ferrari et al., TS (Ugawa et al., 1991; Ugawa et al., 1995). This
2022). Since the cerebellum is anatomically linked is called cerebellum-brain inhibition (CBI). The
to both the visuomotor circuit and the amygdala, reduction in the amplitude of the motor response
it is likely involved in the detection of viola- is interpreted as the consequence of the activa-
tions during the assessment of emotions detected tion of Purkinje cells by the CS. This activation
through the analysis of body expressions. The of the cerebellar cortex inhibits cerebellar nuclei
anterior-superior lobules of the cerebellum are which have themselves a disynaptic excitatory
recruited when perceiving body expressions con- connection with contralateral M1. These obser-
taining emotional contents, likely by a resonance vations have served to explore the effects of the
mechanism (Agnew et al., 2007). Posterior lobules neuromodulation of the cerebellar cortex (tDCS:
are essential to process emotional body language. transcranial direct current stimulation, TMS)
The cerebellum is now considered a master-piece upon CBI.
of the connectome of nodes supporting behavior. In patients presenting focal unilateral cerebellar
Identification of the functional networks open damage, Di Lazzaro et al. have observed reduced
up direct perspectives for clinical applications excitability of the contralateral motor cortex, sug-
both in terms of diagnosis, follow-up and thera- gesting that the cerebellum exerts a facilitating
pies of cerebellar ataxias such as neuromodulation tonic action on motor circuits (Di Lazzaro et al.,
techniques targeting specific areas of the cerebel- 1994a,b, 1995; Restuccia et al., 2001, 2007).
lar cortex. Cerebellar patients experience difficul- This has been confirmed in rodents (Oulad Ben
ties to identify and classify basic emotions and Taib et al., 2005). Application of cerebellar epi-
exhibit difficulties for social and affective behav- dural anodal tDCS decreases the excitability of
ior. Overall, the disorders observed fit with the the motor cortex, reduces the excitability of F
general concept of a mismatch between internal waves, exerts a “smoothing effect” on corticomo-
resources and the environmental demand, leading tor maps and increases the afferent inhibition of
to impaired homeostasis, extending the impact conditioned motor evoked responses (Oulad Ben
of cerebellar dysfunction to structures regulating Taib et al., 2013). This study provided evidence
general metabolic, visceral, and endocrine pro- of a mechanism by which tDCS of the cerebellum
cesses such as the hypothalamus (Schutter, 2022). exerts a remote neuromodulatory effect upon the
motor cortex.
Tesche and Karhu (2000) and Ivry (2000) have
suggested that the cerebellum predicts incoming
somatosensory stimuli and tunes the activity of
CEREBELLO-CORTICAL CIRCUITS: the somatosensory cerebral cortex accordingly.
NEUROPHYSIOLOGICAL ASPECTS In patients with lateralized cerebellar lesions,
Restuccia et al. (2001) have demonstrated that the
Connectivity between the cerebellum and associa- cerebellum influences the activity of inhibitory
tive areas of the cerebral cortex is the anatomical circuitries in the primary somatosensory cortex
substrate of the contribution of the cerebellum in (Restuccia et al., 2001). The cerebellum appears
higher cognitive functions including cognitive and to modulate the excitability of the primary sensory
emotional behavior (Ramnani, 2012; Schmahmann cortex at very early stages of somatosensory input
Cerebellum: Advances in Understanding of Cerebellar Functions 219

processing. Also shown is the implication that the plastic mechanisms is likely one of the neurobio-
cerebellum operates in mismatch negativity. Also, logical substrates for the adaptation to errors. The
cerebellar patients show an impaired capacity to redundancy of information within the cerebellar
correctly process somatosensory information at circuitry is another parameter. Indeed, the mossy
the cortical level. Taken together, these observa- fibers extend widely in a medio-lateral fashion and
tions indicate that the cerebellum exerts a remote parallel fibers run through multiple Purkinje neu-
effect upon both the excitability of the motor cor- rons. In other words, the mossy fibers transmit the
tex and the processing of somatosensory inputs. information simultaneously to multiple functional
units and parallel fibers inform multiple Purkinje
neurons. This redundancy combined with multiple
plasticity mechanisms make of the cerebellum a
unique structure to adapt to the environment and
CEREBELLAR RESERVE to lesions.

Cerebellar ataxias (CAs) result from various inju-


ries and manifest clinically with motor, oculomo-
tor and cognitive/affective deficits (see next
section). Cerebellar symptoms may recover CEREBELLUM AS MODULATOR OF
remarkably, as underlined by Holmes (cf. Mitoma MOTOR AND NON-MOTOR OPERATIONS
and Manto, 2021). Cerebellar reserve is defined as
the capacity for compensation and restoration fol- The mechanisms by which the cerebellum regu-
lowing pathological changes in the cerebellar cir- lates motor control, affect, and emotion is a funda-
cuitry. The structural cerebellar reserve mental topic for neurosciences. The concept of
corresponds to a compensation of a structural motor dysmetria has now been expanded to cogni-
damage in a limited area of the cerebellum (for tive dysmetria (Schmahmann and Sherman, 1998).
instance in case of stroke, traumatic injury or On the basis of the homogeneous organization of
tumor) by another area of the cerebellum (cerebel- the cerebellar cortex, it is presumed that the cere-
lar structures around the lesion, contralateral cer- bellum exerts a similar control over motor and
ebellar hemisphere), which is unaffected. The non-motor behavior. Schmahmann has proposed
functional cerebellar reserve refers to a compensa- the theory of Universal Cerebellar Transform
tion or restoration which occurs within the lesion (UCT) that the major function of the cerebellum is
site (for instance in case of immune ataxias, meta- to maintain homeostasis around a baseline, acting
bolic ataxias, degenerative ataxias) via a func- as a dampener which smooths out performance in
tional reorganization. order to produce harmonious behavior
Experimental studies show that motor recov- (Schmahmann, 2019). The cerebellum would reg-
ery may be substantial despite extensive cerebel- ulate in a similar manner the rate, force and
lar lesions such as hemicerebellectomy (Federico rhythm of motor actions on one hand, and the
et al., 2006). However, a second lesion may appropriateness of cognitive/affective/emotional
decompensate the symptoms, as observed for operations on the other hand. Emotional dysregu-
a subsequent lesion at the level of the sensory lation may be striking both in adults and in chil-
cortex (Mackel, 1987). When the sensory cor- dren. Overall, motor dysmetria, dysmetria of
tex is removed secondarily to a cerebellar lesion thought and emotional dysmetria would share the
and following recovery from the cerebellar defi- same elemental mechanisms. The cerebellum
cits, the initially recovered motor performance is would perform unique computations in a topo-
much worse again (process of decompensation). graphically arranged fashion, according to the
Furthermore, removal of the sensory cortex before loops involved.
the cerebellar lesion increases the cerebellar defi- A leading theory is that the cerebellum/cerebel-
cits and severely limits the recovery, suggesting a lum-like structures play a key-role in the genera-
functional interrelationship between the sensory tion and maintenance of internal models allowing
cortex and the cerebellum. Striatum and vestibulo- predictive computations for motor, cognitive, and
spinal tracts are likely involved also. For instance, affective operations. Predictions are necessary
NMDA blockade with MK-801 impairs the syn- given the inevitable noise and transmission delays
aptic rearrangement in the striatum and impacts within the nervous system (Hardwick et al., 2016).
on motor recovery (Centonze et al., 2008). Thus, For motor control, response latencies are far below
extra-cerebellar lesions are involved in the recov- those that can be achieved using feedback control.
ery process after a cerebellar lesion. There is compelling evidence from psychophysi-
Cerebellar circuitry shows several plasticity cal studies, fMRI studies, and TMS studies that
mechanisms such as LTP and LTD. This set of the cerebellum is involved in motor and non-motor
220 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

predictions/anticipations. A typical example is sequences. A mismatch between the predicted


provided by the anticipatory changes in grip and the actual sensory outcomes would cause
forces during predictable manipulations of objects motor dysmetria. The internal model hypothesis
(Johansson and Cole, 2013). The grip forces are fits very well with the sequencing hypothesis of
scaled without delay and therefore without the use Molinari (Leggio and Molinari, 2015). According
of sensory feedback. The lack of delay means that to this theory, cerebellar microcomplexes compare
the brain predicts the adaptations necessary to the sequences of movements with stored sequences,
changes in viscous and elastic loads applied to the thanks to working memory. If the sequence stored
arm (Ebner, 2013). matches the actual motor plan, cerebellum will
Adaptation of reaching movements to external expect a sequence of elementary events arriving
perturbing forces applied by a robotic manipulan- in its circuitry. In case of mismatch, a signal error
dum or to Coriolis forces also provide a support is generated and the prediction scheme is cor-
for the acquisition and updates of novel inverse rected. The efferent copy originating from M1 and
dynamic models in order to maintain the accuracy the incoming sensations are taken into account. A
(Shadmehr and Mussa-Ivaldi, 1994; Lackner and similar sequencing mechanism with re-calibration
Dizio, 1998). Cerebellar patients cannot compen- would operate for both motor and non-motor oper-
sate to inertial loads (Manto et al., 1994). They ations. This theory is congruent with the obser-
show delayed antagonist muscle responses (errors vation that cerebellar patients exhibit impaired
in timing) and are unable to tune the magnitudes of feedforward control, by contrast with the online
muscle discharges, suggesting that they cannot use or reactive control which is relatively preserved
state estimation to control movement but rather (Ebner, 2013; Manto et al., 1994). The sequenc-
use outdated sensory information from feedback ing hypothesis fits with the presumed role of the
processes (Hardwick et al., 2013). Application cerebellum in predicting somatosensory events
of predictive strategies offer a clear advantage (Bower, 1997). Cerebellar sequencing is not
in term of reactivity as compared to feedback linked to the elaboration of specific content and
strategies. Furthermore, recordings of discharges likely different contents are processed in distinct
of Purkinje neurons show features of internal cerebello-cortical loops.
models. According to Kawato et al. (1999), the For language, cerebellar patients exhibit dif-
mossy fiber-granule cell-Purkinje cell circuit is ficulties to generate lists of words according to
the anatomical substratum of the transformation the phonemic but not the semantic rule. Impaired
of the desired trajectory into the motor command verbal fluency appears specific for phonemically-
(inverse dynamic model). For Ebner (2011), the related retrieval strategies (Molinari and Leggio,
firings of simple spikes fit with a forward inter- 2013). Cerebellar patients show omissions and
nal model, the simple spikes firing preceding the repetitions of strokes and letters when they have
movement kinematics. Cerebellum-like struc- to dictate or copy words. The observation that
tures, for instance the electrosensory lobe in elas- writing deteriorates when the eyes are closed
mobranchs, show similar histological features to argues also for a use of visual feedback for writ-
the cerebellum with parallel fibers originating in ing. Regarding mental representations, Ito (2008)
granule cells (receiving inputs from higher order has suggested that internal models are embedded
motor/sensory areas) and synapsing on principal in the cerebellum with a close interaction with the
cells (analogous to Purkinje neurons). The mossy prefrontal cortex, but this experimental demon-
fiber/granule cell/parallel fibers provide an effer- stration requires validation.
ence copy of the corollary discharge of the drive to The numerous tasks of daily life require an
the electric organ (Bell et al., 2008; Ebner, 2013). integration of the spatial and temporal informa-
A second input reaches the principal cells to relay tion in the msec range (Broersen et al., 2016).
the sensory information from the periphery. This Predictions for the future are intimately correlated
is the equivalent of climbing fibers in the cerebel- to the timing of elemental events. The brain con-
lum. There is evidence that cerebellum-like struc- siders spatio-temporal cues to perform appropriate
tures predict the upcoming sensory state and build anticipatory adjustments and perceptual decisions.
internal forward models (Bodznick et al., 1999). The cerebellum contributes greatly to these com-
Internal models provide representations of the putations as a platform involved in multiple net-
input-output properties of the motor apparatus works including cerebral cortex, basal ganglia,
to adapt movement control (Manto and Habas, hippocampus, and spinal cord (Broersen et al.,
2013). Forward models use the commands for 2016; Manto and Grimaldi, 2012). This is highly
an action and information about the present state demanding given that sensorimotor delays range
to predict the consequences of an action (Ebner, from 50 to 400 msec according to the modal-
2013). Internal models attribute the capacity to the ity and behavioral context (Grimaldi and Manto,
cerebellar circuitry to produce and adapt motor 2012). Cerebellar patients exhibit errors in timing
Cerebellum: Advances in Understanding of Cerebellar Functions 221

both for explicit and implicit tasks (Bares et al., with dysarthria. The typical presentation of stroke
2011). Given the micro-anatomy of the cerebellar in the superior cerebellar artery (SCA) territory
neurons, it is plausible that timing is an intrinsic includes ipsilateral motor dysmetria, dysarthria
property of the circuitry. Unlike motor timing, and gait ataxia/lateropulsion, usually in absence
which is often assessed using rhythm production of cognitive deficits or in association with subtle
tasks, predictive motor timing implies visuo- cognitive errors.
motor coordination with anticipation of a future The vestibulo-cerebellar syndrome (VCS) com-
event, and it is obvious in tasks such as catching prises ocular instability, oculomotor movement
a ball or shooting a moving target (Bares et al., deficits, and ocular misalignment. Oculomotor
2011). Bares and colleagues have studied role of deficits are often prominent in cerebellar patients:
the cerebellum and striatum in predictive motor dysmetria of saccades, saccadic pursuit, impaired
timing in a target interception task in healthy sub- vestibulo-ocular reflex (VOR), deficits in fixa-
jects and in cerebellar patients. The performance tion, errors in vergence. Lesions are observed at
of the control subjects was better than that of the the level of the vermis and/or the flocculo-nod-
cerebellar patients. Successful performance in ular lobe. Although VCS impacts negatively on
both groups was associated with increased activ- the quality of life, it tends to be underestimated
ity in the cerebellum (right dentate nucleus, left by some influential clinical rating scales of cer-
uvula (lobule V), and lobule VI), thalamus, and ebellum-induced ataxia such as the Scale for
in several cortical areas. The higher performance Assessment and Rating of Ataxia (SARA).
in the control subjects was related to activation in
thalamus, putamen (lentiform nucleus), and cer-
ebellum (right dentate nucleus and culmen-lobule
IV), which were not activated either in the ataxic Schmahmann’s Syndrome
patients or within the subgroup of controls with
poor performances. These findings highlight the Lesions of the posterior lobe are associated with
contribution of the cerebellum in visuo-motor various combinations of deficits in the executive
tasks where anticipation of timing is critical. functions, in visuospatial performances, in lin-
guistic processing and regulation of affect
(Schmahmann and Sherman, 1998). Clinically,
this is discernible with the assessment of working
memory, ideational set shifting, perseverations,
CLINICAL SYNDROMES AFTER 250 YEARS drawings, verbal fluency, and prosodia. Severe
OF CEREBELLAR RESEARCH forms present with mutism, as observed in the
posterior fossa syndrome occurring in the imme-
The current clinical view of cerebellar deficits con- diate postoperative phase following resection of a
siders that cerebellar symptoms can be gathered posterior fossa surgery. Cerebellar mutism is typi-
into a cerebellar motor syndrome (CMS), a vestibulo- cally associated with cognitive, behavioral, and
cerebellar syndrome (VCS) and a cerebellar cogni- affective disturbances. Sequences are impaired in
tive affective syndrome or Schmahmann’s syn- the Schmahmann syndrome, including conceptu-
drome (Cabaraux et al., 2021). The core feature of alization of figures. Patients with lesions in the
cerebellar deficits is dysmetria, encompassing both territory of the posterior inferior cerebellar artery
motor dysmetria and dysmetria of thought. (PICA) or with vermian lesions often show a flat-
CMS is typically observed in lesions of the tening of affect, disinhibition, impulsivity, inap-
anterior lobe (lobules I–V), whereas patients with propriate comments which have been clearly
lesions of the lobules VI–X show relatively minor underrecognized in the past. A childhood-like
motor deficits. There is a somatotopic representa- behavior may be observed.
tion as follows: lesions of the vermal/paravermal Pathological laughing and crying also has
lobules III–VI are correlated with ataxia of lower been observed following lesions of the pontocer-
limbs, lesions of vermal/paravermal/hemispheral ebellar tract, supporting the idea that cerebellum
lobules IV–VI are correlated with upper limb contributes to the voluntary control of emotional
ataxia, paravermal/hemispheric lobules V–VI expression. Overall, the general consequence is
are correlated with dysarthria, and lesions of the a lowering of intellectual functions, especially in
superior vermis are correlated with ataxia of pos- case of bilateral and acute lesions. The spectrum
ture/gait (Schoch et al., 2006). Regarding nuclei, of clinical deficits corresponds to a genuine link
lesions of fastigial nuclei are associated with between neurology and psychiatry (Schmahmann
ataxia of stance/gait, lesions of the interposed et al., 2007). Social interactions are highly com-
nuclei/adjacent dentate nuclei cause limb ataxia plex and sophisticated. They require language
and lesions of the dentate nuclei are associated and communication skills. They are also under
222 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

cerebellar control and cerebellar patients may subdivision of the inferior olivary complex which
exhibit an autistic-like behavior. Hypothetically, targets one or more longitudinal zones of Purkinje
the cerebellum builds internal models of human cells. Cerebro-cerebellar connectivity is organized
social interactions, with the goal of optimiz- into multiple parallel loops. The current clinical
ing social behavior. Cerebellum would allow the view of cerebellar deficits considers that cerebel-
anticipation of other person’s behavior, detect lar symptoms can be gathered into a cerebellar
violations in social links and trigger corrections motor syndrome, a vestibulocerebellar syndrome
in the internal models. Such a mechanism would and a cerebellar cognitive affective syndrome. The
have impacts far beyond humans, because it would core feature of cerebellar deficits is dysmetria,
represent a model to understand social interactions encompassing both motor and thought dysmetria.
in the numerous species having a cerebellum. Thus, the cerebellum plays a key-role in the gen-
Furthermore, the model could be used to esti- eration and maintenance of internal models allow-
mate the social cerebellar reserve, with the aim of ing predictive computations for motor, cognitive,
improving social skill set. and affective operations.
Social interactions might represent not only The cerebellum is involved in multiple seg-
adaptative behaviors following a selection pres- regated cerebello-cerebral loops and it com-
sure, but also the sources of selection pressure municates directly with the basal ganglia. The
(Huang and Ricklefs, 2013). Social interactions cerebellum is a critical structure in performing
require and promote communication between motor predictions related to body dynamics and
individuals, which implies a common language to environmental changes. The predictions likely
acquired likely via imitation, a mechanism which apply to both motor and non-motor operations.
(1) recruits many regions of the brain including the Cerebellar cortex sculpts cerebellar nuclei dis-
mirror neurons, and (2) necessitates a high degree charges by a mechanism of inhibition/disinhibi-
of plasticity for perception (Huang and Ricklefs, tion and interacts closely with the inferior olive
2013). The cerebellum, through the numerous cer- through multiple olivo-cortico-nuclear modules.
ebro-cerebello-cerebral channels, is a major node The field of clinical ataxiology is now based upon
for the information coding behind the acquisition three groups of cerebellar symptoms gathered in
of complex behavior including language. the CMS, the VCS, and the Schmahmann’s syn-
drome. All these clinical deficits can be under-
stood as the consequences of impaired predictions
leading to dysmetria.
SUMMARY AND CONCLUSIONS

The human cerebellum contains about 60% of all


neurons of the brain, occupying 10% of brain REFERENCES
volume. Anatomically, the cerebellum is highly
conserved across mammals, with a division of the Adamaszek, M., D’Agata, F., Ferrucci, R., Habas, C.,
cortex by two fissures: (1) primary fissure separat- Keulen, S., Kirkby, & K.C., … Verhoeven, J. (2017).
ing the anterior lobe from the posterior lobe, and Consensus paper: Cerebellum and emotion. Cer-
(2) posterolateral fissure separating the posterior ebellum, 16(2), 552–576.
lobe from the flocculonodular lobe. The cerebellar Agnew, Z. K., Bhakoo, K. K., & Puri, B. K. (2007). The
cortex is folded into 10 lobules (I to X) subdivided human mirror system: A motor resonance theory
into folia. Cerebellar hemispheres are particularly of mind-reading. Brain Research Reviews, 54(2),
developed in mammals. The cerebellar cortex is 286–293.
arranged into three layers: the molecular layer, the Ashida, R., Cerminara, N. L, Edwards, R. J., Apps, R.,
Purkinje cell layer and the granular layer. These & Brooks, J. C. W. (2019). Sensorimotor, language,
three cortical layers contain five main neuronal and working memory representation within the
populations (Purkinje, stellate, basket, Golgi and human cerebellum. Human Brain Mapping,
granule cells). Whereas granule cells are excita- 40(16), 4732–4747.
tory, the other neurons are all inhibitory and use Bares, M., Lungu, O. V., Liu, T., Waechter, T., Gomez,
principally gamma-aminobutyric acid (GABA). C. M., & Ashe, J. (2011). The neural substrate of
Purkinje neurons inhibit cerebellar nuclei, the sole predictive motor timing in spinocerebellar ataxia.
output of the cerebellar circuitry. Cerebellum, 10(2), 233–244.
The cerebellum is divided into a series of Barmack, N. H., & Yakhnitsa, V. (2011). Topsy
olivo-cortico-nuclear modules arranged longitudi- turvy: Functions of climbing and mossy fibers in
nally in the rostro-caudal plane. Each module is the vestibulo-cerebellum. Neuroscientist, 17(2),
defined by its climbing fiber input from a specific 221–236.
Cerebellum: Advances in Understanding of Cerebellar Functions 223

Barmack, N. H. (2016). Vestibular nuclei and their Coco, M., & Perciavalle, V. (2015). Where did the motor
cerebellar connections. In D. L. Gruol, N. Koibuchi, function of the cerebellum come from? Cerebellum
M. Manto, M. Molinari, J. D. Schmahmann & Y. Ataxias, 2(10). doi: 10.1186/s40673-015-0029-8
Shen (Eds.), Essentials of cerebellum and cerebellar Colin, F., Ris, L., & Godaux, E. (2002). Neuroanatomy
disorders (pp. 69–78). Cham, Switzerland: Springer. of the cerebellum. In M. Manto & M. Pandolfo
Bell, C. C., Han, V., & Sawtell, N. B. (2008). Cerebellum- (Eds.), The cerebellum and its disorders (pp 6–29).
like structures and their implications for cerebellar Cambridge, UK: Cambridge University Press.
function. Annual Review of Neuroscience, 31, 1–24. D’Angelo. (2016). Distributed plasticity in the cerebel-
Bernard, J. A., Peltier, S. J., Wiggins, J. L., Jaeggi, S. lar circuit. In D. L. Gruol, N. Koibuchi, M. Manto,
M. Buschkuehl, M., Fling, B. W., & Seidler, R. D. M. Molinari, J. D. Schmahmann & Y. Shen (Eds.),
(2013). Disrupted cortico-cerebellar connectivity in Essentials of cerebellum and cerebellar disorders
older adults. NeuroImage, 83, 103–119. (pp. 285–290). Cham, Switzerland: Springer.
Bernard, J. A., & Seidler, R. D. (2013). Relationships Di Lazzaro, V., Molinari, M., Restuccia, D., Leggio M.
between regional cerebellar volume and sensori- G., Nardone R., Fogli D., & Tonali P. (1994a). Cer-
motor and cognitive function in young and older ebro-cerebellar interactions in man: Neurophysio-
adults. Cerebellum, 12(5), 721–737. logical studies in patients with focal cerebellar
Bodznick, D., Montgomery, J.C., & Carey, M. lesions. Electroencephalography and Clinical Neu-
(1999). Adaptive mechanisms in the elasmo- rophysiology, 93, 27–34.
branch hindbrain. Journal Experimental Biology, Di Lazzaro, V., Restuccia, D., Molinari, M., Leggio, M.
202, 1357–1364. G., Nardone, R., Fogli, D., & Tonali, P. (1994b). Excit-
Bostan, A. C., Dum, R. P., & Strick, P. L. (2010). The ability of the motor cortex to magnetic stimulation
basal ganglia communicate with the cerebellum. in patients with cerebellar lesions. Journal of Neurol-
Proceedings National Academy Sciences of the ogy, Neurosurgery, and Psychiatry, 57, 108–110.
United States of America, 107(18), 8452–8456. Di Lazzaro, V., Restuccia, D., Nardone, R., Leggio, M.
Bower, J. M. (1997). Control of sensory data acquisi- G., Oliviero, A., Profice, P., Tonali, P., & Molinari,
tion. International Review of Neurobiology, 41, M. (1995). Motor cortex changes in a patient with
489–513. hemicerebellectomy. Electroencephalography and
Brissenden, J. A., Levin, E. J., Osher, D. E., Halko, M. Clinical Neurophysiology, 97, 259–263.
A., & Somers, D. C. (2016). Functional evidence for Dykstra, S., & Turner, R.W. (2016). Rebound depolariza-
a cerebellar node of the dorsal attention network. tion and potentiation. In D. L. Gruol, N. Koibuchi, M.
Journal of Neuroscience, 36(22), 6083–6096. Manto, M. Molinari, J. D. Schmahmann & Y. Shen
Brodal, P., & Brodal, A. (1981). The olivocerebellar (Eds.), Essentials of cerebellum and cerebellar disor-
projection in the monkey. Experimental studies ders (pp. 305–309). Cham, Switzerland: Springer.
with the method of retrograde tracing of horse- Ebner, T. J., Hewitt, A. L., & Popa, L. S. (2011). What
radish peroxidase. Journal of Comparative Neurol- features of limb movements are encoded in the
ogy, 201(3), 375–393. discharge of cerebellar neurons? Cerebellum,
Broersen, R., Winkelman, B. H. J., Ozyldirim, O., & De 10(4), 683–693.
Zeeuw, C. I. (2016). Physiology of olivo-cerebellar Ebner, T. J. (2013). Cerebellum and internal models.
loops. In D. L. Gruol, N. Koibuchi, M. Manto, M. In M. Manto, D. L. Gruol, J. D. Schmahmann, N.
Molinari, J. D. Schmahmann & Y. Shen (Eds.), Koibuchi & F. Rossi (Eds.), Handbook of the cere-
Essentials of cerebellum and cerebellar disorders bellum and cerebellar disorders (pp. 1281–1295).
(pp. 323–327). Cham, Switzerland: Springer. Dordrecht: Springer.
Buckner, R. L., Krienen, F. M., Castellanos, A., Diaz, J. Federico, F., Leggio, M. G., Neri, P., Mandolesi, L., &
C., & Yeo, B. T. T. (2011). The organization of the Petrosini, L. (2006). NMDA receptor activity in
human cerebellum estimated by intrinsic func- learning spatial procedural strategies II: The influ-
tional connectivity. Journal of Neurophysiology, ence of cerebellar lesions. Brain Research Bulletin,
106(5), 2322–2345. 70(4–6), 356–367.
Cabaraux, P., Gandini, J., & Manto, M. (2021). The Ferrari, C., Ciricugno, A., & Cattaneo, Z. (2022).
three cornerstones of cerebellar ataxia: Closing Cerebellar contribution to emotional body lan-
the loop of 200 years of cerebellar research. In H. guage perception. In M. Adamaszek, M. Manto &
Mizusawa & S. Kakei (Eds.), Cerebellum as a CNS D. J. L. G. Schutter (Eds.), The emotional cerebel-
Hub (pp. 459–478). Cham, Switzerland. Springer. lum (pp. 141–153). Cham, Switzerland: Springer.
Centonze, D., Rossi, S., De Bartolo, P., De Chiara, V., Grimaldi, G., & Manto, M. (2012). Topography of
Foti, F., Musella, A., … Petrosini, L. (2008) Adapta- cerebellar deficits in humans. Cerebellum, 11(2),
tions of glutamatergic synapses in the striatum 336–351.
contribute to recovery from cerebellar damage. Guell, X., Gabrieli, J. D. E., & Schmahmann, J. D.
European Journal Neuroscience, 27(8), 2188–2196. (2018). Triple representation of language, working
224 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

memory, social and emotion processing in the Kawato, M. (1999) Internal models for motor control
cerebellum: Convergent evidence from task and and trajectory planning. Current Opinion Neurobi-
seed-based resting-state fMRI analyses in a single ology, 9(6), 718–727.
large cohort. NeuroImage, 172, 437–449. Kelly, R. M., & Strick, P. L. (2003). Cerebellar loops
Guell, X., Goncalves, M., Kaczmarzyk, J. R., Gabrieli, with motor cortex and prefrontal cortex of a non-
J. D. E., Schmahmann, J. D., & Ghosh S. S. (2019). human primate. Journal of Neuroscience, 23(23),
LittleBrain: A gradient-based tool for the topo- 8432–8444.
graphical interpretation of cerebellar neuroimag- King, M., Hernandez-Castillo, C. R., Poldrack, R. A.,
ing findings. PLoS One, 14(1). doi: 10.1371/ Ivry, R. B., & Diedrichsen, J. (2019). Functional
journal.pone.0210028 boundaries in the human cerebellum revealed by a
Guell, X., Schmahmann, J., Gabrieli, J., & Ghosh, S. multi-domain task battery. Nature Neuroscience,
(2018). Functional gradients of the cerebellum. 22, 1371–1378.
Elife. 7. doi: 10.7554/eLife.36652 Lackner, J.R., & Dizio, P. (1998). Gravitoinertial force
Habas, C. (2016). Cerebellar closed loops. In D. L. background level affects adaptation to coriolis
Gruol, N. Koibuchi, M. Manto, M. Molinari, J. D. force perturbations of reaching movements. Jour-
Schmahmann & Y. Shen (Eds.), Essentials of cere- nal of Neurophysiology, 80(2), 546–553.
bellum and cerebellar disorders (pp. 343–347). Larsell, O., & Jansen, J. (1973). The comparative
Cham, Switzerland: Springer. anatomy and histology of the cerebellum: The
Habas, C. (2022). Topography of emotions in cere- human cerebellum, cerebellar connections, and
bellum as appraised by functional imaging In M. cerebellar cortex (pp. 1–268). Minneapolis, MN:
Adamaszek, M. Manto & D. J. L. G. Schutter (Eds.), University of Minnesota Press.
The emotional cerebellum (pp. 77–86). Cham, Leggio, M., & Molinari, M. (2015). Cerebellar
Switzerland: Springer. sequencing: A trick for predicting the future. Cer-
Hardwick, R. M., Dagioglou, M., & Miall, R. C. (2013). ebellum, 14(1), 35–38.
State estimation and the cerebellum. In M. Manto, Luo, Y., & Sugihara, I. (2016). The olivocerebellar
D. L. Gruol, J. D. Schmahmann, N. Koibuchi & F. Rossi tract. In D. L. Gruol, N. Koibuchi, M. Manto, M.
(Eds.), Handbook of the cerebellum and cerebellar Molinari, J. D. Schmahmann & Y. Shen (Eds.),
disorders (pp. 1297–1313). Dordrecht: Springer. Essentials of cerebellum and cerebellar disorders
Häusser, M., & Clark, B. A. (1997). Tonic synaptic (pp. 55–61). Cham, Switzerland: Springer.
inhibition modulates neuronal output pattern and Mackel, R. (1987). The role of the monkey sensory
spatiotemporal synaptic integration. Neuron, cortex in the recovery from cerebellar injury.
19(3), 665–678. Experimental Brain Research, 66(3), 638–652.
Herculano-Houzel, S. (2010). Coordinated scaling of Manto, M., Godaux, E., & Jacquy, J. (1994). Cerebel-
cortical and cerebellar numbers of neurons. Fron- lar hypermetria is larger when the inertial load is
tiers in Neuroanatomy, 10(4). doi: 10.3389/ artificially increased. Annals of Neurology, 35(1),
fnana.2010.00012. 45–52.
Huang, C., & Ricklefs, R.E. (2013). Cerebellum and Manto, M., & Habas, C. (2013). Le cervelet: De
human evolution: a comparative and information l’anatomie et la physiologie à la clinique humaine.
theory perspective. In M. Manto, D. L. Gruol, J. D. Paris: Springer.
Schmahmann, N. Koibuchi & F. Rossi (Eds.), Hand- Manto, M., & Mariën, P. (2015). Schmahmann’s syn-
book of the cerebellum and cerebellar disorders drome: Identification of the third cornerstone of
(pp. 1337–1357). Dordrecht: Springer. clinical ataxiology. Cerebellum Ataxias, 2(2). doi:
Ito, M., Sakurai, M., & Tongroach, P. (1982). Climbing 10.1186/s40673-015-0023-1
fibre induced depression of both mossy fibre Marek, S., Siegel, J. S., Gordon, E. M, Raut, R. V.,
responsiveness and glutamate sensitivity of cere- Gratton, C., Newbold, D. J., … Dosenbach, N. U.
bellar Purkinje cells. Journal of Physiology, 324, F. (2018). Spatial and temporal organization of the
113–134. individual cerebellum. Neuron, 100, 977–993.
Ito, M. (2008). Control of mental activities by internal Marshall, S. P., & Lang, E. J. (2009). Local changes in
models in the cerebellum. Nature Reviews: Neuro- the excitability of the cerebellar cortex produce
science, 9(4), 304–313. spatially restricted changes in complex spike syn-
Ivry, R. (2000). Exploring the role of the cerebellum in chrony. Journal of Neuroscience, 29(45),
sensory anticipation and timing: Commentary on 14352–14362.
Tesche and Karhu. Human Brain Mapping, 9(3), Middleton, F. A., & Strick, P. L. (2001). Cerebellar
115–118. projections to the prefrontal cortex of the primate.
Johansson, R. S., & Cole, K. J. (1992). Sensory-motor Journal of Neuroscience, 21(2), 700–712.
coordination during grasping and manipulative Mitoma, H., & Manto, M. (2021). Cerebellar reserve:
actions. Current Opinion in Neurobiology, 2(6), From theoretical framework to therapeutic strat-
815–823. egy. In H. Mizusawa & S. Kakei (Eds.), Cerebellum
Cerebellum: Advances in Understanding of Cerebellar Functions 225

as a CNS hub (pp. 433–444). Cham, Switzerland: Functional changes of the primary somatosensory
Springer. cortex in patients with unilateral cerebellar lesions.
Molinari, M., Chiricozzi, F., Clausi, S., Tedesco, A., De Brain, 124, 757–768.
Lisa, M., & Leggio, M. (2008) Cerebellum and Roth, M. J., Lindner, A., & Their, P. (2016). Visual cir-
detection of sequences, from perception to cogni- cuits. In D. Gruol, N. Koibuchi, M. Manto, M.
tion. Cerebellum, 7, 611–661. Molinari, J. Schmahmann, J. & Y. Shen (Eds.),
Molinari, M., & Leggio, M. G. (2013). Cerebellar Essentials of cerebellum and cerebellar disorders
sequencing for cognitive processing In M. Manto, (pp. 89–100). Cham, Switzerland: Springer.
J. D. Schmahmann, F. Rossi, D. L. Gruol & N. Ruigrok, T. J. (2011). Ins and outs of cerebellar mod-
Koibuchi, N. (Eds.), Handbook of the cerebellum ules. Cerebellum, 10(3), 464–474.
and cerebellar disorders (pp. 1701–1715). Ruigrok, T. J. H. (2016). Spinocerebellar and cerebel-
Dordrecht: Springer lospinal pathways. In D. Gruol, N. Koibuchi, M.
Moore, D. M., D’Mello, A. M., McGrath, L. M., & Manto, M. Molinari, J. Schmahmann, J. & Y. Shen
Stoodley, C. J. (2017). The developmental relation- (Eds.), Essentials of cerebellum and cerebellar dis-
ship between specific cognitive domains and grey orders (pp. 79–88). Cham, Switzerland: Springer.
matter in the cerebellum. Developmental Cogni- Schmahmann, J. D., & Sherman, J. C. (1998). The
tive Neuroscience, 24, 1–11. cerebellar cognitive affective syndrome. Brain,
Otis, T. S. (2016). Simple spikes and complex spikes. 121(4), 561–579.
In D. Gruol, N. Koibuchi, M. Manto, M. Molinari, Schmahmann, J. D., Weilburg, J. B., & Sherman, J. C.
J. Schmahmann, J. & Y. Shen (Eds.), Essentials of (2007). The neuropsychiatry of the cerebellum:
cerebellum and cerebellar disorders (pp. 299– Insights from the clinic. Cerebellum, 6(3), 254–267.
303). Cham, Switzerland: Springer. Schmahmann, J. D. (2016). A brief history of the
Oulad Ben Taib, N., & Manto, M. (2013). Trains of cerebellum. In D. Gruol, N. Koibuchi, M. Manto,
epidural DC stimulation of the cerebellum tune M. Molinari, J. Schmahmann, J. & Y. Shen (Eds.),
corticomotor excitability. Neural Plasticity. doi: Essentials of cerebellum and cerebellar disorders
10.1155/2013/613197 (pp. 5–20). Cham, Switzerland: Springer.
Oulad Ben Taib, N., Manto, M., Pandolfo, M., & Schmahmann, J. D. (2019). The cerebellum and cog-
Brotchi, J. (2005). Hemicerebellectomy blocks the nition. Neuroscience Letters, 688, 62–75.
enhancement of cortical motor output associated Schmahmann, J. D. (2010). The role of the cerebellum
with repetitive somatosensory stimulation in the in cognition and emotion: Personal reflections since
rat. Journal of Physiology, 567, 293–300. 1982 on the dysmetria of thought hypothesis, and
Pierce, J. E., & Péron, J. A. (2022). Reward-based its historical evolution from theory to therapy. Neu-
learning and emotional habit formation in the ropsychological Reviews, 20(3), 236–260.
cerebellum. Advances in Experimental Medicine Schoch, B., Dimitrova, A., Gizewski, E. R., & Tim-
and Biology, 1378, 125–140. mann, D. (2006). Functional localization in the
Pisotta, I., & Molinari, M. (2014). Cerebellar contribu- human cerebellum based on voxelwise statistical
tion to feedforward control of locomotion. Fron- analysis: A study of 90 patients. NeuroImage,
tiers in Human Neuroscience, 8. doi: 10.3389/ 30(1), 36–51.
fnhum.2014.00475 Schutter, D. J. L. G. (2022). The cerebellum and dis-
Ramnani, N., Behrens, T. E., Johansen-Berg, H., Rich- orders of emotion. In M. Adamaszek, M. Manto &
ter, M. C., Pinsk, M. A., Andersson, J. L., … Mat- D. J. L. G. Schutter (Eds.), The emotional cerebel-
thews, P. M. (2006). The evolution of prefrontal lum (pp. 273–283). Cham, Switzerland: Springer.
inputs to the cortico-pontine system: Diffusion Shadmehr, R., & Mussa-Ivaldi, F. A. (1994). Adaptive
imaging evidence from Macaque monkeys and representation of dynamics during learning of a
humans. Cerebral Cortex, 16(6), 811–818. motor task. Journal of Neuroscience, 14(5 Pt 2),
Ramnani, N. (2012). Frontal lobe and posterior pari- 3208–3224.
etal contributions to the cortico-cerebellar system. Shen, Y. (2016). Plasticity of the cerebellum. In D.
Cerebellum, 11(2), 366–383. Gruol, N. Koibuchi, M. Manto, M. Molinari, J.
Ramnani, N. (2006) The primate cortico-cerebellar Schmahmann, J. & Y. Shen (Eds.), Essentials of
system: anatomy and function. Nature Reviews: cerebellum and cerebellar disorders (pp. 317–
Neurosciences, 7(7), 511–522. 321). Cham, Switzerland: Springer.
Restuccia, D., Della, M.G., Valeriani, M., Leggio, Stoodley, C. J., & Schmahmann, J. D. (2009). Func-
M.G., & Molinari, M. (2007) Cerebellar damage tional topography in the human cerebellum: A
impairs detection of somatosensory input changes. meta-analysis of neuroimaging studies. NeuroIm-
A somatosensory mismatch-negativity study. age. 44(2): 489–501.
Brain, 130, 276–287. Stoodley, C. J. (2012). The cerebellum and cognition:
Restuccia, D., Valeriani, M., Barba, C., Le Pera, D., Evidence from functional imaging studies. Cere-
Capecci, M., Filippini, V., & Molinari, M. (2001). bellum, 11(2), 352–365.
226 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Strick, P. L., Dum, R. P., & Fiez, J. A. (2009). Cerebel- the cerebellum in humans. Annals of Neurology,
lum and nonmotor function. Annual Review of 37, 703–713.
Neuroscience, 32, 413–434. Van Overwalle, F., Manto, M., Cattaneo, Z., Clausi,
Sultan, F. (2002). Analysis of mammalian brain archi- S., Ferrari, C., Gabrieli, J. D. E., & Leggio, M.
tecture. Nature, 415, 133–134. (2020). Consensus paper: Cerebellum and social
Tesche, C. D., & Karhu, J. J. (2000). Anticipatory cer- cognition. Cerebellum, 19(6), 833–868.
ebellar responses during somatosensory omission Voogd, J., & Koehler, P.J. (2018). Historic notes on ana-
in man. Human Brain Mapping, 9, 119–142. tomic, physiologic, and clinical research on the cere-
Thier, P. (2012). The oculomotor cerebellum. In S. P. bellum. Handbook of Clinical Neurology, 154, 3–26.
Liversedge, I. Gilchrist, & S. Everling (Eds.), The Witter, L., Canto, C.B., Hoogland, T.M., de Gruijl,
Oxford handbook of eye movements. Oxford, UK: J.R., & De Zeeuw, C.I. (2013). Strength and timing
Oxford University Press. of motor responses mediated by rebound firing in
Thomasson, M., & Péron, J. (2022). Principles of brain the cerebellar nuclei after Purkinje cell activation.
and emotion: beyond the cortico-centric bias. In Frontiers in Neural Circuits, 21(7). doi: 10.3389/
M. Adamaszek, M. Manto & D. J. L. G. Schutter fncir.2013.00133
(Eds.), The emotional cerebellum (pp. 13–24). Yamazaki, T. (2021). Evolution of the Marr-Albus-Ito
Cham, Switzerland: Springer. model. In H. Mizusawa & S. Kakei (Eds.), Cerebel-
Ugawa, Y., Day, B. L., Rothwell, J. C., Thompson, P. lum as a CNS Hub (pp. 239–255). Cham, Switzer-
D., Merton, P. A., & Marsden, C. D. (1991). Modu- land. Springer.
lation of motor cortical excitability by electrical Zanatta, A., Cherici, C., Bargoni, A., Buzzi, S., Cani,
stimulation over the cerebellum in man. Journal of V., Mazzarello, P., & Zampieri, F. (2018). Vincenzo
Physiology, 441, 57–72. Malacarne (1744–1816) and the first description
Ugawa, Y., Uesaka, Y., Terao, Y., Hanajima, R., & of the human cerebellum. Cerebellum, 17(4),
Kanazawa, I. (1995). Magnetic stimulation over 461–464.
15
Amygdaloid Complex
Hans J. Markowitsch, Jens Borgelt, and
Angelica Staniloiu

INTRODUCTION AMYGDALA AND LIMBIC SYSTEM

Karl Friedrich Burdach introduced in 1822 the The amygdaloid complex constitutes one of the
term amygdala to describe a nuclear mass in the major hubs (cf. Pessoa, 2008, Fig. 1) of the so-
anterior portion of the human temporal lobes. called Limbic System. Though already introduced
Being existent in probably all vertebrates by Broca in 1878 as le grand lobe limbique, it was
(de Olmos, 2004), it is – in the human brain and only MacLean in 1952 who coined the term
that of higher mammals – composed of 13 telen- Limbic System for describing a combination of
cephalic nuclei which are grouped in three phylogenetically older structures in the brain
­clusters: the basolateral, the cortical-like, and the (older cortical areas, nuclei, and interconnecting
centromedial (Hortensius et al., 2017; Pabba, fiber systems), which, so to say, constitute an
2013; Sims and Williams, 1990; cf. Markowitsch interface between the brain stem and the phyloge-
and Staniloiu, 2012b, Fig. 4, or Sarter and netically recent neocortex (Figure 15.1). In 1970,
Markowitsch, 1985b, Fig. 2, for a delineation of MacLean refined this idea by proposing a pro-
amygdalar subregions). As it is a conglomerate of toreptilian portion (principally from the spinal
several nuclei, one also speaks of the Amygdaloid cord to the basal ganglia), a paleomammalian one
Complex, in order to emphasize this. Another (the Limbic System), and a neomammalian one
expression is corpus amygdaloideum. From the (the cortical mantle). Within the Limbic System
functional side ‘emotional, olfactory, visceral, there are two major circuits which interact which
somatosensory functions, an involvement in gen- each other. One is the Papez Circuit (Papez,
eral arousal and orienting reactions, and in sleep, 1937), the other the Basolateral Limbic Circuit
fight, defense, feeding and sexual activities have (Figure 15.2) (cf. Markowitsch and Staniloiu,
been attributed to amygdaloid nuclei’ (Sarter and 2012a, b). While the first circuit has the hip-
Markowitsch, 1985). From the first half of the pocampal formation in its center, the second cir-
twentieth century, it was proposed that the amyg- cuit includes the amygdala, the septal nuclei/basal
dala processes sensory stimuli that already have forebrain, and the thalamic mediodorsal nucleus
been pre-analyzed and modified by other centers (Sarter and Markowitsch, 1983, 1984). The (prin-
of the brain (Hilpert, 1928; Mittelstrass, 1937). cipally) unidirectional ventral amygdalofugal
228 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 15.1 Schematical sagittal section through the human brain, showing the three divi-
sions, proposed by MacLean (1952, 1970): the neomammalian, paleomammalian, and reptilian
brain.

Figure 15.2 The basolateral limbic circuit. It links the amygdala, mediodorsal thalamic
nucleus, and area subcallosa together by distinct fiber projections, namely the ventral
­amygdalofugal pathway, the inferior thalamic peduncle, and the bandeletta diagonalis.

pathway leads from the amygdala to the medio- circuit evaluates the affective side of incoming
dorsal thalamus, from there fibers reach the sub- information and interacts with the second, the
callosal area, which then projects via the bandeletta medial circuit. Amygdala and septal nuclei act in
diagonalis back into the amygdaloid body. This opposition: activation of the amygdala increases
Amygdaloid Complex 229

emotionality, activation of the septal nuclei control network is composed of the lateral pre-
decreases it (and vice versa) (Cramon and frontal, the orbitofrontal and anterior cingular
Markowitsch, 2000; Cramon et al., 1993; cortices as well as of the nucleus accumbens and
Markowitsch and Staniloiu, 2012c). The medio- the ventral tegmental area, which all are con-
dorsal nucleus (or – again – nuclear complex; cf. trolled by in- or outputs from the amygdala.
Hassler, 1982) is implicated in many functions
from sleep and consciousness to memory and
decision making (Guilleminault et al., 1994;
Markowitsch, 1982, 2005; Markowitsch and AMYGDALA: FUNCTIONAL
Staniloiu, 2012a; Mitchell, 2015). INVOLVEMENTS
Mesulam (2000) described the limbic system as
a group of interconnected cortical and subcortical
structures that As already stated above the large number of fiber
connections of the amygdala of course implies a
similarly large functional involvement.
• bind “distributed information related to recent
Furthermore, damage or hyperexcitation of the
events and experiences in a manner that sup- amygdala lead to distinct disease pictures.
ports declarative/episodic/explicit memory” Though a bit overgeneralized, older portions of
• channels “emotion and drives (…) to extraper- the amygdala are more closely related to basic
sonal events and mental content” functions such as olfaction and drive control
• links “mental activity with autonomic, hormonal, (Kose et al., 2021; Leitão et al., 2022), while
and immunological states” more recently evolved portions are engaged in
• coordinates “affiliative behaviors related to higher functions such as charging emotions to
social cohesion” memory (Cahill et al., 1995; McGaugh, 2005,
• and is involved in the “[p]erception of smell, 2015; Siebert et al., 2003). Over more than 100
years, theories on emotional processing have
taste, and pain.”
been proposed and can be found in every text-
book on biopsychology (e.g., Cannon-Bard
A nicely illustrated review of the structure and theory of emotions, James-Lange theory of emo-
function of the limbic system was written by tions, Schachter-Singer and Lazarus’ theories of
Catani et al. (2013). The connectivity and conse- emotions and cognitive appraisal).
quently the functional involvement of the amyg-
dala is manifold. One example of input–output
relations of different amygdalar portions is given
in Table 15.1, another – more refined one – can Amygdala and Fear
be found in Pessoa (2008). This author, for
instance, stresses sensory (visual) connections Especially fear conditioning and other forms of
and others which show that the amygdala partici- processing fear have been a major research topic
pates in executive functions which makes sense related to the amygdala. As modified in Figure 15.3,
as from a developmental perspective the amyg- the sketch of LeDoux (1994) is well-known
dala belongs to the basal ganglia. The executive among researchers into the physiology of

Table 15.1 Connections of different portions of the amygdala


Nuclear mass Inputs Outputs

Lateral gustatory and visceral, somatosensory, auditory, prefrontal cortex medial temporal lobe nucleus
visual cortices thalamic and hypothalamic nuclei accumbens
Basal gustatory and visceral, somatosensory, auditory, prefrontal cortex striatum medial temporal lobe
visual cortices thalamic and hypothalamic nuclei
Central gustatory and visceral, somatosensory, auditory, bed nucleus of the stria terminalis, hypothalamus
visual cortices thalamic and hypothalamic nuclei midbrain, pons, medulla ascending cholinergic
brainstem and monoaminergic systems
Medial gustatory and visceral cortices olfactory system, bed nucleus of the stria
terminalis, hypothalamic, and thalamic nuclei
Mixed nuclei olfactory input

This list was created after Figure 1A of Pabba (2013).


230 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

emotions. Evidence for the repeatedly proposed in numerous publications (Bierbrauer et al., 2021;
assumption of LeDoux (1994, 1996, 2000) that Pare and Duvari, 2012; Simic et al., 2021). Though
there exists a subcortical route from the thalamus a number of studies used sophisticated techniques
to the amygdala is still sparse, although a mode- to trace the wiring of fear conditioning within indi-
ling study, based on the recording of magnetoen- vidual amygdalar nuclei and circuit configurations
cephalographic activity in human beings, has (Lee et al., 2017; Pare and Duvari, 2012), a clear
provided evidence for its existence (Garrido et al., picture is still not available, as was stressed in the
2012). Other authors argue for a primary role of reviews cited before. This is in part attributable to
the cortical structures (Pessoa and Adolphs, 2010), the high interconnectivity of the amygdalar complex
or find at best, indirect evidence, as with the (“hub”; see above); Carlson et al. (2013), for exam-
method of diffusion tensor imaging, the direction ple, noted that individuals with a high attentional
of fiber projections cannot be determined une- predisposition to threat demonstrated a particu-
quivocally (Tamietto et al., 2012). larly high anterior cingulate-amygdala connectiv-
Fear-and stress-related responses of the amyg- ity. Similarly, Mao et al. (2020) found that social
dala – in particular its basolateral portion (Adhikari anxiety is positively correlated with functional
et al., 2015; Sun et al., 2020; Yau et al., 2021) and amygdala-orbitofrontal connectivity, and Ferri et al.
serotonin as an important transmitter in fear learn- (2016) established that attentional modulation is
ing (Bocchio et al., 2016) – have been documented related to amygdala-precuneus connectivity.

Figure 15.3 Proposed role of the amygdala in processing anxiety-related stimuli. Seeing a
tarantula implies visual information to pass from the retinae to the visual thalamus (lateral
geniculate nucleus) and then to the (primary) visual cortex. From there they reach via the
ventral visual pathway the polymodal regions of the temporal lobe and then the amygdala.
LeDoux (1994) proposed that there is an additional pathway from the visual thalamus to
the amygdala, providing a shallow, diffuse, coarse picture of the relevant environmental
stimulus, which nevertheless already alerts the amygdala to induce a counter-reaction – even
before the object becomes conscious (via the cortical processing mechanisms). For LeDoux
this stresses the role of the amygdala in fear conditioning.
Amygdaloid Complex 231

Amygdala and Positive Emotions via amygdala – ventromedial prefrontal cortical


connectivity emotion regulation is controlled.
While especially earlier investigations and studies Together, these data confirm the conclusions of
in animals emphasized the amygdala’s role in fear reviews published a decade ago, stating that the
and threat, later reports also established a substan- amygdala is necessary for processes of updating
tial role of the amygdala in processing positive representations of value (e.g., fear extinction or
emotions (Baxter and Murray, 2002). The early reinforcer devaluation) (Morrison and Salzman,
dominance of findings on fear and the amygdala 2010). The authors commented that “The encod-
can also be explained from an evolutionary point ing of affective significance by the amygdala
of view, as it is more important for survival of might be best described as a representation of state
individuals and species, to correctly analyze and value – a representation that is useful for coordi-
interpret threat signaling stimuli than those related nating physiological, behavioral, and cognitive
to joy and happiness. In fact, a study using a com- responses in an affective/emotional context.”
bination of single unit recordings and fluores-
cence labeling of individual neurons in mice
found that there are antagonistic populations of
neurons in the basolateral amygdala that are Other Functions of the Amygdala
engaged in valence-specific behaviors (Kim et al.,
2016). This finding is in line to a related one in the Smell accounts for 95–99% of chemosensation,
same species, also using a combination of meth- while the rest is processed as taste (Li and Lui,
ods (morphological, electrophysiological, and 2021). One of the major pathways of smell passes
optogenetic), and showing separate clusters of from the piriform cortex to the entorhinal cortex
basolateral amygdalar neurons for fear and reward and amygdala to the hippocampus. Any destruction
conditioning (Namburi et al., 2015). of one of these regions results – according to
Similarly, there are several findings in human Li and Lui (2021) – in anosmia. The medial amyg-
beings pointing to an amygdalar involvement in dala seems to be the principal area involved in
positive emotions (though in humans, the orbito- threat-related odor perception, as a review of
frontal cortex “phylogenetically” took over some Takahashi et al. (2005) suggests. A bit unexpected is
of these functions, as Edmund Rolls argued in that also taste, namely oral texture features of vis-
2021; cf. also Markowitsch et al., 2003, for emo- cosity, fat texture, and temperature are processed via
tion-memory-related orbitofrontal activations and the amygdala (and the orbitofrontal cortex) (Rolls,
the findings of Gee et al., 2022, indicating an 2020; Rolls et al., 2018). While these findings were
amygdala-frontal processing in childhood which obtained in the non-human primate, there is also
is reversed to top-down [fronto-amygdalar] pro- evidence that the human amygdala is activated by
cessing in adulthood). the taste of glucose (Francis et al., 1999), as well as
Results of a resting-state functional magnetic in taste in general (Petekkaya et al., 2022).
resonance (fMRI) study in humans demonstrated There is also ample evidence for an amygdala
a direct relation between reduced left amygdalar involvement in pain perception (e.g., Cai et al.,
activity and a heightened right orbitofrontal and 2018; Thompson and Neugebauer, 2017) and in
a bi-hemispheric precuneus connectivity for per- social perception – particularly in the perception
ceived social support (Sato et al., 2020) – again of facial affect (Rutishauser et al., 2015; Wang
emphasizing the hub function of the amygdala et al., 2014, 2021), but also in emotional scene
(see above). Related to the findings of Sato et al. perception (Frank et al., 2019; Oba et al., 2020).
(2020) on left amygdalar involvement in emotion Particularly the right amygdala seems to play in
processing, Puccetti et al. (2021) noted similarly scene perception a more basic “ecological” role,
in normal individuals (also using fMRI) that “indi- analyzing familiarity dimensions of the scene,
viduals demonstrating less persistent activation while temporal neocortical areas are engaged in
patterns in the left amygdala to aversive stimuli more integrative understanding of scenes, recruit-
reported more positive and less negative affect in ing the social role of the viewer (Oba et al., 2020).
daily life”. Aquino et al. (2020) studied via sin- The left amygdala, on the other hand, is related
gle unit recording in the amygdala of patients to perceived social support, in particular its lat-
with epilepsy that social cognition and reward erobasal and superficial subregions. This was
learning engage the amygdala and also results found by Sato et al. (2016) in human beings on
on mediation training (Kral et al., 2018), as well the basis of applying to 49 healthy participants
as a meta-analysis on functional magnetic reso- a so-called Multidimensional Scale of Perceived
nance imaging studies that examined emotion reg- Social Support and measuring volume and shape
ulation-modulated connectivity of the amygdala of the amygdala with structural magnetic reso-
(Berboth and Morawetz, 2021) demonstrated that nance imaging.
232 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Interestingly, a number of the studies, men- PATHOLOGY-RELATED ASPECTS


tioned up to now, pointed to the fact that there AND DISEASE CONDITIONS OF THE
is a dominance of one amygdala over the other
with respect to a number of functions. This was
AMYGDALA
already addressed in a review from the end of the
last century (Markowitsch, 1998/1999). In this it Various forms of pathological conditions, signifi-
was stated that “the left amygdala is more closely cant deviations from the normal and diseases are
related to affective information encoding with a linked to the amygdaloid complex. Psychopathy-
higher affinity to language and to detailed fea- related conditions – such as conduct disorder,
ture extraction, and the right amygdala to affec- callous-unemotional behavior, and antisocial per-
tive information retrieval with a higher affinity to sonality disorder – have been linked to changes in
pictorial or image-related material. Furthermore, the amygdala and its connections with anterior
the right amygdala may be more strongly engaged cortical areas (Aghajani et al., 2016; Cardinale
than the left one in a fast, shallow or gross analysis et al., 2019; Fairchild et al., 2019; Kaya et al.,
of affect-related information.” 2020; Yoder et al., 2015).
This statement directs to memory modula-
tion as another function in which the amygdala is
implicated. It has been known since long that the
amygdala modulates memory performance – both Amygdala and Aggression
during encoding and during retrieval (Sarter and As mainly based on data in rats and cats, Haller
Markowitsch, 1985a, b; Cahill et al., 1995). For epi- (2018) argued that predatory aggression is con-
sodic-autobiographical memory, emotional tagging trolled by the central amygdala, rivalry aggression
is mentioned as a core feature of this memory system by the medial amygdala, and violent aggression
(Markowitsch and Staniloiu, 2012a–c). Retrieving by both medial and central amygdala. Whole and
episodic-autobiographical memories reactivates centromedial amygdala–insular connectivity is
(among other structures) the right amygdala as was related to maternal aggressive behavior and later
shown in a fMRI study on episodic-autobiograph- major depressive disorder (Callaghan et al., 2017).
ical memory retrieval in normal individuals (Fink And also on the human level, there is a reduced
et al., 1996). This finding was confirmed in a later amygdala-orbitofrontal connectivity in youth with
study in which, in addition, it was found that only disruptive behavior disorders (Marsh et al., 2011) –
true, but not fictitious memories engage the right pointing to a role of the amygdala in neuromoral
amygdala (Markowitsch et al., 2000). behavior (Raine, 2019), and again, to the hub
Using a different approach, Rutishauser et al. function of the amygdala in concert with areas of
(2008) observed with single unit recordings the anterior limbic cortex.
that there was a substantial activation within the Psychotic spectrum conditions such as schiz-
human amygdala during retrieval of what they ophrenia and bipolar disorders lead to several
termed declarative memory in the title and epi- changes in amygdala shape, volume, and connec-
sodic memory in the text (cf. Rutishauser et al., tivity (Ho et al., 2019; Sabharwal et al., 2021). Ho
2015). Based on a study comparing the results of et al. (2019) especially emphasized changes in the
temporal lobe-damaged patients and non-brain- uncinate fascicle (reduced fractional anisotropy in
damaged individuals Buchanan et al. (2005) con- diffusion tensor imaging), a bidirectional pathway
cluded that the amygdala and surrounding cortices interconnecting frontal and temporal regions of the
of the medial temporal lobe may be a necessary brain (e.g., see Staniloiu and Markowitsch, 2010,
component in the neural circuitry necessary for Fig. 1). Corresponding findings – that is, a reduced
vivid recollection of unpleasant emotional events. amygdala-prefrontal connectivity – were obtained
The interaction between emotion and explicit epi- by Tang et al. (2018) for individuals between 26
sodic memory was confirmed in a fMRI-study by and 45 years of age and bipolar depression. Not
Katsumi and Dolcos (2020), while Bocchio et al. only in patients with bipolar depression, but also
(2017) concluded that “The neuronal circuits of in those with major depressive disorder (MDD),
the basolateral amygdala (BLA) are crucial for functional connectivity changes between the
acquisition, consolidation, retrieval, and extinction amygdala and other brain regions, in particular
of associative emotional memories.” Canli et al. also with the so-called default mode network were
(2000) identified a correlation between amyg- obtained (Wackerhagen et al., 2020; cf. Redlich
dala activation and episodic memory for highly et al., 2017, for therapy-related changes in the
emotional, but not for neutral stimuli. Thus, the amygdala in patients with MDD). Related find-
processing of memories with biological or social ings on a reduced functional connectivity in the
significance via the amygdala is well established medial temporal and frontoparietal network were
(Markowitsch and Staniloiu, 2011). obtained in patients with MDD and an anxiety
Amygdaloid Complex 233

symptomatology, using resting resting-state MRI to differences in the sociobehavioral phenotypes


(He et al., 2019). Other psychiatric disease con- of the two syndromes.
ditions in which the amygdala is involved are Furthermore, epileptic attacks (Beh et al.,
OCD (Paul et al., 2019; Rus et al., 2016), PTSD 2016), the Klüver-Bucy’s syndrome (Caro and
(Bijanki et al., 2020; Morey et al., 2020), and BPD Jiminez, 2016; Lanska, 2018), Alzheimer’s dis-
(Mancke et al., 2018). ease (Kim et al., 2020; Murray et al., 2021), and
Urbach-Wiethe disease (Hurlemann et al., 2007;
Koen et al., 2016; Markowitsch et al., 1994) are
accompanied by damage to or degeneration of the
Amygdala and Neurological Disorders amygdala (see below).

On the neurological disease side, there are a


number of disorders associated with the amyg-
dala. Frequently, autism-spectrum disorders Amygdala and Functional
(Dziobek et al., 2010; Hennessey et al., 2018; Sato Neurological Disorders
and Uono, 2019) are related to the amygdala. It
seems that the development of the amygdala from So-called functional neurological disorders
childhood to adulthood differs in individuals with (Carson et al., 2016; Edwards, 2016; Hallett,
autism-spectrum disorders in comparison to 2016) constitute a bridge between neurological
normals; the amygdala suddenly starts to signifi- and psychiatric disorders and involve the amyg-
cantly increase in volume at around age 6.5 to 12 dala as well (Jungilligens et al., 2022; Voon et al.,
years in children with autism-spectrum disorders 2010, 2011). This is also the case in dissociative
(Xu et al., 2020). amnesia (also named functional amnesia or mnes-
Other gene-related disorders, associated with tic block syndrome (Markowitsch and Staniloiu,
changes in the amygdala and/or its connectivity, 2016; Staniloiu and Markowitsch, 2014), where
are the fragile-X syndrome (Bruno et al., 2017; we showed a reduced amygdala metabolism in
Hessl et al., 2007; Olmos-Serrano and Corbin, affected patients (Brand et al., 2009; Staniloiu
2011; Suvrathan and Chattarji, 2011), Williams- et al., 2011) and a lacking synchrony between
Beuren’s syndrome, and Down’s syndrome. emotion- and fact-processing regions of the brain
Both the Williams’ and Down’s syndromes (cf. Markowitsch and Staniloiu, 2022). This syn-
(Murphy and Ellis, 1991; Porter et al., 2007; Pujol chrony is normally provided by the uncinate fasci-
et al., 2015) have been associated with changes in cle, a fiber system interconnecting anterior
the amygdala and/or its connectivity. Williams’ temporal regions (including the amygdala) with
syndrome is an especially interesting condition, the ventrolateral prefrontal cortex. Von der Heide
referring (as does Down’s syndrome) to a specific et al. (2013) pointed out that the uncinate fascicle
gene defect. This effect leads during develop- has prominent roles in mnestic processing, social
ment to some intellectual disabilities, in particu- cognition, and emotional valence. Most likely, the
lar learning problems, distinct facial features, so-called overgeneral memory effect, which
and cardiovascular and distinctive personality implies that autobiographical memories lack the
features. Especially the last are probably related usual emotional colorization and have a reduced
to the amygdala, as in common sense, they are specificity, can be attributed to microstructural
characterized as particularly “social.” This implies changes in the amygdala and the uncinate fascicle
that they have a major tendency to approach other (Govindan et al., 2010).
persons, including strangers, a decreased social
anxiety and an increased attention for faces (e.g.,
Bellugi et al., 1999, 2000; Jones et al., 1999). Lew
et al. (2020) found in post-mortem amygdala tis-
sue opposing directions of change in the serotoner-
AMYGDALA AND URBACH-WIETHE
gic innervation in autism-spectrum disorder brains DISEASE
and in those of individuals with Williams’ syn-
drome. The brains of the individuals with autism- Since 1993 we have studied patients with Urbach-
spectrum disorder had an increase in serotonergic Wiethe disease – first in Germany (Babinsky
axons (compared to neurotypical brains), and the et al., 1993; Brand et al., 2007; Cahill et al., 1995;
brains of former Wilson’s syndrome individu- Markowitsch et al., 1994) and later in South
als displayed a decrease. These opposite changes Africa (Siebert et al., 2003). These patients are of
were significant between the two syndromes special interest, as they have quite selective – and
across multiple amygdaloid nuclei. The authors bilateral symmetrical – damage (calcification) of
assume that these differential changes contribute the amygdaloid region; therefore, they provide a
234 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

unique “model” of the behavioral consequences of judge unapproachability, or to detect potentially


bilateral amygdala damage in human beings. dangerous or threatening people. Again, this is in
A still unresolved question is whether the cal- conformity with our data that showed a remark-
cifications of the amygdalae are already present able lack of fear in the Urbach-Wiethe patients
at birth or only develop in youth or young adult- (Markowitsch et al., 1994; Siebert et al., 2003).
hood. We (Markowitsch et al., 2023) studied a An impressive description of the lack of fear
young patient both at age 9 years and at age 19 by the Urbach-Wiethe patient of Adolphs et al.
years and found that he – in spite of genetically (1998) was given by Feinstein et al. (2011). As
proven Urbach-Wiethe disease – did not show the an example, they reported that the patient repeat-
typical calcifications in his amygdalae, suggesting edly wanted to touch snakes in an exotic pet house
that these develop only in adulthood and may vary (in spite of saying before that she disliked snakes
in density between individuals. Two other studies and spiders). Lilienfeld and co-workers (2018)
addressed this question as well with in part nega- examined patient S.M. with Urbach-Wiethe dis-
tive evidence (Appenzeller et al., 2006; Dertlioğlu ease (the same patient as studied in Adolphs et al.,
et al., 2014). While the paper of Appenzeller et al. 1998; Feinstein et al., 2011; Tranel and Hyman,
(2006) only reported that calcification was more 1990 and in other papers) and found that though
evident in patients with longer disease duration, she seemed to be pretty fearless and deficient in
that of Dertlioğlu et al. (2014) found absence of self-control, she did not fulfill other criteria of
brain calcifications in nine patients, aged between psychopathy, such as cold-heartedness and mean-
2 and 20 years (mean age: 9 years, median age: ness. The authors concluded that extreme fear
11 years), but positive evidence in one girl of deficits can co-exist with an absence of other
11 years of age. A third study, that of Pishnamazi main psychopathic attributes. Pishnamazi et al.
et al. (2016), demonstrated bilateral symmetrical (2016) noted from their single case report of a
calcifications of the amygdalae in a 14.5-year-old young female patient with Urbach-Wiethe disease
girl. Consequently, the factors favoring or inhib- that rapid attention to emotions was unchanged
iting early amygdala calcification, have not been compared to ten well-matched control individu-
unraveled. als, but appeared to be crucial for assessing the
We observed deficits with respect to the typical biological relevance of sensory events, and for an
amygdalar functions – from deficits in olfaction efficient allocation of perceptual resources. This
(Siebert et al., 2003) to deficits in episodic-auto- was also the result of a study with two twin sisters
biographical memory performance (Cahill et al., with Urbach-Wiethe disease, who showed faster
1995) – but also deficits in decision making both illusion onset and increased vividness ratings
under ambiguity and under risk (Brand et al., towards the rubber-hand illusion (Spengler et al.,
2007). Patients with Urbach-Wiethe disease were 2019). The authors concluded from their study
unable to keep social distancing and had a lack in (which also involved oxytocin-based dampening
theory-of-mind functions. Interestingly, Bellugi of the amygdala reactivity in normal individuals)
et al. (1999) compared the behavior of 25 individ- that a healthy amygdala protects from interpre-
uals with Williams’ syndrome with that of three tating potentially fatal threats due to a distorted
individuals with bilateral amygdala damage (one body-self.
with Urbach-Wiethe disease, two with encepha- Taken together, a malfunctioning amygdala not
litis; Adolphs et al., 1998). They found both only leads to severe changes in cognitive-affective
groups to be friendly in normal life and tending to behavior, but also to long-lasting alterations in the
approach people that would normally be avoided. personality structure and consequently in well-
(One of us [HJM] had similar experiences with being, and in interpreting sensory signals. This all
Urbach-Wiethe individuals from South Africa. the more, as a well-functioning amygdala is also
One of them, after just having been introduced to essential for social interaction, a capacity which is
me, said: “Hans, you come from Germany. Why the main feature of human beings.
didn’t you bring me a beer stein?”) In a face rat-
ing task, both groups gave abnormally positive
approachability ratings to unfamiliar faces (Bellugi
et al., 1999). However, while the individuals with
Wilson’s syndrome gave abnormally positive rat- SUMMARY AND CONCLUSIONS
ings for all faces (approachable and unapproach-
able ones), the patients with complete amygdala Within the limbic system, the amygdala consti-
damage gave abnormally positive ratings only for tutes the largest nuclear group. As a non-cortical
those faces that normally received the most nega- structure (i.e., in opposition to the hippocampal
tive ratings. The authors interpret these findings formation, which is three-layered archicortex), it
as an inability of amygdala-damaged patients to is phylogenetically old and existent even in
Amygdaloid Complex 235

teleosts, where it apparently already is dividable duration in lipoid proteinosis. Journal of Neuroim-
into thirteen telencephalic territories – similarly as aging, 16, 154–156.
in tetropods (Porter and Mueller, 2020). As for the Aquino, T. G., Minxha, J., Dunne, S., Ross, I. B.,
hippocampus, research from the nineteenth cen- Mamelak, A. N., Rutishauser, U., & O’Doherty, J. P.
tury viewed olfactory processing as the primary (2020). Value-related neuronal responses in the
function of the amygdala. Later, a number of human amygdala during observational learning.
additional functions were found showing that the Journal of Neuroscience, 40, 4761–4772.
amygdala is central for emotional and social pro- Babinsky, R., Calabrese, P., Durwen, H. F., Markow-
cessing and is indeed a central hub not only within itsch, H. J., Brechtelsbauer, D., Heuser, L. &
the limbic system, but within the central nervous Gehlen, W. (1993). On the possible contribution of
system as a whole. the amygdala in memory. Behavioural Neurology,
The term ‘amygdaloid complex’ refers to the 6, 167–170.
fact that the subcortical nuclear mass in the ante- Baxter, M.G., & Murray, E. A. (2002). The amygdala
rior portion of the temporal cortex of the human and reward. Nature Reviews: Neuroscience, 3,
brain (commonly named ‘amygdala’) is composed 563–573.
of more than a dozen nuclei which usually are Beh, S., Cook, M. J., & D’Souza, W. J. (2016). Isolated
combined within three subdivisions. Being heav- amygdala enlargement in temporal lobe epilepsy:
ily interconnected with cortical, diencephalic and A systematic review. Epilepsy and Behavior, 60,
brain stem structures, the amygdala is described as 33–41.
a hub with multiple cognitive and memory func- Bellugi, U., Adolphs, R., Cassady, C., & Chiles, M.
tions – from the interpretation of preprocessed (1999). Towards the neural basis for hypersociabil-
sensory information to the analysis of emotional ity in a genetic syndrome. Neuroreport, 10,
expressions and the colorization of memories. The 1653–1657.
amygdala analyzes the biological and social sig- Bellugi, U., Lichtenberger, L., Jones, W., Lai, Z., & St.
nificance of incoming information and provides George, M. (2000). I. The neurocognitive profile of
valence or value to incoming sensory signals. Williams syndrome: A complex pattern of strengths
On the output side, amygdala activity influences and weaknesses. Journal of Cognitive Neurosci-
behavioral reactions, especially, but not exclu- ence, 12(S1), 7–29.
sively, with respect to emotional expressions. Berboth, S., & Morawetz, C. (2021). Amygdala-pre-
Damage to or malfunctioning of the amygdala frontal connectivity during emotion regulation: A
can lead to numerous complex disease conditions meta-analysis of psychophysiological interactions.
from epileptic attacks to syndromes which may Neuropsychologia, 153. doi: 10.1016/j.
result in persistent and severe changes in the life neuropsychologia.2021.107767.
of the individual so affected. Bierbrauer, A., Fellner, M.-C., Heinen, R., Wolf, O. T.,
& Axmacher, N. (2021). The memory trace of a
stressful episode. Current Biology, 31. doi:
10.1016/j.cub.2021.09.044
Bijanki, K. R., van Rooij, S. J. H., Ely, T. D., Stevens, J.
REFERENCES S., Inman, C. S., Fasano, R. …Willie, J. T. (2020).
Case series: Unilateral amygdala ablation amelio-
Adhikari, A., Lerner, T. N., Finkelstein, J., Pak, S., Jen- rates post-traumatic stress disorder symptoms and
nings, J. H., Davidson, T. J., … Deisseroth, K. biomarkers. Neurosurgery, 87(4), 796–802.
(2015). Basomedial amygdala mediates top-down Bocchio, M., McHugh, S. B., Bannerman, D. M.,
control of anxiety and fear. Nature, 527, 179–185. Sharp, T., & Capogna, M. (2016). Serotonin,
Adolphs, R., Tranel, D., & Damasio, A. R. (1998). The amygdala and fear: Assembling the puzzle. Fron-
human amygdala in social judgment. Nature, 393, tiers in Neural Circuits, 10. doi: 10.3389/
470–474. fncir.2016.00024
Aghajani, M., Colins. O. F., Klapwijk, E. T., Veer, I. M., Bocchio, M., Nabavi, S., & Capogna, M. (2017) Syn-
Andershed, H., Popma, A., van der Wee, N. J., & aptic plasticity, engrams, and network oscillations
Vermeiren, R. R. (2016). Dissociable relations in amygdala circuits for storage and retrieval of
between amygdala subregional networks and emotional memories. Neuron, 94, 731–743.
psychopathy trait dimensions in conduct-disor- Brand, M., Eggers, C., Reinhold, N., Fujiwara, E., Kes-
dered juvenile offenders. Human Brain Mapping, sler, J., Heiss, W.-D. & Markowitsch, H.J. (2009).
37, 4017–4033. Functional brain imaging in fourteen patients with
Appenzeller, S., Chaloult, E., Velho, P., de Souza, E. dissociative amnesia reveals right inferolateral pre-
M., Araújo, V. Z., Cendes, F., & Li, L. M. (2006). frontal hypometabolism. Psychiatry Research:
Amygdalae calcifications associated with disease Neuroimaging Section, 174, 32–39.
236 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Brand, M., Grabenhorst, F. Starcke, K., Vandekerck- neurological disorders (pp. 105–120). Amsterdam:
hove, M. M. P., & Markowitsch, H. J. (2007). Role Elsevier.
of the amygdala in decisions under ambiguity and Catani, M., Dell’Acqua, F. D., & Thibaut de Schotten,
decisions under risk: evidence from patients with M. (2013). A revised limbic system model for
Urbach-Wiethe disease. Neuropsychologia, 45, memory, emotion and behaviour. Neuroscience
1305–1317. and Biobehavioral Reviews, 37, 1724–1737.
Bruno, J. L., Hosseini, S. M. H., Saggar, M., Quintin, Ciocchi, S., Herry, C., Grenier, F., Wolff, S. B., Letz-
E. M., Raman, M. M., & Reiss, A. L. (2017). Altered kus, J. J., Vlachos, I., … Lüthi, A. (2010). Encoding
brain network segregation in fragile X syndrome of conditioned fear in central amygdala circuits.
revealed by structural connectomics. Cerebral Nature, 468, 277–282.
Cortex, 27, 2249–2259. Cramon, D. Y. von, & Markowitsch, H. J. (2000).
Buchanan, T. W., Tranel, D., & Adolphs, R. (2005). Human memory dysfunctions due to septal lesions.
Emotional autobiographical memories in amnesic In R. Numan (Ed.), The behavioral neuroscience of
patients with medial temporal lobe damage. Jour- the septal region (pp. 380–413). Berlin: Springer.
nal of Neuroscience, 25, 3151–3160. Cramon, D.Y. von, Markowitsch, H. J., & Schuri, U.
Burdach, K. F. (1819–1822). Vom Baue und Leben (1993). The possible contribution of the septal region
des Gehims. (2 Vols.). Leipzig: Dyk’sche to memory. Neuropsychologia, 31, 1159–1180.
Buchhandlung. de Olmos, J. S. (2004). Amygdala. In G. Paxinos & J.
Cahill, L., Babinsky, R., Markowitsch, H. J., & K. Mai (Eds.), The human nervous system (2nd ed.,
McGaugh, J. L. (1995). Involvement of the amyg- pp. 739–868). San Diego: Elsevier/Academic.
daloid complex in emotional memory. Nature, Dertlioğlu, S. B., Çalık, M., & Çiçek, D. (2014). Demo-
377, 295–296. graphic, clinical, and radiologic signs and treat-
Cai, Y. Q., Wang, W., Paulucci-Holthauzen, A., & ment responses of lipoid proteinosis patients: A
Pan, Z. Z. (2018). Brain circuits mediating oppos- 10-case series from Şanlıurfa. International Journal
ing effects on emotion and pain. Journal of Neu- of Dermatology, 53, 516–523.
roscience, 38, 6340–6349. Duvarci, S., Popa, D., & Pare, D. (2011). Central
Callaghan, B. L., Dandash, O., Simmons, J. G., amygdala activity during fear conditioning. Journal
Schwartz, O., Byrne, M. L., Sheeber, L., Allen, N. B., of Neuroscience, 31, 289–294.
& Whittle, S. (2017). Amygdala resting connectivity Dziobek, I., Bahnemann, M., Convit, A., & Heekeren,
mediates association between maternal aggression H. R. (2010) The role of the fusiform-amygdala
and adolescent major depression: A 7-year longitu- system in the pathophysiology of autism. Archives
dinal study. Journal of the American Academy of of General Psychiatry, 67, 397–405.
Child and Adolescent Psychiatry, 56, 983–991. Edwards, M. J. (2016). Neurobiological theories of
Canli, T., Zhao, Z., Brewer, J., Gabrieli, J. D. E., & Cahill, functional neurologic disorders. In M. Hallett, J.
L. (2000) Event-related activation in the human Stone & A. Carson (Eds.), Handbook of clinical
amygdala associates with later memory for individ- neurology (3rd series): Functional neurological dis-
ual emotional experience. Journal of Neuroscience, orders (pp. 131–137). Amsterdam: Elsevier.
20. doi: 10.1523/JNEUROSCI.20-19-j0004.2000 Fairchild, G., Hawes, D. J., Frick, P. J., Copeland, W.
Cardinale, E. M., O’Connell, K., Robertson, E. L., E., Odgers, C. L., Franke, B., Freitag, C. M., & De
Meena, L. B., Breeden, A. L., Lozier, L. M., VanMe- Brito, S. A. (2019). Conduct disorder. Nature
ter, J. W., & Marsh, A. A. (2019). Callous and Reviews: Disease Primers, 5. doi: 10.1038/s41572-
uncaring traits are associated with reductions in 019-0095-y
amygdala volume among youths with varying Feinstein, J. S., Adolphs, R., Damasio, A., & Tranel, D.
levels of conduct problems. Psychological Medi- (2011). The human amygdala and the induction
cine, 49, 1449–1458. and experience of fear. Current Biology, 21, 34–38
Carlson, J. M., Cha, J., & Mujica-Parodi, L. R. (2013). Ferri, J., Schmidt, J., Hajcak, G., & Canli, T. (2016).
Functional and structural amygdala: Anterior cin- Emotion regulation and amygdala-precuneus con-
gulate connectivity correlates with attentional bias nectivity: Focusing on attentional deployment.
to masked fearful faces Cortex, 49, 2595–2600. Cognitive, Affective and Behavioral Neuroscience,
Caro, M. A., & Jimenez, X. F. (2016). Mesiotemporal 16, 991–1002.
disconnection and hypoactivity in Klüver-Bucy Fink, G. R., Markowitsch, H. J., Reinkemeier, M.,
syndrome: Case series and literature review. Jour- Bruckbauer, T., Kessler, J., & Heiss, W.-D. (1996).
nal of Clinical Psychiatry, 77. doi: 10.4088/ Cerebral representation of one’s own past: neural
JCP.14r09497 networks involved in autobiographical memory.
Carson, A., Ludwig, L., & Welch, K. (2016). Psycho- Journal of Neuroscience, 16, 4275–4282.
logic theories in functional neurologic disorders. In Francis, S., Rolls, E. T., Bowtell, R., McGlone, F.,
M. Hallett, J. Stone & A. Carson (Eds.), Handbook O’Doherty, J., Browning, A., Clare, S., & Smith, E.
of clinical neurology (3rd series): Functional (1999). The representation of pleasant touch in
Amygdaloid Complex 237

the brain and its relationship with taste and olfac- with the fragile X permutation. Brain, 130,
tory areas. Neuroreport, 10, 453–459. 404–416.
Frank, D. W., Costa, V. D., Averbeck, B. B., & Hilpert, P. (1928). Der Mandelkern des Menschen: I.
Sabatinelli, D. (2019). Directional interconnectivity Cytoarchitektonik und Faserverbindungen. Journal
of the human amygdala, fusiform gyrus, and für Psychologie und Neurologie, 36, 44–74.
orbitofrontal cortex in emotional scene perception. Ho, N. F., Li Hui Chong, P., Lee, D. R., Chew, Q. H.,
Journal of Neurophysiology, 122, 1530–1537. Chen, G., & Sim, K. (2019). The amygdala in
Garrido, M. I., Barnes, G. R., Sahani, M., & Dolan, R. schizophrenia and bipolar disorder: A synthesis of
J. (2012). Functional evidence for a dual route to structural MRI, diffusion tensor imaging, and rest-
amygdala. Current Biology, 22, 129–134. ing-state functional connectivity findings. Harvard
Gee, D. G., Hanson, C., Roksan Caglar, L., Fareri, D. Reviews in Psychiatry, 27, 150–164.
S., Gabard-Durnam, L. J., Mills-Finnerty, C., …Tot- Hortensius, R., Terburg, D., Morgan, B., Stein, D. J.,
tenham, N. (2022). Experimental evidence for a van Honk, J., & de Gelder, B. (2017). The dynamic
child-to-adolescent switch in human amygdala- consequences of amygdala damage on threat pro-
prefrontal cortex communication: A cross-­sectional cessing in Urbach-Wiethe disease. A commentary
pilot study. Developmental Science, 26. doi: on Pishnamazi et al. (2016). Cortex, 88,192–197.
10.1111/desc.13238 Hurlemann, R., Wagner, M., Hawellek, B., Reich, H.,
Govindan, R. M., Behen, M. E., Helder, E., Makki, M. Pieperhoff, P., Amunts, K., … Dolan, R. J. (2007).
I., & ChuganI, H. T. (2010). Altered water diffusiv- Amygdala control of emotion-induced forgetting
ity in cortical association tracts in children with and remembering: Evidence from Urbach-Wiethe
early deprivation identified with tract-based spatial disease. Neuropsychologia, 45, 877–884.
statistics. Cerebral Cortex, 20, 561–569. Jones, W., Bellugi, U., Lai, Z., Chiles, M., Reilly, J.,
Guilleminault, C., Lugaresi, E., Montagnu, P., & Gam- Lincoln, A., & Adolps, R. (2000). II. Hypersociability
betti, P.-L. (Eds.) (1994). Fatal familial insomnia: in Williams syndrome. Journal of Cognitive
Inherited prion diseases, sleep, and the thalamus. Neuroscience,12(S1), 30–46.
New York: Raven. Jungilligens, J., Paredes-Echeverri, S., Popkirov, S., Feld-
Haller, J. (2018). The role of central and medial amyg- man Barrett, L., & Perez, D. L. (2022). A new science
dala in normal and abnormal aggression: A review of emotion: Implications for functional neurological
of classical approaches. Neuroscience and Biobe- disorder. Brain, 145, 2648–2663. doi: 10.1093/
havioral Reviews, 85, 34–43. brain/awac204.
Hallett, M. (2016). Neurophysiologic studies of func- Katsumi, Y., & Dolcos, S. (2020). Suppress to feel and
tional neurologic disorders. In M. Hallett, J. Stone remember less: Neural correlates of explicit and
& A. Carson (Eds.) Handbook of clinical neurology implicit emotional suppression on perception and
(3rd series): Functional neurological disorders memory. Neuropsychologia, 145. doi: 10.1016/j.
(pp. 61–67). Amsterdam: Elsevier. neuropsychologia.2018.02.010.
Hassler, R. (1982). Architectonic organization of the Kaya, S., Yildirim, H., & Atmaca, M. (2020). Reduced
thalamic nuclei. In G. Schaltenbrand & A. E. hippocampus and amygdala volumes in antisocial
Walker (Eds.), Stereotaxy of the human brain personality disorder. Journal of Clinical Neurosci-
(pp. 140–180). Stuttgart: Thieme. ence, 75, 199-203.
Haubensak, W., Kunwar, P. S., Cai, H., Ciocchi, S., Kim, H.-G., Park, S., Rhee, H. Y., Lee, K. M., Ryu, C.-
Wall, N. R., Ponnusamy, R., … Anderson, D. J. W., Lee, S. Y., Kim, E. J., Wang, Y., & Jahng, G.-H.
(2010). Genetic dissection of an amygdala micro- (2020). Evaluation and prediction of early Alzhei-
circuit that gates conditioned fear. Nature, 468, mer’s disease using a machine learning-based
270–276. optimized combination-feature set on gray matter
He, C., Gong, L., Yin, Y., Yuan, Y., Zhang, H., Lv, L., volume and quantitative susceptibility mapping.
Soares, J. C., Zhang, H., Xie, C., & Zhang, Z. Current Alzheimer Research, 17, 428–437.
(2019). Amygdala connectivity mediates the asso- Kim, J., Pignatelli, M., Xu, S., Itohara, S., & Tone-
ciation between anxiety and depression in patients gawa, S. (2016). Antagonistic negative and posi-
with major depressive disorder. Brain Imaging and tive neurons of the basolateral amygdala. Nature
Behavior, 13, 1146–1159. Neuroscience, 19, 1636–1646.
Hennessey, T., Andari, E., & Rainnie, D. G. (2018). Koen, N., Fourie, J., Terburg, D., Stoop, R., Morgan,
RDoC-based categorization of amygdala functions B., Stein, D. J., & van Honk, J. (2016). Translational
and its implications in autism. Neuroscience and neuroscience of basolateral amygdala lesions:
Biobehavioral Reviews, 90, 115–129. Studies of Urbach-Wiethe disease. Journal of Neu-
Hessl, D., Rivera, S., Koldewyn, K., Cordeiro, L., roscience Research, 94, 504–512.
Adams, J., Tassone, F., Hagerman, P. J., & Hager- Kose, Y., Hatamoto, Y., Takae, R., Tomiga, Y., Yasukata,
man, R. J. (2007). Amygdala dysfunction in men J., Komiyama, T., & Higaki, Y. (2021). Association
238 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

between the inability to identify particular odors and Markowitsch, H. J. (1982). Thalamic mediodorsal
physical performance, cognitive function, and/or nucleus and memory: A critical evaluation of stud-
brain atrophy in community-dwelling older adults ies in animals and man. Neuroscience and Biobe-
from the Fukuoka Island City study. BMC Geriatrics, havioral Reviews, 6, 351–380.
21. doi: 10.1186/s12877-021-02363-y Markowitsch, H.J. (1998/99). Differential contribu-
Kral, T. R. A., Schuyler, B. S., Mumford, J. A., Rosen- tion of the right and left amygdala to affective
kranz, M. A., Lutz, A., Davidson, R. J. (2018). information processing. Behavioural Neurology,
Impact of short- and long-term mindfulness medi- 11, 233–244.
tation training on amygdala reactivity to emotional Markowitsch, H. J. (2005). Time, memory, and con-
stimuli. NeuroImage, 181, 301–313. sciousness. A view from the brain. In R. Buccheri,
Lanska, D. L. (2018). The Klüver-Bucy syndrome. Fron- A. C. Elitzur & M. Saniga (Eds.), Endophysics, time,
tiers of Neurology and Neuroscience, 41, 77–89. quantum, and the subjective: Proceedings of the
LeDoux, J. E. (1994). Emotion, memory and the ZiF interdisciplinary research workshop (pp. 131–
brain. Scientific American, 270, 32–39. 147). Bielefeld: World Scientific.
LeDoux, J. E. (1996). The emotional brain. New York: Markowitsch, H. J., Calabrese, P., Würker, M.
Simon & Schuster. Durwen, H. F., Kessler, J., Babinsky, R., … Gehlen,
LeDoux, J. E. (2000). Emotion circuits in the brain. W. (1994). The amygdala’s contribution to
Annual Review of Neuroscience, 23, 155–184. memory: A PET-study on two patients with Urbach-
Lee, S.-C., Amir, A., Haufler, D., & Pare, D. (2017). Wiethe disease. NeuroReport, 5, 1349–1352.
Differential recruitment of competing valence- Markowitsch, H. J., & Staniloiu, A. (2011). Amygdala
related amygdala networks during anxiety. in action: Relaying biological and social signifi-
Neuron, 96, 81–88. cance to autobiographical memory. Neuropsycho-
Leitão, J., Burckhardt, M., & Vuilleumier, P. (2022). logia, 49, 718–733.
Amygdala in action: Functional connectivity during Markowitsch, H. J., & Staniloiu, S. (2012a). Amnesic
approach and avoidance behaviors. Journal of disorders. Lancet, 380, 1429–1440.
Cognitive Neuroscience, 34, 729–747. Markowitsch, H. J., & Staniloiu, A. (2012b). The con-
Lew, C. H., Groeniger, K. M., Hanson, K. L., Cuevas, tribution of the amygdala for establishing and
D., Greiner, D. M. Z., Hrvoj-Mihic, B., … Semende- maintaining an autonomous self and autobio-
feri, K. (2020). Serotonergic innervation of the graphical memory. In D. Yilmazer-Hanke (Ed.),
amygdala is increased in autism spectrum disorder Insights into the amygdala: Structure, function
and decreased in Williams syndrome. Molecular and implications for disorders (pp. 277–318).
Autism 11. doi: 10.1186/s13229-019-0302-4. Hauppauge, NY: Nova Science.
Li, X., & Lui, F. (2021). StatPearls. Treasure Island, FL: Markowitsch, H. J., & Staniloiu, A. (2012c). A rap-
StatPearls. prochement between emotion and cognition:
Lilienfeld, S. O., Sauvigné, K. C., Reber, J., Watts, A. Amygdala, emotion and self-relevance in episodic-
L., Hamann, S., Smith, S. F., …Tranel, D. (2018). autobiographical memory. Behavioral and Brain
Potential effects of severe bilateral amygdala Sciences, 35, 164–166.
damage on psychopathic personality features: A Markowitsch, H. J. & Staniloiu, A. (2016). Functional
case report. Personality Disorders, 9, 112–121. (dissociative) retrograde amnesia. In M. Hallett, J.
MacLean, P. D. (1952). Some psychiatric implications Stone & A. Carson (Eds.), Handbook of clinical
of physiological studies of frontotemporal portion neurology (3rd series): Functional neurological dis-
of limbic system (visceral brain). Electroencephalog- orders (pp. 419–445). Amsterdam: Elsevier.
raphy and Clinical Neurophysiology, 4, 407–418. Markowitsch, H. J. & Staniloiu, A. (2022). The impor-
MacLean, P. D. (1970). The triune brain, emotion, tance of a synchrony between emotion and memory:
and scientific bias. In F. O. Schmitt (Ed.), The neu- Cases with dissociative amnesia. In C. Pracana & M.
rosciences: Second study program (pp. 336–348). Wang (Eds.), Psychological applications and trends
New York: Rockefeller University Press. (pp. 47–51). Lisboa, Portugal: inScience Press.
Mancke, F., Herpertz, S., Hirjak, D., Knies, R., & Markowitsch, H. J., Staniloiu, A., & Wahl-Kordon, A.
Bertsch, K. (2018). Amygdala structure and (2023). Urbach-Wiethe disease in a young patient
aggressiveness in borderline personality disorder. without apparent amygdala calcification. Neu-
European Archives of Psychiatry and Clinical Neu- ropsychologia, 183, 108505. https://doi.
roscience, 268, 417–427. org/10.1016/j.neuropsychologia.2023.108505
Mao, Y., Zuo, X. N., Ding, C., & Qiu, J. (2020). OFC Markowitsch, H. J., Thiel, A., Reinkemeier, M., Kes-
and its connectivity with amygdala as predictors sler, J., Koyuncu, A., & Heiss, W.-D. (2000). Right
for future social anxiety in adolescents. Develop- amygdalar and temporofrontal activation during
mental and Cognitive Neuroscience, 44. doi: autobiographic, but not during fictitious memory
10.1016/j.dcn.2020.100804. retrieval. Behavioural Neurology, 12, 181–190.
Amygdaloid Complex 239

Markowitsch, H. J., Vandekerckhove, M. M. P., Lan- Olmos-Serrano, J. L., & Corbin, J. G. (2011). Amyg-
fermann, H., & Russ, M. O. (2003). Engagement of dala regulation of fear and emotionality in fragile
lateral and medial prefrontal areas in the ecphory X syndrome. Developmental Neuroscience, 33,
of sad and happy autobiographical memories. 365–378.
Cortex, 39, 643–665. Pabba, M. (2013). Evolutionary development of the
Marsh, A. A., Finger, E. C., Fowler, K. A., Jurkowitz, I. T. amygdaloid complex. Frontiers in Neuroanatomy,
N., Schechter, J. C., Yu, H. H., Pine, D. S., & Blair, R. 7. doi: 10.3389/fnana.2013.00027
J. R. (2011). Reduced amygdala–orbitofrontal con- Pare, D., & Duvarci, S. (2012). Amygdala microcircuits
nectivity during moral judgments in youths with mediating fear expression and extinction. Current
disruptive behavior disorders and psychopathic traits. Opinion in Neurobiology, 22, 717–723.
Psychiatry Research: Neuroimaging, 194, 279–286. Paul, S., Beucke, J. C., Kaufmann, C., Mersov, A.,
McGaugh, J. L. (2005). Emotional arousal and Heinzel, S., Kathmann, N., & Simon, D. (2019).
enhanced amygdala activity: new evidence for the Amygdala-prefrontal connectivity during appraisal
old perseveration–consolidation hypothesis. Learn- of symptom-related stimuli in obsessive-compul-
ing & Memory 12, 77–79. sive disorder. Psychological Medicine, 49,
McGaugh J. L. (2015). Consolidating memories. 278–286.
Annual Review of Psychology, 66, 1–24. Pessoa, L. (2008). On the relationship between emo-
Mesulam, M.-M. (2000). Behavioral neuroanatomy: tion and cognition. Nature Reviews: Neuroscience,
Large-scale networks, association cortex, frontal 9, 148–158.
syndromes, the limbic system and hemispheric spe- Pessoa, L. (2010). Emotion and cognition and the
cializations. In M.-M. Mesulam (Ed.), Principles of amygdala: From “what it is?” to “what’s to be
behavioural and cognitive neurology (pp. 1–120). done?” Neuropsychologia, 48, 3416–3429.
New York: Oxford University Press. Pessoa, L., & Adolphs, R. (2010). Emotion processing
Mitchell, A. S. (2015). The mediodorsal thalamus as and the amygdala: From a ‘low road’ to ‘many
a higher order thalamic relay nucleus important roads’ of evaluating biological significance. Nature
for learning and decision-making. Neuroscience Reviews: Neuroscience, 11, 773–783.
and Biobehavioral Reviews, 54, 76–88. Petekkaya, E., Kuş, B., Doğan, S., Bayaroğulları, H.,
Mittelstrass, H. (1937). Vergleichend-anatomische Mutlu, T., Murat Melek, İ., Arpacı, A. J. (2022).
Untersuchungen über den Mandelkern der Säuge- Possible role of endocannabinoids in olfactory and
tiere. Zeitschrift für Anatomie und Entwicklungs- taste dysfunctions in Alzheimer’s and Parkinson’s
geschichte, 106, 717–738. patients and volumetric changes in the brain.
Morey, R. A., Clarke, E. K., Haswell, C. C., Phillips, R. Clinical Neuroscience, 100, 52–58.
D., Clausen, A. N., Mufford, M. S., … LaBar, K. S. Pishnamazi, M., Tafakhori, A., Loloee, S., Modabber-
(2020). Amygdala nuclei volume and shape in mili- nia, A., Aghamollaii, A., Bahrami, B., & Winston, J.
tary veterans with posttraumatic stress disorder. S. (2016). Attentional bias towards and away from
Biological Psychiatry and Cognitive Neuroscience: fearful faces is modulated by developmental
Neuroimaging, 5, 281–290. amygdala damage. Cortex, 81, 24–34.
Morrison, S. E., & Salzman, C. D. (2010). Re-valuing Porter, B. A., & Mueller, T. (2020). The zebrafish
the amygdala. Current Opinion in Neurobiology, amygdaloid complex: Functional ground plan,
20, 221–230. molecular delineation, and everted topology.
Murphy, G. M. Jr., & Ellis, W. G. (1991). The amyg- Frontiers in Neuroscience, 14. doi: 10.3389/
dala in Down’s syndrome and familial Alzheimer’s fnins.2020.00608
disease: Four clinicopathological case reports. Porter, M. A., Coltheart, M., & Langdon, R. (2007).
Biological Psychiatry, 30, 92–106. The neuropsychological basis of hypersociability in
Murray, A. N., Chandler, H. L., & Lancaster, T. M. Williams and Down syndrome. Neuropsychologia,
(2021). Multimodal hippocampal and amygdala 45, 2839–2849.
subfield volumetry in polygenic risk for Alzheimer’s Puccetti, N. A., Schaefer, S. M., van Reekum, C. M.,
disease. Neurobiology of Aging, 98, 33–41. Ong, A. D., Almeida, D. M., Ryff, C. D., Davidson,
Namburi, P., Beyeler, A., Yorozu, S., Calhoon, G. G., R. J., & Heller, A. S. (2021). Linking amygdala per-
Halbert, S. A., Wichmann, R., … Tye, K. M. (2015). sistence to real-world emotional experience and
A circuit mechanism for differentiating positive psychological well-being. Journal of Neuroscience,
and negative associations. Nature, 520, 675–678. 41, 3721–3730.
Oba, K., Sugiura, M., Hanawa, S., Suzuki, M., Jeong, Pujol, J., del Hoyo, L., Blanco-Hinojo, L., de Sola, S.,
H., Kotozaki, Y., … Kawashima, R. (2020). Differ- Macià, D., Martínez-Vilavella, G., … de la Torre, R.
ential roles of amygdala and posterior superior (2015). Anomalous brain functional connectivity
temporal sulcus in social scene understanding. contributing to poor adaptive behavior in Down
Social Neuroscience, 15, 516–529. syndrome. Cortex, 64, 148–156.
240 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Raine, A. (2019). The neuromoral theory of antiso- The association between perceived social support
cial, violent, and psychopathic behavior. Psychiatry and amygdala structure. Neuropsychologia, 85,
Research, 277, 64–69. 237–244.
Redlich, R., Bürger, C., Dohm, K., Grotegerd, D., Sato, W., Kochiyama, T., Uono, S., Sawada, R., &
Opel, N., Zaremba, D., … Dannlowski, U. (2017). Yoshikawa, S. (2020). Amygdala activity related to
Effects of electroconvulsive therapy on amygdala perceived social support. Scientific Reports, 10.
function in major depression: A longitudinal func- doi: 10.1038/s41598-020-59758-x. PMID:
tional magnetic resonance imaging study. Psycho- 32076036
logical Medicine, 47, 2166–2176. Sato, W., & Uono, S. (2019). The atypical social brain
Rolls, E. T. (2020). The texture and taste of food in network in autism: advances in structural and
the brain. Journal of Texture Studies, 51, 23–44. functional MRI studies. Current Opinion in Neurol-
Rolls, E. T. (2021). The neuroscience of emotional ogy, 32, 617–621.
disorders. In K. M. Heilman and S. E. Nadeau Siebert, M., Markowitsch, H. J., & Bartel, P. (2003).
(Eds.), Handbook of clinical neurology (Vol. 183, Amygdala, affect, and cognition: Evidence from
3rd series): Disorders of emotion in neurologic ten patients with Urbach-Wiethe disease. Brain,
disease (pp. 1–26). Amsterdam: Elsevier. 126, 2627–2637.
Rolls, E. T., Mills, T., Norton, A., Lazidis, A., & Norton, Šimic, G., Tkalcic, M., Vukic, V., Mulc, D., Španic, E.,
I. T. (2018). Neuronal encoding of fat using the Šagud, M., … Hof, P. (2021). Understanding emo-
coefficient of sliding friction in the cerebral cortex tions: Origins and roles of the amygdala. Biomol-
and amygdala. Cerebral Cortex, 28, 4080–4089. ecules, 11. doi: 10.3390/biom11060823
Rus, O. G., Reess, T. J., Wagner, G., Zimmer, C., Spengler, F. B., Scheele, D., Kaiser, S., Heinrichs, M.,
Zaudig, M., & Koch. K. (2016). Functional and & Hurlemann, R. (2019). A protective mechanism
structural connectivity of the amygdala in obses- against illusory perceptions is amygdala-depend-
sive-compulsive disorder. NeuroImage: Clinical, 13, ent. Journal of Neuroscience, 39, 3301–3308.
246–255. Staniloiu, A., & Markowitsch, H. J. (2010). Looking at
Rutishauser, U., Mamelak, A. N., & Adolphs, R. comorbidity through the glasses of neuroscientific
(2015). The primate amygdala in social perception: memory research: a brain network perspective.
From electrophysiological recordings and stimula- Behavioral and Brain Sciences, 33, 171–172.
tion. Trends in Neurosciences, 38, 295–306. Staniloiu, A., & Markowitsch, H. J. (2014). Dissocia-
Rutishauser, U., Schuman, E. M., Mamelak, A.N. tive amnesia. Lancet Psychiatry, 1, 226–241.
(2008). Activity of human hippocampal and amyg- Staniloiu, A., & Markowitsch, H. J. (2020). Episodic
dala neurons during retrieval of declarative memo- memory is emotionally-laden memory, requiring
ries. Proceedings of the National Academy of amygdala involvement. Behavioral and Brain Sci-
Sciences of the United States of America, 105, ences, 42, 34–36.
329–334. Staniloiu, A., Vitcu, I., & Markowitsch, H. J. (2011).
Sabharwal, A., Kotov, R., & Mohanty, A. (2021). Neuroimaging and dissociative disorders. In V.
Amygdala connectivity during emotional face per- Chaudhary (Ed.), Advances in brain imaging (pp.
ception in psychotic disorders. Schizophrenia 11–34). London: IntechOpen.
Research, 228, 555–566. Sun, Y., Gooch, H., & Sah, P. (2020). Fear condition-
Sarter, M., & Markowitsch, H. J. (1983). On the con- ing and the basolateral amygdala. F1000Research,
vergence of basolateral amygdaloid and medio- 9. doi: 10.12688/f1000research.21201.1
dorsal thalamic projections into different areas of Suvrathan, A., & Chattarji, S. (2011). Fragile X syn-
the frontal cortex of the rat. Brain Research Bulle- drome and the amygdala. Current Opinion in
tin, 10, 607–622. Neurobiology, 21, 509–515.
Sarter, M., & Markowitsch, H. J. (1984). Collateral Takahashi, L. K., Nakashima, B. R., Hong, H., & Wata-
innervation of the medial and lateral prefrontal nabe K. (2005). The smell of danger: A behavioral
cortex by amygdaloid, thalamic, and brain stem and neural analysis of predator odor-induced fear.
neurons. Journal of Comparative Neurology, 224, Neuroscience and Biobehavioral Reviews, 29,
445–460. 1157–1167.
Sarter, M., & Markowitsch, H. J. (1985a). The amyg- Tamietto, M., Pullens, P., de Gelder, B., Weiskrantz,
dala’s role in human mnemonic processing. Cortex, L., & Goebel, R. (2012). Subcortical connections to
21, 7–24. human amygdala and their changes following
Sarter, M., & Markowitsch, H. J. (1985b). The involve- destruction of the visual cortex. Current Biology,
ment of the amygdala in learning and memory: A 22, 1449–1455.
critical review with emphasis on anatomical rela- Tang, Y., Ma, Y., Chen, X., Fan, X., Jiang, X., Zhou,
tions. Behavioral Neuroscience, 99, 342–380. Y., Wang, F., & Wei, S. (2018). Age-specific effects
Sato, W., Kochiyama, T., Kubota, Y., Uono, S., of structural and functional connectivity in pre-
Sawada, R., Yoshimura, S., & Toichi, M. (2016). frontal-amygdala circuitry in women with bipolar
Amygdaloid Complex 241

disorder. BMC Psychiatry, s18. doi: 10.1186/ Wackerhagen, C., Veer, I. M., Erk, S., Mohnke, S.,
s12888-018-1732-9 Lett, T. A., Wüstenberg, T., … Walter, H. (2020).
Thompson, J. M., & Neugebauer, V. (2017). Amyg- Amygdala functional connectivity in major
dala plasticity and pain. Pain Research and Man- depression: Disentangling markers of pathology,
agement. doi: 10.1155/2017/8296501 risk and resilience. Psychological Medicine, 50,
Tranel, D., & Hyman, B. T. (1990). Neuropsychological 2740–2750.
correlates of bilateral amygdala damage. Archives Wang, S., Tudusciuc, O., Mamelak, A. N., Ross, I. B.,
of Neurology, 47, 349-355. Adolphs, R., & Rutishauser, U. (2014). Neurons in
Von der Heide, R. J., Skipper, L. M., Klobusicky, E., & the human amygdala selective for perceived emo-
Olson, I. R. (2013). The ventral pathway of the tion. Proceedings of the National Academy of Sci-
human brain: A continuous association tract ences of the United States of America, 111,
system. Brain, 136, 1692–1707. E3110–E3119.
Voon, V., Brezing, C., Gallea, C., Ameli, R., Roelofs, Xu, Q., Zuo, C., Liao, S., Long, Y., & Wang, Y. (2020).
K., Curt LaFrance, W., & Hallett, M. (2010). Emo- Abnormal development pattern of the amygdala
tional stimuli and motor conversion disorder. and hippocampus from childhood to adulthood
Brain, 133, 1526–1536. with autism. Journal of Clinical Neuroscience, 78,
Voon, V., Brezing, C., Gallea, C., & Hallett, M. (2011). 327–332.
Aberrant supplementary motor complex and Yoder, K. J., Harenski, C., Kiehl, K. A., & Decety, J.
limbic activity during motor preparation in motor (2015). Neural networks underlying implicit and
conversion disorder. Movement Disorders, 26, explicit moral evaluations in psychopathy. Transla-
2396–2403. tional Psychiatry, 5. doi: 10.1038/tp.2015.117
16
Hippocampal Formation
Rosanna K. Olsen and Mrinmayi Kulkarni

INTRODUCTION histological evidence (Duvernoy et al., 2013;


Insausti and Amaral, 2012; Lorente De Nó, 1934).
The hippocampal formation, a bilateral neuroana- A parcellation scheme used by many neuroanato-
tomical structure located deep within the temporal mists separates the hippocampus into the cornu
lobe, is one of the most well studied regions of the ammonis (CA) regions 1–4,1 the dentate gyrus,
brain due to its central importance in critical cog- and the subiculum. These hippocampal subre-
nitive functions such as memory and spatial navi- gions differ in their cellular composition, laminar
gation. In addition, this structure’s vulnerability to structure, as well as their internal and external
damage and its relevance to many types of psychi- structural connections. The cytoarchitectural dif-
atric, neurodevelopmental, and neurodegenerative ferences across subregions can best be appreciated
conditions, have made it a target of extensive using post-mortem histology staining techniques
research (Boyle et al., 2012, 2023a,b; Li et al., (Ding and Van Hoesen, 2015; see Figure 16.1). In
2019; Terreros-Roncal et al., 2021). In this chap- addition to these structural distinctions, detailed
ter, we describe in detail both the macro- and study of particular subregions of the hippocampal
microstructural neuroanatomy of the hippocam- formation using in vivo neuroimaging methods
pus, as well as the proposed neuroscientific func- has now provided evidence for distinct functional
tions of its subregions. This chapter concludes contributions to different cognitive processes
with a summary of the role of the hippocampus in (Carr et al., 2010; Olsen and Robin, 2020). For
different types of memory and other cognitive example, high resolution functional neuroimag-
functions. ing studies have indicated that the dentate gyrus
While the hippocampus is often treated as a subregion may play a selective role in mnemonic
distinct anatomical unit, it is important to appre- discrimination of visually similar scenes (Aly and
ciate that the hippocampal formation is not a Turk-browne, 2016; Berron et al., 2016).
homogenous, unitary structure. Traditionally, Another difference between the subregions of
the hippocampal formation has been subdivided the human hippocampal formation is that they
into several distinct subregions, which can be are differentially affected by distinct pathological
defined by their microstructural anatomical prop- processes in a time-dependent manner (Braak and
erties, most commonly derived from post-mortem Tredici, 2018). For example, it is well established
Hippocampal Formation 243

Figure 16.1 Histological staining of post-mortem hippocampal tissue.


Left Panel: Two representative coronal slices through the hippocampal head which demon-
strate the internal and external digitations on the superior part of the head. Note that the
dentate gyrus is absent from the more anterior slice (upper) slice. The lower slice contains
the “vertical section” of the hippocampal head as well as uncal sections. The middle panel
is zoomed in so that the cell bodies can be visualized. The different size, shape, and density
of cells allow for differentiation of the CA subfields 1–4. Right panel: One representative
coronal slice through the hippocampal body, with subfields labeled based on histological
features of the different regions.
Source: Left image is adapted with permission from Ding and Van Hoesen (2015); Middle panel adapted from Duvernoy
(2005). Right panel adapted from Olsen et al. (2019).

that in people with Alzheimer’s disease, tau Wael et al., 2018), as well as differential neural
pathology selectively affects particular hippocam- responses in these regions (Grady, 2020; Robin
pal subregions (CA1) in the earliest stages of the et al., 2019), and differential genetic expression
disease (Braak and Braak, 1991). High resolution (Cembrowski and Spruston, 2019; Dong et al.,
structural neuroimaging studies have provided 2009; Vogel et al., 2020) has led to novel insights
complementary in vivo evidence for selective atro- into this long-axis organization of the hippocam-
phy. These studies have used magnetic resonance pal formation (Genon et al., 2021). This chapter
imaging (MRI) to identify grey matter volume loss provides an overview of the current understanding
in the CA1 region of people who have mild cog- of the macro- and microstructural properties of the
nitive impairment, a condition that often precedes hippocampal formation, as well as current theo-
Alzheimer’s disease (de Flores et al., 2015). CA1 ries of its functional organization, with a special
is also thought to be more susceptible to hypoxic- emphasis on the cognitive functions supported by
ischemic damage than more resistant areas such as distinct hippocampal subregions in humans.
CA3 (Schmidt-Kastner and Freund, 1991).
In addition to the variability within the hip-
pocampal formation along its transverse axis
described above, the structural and functional
properties of the hippocampal formation also are ANATOMY OF THE HIPPOCAMPUS
thought to differ significantly along its longitu-
dinal axis (Poppenk et al., 2013; Strange et al., Macrostructural Definitions
2014). There is considerable evidence from mul-
tiple lines of research that indicates anterior and The hippocampus is comprised of two cortical
posterior human hippocampal formation regions laminae (i.e. layered tissue) rolled up one inside
play variable roles in different types of cognition the other: the cornu ammonis and the gyrus denta-
and are also differentially affected in development tus (or “dentate gyrus,” also referred to as the
and by disease (Lladó et al., 2018; Riggins et al., “fascia dentata”), separated from each other by the
2018). Emerging research on the structural and hippocampal sulcus (Figure 16.2A). The term
functional connectivity of the anterior and pos- “ram’s horn” was adopted to describe the shape of
terior hippocampus (Libby et al., 2012; Vos De the cornu ammonis regions, and accordingly,
244 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 16.2 Folding patterns in development and in the adult hippocampus.


The curved structure of the hippocampus originates from an initially flat piece of tissue that
folds medially during development. This fold forms the hippocampal sulcus, denoted by the
black arrow. The green arrow demonstrates the proximal-distal dimension of the folding pat-
tern, and the red arrow demonstrates the folding along the long-axis. The fully developed
hippocampus is shown on the right (pictured from above). Dotted lines indicate commonly
used long-axis divisions of the hippocampus into the head (uncal section), head (body-like
section), body, and tail. A = anterior, L = lateral, M = medial, P = posterior
Source: From DeKraker et al., 2018; adapted from Duvernoy et al., 2013.

these regions are named after the Egyptian god dentate gyrus (DG), and subiculum portions as
Ammon (Ammun Kneph). Many anatomists and “allocortex” (sometimes also called archicortex;
neuroscience researchers also include the subicu- cf. Insausti et al., 2017)
lum (which is located between the cornu ammonis In the human, the hippocampus is located in the
regions and the adjacent entorhinal regions in the medial aspect of the temporal lobe and bordered
medial temporal lobe cortex) as part of the hip- anteriorly by the amygdala. Parts of the thalamus
pocampal formation, although some view the (e.g., the lateral geniculate nucleus) border the
subiculum as a transitional region between the superior aspect of the hippocampal formation. The
hippocampus and the rest of the temporal lobe. lateral part of the hippocampus (mostly CA1) is
The hippocampus proper, when dissected away bordered by the temporal horn of the lateral ventri-
from the rest of the brain, resembles a seahorse cle. The subiculum extends laterally and inferiorly
(Figure 16.2). The term “hippocampus” is the into the parahippocampal gyrus, which is a mac-
Latin word for seahorse, in line with the Greek rostructural landmark for the inferomedial aspect
term “hippokampos”. The cortical tissue that of the hippocampus. The tail of the hippocampus
makes up the hippocampus contains three pri- is described as “disappearing” (i.e., narrowing)
mary layers of cells rather than the more common beneath the splenium. The indusium griseum is a
regions of neocortex that contain six layers of structure thought to be a dorsal extension of the
cells. This laminar structure of the hippocampal hippocampus, narrow strip of grey matter running
formation classifies the cornu ammonis (CA) 1-4, dorsal to the corpus callosum in the subcingulate
Hippocampal Formation 245

region. It is now recognized that the subiculum belonging to the CA, DG, and subiculum. The
can be separated from two distinct adjacent subre- position of these subfields within the uncus can be
gions (e.g., the presubiculum, and the parasubicu- explained by the folding pattern of the hippocam-
lum; Insausti et al., 2017). The presubiculum and pus during development (DeKraker et al., 2018;
parasubiculum are referred to as “periallocortex” Ding and Van Hoesen, 2015; Duvernoy et al.,
as they contain six layers, although one layer is 2013; see Figure 16.2).
“cell-free” and is referred to as the lamina disse- The superior part of the hippocampal head
cans (Insausti et al., 2017). contains “elevations” or “bumps,” which have
been described by Duvernoy as “the digitationes
hippocampi.” Ding and Van Hoesen (2015) have
Hippocampal Head described these digitations in detail, and also make
the distinction between the internal digitations
The anterior, middle, and posterior parts of the (those that extend inferiorly) and external digita-
human hippocampus are dramatically different tions (those that extend superiorly). Ding and oth-
from each other in terms of macrostructural shape. ers have also described the “vertical digitation”
The anterior part of the hippocampus contains a which is the most medial digitation. This part of
rounded “bulge” that extends medially; this part the hippocampus gets its name from its vertical
of the hippocampal head is referred to as the orientation (Figures 16.1 and 16.2). The number
“uncus” which is named for its hook-like structure and size of the digitations can vary across sub-
(Figures 16.2 and 16.3). Like the other parts of the jects (DeKraker et al., 2021). There are also some
hippocampal formation, the uncus is not a single “transitional” structures in the anterior hippocam-
homogenous region; it contains different types of pus that connect it to the amygdala and entorhinal
hippocampal tissue, including gray matter cortex. One region, the hippocampal-amygdala

Figure 16.3 Hippocampal shape varies greatly across individuals. A: 3-dimensional render-
ings of different hippocampi, which demonstrate the variability of the hippocampal head
digitations, curvature, and overall shape. B: Two examples of two different individuals with
variable digitations in the hippocampal head. C and D: Two examples of hippocampal denta-
tions on the inferior surface of the hippocampus. These two individuals vary by the number
and depth of the dentations.
Source: A and B adapted from DeKraker et al. (2021). C & D adapted from Beattie et al. (2017).
246 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

transition area (abbreviated “HATA”), is often layers of the CA are the stratum oriens, the stra-
located in or near the vertical digitation. tum pyramidale, and a third layer that combines
Compared with the hippocampal head, the the stratum radiatum, lacunosum, and molecu-
body of the hippocampus is more tubular or cylin- lare, which is sometimes abbreviated as “SRLM”
drically-shaped, with a further narrowing that typ- (Figure 16.4), providing a useful landmark for
ically continues posteriorly into the hippocampal neuroimagers to separate CA layers from the
tail. Like the hippocampal head, the anatomical DG layers of the hippocampal formation (de
features (i.e., size and shape) of the hippocampal Flores et al., 2020; Wisse et al., 2020). For
tail vary from person to person, and in some indi- example, in high-resolution T2-weighted MRI
viduals the hippocampal tail curves medially and scans, the SRLM can be visualized as a c-shaped
sometimes superiorly (Figure 16.3A). “dark band” in coronal sections (as shown in
An additional macrostructural feature of the Figure 16.4).
hippocampus that has recently been recognized The pyramidal layer of cornu ammonis shows
is the presence of “dentations” or “bumps” on regional variations, which has been used by neu-
the inferior surface of the hippocampal body and roanatomists to determine the different numbers/
tail (Figure 16.3C and 16.3D; Chang et al., 2018; names for the different subfields. Lorente de No
Fleming Beattie et al., 2017). Note that these struc- in the early 1900s described four fields: CA1-CA4
tures are thought to be distinct from the digitations (Figure 16.1, middle panel). CA1 contains pyram-
present on the superior aspect of the hippocampal idal cells that are small, triangular, and more
head. The functional relevance of these dentations “scattered” or widely spaced compared to CA2
is not yet well characterized, especially because and CA3. Conversely, the cells in CA2 and 3 are
they are difficult to visualize using current in vivo larger and more densely packed compared to those
neuroimaging protocols. of CA1. The CA2 region has the most compact
and narrow pyramidal cell layer of the hippocam-
pus. Its cells are almost as large as those in CA3,
but there is far less space between the cell bod-
ies. The CA3 pyramidal cells are the largest of the
MICROSTRUCTURAL DEFINITIONS hippocampus and stain darkly in Nissl stains. CA3
also contains the unmyelinated mossy fibers of the
Cellular Composition DG which form a supplementary layer in between
the statum radiatum and statrum moleculare,
As described above, the CA, DG, and subiculum called the stratum lucidum. The stratum pyrami-
subregions of the hippocampus are composed of dale layer of CA3 is approximately ten cells thick
three major layers of cells. The three major and it has a relatively homogeneous appearance.

Figure 16.4 Left panel demonstrates the different types of layers in fascia dentata (dentate
gyrus) and cornu ammonis.
Right panel demonstrates the layers on high resolution postmortem MRI and on ­postmortem
histology. The SRLM layers appear as a dark band on T2-weighted MRI.
Source: Left panel adpated from Duvernoy, 2005; Right panel adapted from de Flores et al., 2019.
Hippocampal Formation 247

Finally, the pyramidal cell layer of the subiculum efferent white matter structures within and adja-
is 30 or more cells thick, and can be divided into cent to the human hippocampus (Amaral et al.,
at least two sublaminae (sub-layers). Relative 2018; Beaujoin et al., 2018; Ke et al., 2020;
to CA1, the subicular pyramidal cells tend to be Parekh et al., 2015; Zeineh et al., 2012, 2017).
somewhat larger and more widely spaced. These include the perforant path system, which
Turning to the dentate gyrus, this subregion of connects the entorhinal cortex to several differ-
the hippocampus also contains three major layers. ent regions of the hippocampus, including the
The layers of the dentate gyrus are: the stratum dentate gyrus, CA3, and CA1. The perforant
moleculare (molecular layer), the stratum granu- path system has many components, including
losum (granular layer) and the polymorphic (or both transverse and longitudinal components
multiform) layer. The stratum granulosum is con- (Zeineh et al., 2017) which connect the more
sidered the main layer of this structure and it con- anterior and posterior regions of the hippocam-
tains the somata of granular neurons, which are pal formation.
small and round but densely packed, making the Current evidence of these white matter struc-
layer easy to distinguish in post-mortem stained tures and their associated intra-hippocampal net-
(e.g., Nissl) slices. Mossy fibers are the axons from works has influenced theories about hippocampal
these granular neurons and they traverse the poly- functions. Until recently, the field has relied heav-
morphic layer into CA4 and CA3 (Figure 16.4). ily on tracer studies in non-human animal mod-
els (Bubb et al., 2017; Witter and Amaral, 1991).
Future improvements in neuroimaging technology
will help update any cross-species differences that
Neurochemical Composition of have evolved in humans. For example, the end-
Hippocampal Subregions folial pathway, which is an output pathway from
CA4, projecting to the Schaffer collaterals of
The cells within the hippocampus contain numer- CA3, seems to exist only in primates (Lim et al.,
ous receptor types which are co-localized in differ- 1997; Parekh et al., 2015).
ent regions and layers of the hippocampus.
Neurotransmitter types include noradrenaline/nor-
epinephrine, serotonin, acetylcholine, dopamine,
glutamate, and GABA. Peptides and other chemi-
HIPPOCAMPAL AFFERENTS
cal markers have also been shown to localize cer-
tain populations of interneurons in the hippocampus.
Moreover, different calcium-binding proteins
Entorhinal Cortex Inputs
including parvalbumin, calbindin, and calretinin
also bind to different subsets of GABAergic neu- The entorhinal cortex provides most of the input
rons. Recent investigations have combined classic into the hippocampus from both subcortical and
cytoarchitectonic methods with receptor-mapping neocortical sites. However, there is a well-charac-
methods to understand the chemoarchitecture of terized topographical pattern of projections from
the hippocampal subregions (Ding and Van distinct anatomical subregions of the entorhinal
Hoesen, 2015; Palomero-Gallagher et al., 2020; cortex into the hippocampus (Witter, 2007). There
Zeineh et al., 2017). These investigations have are at least two patterns that have been identified.
provided unique evidence for the existence of First, the lateral entorhinal cortices project to the
unique subregions (e.g., CA4 and pro-subiculum) more distal part of CA1 (closer to the subiculum)
not universally recognized by other neuroanato- and the medial entorhinal cortices project to the
mists who rely on more traditional mapping meth- more proximal part of CA1 (closer to CA2).
ods which rely more heavily on cytoarchitecture. Recent studies have also indicated that the differ-
ent layers of CA1 receive different inputs from
lateral and medial entorhinal cortex. The lateral
entorhinal cortices contribute excitatory synapses
INTRINSIC AND EXTRINSIC in the superficial CA1 layers; whereas the medial
CONNECTIVITY OF THE HIPPOCAMPUS entorhinal cortex excite the deeper layers of CA1
(Masurkar et al., 2017). This layer-specific pattern
of connectivity suggests that the well-established
White Matter Structures Within “tri-synaptic circuit” is more complex than origi-
the Hippocampus nally proposed and that new models of hippocam-
pal function should evolve to incorporate the
Recent advances in high-resolution imaging heterogeneity of these circuits (Valero et al.,
techniques have helped define the afferent and 2018).
248 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Hippocampal Efferents The effect of hippocampal vascularization on neu-


rodegeneration is still an area of active investiga-
The fornix is a major output structure of the hip- tion. Several recent in vivo studies in older adults
pocampus, projecting from the subiculum to the have demonstrated that “mixed” or “dual supply”
mammillary bodies, anterior thalamus and the to the hippocampus (i.e. blood supply arising from
septum (Bubb et al., 2017). Distinct components both the AChA AChA and PCA) might be protec-
of fornix, including the pre-commissural fornix, tive against cognitive decline and hippocampal
have been further characterized using diffusion atrophy (Perosa et al., 2020).
tensor imaging (Choi et al., 2019; Hodgetts et al.,
2020; Wei et al., 2017; Williams et al., 2020;
Zeineh et al., 2012). The alveus and the fimbria
are longitudinal fibers, which merge with the Hippocampal Long-axis Organization
fornix, which then projects to the mammillary
bodies, as well as the anterior nucleus of the thala- While studies into differences in cytoarchitecture
mus. The alveus covers the intraventricular sur- have shed light on subfield organization along the
face and contains axons of the hippocampal and proximal-distal/medial-lateral axis, there is sub-
subicular neurons. Similar to the gray matter of stantial evidence supporting systematic variation
the hippocampus, these white matter structures in structure and function along the long-axis of the
can be measured volumetrically by using high- hippocampus. In both rodent and primate studies,
resolution structural MRI. In Alzheimer’s disease, the hippocampus is often divided into two seg-
the fimbria and fornix are reduced in volume, but ments, transitioning from the anterior hippocam-
the alveus is not affected (Amaral et al., 2018). pus in primates (ventral hippocampus in rodents)
to the posterior hippocampus (dorsal hippocam-
pus in rodents) at the last slice containing the
uncal apex (Poppenk et al., 2013). In humans,
Vascular Supply several dichotomies for the functional specializa-
tion of the anterior and posterior hippocampus
The vascular supply of the hippocampus has been have been proposed (Grady, 2020). Some have
well-studied in post-mortem studies (Erdem et al., suggested that the anterior hippocampus, through
1993) and more recently using MRI-angiography its direct and indirect connections with the amyg-
(Spallazzi et al., 2019). The hippocampus is pri- dala, nucleus accumbens, hypothalamus, and ven-
marily supplied by the posterior cerebral artery tral tegmental area, is specialized for emotional
(PCA), and to a lesser extent, the anterior choror- processing, whereas the posterior hippocampus
idal artery (AChA; Duvernoy et al., 2013; Erdem supports cognitive processing (Murty et al., 2011).
et al., 1993). It is believed that there are significant Some neuroimaging studies have reported selec-
individual differences in the patterns of the vascu- tive activity in the anterior and posterior hip-
lar supply to the hippocampus by the hippocampal pocampus during memory encoding and retrieval,
arteries that arise from these two main vessels. respectively (Lepage et al., 1998). Still others
Post-mortem studies originally demonstrated that have suggested posterior hippocampal specializa-
the contribution of the AChA to the hippocampal tion in the representation of spatial information
arteries is quite variable and is sometimes absent and navigation (Woollett and Maguire, 2011).
(Marinković, Milisavljević, and Puškaš, 1992). However, evidence in support of these dichoto-
This vascular variability has now been docu- mies has been mixed, suggesting that differences
mented in vivo using MR angiography (Perosa in functional specialization may be dynamic (i.e.,
et al., 2020; Spallazzi et al., 2019). dependent on moment-to-moment input from the
It has long been recognized that the hip- MTL and neocortex to the anterior and posterior
pocampus, in general, is vulnerable to hypoxia regions). Thus, the ability to probe this functional
and other conditions causing cerebral hypoper- specialization may be sensitive to the specific
fusion (Duvernoy et al., 2013; Schmidt-Kastner tasks used to measure cognitive processes
and Freund, 1991). It has also been recognized (Poppenk et al., 2013).
that certain subregions of the hippocampus are In addition to differences in cognitive processes,
more sensitive to hypoxia than other subregions. the anterior and posterior hippocampal subregions
For example, the CA1 region was described by also exhibit different profiles of connectivity with
Wilhelm Sommer in the late 1800s as the “vulner- the rest of the brain. Distinct brain networks have
able sector” and the CA3 region as the “resistant been identified based on their correlated fluctua-
sector.” The reason for the differential sensitivity tions in activity (“functional connectivity”) with
to hypoxia among subregions may be explained by other regions during task and rest. Regions in the
the arrangement of the hippocampal vasculature. MTL cortex (adjacent to the hippocampus) such
Hippocampal Formation 249

as the PRC and PHC are part of distinct functional capable of representing fine-grained, detailed
networks (Ranganath and Ritchey, 2012). The information (Poppenk et al., 2013). This distinc-
PRC, considered a core component of the ante- tion is also mirrored in humans. Activity in the
rior temporal (AT) network, exhibits connectivity posterior hippocampus is higher for tasks relying
with the amygdala, lateral orbitofrontal cortex, on retrieval of fine spatial details of an experience
and the anterior temporal lobe. The PHC, on the (Hirshhorn et al., 2012), and conversely, more
other hand, is part of the posterior medial (PM) activity in the anterior hippocampus is observed
network, along with the retrosplenial cortex, pre- when approximate or relative spatial information
cuneus, angular gyrus, posterior cingulate cortex, is required (Poppenk et al., 2013; Strange et al.,
and medial prefrontal cortex. 2014). Extending this dichotomy to the represen-
These networks contribute to different aspects tation of non-spatial information, studies have
of episodic memory. Substantial evidence has reported that retrieval of episodic details recruits
linked regions in the AT network to familiarity- the posterior hippocampus, whereas retrieval of
based, item memory, emotional and motivational gist or schematic representations is associated
processing, and representation of conceptual and with anterior hippocampal activity (Addis et al.,
semantic information. Together, these regions have 2004). Thus, differences in local circuit organi-
been proposed to support the use of prior experi- zation across the hippocampus long-axis seem to
ence to guide future behaviour (Ranganath and the reflected in the high-level representation of
Ritchey, 2012). In contrast, studies have shown information.
the involvement of regions in the PM network in Overall, mounting evidence indicates differen-
recollection-based memory (i.e., memory for con- ces in local information processing, intra-hippo-
textual information, Yonelinas et al., 2010), spa- campal and neocortico-hippocampal connectivity,
tial navigation, and recall of event detail. Activity as well as functional specialization along the long
in the PM network scales with the accuracy and axis of the hippocampus. These studies, in addi-
vividness of retrieved information (Cooper and tion to those demonstrating different trajecto-
Ritchey, 2019; Kuhl and Chun, 2013; Richter ries of decline in normal and pathological aging
et al., 2016). Additionally, multivariate patterns (Blujus et al., 2022; Langnes et al., 2020; Lladó
of activity in the PM network carry information et al., 2018; Martin et al., 2010), suggest that the
about the content of retrieved episodic memo- long-axis is an important organizing principle
ries (Robin et al., 2018). Moreover, studies have within the hippocampus.
shown the involvement of the PM network not
only in memory retrieval, but also episodic future
thinking and simulation (Cooper and Ritchey,
2020). Importantly, these networks exhibit differ- Structure–Function Relationships
ential connectivity with the anterior and posterior
hippocampus. The anterior hippocampus is more Experimental evidence from hippocampal amne-
highly functionally connected to the AT network, sic cases (like the famous hippocampal amnesic,
whereas the posterior hippocampus is functionally “H.M.”) as well as lesions in non-human animal
more connected to the PM network (Barnett et al., models has indicated that the hippocampus plays
2021; Ranganath and Ritchey, 2012). Thus, the a critical role in the formation of new memories.
anterior and posterior hippocampus may subserve Certain types of memory, including autobiograph-
distinct cognitive processes through their differen- ical or episodic memory, are adversely affected by
tial connections with the rest of the brain. damage to the hippocampus, which can occur as a
Another feature of the hippocampus that has result of surgical procedures (e.g., temporal lobec-
been theorized to drive the long-axis organiza- tomy), strokes, anoxia, encephalitis, or neurologi-
tion of hippocampal function relates to different cal conditions such as epilepsy and dementia.
responsivity of neurons in the anterior and pos- Age-related atrophy or neurodegeneration in the
terior hippocampus. Neural responses along the hippocampus also occurs as a natural consequence
long axis of the hippocampus differ in the granu- of healthy aging in humans (Raz et al., 2004a,b).
larity of their representation. In rodents, the spa- Researchers have demonstrated that damage or
tial receptive field sizes of neurons increase from atrophy to the hippocampus results in changes in
dorsal to ventral hippocampus (Kjelstrup et al., certain types of memory function, although the
2008). This receptive field pattern, in conjunction extent of the impairment can vary across patients.
with findings about the type of input that subre- The types of memory that are impaired and spared
gions receive from the entorhinal and perirhinal due to hippocampal damage/atrophy has led to our
cortices, suggests that the anterior hippocampus understanding of how memory is organized in the
supports coarse-grained representations of infor- brain. Most prominently, changes in long-term,
mation whereas the posterior hippocampus is more remote, episodic, or autobiographical memory
250 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

have been reported (Spiers et al., 2001), and other following initial learning (Gabrieli et al., 1993),
types of memory (e.g., semantic memory, motor indicating that regions outside the hippocampus
learning) are either spared or less impaired. It is may support forms of memory that do not require
important to recognize, however, that damage to conscious awareness.
the hippocampus can also affect other cognitive More recently, researchers have argued that
functions, outside of memory (see below). non-conscious forms of memory are also critically
dependent on the hippocampus (Henke, 2010;
Moscovitch, 2008). For example, humans with
hippocampal damage do not demonstrate the same
Role of Hippocampus in Episodic type of implicit learning of object-location associ-
Memory ations compared with people without hippocampal
damage (Chun and Phelps, 1999).
Episodic memory, which is thought to rely criti- Similarly, the expression of memory in indirect
cally on the hippocampus, refers to memories of measures, such as viewing behavior, is absent in
unique events, which occur at a particular time patients with damage limited to the hippocampus
and place, and are typically only experienced once (Hannula et al., 2007; Ryan and Cohen, 2004),
(Tulving, 2002). Autobiographical memory refers whereas in neurologically intact individuals, this
to memories of life events for a particular indi- effect is observed even without conscious aware-
vidual and this type of memory is severely ness of learned materials (Ryan et al., 2000). In
impaired in persons with damage to the hip- fact, in healthy young adults, hippocampal activity
pocampus (Addis et al., 2007). Furthermore, epi- is associated with the memory based eye-move-
sodic memory is associated with the ability to ments even when explicit responses are incorrect
experience what Tulving called, “mental time (Hannula and Ranganath, 2009). These results
travel.” This form of memory allows someone to suggest that implicit memory is tied to hippocam-
“re-experience” a previous event with high fidel- pal processing both during encoding and retrieval
ity, including associated thoughts and feelings, the (Hannula and Greene, 2012).
spatial context in which the episode occurred, and
other associated sensory details (sights, sounds,
smells). The hippocampus is thought to perform
important binding and indexing functions to form Role of Hippocampus in Relational
these autobiographical memory associations and Memory
later retrieve them.
A key feature of episodic memory is that it con-
tains memory for individual elements of an epi-
sode, as well as the relations between the elements.
Do Only Conscious Forms of Memory Thus, one’s memory for an event such as a music
Depend on the Hippocampus? concert, may consist of a bound representation of
elements such as the venue of the show, the band
Autobiographical and episodic memory are typi- that performed, the songs they showcased, and the
cally grouped under the umbrella category of location of one’s seat in the audience with respect
“long-term memory.” Furthermore, these memo- to the stage. Studies have shown that this type of
ries have been categorized by some researchers as memory, termed relational memory, critically
“declarative” or “explicit” types of memory as depends on the hippocampus (Cohen and
opposed to “non-declarative” or “implicit.” These Eichenbaum, 1993; Davachi, 2006). Compelling
definitions of memory loss have focused on the evidence for the involvement of the hippocampus
notion that hippocampal damage primarily affects in relational memory comes from a neuropsycho-
memory that is available to our conscious aware- logical study in which participants completed a
ness (Squire and Zola, 1996). For example, indi- task which tested memory for individual items, as
viduals with hippocampal amnesia are impaired well as memory for spatial, temporal, and associa-
when asked to consciously report a recent experi- tive relationships between items. Relative to age-
ence (e.g., verbally recall a list of words or recog- matched controls, patients with hippocampal
nize a list of pictures). Other forms of memory amnesia were impaired on all tests of relational
that are considered implicit, or non-declarative, memory, whereas their memory for individual
like skill learning (Milner, 1970), are not as criti- items was relatively spared (Konkel et al., 2008;
cally impaired in hippocampal amnesia (Corkin, cf. Giovanello et al., 2003; Hannula et al., 2007;
2002). Interestingly, improvements in perfor- Ryan et al., 2000). These findings are supple-
mance of patients with hippocampal damage on mented by results from neuroimaging studies that
these tasks were evident up to several months show greater hippocampal activation during
Hippocampal Formation 251

encoding and retrieval of associations supported Similarly, neuroimaging studies have also
by relational memory compared with encoding documented the involvement of the hippocam-
and retrieval of item memory (Giovanello et al., pus in the retrieval of recent episodic memories
2004; Kirwan and Stark, 2004; Prince et al., (Greicius et al., 2003; Prince et al., 2005). The
2005). Consequently, it has been proposed that the extent to which the hippocampus is involved in the
role that the hippocampus plays in memory for- recall of remote memories, on the other hand, is
mation is that of binding disparate elements of an less clear. There have been reports that damage to
episode to form a coherent, bound representation the hippocampus affects the ability to recall mem-
(Konkel and Cohen, 2009), with regions in the ories which were experienced before the damage
surrounding MTL cortex (e.g., perirhinal cortex) occurred (i.e., retrograde amnesia; Squire et al.,
being recruited to support memory for individual 2001). In several of these cases retrograde amnesia
items (Davachi, 2006). is limited to memories only from a few years prior
to the hippocampal damage (Kapur and Brooks,
1999; cf. Argyropoulos and Butler, 2020), which
suggests that the hippocampus plays a time-lim-
Episodic Versus Semantic Memory ited role in the consolidation and retrieval of past
events (Bayley et al., 2003; Boccia et al., 2019).
Based on patterns of impaired and spared memory However, other evidence has indicated the hip-
in people with hippocampal damage, memory pocampus is necessary for the retrieval of per-
researchers have often distinguished between epi- sonally experienced detailed events even from
sodic and semantic memory. Episodic memory the distant past (Moscovitch et al., 2006). For
refers to memories for information that has been instance, results from neuroimaging studies have
experienced only once and occurs in a particular shown hippocampal activation during retrieval
place and time. Semantic memory refers to infor- regardless of the age of the memory (Addis et al.,
mation that has been experienced multiple times 2004; Ryan et al., 2001). For this reason, it has
(i.e., knowledge that is context-free). Some exam- been argued that episodic memories, in particular
ples of semantic memory are memory for facts memories tied to a particular spatiotemporal con-
about the world and knowledge of word meaning text, are always dependent upon the hippocampus,
(i.e., vocabulary). Semantic memory is relatively no matter how much time has passed since the epi-
spared in amnesia, especially in cases where the sode (Moscovitch and Gilboa, 2021).
damage is restricted to the hippocampus. However,
there is some retrograde amnesia for more recently
acquired semantic knowledge. This suggests that
the hippocampus plays a more time-limited role in Role of Hippocampus in Short- and
semantic memory compared to episodic memory Long-term Memory
and that there is an important distinction between
episodic and semantic memory (Rosenbaum et al., While the role of the hippocampus in long-term
2005). memory is well-established, more recent research
has provided evidence for its role in other types of
cognition, such as visual perception and “short-
term” or “working memory” (Graham et al.,
Role of Hippocampus in Retrieval of 2010). Some scholars have proposed that the
Recent and Remote Memories underlying computations performed by the hip-
pocampal formation support a binding mechanism
As mentioned above, it is well established that that is critical for multiple forms of cognition,
damage to the hippocampus affects the ability to including short-term memory and perception
form new episodic or long-term memories (Cohen (Olsen et al., 2012). Binding refers to the forma-
and Eichenbaum, 1993). Memory loss for events tion of an association among distinct elements of
that occur after hippocampal damage is referred to an experience (e.g., a face and name).
as anterograde memory loss. In addition to antero- The role of the hippocampus in the binding of
grade memory loss in patients with hippocampal elements in service of long-term memory has been
damage, numerous neuroimaging studies have outlined above. However, results from several
demonstrated consistent activity in the hippocam- studies indicate that this computation performed
pus during memory encoding (Davachi et al., by hippocampus may be critical even at shorter
2003; Greicius et al., 2003; Prince et al., 2005). delays (Olsen et al., 2012). For instance, patients
Taken together, these results indicate that the hip- with hippocampal amnesia are impaired on work-
pocampus plays a central and pivotal role in the ing memory tasks that require binding of multi-
formation of new memories. ple features of a visual display (e.g., the color of
252 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

items and their locations), even when tested after further allow for the analysis of both subfield and
a 1s delay (Borders et al., 2022). In line with this anterior-posterior organization (Hrybouski et al.,
finding, among healthy young adults, the level of 2019).
univariate activity in the head and body of the hip-
pocampus was predictive of the precision of work-
ing memory decisions.
In addition to this binding mechanism, which Material Specificity of Hippocampal
may play a more critical role during memory Representations
encoding, the hippocampus also likely performs a
“comparison” mechanism during memory retrieval Other groups have focused on the type of materi-
(Chen et al., 2011; Kumaran and Maguire, 2007; als used as memory stimuli in different memory
Vinogradova, 2001). Comparison refers to the pro- experiments to better understand the contributions
cess of evaluating the current information with the of the hippocampus to these different aspects of
associated information stored in the brain. Thus, memory. While the adjacent MTL cortex shows
the hippocampus contributes to multiple cogni- strong category selectivity, with the anterior MTL
tive processes that require several pieces of infor- cortex responding to faces and objects and the
mation to be bound together or when currently posterior MTL cortex responding to scenes
observed information needs to be compared to (Litman et al., 2009), the hippocampus does not
information stored in memory. demonstrate a similar dissociation along its long
axis (Liang et al., 2013). Instead, the anterior hip-
pocampus shows greater activity during the
encoding of visual activity across different stimu-
Role of Hippocampus in Encoding and lus categories (faces, scenes, objects) overall
Retrieval of Recent Memories compared to the posterior hippocampus. Note that
some studies have suggested that the role of the
While some studies have focused on understand- anterior and posterior hippocampus may vary by
ing the particular mnemonic processes performed brain hemisphere. For example, studies have
by the hippocampus, such as binding and com- found that verbal memory was associated with
parison, other groups have investigated whether left-lateralized anterior hippocampus activation,
the hippocampus is more involved during particu- compared to pictoral memory which was associ-
lar task phases such as encoding (i.e., the initial ated with activity in the right posterior hippocam-
study episode) or retrieval (i.e., during the recog- pus (Persson and Söderlund, 2015).
nition or recall stage). As mentioned above, meta-
analyses (Grady, 2020; Kim, 2011) have found
that the hippocampus is more active during
memory encoding compared to memory retrieval. ROLE OF HIPPOCAMPUS OUTSIDE OF
Moreover, the anterior hippocampus was more LONG-TERM DECLARATIVE MEMORY
active during encoding than the posterior hip-
pocampus (Grady, 2020; Lepage et al., 1998).
During memory retrieval the opposite pattern has Role of Hippocampus in Spatial
been observed, where the posterior hippocampus Navigation
is more active than the anterior hippocampus. It is
currently unclear if these patterns are reflecting The discovery of “place cells”, which are cells
the relative position of the hippocampal subfields located in the CA1 and CA3 subfields that fire
along the hippocampus long-axis, or another when an animal is in a particular spatial location
organizing principle (e.g., connectivity) of these or “place field” has led to several prominent theo-
anterior and posterior hippocampal subregions ries of the role of the hippocampus in spatial
(Olsen and Robin, 2020). Empirical studies using memory and the nature of hippocampal represen-
high resolution imaging have reported conflicting tations and the formation of a “cognitive map”
results regarding which of the hippocampal sub- (O’Keefe and Nadel, 1978). While place cells
fields are more involve in memory encoding represent one important dimension of the hip-
versus memory retrieval (Hrybouski et al., 2019). pocampus’s role in cognition, it is clear that cells
Some studies have reported greater activity in DG within the hippocampus represent other types of
and CA2/3 during encoding and greater activity in information besides spatial location of the animal
the subiculum during retrieval (Eldridge et al., (Eichenbaum, 2000). Furthermore, hippocampal
2005; Suthana et al., 2015; Zeineh et al., 2003), cell firing reflects both the relevant spatial and
but this pattern has not been consistently reported nonspatial features of a particular task or experi-
in studies that have used thinner slices, which ence (Cohen and Eichenbaum, 1993; Eichenbaum
Hippocampal Formation 253

and Cohen, 2001). In fact, research has demon- Note


strated that some cells within the hippocampus are
sensitive to particular combinations of experiences 1  Some anatomists do not recognize the CA4
(e.g. encountering a particular odor or item in a region as a distinct subfield of the hippocampus,
particular location). Thus, it has been argued that and refer to this region instead as the “hillus”
the cognitive map theory of hippocampal function region of the DG.
does not account for the diversity of representa-
tions which are supported by the hippocampus.

REFERENCES
SUMMARY AND CONCLUSIONS Addis, D. R., Moscovitch, M., Crawley, A. P., &
McAndrews, M. P. (2004). Recollective qualities
The careful study of patients with hippocampal modulate hippocampal activation during autobio-
damage has set the stage for the modern neuroscien- graphical memory retrieval. Hippocampus, 14(6),
tific understanding of hippocampal function. 752–762.
Recently, the study of this complicated brain struc- Addis, D. R., Moscovitch, M., & Mcandrews, M. P.
ture using modern imaging tools such as high-reso- (2007). Consequences of hippocampal damage
lution structural and functional MRI have allowed across the autobiographical memory network
for a more thorough investigation of hippocampal in left temporal lobe epilepsy. Brain, 130,
function in humans. However, there is still consider- 2327–2342.
able uncertainty regarding (1) the particular types of Aly, M., & Turk-browne, N. B. (2016). Attention sta-
cognitive function affected by hippocampal damage bilizes representations in the human hippocam-
and (2) the contributions of specific subfields to dif- pus. Cerebral Cortex, 26, 783–796.
ferent types of memory. Thus, future research Amaral, R. S. C., Park, M. T. M., Devenyi, G. A., Lynn,
should adopt a variety of approaches, including V., Pipitone, J., Winterburn, J., … Chakravarty, M.
studies of patients and infra-human species with M. (2018). Manual segmentation of the fornix,
targeted focal lesions, using high resolution imag- fimbria, and alveus on high-resolution 3T MRI:
ing that allows for the delineation of particular brain Application via fully-automated mapping of the
subregions, as well as data driven methods that may human memory circuit white and grey matter in
uncover critical novel organization of this important healthy and pathological aging. NeuroImage. doi:
brain structure (DeKraker et al., 2021; Genon et al., 10.1016/j.neuroimage.2016.10.027
2021; Olsen and Robin, 2020). Argyropoulos, G. P., & Butler, C. R. (2020). Does hip-
pocampal atrophy explain anterograde and retro-
grade amnesia following autoimmune limbic
encephalitis? Hippocampus, 30(9), 1013–1017.
ABBREVIATION LIST Barnett, A. J., Reilly, W., Dimsdale-Zucker, H. R.,
Mizrak, E., Reagh, Z., & Ranganath, C. (2021).
Intrinsic connectivity reveals functionally distinct
cortico-hippocampal networks in the human
AChA = Anterior choroidal artery
brain. PLoS Biology, 19(6). doi: 10.1371/journal.
CA = cornu ammonis pbio.3001275
DG = dentate gyrus (also called gyrus dentatus) Bayley, P. J., Hopkins, R. O., & Squire, L. R. (2003).
ERC = entorhinal cortex Successful recollection of remote autobiographical
FD = fascia dentata (part of the dentate gyrus) memories by amnesic patients with medial tempo-
HATA = hippocampal-amygdala transition area ral lobe lesions. Neuron, 38, 135–144.
MTL = medial temporal lobe Beaujoin, J., Palomero-Gallagher, N., Boumezbeur, F.,
MRI = magnetic resonance imaging Axer, M., Bernard, J., Poupon, F., … Poupon, C.
PaS = parasubiculum (2018). Post-mortem inference of the human hip-
PCA = posterior cerebral artery pocampal connectivity and microstructure using
ultra-high field diffusion MRI at 11.7 T. Brain Struc-
PHC = parahippocampal cortex
ture and Function, 223(5), 2157–2179.
PRC = perirhinal cortex Berron, D., Schütze, H., Maass, A., Cardenas-Blanco,
PreS = presubiculum A., Kuijf, H. J., Kumaran, D., & Düzel, E. (2016).
ProS = prosubiculum Strong evidence for pattern separation in human
SRLM = stratum radiatum, lacunosum, and moleculare dentate gyrus. Journal of Neuroscience, 36(29),
Sub = subiculum 7569–7579.
254 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Blujus, J., Cole, M. W., Festa, E. K., Buka, S. L., Sal- brain with 7.0T MRI super-resolution track density
loway, S. P., Heindel, W. C., & Oh, H. (2022). imaging and track tracing. Frontiers in Neuro-
Functional redundancy of the posterior hip- anatomy, 13. doi: 10.3389/fnana.2019.00017
pocampi, but not anterior hippocampi or left Chun, M. M., & Phelps, E. A. (1999). Memory deficits
frontal cortex, is disrupted in pathological brain for implicit contextual information in amnesic sub-
aging. bioRxiv. doi: 10.1101/2022.06.18.496543 jects with hippocampal damage. Nature Neurosci-
Boccia, M., Teghil, A., & Guariglia, C. (2019). Looking ence, 2(9), 844–847.
into recent and remote past: Meta-analytic evi- Cohen, N. J., & Eichenbaum, H. (1993). Memory,
dence for cortical re-organization of episodic amnesia and the hippocampal system. Cambridge,
autobiographical memories. Neuroscience & MA: MIT Press.
Biobehavioral Reviews, 107, 84–95. Cooper, R. A., & Ritchey, M. (2019). Cortico-hip-
Borders, A. A., Ranganath, C., & Yonelinas, A. P. pocampal network connections support the multi-
(2022). The hippocampus supports high-precision dimensional quality of episodic memory. Elife, 8.
binding in visual working memory. Hippocampus, doi: 10.7554/eLife.45591
32(3), 217–230. Corkin, S. (2002). What’s new with the amnesic
Boyle, G. J., Saklofske, D. H., & Matthews, G. (2012). patient H.M.? Nature Reviews: Neuroscince, 3(2),
(Eds.), SAGE benchmarks in psychology: Psycho- 153–160.
logical assessment, Vol. 3: Clinical neuropsycho- Davachi, L. (2006). Item, context and relational epi-
logical assessment. London: Sage. sodic encoding in humans. Current Opinion in
Boyle, G. J., Stern, Y., Stein, D. J., Sahakian, B. J., Neurobiology, 16(6), 693–700.
Golden, C. J., Lee, T. M.-C., & Chen, S.-H. A. Davachi, L., Mitchell, J. P., & Wagner, A. D. (2003).
(2023a). (Eds.), The SAGE handbook of clinical Multiple routes to memory: Distinct medial tempo-
neuropsychology, Vol. 1: Clinical neuropsychologi- ral lobe processes build item and source memo-
cal disorders. London: Sage. ries. Proceedings of the National Academy of
Boyle, G. J., Stern, Y., Stein, D. J., Sahakian, B. J., Sciences of the United States of America, 100(4),
Golden, C. J., Lee, T. M.-C., & Chen, S.-H. A. 2157–2162.
(2023b). (Eds.), The SAGE handbook of clinical de Flores, R., Berron, D., Ding, S. L., Ittyerah, R.,
neuropsychology, Vol. 2: Clinical neuropsychologi- Pluta, J. B., Xie, L., … Wisse, L. E. M. (2020). Char-
cal assessment and diagnosis. London: Sage. acterization of hippocampal subfields using ex vivo
Braak, H., & Braak, E. (1991). Neuropathological MRI and histology data: Lessons for in vivo seg-
stageing of Alzheimer-related changes. Acta Neu- mentation. Hippocampus, 30(6), 545–564.
ropathologica, 82(4), 239–259. de Flores, R., La Joie, R., & Chetelat, G. (2015). Struc-
Braak, H., & Tredici, K. D. (2018). Spreading of tau tural imaging of hippocampal subfields in healthy
pathology in sporadic Alzheimer’s disease along aging and Alzheimer’s disease. Neuroscience, 309,
cortico-cortical top-down connections. Cerebral 29–50.
Cortex, 28(9), 3372–3384. DeKraker, J., Ferko, K. M., Lau, J. C., Köhler, S., &
Bubb, E. J., Kinnavane, L., & Aggleton, J. P. (2017). Khan, A. R. (2018). Unfolding the hippocampus:
Hippocampal-diencephalic-cingulate networks for An intrinsic coordinate system for subfield seg-
memory and emotion: An anatomical guide. Brain mentations and quantitative mapping. NeuroIm-
and Neuroscience Advances, 1. doi: 10.1177/ age, 167, 408–418.
2398212817723443 DeKraker, J., Köhler, S., & Khan, A. R. (2021). Sur-
Carr, V. A., Rissman, J., & Wagner, A. D. (2010). face-based hippocampal subfield segmentation.
Imaging the human medial temporal lobe with Trends in Neurosciences, 44(11), 856–863.
high-resolution fMRI. Neuron, 65(3), 298–308. Ding, S.-L., & Van Hoesen, G. W. (2015). Organiza-
Cembrowski, M. S., & Spruston, N. (2019). Heteroge- tion and detailed parcellation of human hip-
neity within classical cell types is the rule: Lessons pocampal head and body regions based on a
from hippocampal pyramidal neurons. Nature combined analysis of cyto- and chemoarchitec-
Reviews: Neuroscience, 20(4), 193–204. ture. Journal of Comparative Neurology, 523(15),
Chang, C., Huang, C., Zhou, N., Li, S. X., Ver Hoef, L., 2233–2253.
& Gao, Y. (2018). The bumps under the hippocam- Dong, H. W., Swanson, L. W., Chen, L., Fanselow, M.
pus. Human Brain Mapping, 39(1), 472–490. S., & Toga, A. W. (2009). Genomic-anatomic evi-
Chen, J., Olsen, R. K., Preston, A. R., Glover, G. H., & dence for distinct functional domains in hip-
Wagner, A. D. (2011). Associative retrieval pro- pocampal field CA1. Proceedings of the National
cesses in the human medial temporal lobe: Hip- Academy of Sciences of the United States of
pocampal retrieval success and CA1 mismatch America, 106(28), 11794–11799.
detection. Learning and Memory, 18(8), 523–528. Duvernoy, H. M., Cattin, F., Risold, P. Y., Vannson, J.
Choi, S.-H., Kim, Y.-B., Paek, S.-H., & Cho, Z.-H. L., & Gaudron, M. (2013). The human hippocam-
(2019). Papez circuit observed by in vivo human pus: Functional anatomy, vascularization and serial
Hippocampal Formation 255

sections with MRI (4th ed.) doi: 10.1007/978-3- Human Neuroscience, 6(80). doi: 10.3389/fnhum.
642-33603-4 2012.00080
Eichenbaum, H. (2000). A cortical-hippocampal Hannula, D. E., & Ranganath, C. (2009). The eyes
system for declarative memory. Nature Reviews: have it: Hippocampal activity predicts expression
Neuroscience, 1, 41–50. of memory in eye movements. Neuron, 63(5),
Eichenbaum, H., & Cohen, N. J. (2001). From condi- 592–599.
tioning to conscious recollection: Memory systems Hannula, D. E., Ryan, J. D., Tranel, D., & Cohen, N. J.
of the brain. New York: Oxford University Press. (2007). Rapid onset relational memory effects are
Eldridge, L. L., Engel, S. A., Zeineh, M. M., evident in eye movement behavior, but not in hip-
Bookheimer, S. Y., & Knowlton, B. J. (2005). A dis- pocampal amnesia. Journal of Cognitive Neurosci-
sociation of encoding and retrieval processes in ence, 19(10), 1690–1705.
the human hippocampus. Journal of Neurosci- Henke, K. (2010). A model for memory systems
ence, 25(13), 3280–3286. based on processing modes rather than conscious-
Erdem, A., Yaşargil, M. G., & Roth, P. (1993). Micro- ness. Nature Review: Neuroscience, 11, 523–532.
surgical anatomy of the hippocampal arteries. Hirshhorn, M., Grady, C., Rosenbaum, R. S., Wino-
Journal of Neurosurgery, 79(2), 256–265. cur, G., & Moscovitch, M. (2012). Brain regions
Fleming Beattie, J., Martin, R. C., Kana, R. K., Desh- involved in the retrieval of spatial and episodic
pande, H., Lee, S., Curé, J., & Ver Hoef, L. (2017). details associated with a familiar environment:
Hippocampal dentation: Structural variation and An fMRI study. Neuropsychologia, 50(13),
its association with episodic memory in healthy 3094–3106.
adults. Neuropsychologia, 101, 65–75. Hodgetts, C. J., Stefani, M., Williams, A. N., Kolarik,
Gabrieli, J. D., Corkin, S., Mickel, S. F., & Growdon, J. B. S., Yonelinas, A. P., Ekstrom, A. D., … Graham,
H. (1993). Intact acquisition and long-term reten- K. S. (2020). The role of the fornix in human navi-
tion of mirror-tracing skill in Alzheimer’s disease gational learning. Cortex, 124, 97–110.
and in global amnesia. Behavioral Neuroscience, Hrybouski, S., MacGillivray, M., Huang, Y., Madan, C.
107(6), 899–910. R., Carter, R., Seres, P., & Malykhin, N. V. (2019).
Genon, S., Bernhardt, B. C., La Joie, R., Amunts, K., Involvement of hippocampal subfields and anterior-
& Eickhoff, S. B. (2021). The many dimensions of posterior subregions in encoding and retrieval of
human hippocampal organization and (dys)func- item, spatial, and associative memories: Longitudinal
tion. Trends in Neurosciences, 44(12), 977–989. versus transverse axis. NeuroImage, 191, 568–586.
Giovanello, S. K., Schnyer, D. M., & Verfaellie, M. Insausti, R., & Amaral, D. G. (2012). Hippocampal
(2004). A critical role for the anterior hippocampus formation. In J. K. Mai & G. Paxinos (Eds.), The
in relational memory: Evidence from an fMRI study human nervous system (3rd ed., pp. 896–942). San
comparing associative and item recognition. Hip- Diego: Elsevier/Academic.
pocampus, 14(1), 5–8. Insausti, R., Muñoz-López, M., Insausti, A. M., &
Giovanello, K. S., Verfaellie, M., & Keane, M. M. Artacho-­Pérula, E. (2017). The human periallocor-
(2003). Disproportionate deficit in associative rec- tex: Layer pattern in presubiculum, parasubiculum
ognition relative to item recognition in global and entorhinal cortex: A review. Frontiers in Neuro-
amnesia. Cognitive, Affective, & Behavioral Neuro- anatomy, 11(84). doi: 10.3389/fnana.2017.00084
science, 3(3), 186–194. Kapur, N., & Brooks, D. J. (1999). Temporally-specific
Grady, C. L. (2020). Meta-analytic and functional retrograde amnesia in two cases of discrete bilat-
connectivity evidence from functional magnetic eral hippocampal pathology. Hippocampus, 9(3),
resonance imaging for an anterior to posterior 247–254.
gradient of function along the hippocampal axis. Ke, J., Foley, L. M., Hitchens, T. K., Richardson, R. M.,
Hippocampus, 30(5), 456–471. & Modo, M. (2020). Ex vivo mesoscopic diffusion
Graham, K. S., Barense, M. D., & Lee, A. C. H. (2010). MRI correlates with seizure frequency in patients
Going beyond LTM in the MTL: A synthesis of neu- with uncontrolled mesial temporal lobe epilepsy.
ropsychological and neuroimaging findings on the Human Brain Mapping, 41(16), 4529–4548.
role of the medial temporal lobe in memory and Kim, H. (2011). Neural activity that predicts subse-
perception. Neuropsychologia, 48(4), 831–853. quent memory and forgetting: A meta-analysis of
Greicius, M. D., Krasnow, B., Boyett-Anderson, J. M., 74 fMRI studies. NeuroImage, 54(3), 2446–2461.
Eliez, S., Schatzberg, A. F., Reiss, A. L., & Menon, Kirwan, C. B., & Stark, C. E. (2004). Medial temporal
V. (2003). Regional analysis of hippocampal activa- lobe activation during encoding and retrieval of novel
tion during memory encoding and retrieval: fMRI face-name pairs. Hippocampus, 14(7), 919–930.
study. Hippocampus, 13(1), 164–174. Kjelstrup, K. B., Solstad, T., Brun, V. H., Hafting, T.,
Hannula, D. E., & Greene, A. J. (2012). The hip- Leutgeb, S., Witter, M. P., Moser, E. I., & Moser, M.
pocampus reevaluated in unconscious learning B. (2008). Finite scale of spatial representation in
and memory: At a tipping point? Frontiers in the hippocampus. Science, 321, 140–143.
256 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Konkel, A., & Cohen, N. J. (2009). Relational memory destined for mild cognitive impairment. Neurobiol-
and the hippocampus: Representations and meth- ogy of Aging, 31(7), 1099–1106.
ods. Frontiers in Neuroscience, 3(2), 166–174. Masurkar, A. V, Srinivas, K. V, Brann, D. H., Warren,
Konkel, A., Warren, D. E., Duff, M. C., Tranel, D. N., R., Lowers, D. C., & Siegelbaum, S. A. (2017).
& Cohen, N. J. (2008). Hippocampal amnesia Medial and lateral entorhinal cortex differentially
impairs all manner of relational memory. Frontiers excite deep versus superficial CA1 pyramidal arti-
in Human Neuroscience, 2(15). doi: 10.3389/ cle. Cell Reports, 18(1), 148–160.
neuro.09.015.2008 Milner, B. (1970). Memory and the medial temporal
Kuhl, B. A., & Chun, M. M. (2014). Successful regions of the brain. Biology of Memory, 23,
remembering elicits event-specific activity patterns 31–59.
in lateral parietal cortex. Journal of Neuroscience, Moscovitch, M. (2008). The hippocampus as a”
34(23), 8051–8060. stupid,” domain-specific module: Implications for
Kumaran, D., & Maguire, E. (2007). Match mismatch theories of recent and remote memory, and of
processes underlie human hippocampal responses imagination. Canadian Journal of Experimental
to associative novelty. Journal of Neuroscience, Psychology, 62(1), 62–79.
27(32), 8517–8524. Moscovitch, M., & Gilboa, A. (2021). Systems con-
Langnes, E., Sneve, M. H., Sederevicius, D., Amlien, I. solidation, transformation and reorganization:
K., Walhovd, K. B., & Fjell, A. M. (2020). Anterior Multiple trace theory, trace transformation theory
and posterior hippocampus macro- and microstruc- and their competitors. PsyArXiv. doi: 10.31234/
ture across the lifespan in relation to memory: A osf.io/yxbrs
longitudinal study. Hippocampus, 30(7), 678–692. Moscovitch, M., Nadel, L., Winocur, G., Gilboa, A., &
Lepage, M., Habib, R., & Tulving, E. (1998). Hip- Rosenbaum, R. S. (2006). The cognitive neuroscience
pocampal PET activations of memory encoding of remote episodic, semantic and spatial memory.
and retrieval: The HIPER model. Hippocampus, Current Opinion in Neurobiology, 16, 179–190.
8(4), 313–322. Murty, V. P., Ritchey, M., Adcock, R. A., & LaBar, K. S.
Li, Y., Shen, M., Stockton, M. E., & Zhao, X. (2019). (2011). Reprint of: fMRI studies of successful emo-
Hippocampal deficits in neurodevelopmental dis- tional memory encoding: A quantitative meta-
orders. Neurobiology of Learning and Memory, analysis. Neuropsychologia, 49(4), 695–705.
165. doi: 10.1016/j.nlm.2018.10.001 O’Keefe, J. A., & Nadel, L. (1978). The hippocampus
Liang, J. C., Wagner, A. D., & Preston, A. R. (2013). as a cognitive map. London: Clarendon.
Content representation in the human medial tem- Olsen, R. K., Moses, S. N., Riggs, L., & Ryan, J. D.
poral lobe. Cerebral Cortex, 23(1), 80–96. (2012). The hippocampus supports multiple cogni-
Libby, L. A, Ekstrom, A. D., Ragland, J. D., & Ranga- tive processes through relational binding and
nath, C. (2012). Differential connectivity of perirhi- comparison. Frontiers in Human Neuroscience, 6.
nal and parahippocampal cortices within human doi: 10.3389/fnhum.2012.00146
hippocampal subregions revealed by high-resolu- Olsen, R. K., & Robin, J. (2020). Zooming in and
tion functional imaging. Journal of Neuroscience, zooming out: the importance of precise anatomi-
32(19), 6550–6560. cal characterization and broader network under-
Lim, C., Mufson, E. J., Kordower, J. H., Blume, H. W., standing of MRI data in human memory
Madsen, J. R., & Saper, C. B. (1997). Connections experiments. Current Opinion in Behavioral Sci-
of the hippocampal formation in humans: II. The ences, 32, 57–64.
endfolial fiber pathway. Journal of Comparative Palomero-Gallagher, N., Kedo, O., Mohlberg, H.,
Neurology, 385(3), 352–371. Zilles, K., & Amunts, K. (2020). Multimodal map-
Litman, L., Awipi, T., & Davachi, L. (2009). Category- ping and analysis of the cyto- and receptorarchi-
specificity in the human medial temporal lobe tecture of the human hippocampus. Brain
cortex. Hippocampus, 19(3), 308–319. Structure and Function, 225(3), 881–907.
Lladó, A., Tort-Merino, A., Sánchez-Valle, R., Falgàs, Parekh, M. B., Rutt, B. K., Purcell, R., Chen, Y., &
N., Balasa, M., Bosch, B., … Hornberger, M. Zeineh, M. M. (2015). Ultra-high resolution in-vivo
(2018). The hippocampal longitudinal axis-rele- 7.0T structural imaging of the human hippocam-
vance for underlying tau and TDP-43 pathology. pus reveals the endfolial pathway. NeuroImage,
Neurobiology of Aging, 70. doi: 10.1016/j. 112. doi: 10.1016/j.neuroimage.2015.02.029
neurobiolaging.2018.05.035 Perosa, V., Priester, A., Ziegler, G., Cardenas-Blanco,
Marinković, S., Milisavljević, M., & Puškaš, L. (1992). A., Dobisch, L., Spallazzi, M., … Düzel, E. (2020).
Microvascular anatomy of the hippocampal for- Hippocampal vascular reserve associated with
mation. Surgical Neurology, 37(5), 339–349. cognitive performance and hippocampal volume.
Martin, S. B., Smith, C. D., Collins, H. R., Schmitt, F. Brain, 143(2), 622–634.
A., & Gold, B. T. (2010). Evidence that volume of Persson, J., & Söderlund, H. (2015). Hippocampal
anterior medial temporal lobe is reduced in seniors hemispheric and long-axis differentiation
Hippocampal Formation 257

of stimulus content during episodic memory Hippocampal complex and retrieval of recent and
encoding and retrieval: An activation likelihood very remote autobiographical memories: Evidence
estimation meta-analysis. Hippocampus, 25(12), from functional magnetic resonance imaging in
1614–1631. neurologically intact people. Hippocampus, 11(6),
Poppenk, J., Evensmoen, H. R., Moscovitch, M., & 707–714.
Nadel, L. (2013). Long-axis specialization of the Schmidt-Kastner, R., & Freund, T. F. (1991). Selective
human hippocampus. Trends in Cognitive Sci- vulnerability of the hippocampus in brain ischemia.
ences, 17(5). doi: 10.1016/j.tics.2013.03.005 Neuroscience, 40(3), 599–636.
Prince, S. E., Daselaar, S. M., & Cabeza, R. (2005). Spallazzi, M., Dobisch, L., Becke, A., Berron, D.,
Neural correlates of relational memory: successful Stucht, D., Oeltze-Jafra, S., … Düzel, E. (2019).
encoding and retrieval of semantic and perceptual Hippocampal vascularization patterns: A high-res-
associations. Journal of Neuroscience, 25(5), olution 7 Tesla time-of-flight magnetic resonance
1203–1210. angiography study. NeuroImage: Clinical, 21. doi:
Ranganath, C., & Ritchey, M. (2012). Two cortical 10.1016/j.nicl.2018.11.019
systems for memory-guided behaviour. Nature Spiers, H. J., Maguire, E. A., & Burgess, N. (2001).
Reviews: Neuroscience, 13(10), 713–726. Hippocampal amnesia. Neurocase, 7, 357–382.
Raz, N., Gunning-Dixon, F., Head, D., Rodrigue, K. M., Squire, L. R., Clark, R. E., & Knowlton, B. J. (2001).
Williamson, A., & Acker, J. D. (2004a). Aging, sexual Retrograde amnesia. Hippocampus, 11(1), 50–55.
dimorphism, and hemispheric asymmetry of the Squire, L. R., & Zola, S. M. (1996). Structure and
cerebral cortex: Replicability of regional differences in function of declarative and nondeclarative memory
volume. Neurobiology of Aging, 25(3), 377–396. systems. Proceedings of the National Academy of
Raz, N., Rodrigue, K. M., Head, D., Kennedy, K. M., Sciences of the United States of America, 93(24),
& Acker, J. D. (2004b). Differential aging of the 13515–13522.
medial temporal lobe: A study of a five-year Strange, B. A., Witter, M. P., Lein, E. S., & Moser, E. I.
change. Neurology, 62(3), 433–438. (2014). Functional organization of the hippocam-
Richter, F. R., Cooper, R. A., Bays, P. M., & Simons, J. pal longitudinal axis. Nature Reviews: Neurosci-
S. (2016). Distinct neural mechanisms underlie the ence, 15(10), 655–669.
success, precision, and vividness of episodic Suthana, N. A., Donix, M., Wozny, D. R., Bazih, A.,
memory. Elife, 5. doi: 10.7554/eLife.18260 Jones, M., Heidemann, R. M., … Neuroscience, F.
Riggins, T., Geng, F., Botdorf, M., Canada, K., Cox, (2015). High-resolution 7T fMRI of human hip-
L., & Hancock, G. R. (2018). Protracted hippocam- pocampal subfields during associative learning. Jour-
pal development is associated with age-related nal of Cognitive Neuroscience, 27(6), 1194–1206.
improvements in memory during early childhood. Terreros-Roncal, J., Moreno-Jiménez, E. P., Flor-
NeuroImage, 174(5), 127–137. García, M., Rodríguez-Moreno, C. B., Trinchero,
Ritchey, M., & Cooper, R. A. (2020). Deconstructing M. F., Cafini, F., Rábano, A., & Llorens-Martín, M.
the posterior medial episodic network. Trends in (2021). Impact of neurodegenerative diseases on
Cognitive Sciences, 24(6), 451–465. human adult hippocampal neurogenesis. Science,
Robin, J., Buchsbaum, B. R., & Moscovitch, M. 374, 1106–1113.
(2018). The primacy of spatial context in the Tulving, E. (2002). Episodic memory: From mind to
neural representation of events. Journal of Neuro- brain. Annual Review of Psychology, 53(1), 1–25.
science, 38(11), 2755–2765. Valero, M., Menendez, L., & Prida, D. (2018). The
Robin, J., Rai, Y., Valli, M., & Olsen, R. K. (2019). hippocampus in depth: A sublayer-specific per-
Category specificity in the medial temporal lobe: A spective of entorhinal-hippocampal function. Cur-
systematic review. Hippocampus, 29(4). doi: rent Opinion in Neurobiology, 52, 107–114.
10.1002/hipo.23024 Vinogradova, O. S. (2001). Hippocampus as compara-
Rosenbaum, R. S., Stefan, K., Schacter, D. L., Mosco- tor: Role of the two input and two output systems
vitch, M., Westmacott, R., Black, S. E., … Tulving, of the hippocampus in selection and registration of
E. (2005). The case of K. C.: Contributions of a information. Hippocampus, 11(5), 578–598.
memory-impaired person to memory theory. Neu- Vogel, J. W., La Joie, R., Grothe, M. J., Diaz-Papko-
ropsychologia, 43, 989–1021. vich, A., Doyle, A., Vachon-Presseau, E., … Evans,
Ryan, J. D., Althoff, R. R., Whitlow, S., & Cohen, N. A. C. (2020). A molecular gradient along the lon-
J. (2000). Amnesia is a deficit in relational memory. gitudinal axis of the human hippocampus informs
Psychological Science, 11(6), 454–461. large-scale behavioral systems. Nature Communi-
Ryan, J. D., & Cohen, N. J. (2004). Processing and cations, 11(1). doi: 10.1038/s41467-020-14518-3
short-term retention of relational information in Vos De Wael, R., Larivière, S., Caldairou, B., Hong, S.
amnesia. Neuropsychologia, 42(4), 497–511. J., Margulies, D. S., Jefferies, E., … Bernhardt, B.
Ryan, L., Nadel, L., Keil, K., Putnam, K., Schnyer, D., C. (2018). Anatomical and microstructural deter-
Trouard, T., & Moscovitch, M. (2001). minants of hippocampal subfield functional
258 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

connectome embedding. Proceedings of the gyrus, hippocampus, and subicular complex. Jour-
National Academy of Sciences of the United States nal of Comparative Neurology, 307(3), 437–459.
of America, 115(40), 10154–10159. Woollett, K., & Maguire, E. A. (2011). Acquiring “the
Wei, P. H., Mao, Z. Q., Cong, F., Yeh, F. C., Wang, B., knowledge” of London’s layout drives structural
Ling, Z. P., … Yu, X. G. (2017). In vivo visualization brain changes. Current Biology, 21(24),
of connections among revised Papez circuit hubs 2109–2114.
using full q-space diffusion spectrum imaging Yonelinas, A. P., Aly, M., Wang, W. C., & Koen, J. D.
tractography. Neuroscience, 357, 400–410. (2010). Recollection and familiarity: Examining
Williams, A. N., Ridgeway, S., Postans, M., Graham, controversial assumptions and new directions.
K. S., Lawrence, A. D., & Hodgetts, C. J. (2020). Hippocampus, 20(11), 1178–1194.
The role of the pre-commissural fornix in episodic Zeineh, M. M., Engel, S. A., Thompson, P. M., &
autobiographical memory and simulation. Bookheimer, S. Y. (2003). Dynamics of the hip-
Neuropsychologia, 142. doi: 10.1016/j. pocampus during encoding and retrieval of face-
neuropsychologia.2020.107457 name pairs. Science, 299, 577–580.
Wisse, L. E. M., Chételat, G., Daugherty, A. M., de Zeineh, M. M., Holdsworth, S., Skare, S., Atlas, S. W.,
Flores, R., la Joie, R., Mueller, S. G., … Carr, V. A. & Bammer, R. (2012). Ultra-high resolution diffu-
(2020). Hippocampal subfield volumetry from sion tensor imaging of the microscopic pathways
structural isotropic 1 mm3 MRI scans: A note of of the medial temporal lobe. NeuroImage, 62(3),
caution. Human Brain Mapping. doi: 10.1002/ 2065–2082.
hbm.25234 Zeineh, M. M., Palomero-Gallagher, N., Axer, M.,
Witter, M. P. (2007). The perforant path: projections Gräßel, D., Goubran, M., Wree, A., … Zilles, K.
from the entorhinal cortex to the dentate gyrus. (2017). Direct visualization and mapping of the
Progress in Brain Research, 163, 43–61. spatial course of fiber tracts at microscopic resolu-
Witter, M. P., & Amaral, D. G. (1991). Entorhinal tion in the human hippocampus. Cerebral Cortex,
cortex of the monkey: V. Projections to the dentate 27(3), 1779–1794.
17
Cerebral Lateralization and
Hemispheric Specialization
Luis M. García-Marín, Zuriel Ceja, Miguel E.
Rentería, and Albert M. Galaburda

INTRODUCTION For instance, work on birds has suggested that the


brain operates as a unity, with the corollary that
The discovery of cerebral dominance that one sequential focal brain damage, irrespective of
hemisphere predominates over the other for the location, results in the gradual weakening of all
control of a given motor, sensory, perceptual, cog- functions. Nonetheless, the work of anatomists
nitive, or metacognitive function (Bear et al., 1996; has contributed by showing that underlying gyri
Finger, 2012; White et al., 1994), in turn, has given and sulci commonly are areas with distinctive
rise to the concept of cerebral lateralization, the microscopic morphology (and distinctive func-
notion that brain functions are differentially dis- tion?), which has led to the creation of architec-
tributed between the cerebral hemispheres (e.g., tonic maps, one of which (Brodmann’s) is still in
Paul Broca’s nineteenth-century identification of common use today (Vanderah and Gould, 2020).
the left-hemisphere speech motor articulation These detailed maps of the human brain have
center). Despite more than 150 years of neuroana- underscored the focally differentiated, non-uni-
tomical research (cf. Harris, 1984; Vanderah and form low-magnification microscopic anatomy of
Gould, 2020), the fundamental origins of cerebral the cerebral cortex, with areas displaying different
lateralization and its raison d’être still remain arrangements and sizes of neurons. Later, similar
essentially unknown. mapping approaches were used to show size dif-
Historically, cerebral lateralization has been ferences in several of these areas between left and
richly discussed. In early thinking, the size of each right hemispheres. From quite early with blunt
part of the brain, in gross anatomical terms, was dissection of the fixed post-mortem human brain
thought to correspond to the strength of a particu- (Heimer, 2005), to the demonstration of white
lar mental faculty – phrenology was born! It is fair matter connectivity by staining for degenerating
to say that this set of assumptions, as competing axons following damage (Talamonti et al., 2013),
theories emerged, remained viable in scientific cir- followed by improved methods for tracing axonal
cles until the second half of the twentieth century connections in the monkey (Heimer and Robards,
and, in clinical settings, even until today. At the 1981; Lanciego and Wouterlood, 2020), and cul-
very outset, there was pushback against localiza- minating with the current methods of imaging
tion from both a scientific and social perspective. tractography in living human beings, an extensive,
260 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

albeit directed and non-diffuse organization of sylvian fissure is shorter and curves upwards
fiber pathways has been highlighted. While the more sharply at its posterior end, whereas the
notion of individual focal brain areas support- left is straighter and reaches further posteriorly
ing individual functions has largely given way (Sherman and Galaburda, 1985; also see Figure
to circuits and networks composed of nodes and 17.1). In addition, the banks of the posterior por-
connections, some asymmetric but mostly distrib- tion of the sylvian fissure comprising the planum
uted between the two hemispheres (Behrens and temporale and parietal operculum, including the
Johansen-Berg, 2009; Fox, 2018; Liu et al., 2009), supramarginal gyrus, are associated with phono-
the notion that the brain works essentially as a logical encoding, speech notion, analyzing the
general-purpose computer supporting all functions amplitude and frequency of sounds (Hickok and
with the same machinery distributed throughout Poeppel, 2007).
the hemispheres has been discarded. Indeed, over The finding of a larger left planum temporale in
the past century, it has become established, for most people also corresponds to a longer left syl-
example, that the right hemisphere is dominant for vian fissure. The left planum temporale can be up
many aspects of vision and attention, as well as to ten times larger than the right. In fact, this is the
for emotional behaviors (Stone and Aybek, 2016). most striking asymmetry seen in the human brain
Research into localization and lateralization has (Amunts et al., 1996). However, the relationship
continued until now, but there has been a growing of the sylvian and planum asymmetries to lan-
emphasis on describing the anatomy and physiol- guage and handedness is not entirely straightfor-
ogy of perception and cognition in terms of coop- ward (Dorsaint-Pierre et al., 2006; Foundas et al.,
erating neural networks containing multiple and 1994). This may reflect a tendency to assume that
bilaterally located nodes and connections (Fox, all right-handers are the same and all left-handers
2018). However, examining these networks still are the same (Geschwind and Galaburda, 1985),
supports a skewed distribution between the hemi- as well as the fact that different methodologies are
spheres for some functions, with more nodes on one often employed to circumscribe and measure the
side than on the other for a given function. Although planum temporale in different studies (Kulynych
it remains undeniable that lateralized injury to the et al., 1993).
brain, whether on the left or the right side of the The planum temporale appears in higher non-
brain, leads to distinct syndromes, it is equally clear human primates and old-world monkey species,
that strict localization and lateralization of single, including baboons (Becker et al., 2022), as does
focal brain areas as responsible for discrete cogni- the sylvian asymmetry. The fact that these non-
tive function is no longer a tenable concept. human primates lack language has cast doubt
on their validity for language capacity, although
it is likely that the asymmetry may be necessary
Brain Lateralization and Hemispheric but not sufficient for language, in a majority,
Specialization although not in all humans. Thus, although the
posterior sylvian/planum temporale asymmetry
The human brain is one of the most intensely stud- is a common accompaniment of linguistic capac-
ied organs in the body and one of the least well ity in right-handers, there is variability within the
understood, reflecting its enormous complexity human population, so that some humans, more so
and individual variability. One of the main charac- left-handers, may not be organized asymmetri-
teristics of the human brain is its profound func- cally for language in the posterior temporoparietal
tional lateralization. Nevertheless, anatomically regions (Annett, 2013).
speaking, the brain appears quite symmetrical on Petalias comprise cerebral lobes that pro-
quick inspection, even as a closer look clearly trude beyond the extension of their contralateral
shows structural differences between the two hem- homologs, leading to a stronger impression on the
ispheres (Vanderah and Gould, 2020). In this sec- inner table of the skull (i.e., protrusions of por-
tion, we list some of the prominent structural brain tions of one cerebral hemisphere toward the other
asymmetries and their functional correlations (an hemisphere). Petalias have been demonstrated
exhaustive catalog of asymmetric areas and their in computerized tomography, magnetic reso-
variability is beyond the scope of this chapter). nance imagies, and endocasts of the neanderthal
man. Right frontal/left occipital petalias are more
prominent in right-handers, and also have been
Prominent Structural Brain documented in ancestral human skulls and some
Asymmetries non-human primates (Amunts et al., 1996). Such
petalias are associated with an incursion of the
Brain asymmetries have been studied for well right frontal lobe into the left side of the skull and
over a century, and findings show that the right the left occipital lobe to the right, thus conforming
Cerebral Lateralization and Hemispheric Specialization 261

Figure 17.1 Cerebral features of the fetal brain.

to the so-called Yakovlevian counter-clockwise seen in the human brain. Another issue regarding
torque. Additional studies have reported the asym- non-human brain asymmetry that is not widely
metry present in non-human primates of the equiv- discussed in the literature is that handedness,
alent of Broca’s area in the frontal operculum language lateralization, and brain asymmetry are
(Cantalupo and Hopkins, 2001). However, these not evenly distributed in the human population,
asymmetries are subtle by comparison to those with a strong bias to some patterns of asymmetry
262 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

occurring more frequently than others; such strong in ability to experience emotions, commonly pre-
population biases do not seem to be present in served among patients who have experienced right
non-human animals (see below). hemisphere trauma (Ross, 2021).
Emotional indicators include autonomic or
hypothalamic responses inducing changes in heart
Lateralization of Emotions and rate, pupillary size, sweating, nausea, changes
Cognition in neuroendocrine secretions; somatic motor
responses such as arousal and fight–flight behav-
The lateralization of emotions and cognition is of iors; changes in verbal-linguistic communication
interest in neuroscience. Based mainly on effects of arising mainly in left hemisphere; and affective-
brain injury, cognitive abilities are more dependent prosodic communication disclosing right hemi-
on intactness of the left hemisphere, whereas emo- sphere dominance (Ross, 2021). Some of these
tional integrity depends more on right hemisphere responses implicate the anterior cingulate, medial
intactness (Corballis, 2014). Two neural hypothe- prefrontal cortex, dorsal prefrontal cortex, amyg-
ses explain emotional lateralization (Table 17.1). dala, anterior insula, as well as areas associated
Right hemisphere hypothesis (RHH) refers to the with empathy and theory of mind (TOM), where
proposed right-hemisphere dominance for emotion laterality to the right hemisphere is often, but not
perception and emotional behavior, which includes consistently, observed (Hillis, 2014). Fear and
processing of social interactions, and processing of pain mechanisms, which underlie much emotional
spatial information. Such lateralization has been behavior, seem to be lateralized to the right amyg-
documented in primates, suggesting that there is an dala (Allen et al., 2021; Guadagno et al., 2020).
evolutionary continuity dating back 30–40 million One RHH limitation is the apparent symmetric
years (Lindell, 2013). RHH is supported by obser- neuroanatomy of structures including the amyg-
vations of impairment due to brain damage to the dala, hippocampus, and insula. This symmetry
right hemisphere, including perception and inter- might signify that both hemispheres participate
pretation of, and response to, emotional stimuli in different aspects of emotion, as per the valence
through diverse sensory channels, such as speech hypothesis. It has been suggested that the modu-
prosody, facial expressions, and intrinsic arousal lating influence of the right hemisphere on affec-
responses to emotional stimuli (Ross, 2021). tive-prosodic features of communication could be
However, lateralization can be violated in prosop- complementary to left hemisphere dominance for
agnosia, commonly seen with right posterior modulation of verbal-linguistic features (Ross,
infero-temporal lesions, whereby the cognitive 2010). Whether RHH or VH can explain emotional
content of familiar faces is degraded while emo- lateralization more accurately has been discussed
tional content is preserved. (Killgore and Yurgelun-Todd, 2007; Ross, 2021;
The Valence Hypothesis (VH) is based on obser- Sharma et al., 2020). Findings suggest that both
vations in patients during left and right-sided Wada theories can potentially explain different aspects
tests, and on results of frontal-lobe strokes vis-à- of a complex distributed emotion processing sys-
vis depression (Harciarek and Mańkowska, 2021). tem (Killgore and Yurgelun-Todd, 2007).
Technologies commonly used to evaluate this
hypothesis also include magnetic resonance imaging
(MRI) and neurophysiological studies. VH suggests Lateralization of Language
that there is a right hemisphere bias toward negative
emotions and a left hemisphere bias toward positive Language lateralization to the left hemisphere is
emotions (Gainotti, 2019). VH refers mostly to a one of the least disputed examples of functional
loss of emotional indicators, instead of a reduction asymmetry in the human brain, although the

Table 17.1 Right hemisphere and valence hypotheses


Emotions and cognition lateralization theories
Right Hemisphere Hypothesis (RHH) Valence Hypothesis (VH)

Description • Left hemisphere for cognitive abilities Opposite dominance:


• Right hemisphere for emotion processing, geometric • Left hemisphere for positive emotions
processing, and social interaction development • Right hemisphere for negative emotions
Supported by Observation of damage to right hemisphere Observation in patients during left and
right-sided Wada test
Author Corballis, 2014 Gainotti, 2019
Cerebral Lateralization and Hemispheric Specialization 263

origin of this common phenotype is not known. formal psycholinguistic parameters, such as gram-
Language is a late arrival in evolution, at least in mar, lexical knowledge, and pragmatic use of lan-
its human form, so its left-sided localization may guage are further analyzed separately with regard
reflect evolutionarily earlier lateralization of other to cerebral lateralization (Heine et al., 2014).
skills to the right in non-human vertebrates. For
instance, Denenberg et al. (1981) reported right-
sided lateralization for emotional behaviors in Handedness
rats, suggesting that, evolutionarily, this lateraliza-
tion preceded language lateralization. Language Handedness, also known as hand preference, is the
lateralization to the left hemisphere may have propensity to learn more easily and achieve greater
been necessitated by the previous lateralization of skill and comfort using one hand instead of the
emotions and attention to the right. On the other other in the performance of tasks such as writing,
hand, language lateralization to the left may have grasping an object, throwing a ball, or handling a
been an adaptation of an earlier lateralization to violin bow, for instance. Genetic and epigenetic
the left for conspecific vocalizations and motor factors, including cultural factors, modulate the
preference (Siniscalchi et al., 2018). expression of handedness in a given population
Initial findings related to language lateraliza- (Ocklenburg et al., 2017). Hand preferences,
tion were made in patients with brain injury as whether right, left, or ambidextrous, are not abso-
well as epileptic patients. Defined pathologically, lute categories, and instead, the degree of right-
Broca’s area usually includes the pars opercularis handedness is commonly assessed by handedness
and pars triangularis of the frontal operculum, as questionnaires such as the Edinburgh Handedness
well as the subcentral cortex, but more extensive Inventory (Schachter, 2000). Whereas the propor-
involvement is also implicated. Wernicke’s area tion of right versus left-handers varies according to
includes the planum temporale, plus various quan- the criteria used to determine handedness (as well
tities of the superior and middle temporal gyri and as according to culture, country, ethnic groups, and
the parietal operculum. Some individuals with other factors in western countries), 70% to 95% of
extensive injury to these regions, most of them people are right-handed, whereas only 5% to 30%
being left-handed, show no language deficits. of people are left-handed.
Lateralization of normal language function is Handedness occurs at the individual level (e.g.,
somewhat less striking than that documented by a person is left-handed) and at a population level
acute brain injury, whereby most contemporary (e.g., 15% of humans are left-handed). Handedness
functional studies implicate the participation of (or pawedness or clawedness) occurs in individual
the right hemisphere, even in right-handers and non-human species (Giljov et al., 2015; Ocklenburg
especially in the absence of literacy (Dehaene et al., 2013, 2014), but a population bias for hand
et al., 2010; Lecours et al., 1988). This is impor- preference is usually associated with humans and
tant because although there is evidence that hominids, although more recently, population-
lateralization reflects hardwired, biological level asymmetries in non-human primates have
underpinnings, its full expression is also a reflec- been reported (Lonsdorf and Hopkins, 2005),
tion of learning. Age alone may affect language especially favoring female right-handedness, both
lateralization, the mechanisms for which are not in captivity and in the wild (Llorente et al., 2011).
understood, whereby lateralization first increases Archaeological evidence suggests that human
in childhood, then remains stable in young adult ancestors such as Homo Heidelbergensis (Lozano
life, to gradually decrease in senescence (Brown et al., 2009) and Homo Habilis (Frayer et al., 2016)
et al., 2005; Holland et al., 2001). However, not might also have had a hand-side preference.
everyone has reported this age-related change Lateralization is a conserved feature of ver-
(Wood et al., 2004). PET, transcranial Doppler, tebrates, and recent studies suggest lateraliza-
and fMRI studies have evaluated language locali- tion can be a feature of invertebrate species, too
zation and lateralization in healthy subjects. These (Frasnelli et al., 2012). In addition to the few clues
noninvasive studies in healthy adults indicate that to lateralization’s phylogenetic history, some work
approximately 95% of right-handers display left- has been done on the ontogenetic development of
hemispheric language dominance (Knecht et al., asymmetry. For instance, there has been discus-
2000), keeping in mind the variability reported sion about the possible genetic origin of handed-
above, as well as factors such as handedness, fam- ness. Nonetheless, genome studies have failed to
ily history, and sex (Holland et al., 2001; Josse detect a simple genetic model able to explain all
and Tzourio-Mazoyer, 2004; Knecht et al., 2000; of the lateralization (Armour et al., 2014), and
Tzourio-Mazoyer et al., 2004). We anticipate that instead, it is more likely that many genes can
some of the variability in the results of language explain a fraction of the variance in handedness
lateralization studies will be resolved when more data. Some studies suggest just one-quarter of the
264 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

variance can be explained by genetic variation, However, at five gestational weeks, symmetry is
among them the androgen receptor (AR) gene disrupted as the heart, originally a straight tube in
(Arning et al., 2015), leucine-rich repeat trans- the midline loops and settles at its characteristic
membrane neuronal 1 (LRRTM1) (Francks et al., left-sided position (Schmitz et al., 2019).
2007), and proprotein convertase subtilisin/Kexin Comparable twists result in the liver becoming
type 6 (PCSK6) (Scerri et al., 2011). right-sided and the stomach left-sided by the sixth
Most of the laterality research has focused on gestational week (Schmitz et al., 2019). By seven
the cerebral cortex. This is not unreasonable, since gestational weeks, the lung divides into right and
the cortex is the brain system that differs most left buds. These buds will eventually split into two
between human and non-human animals. However, lobes on the left side of the midline and three
at least part of lateralization is learned, and a fair lobes on the right side (Schmitz et al., 2019).
amount of learning involves subcortical systems. Development of visceral asymmetries reaches
Ontogenetically, handedness starts early in devel- completion by the tenth gestational week, which
opment (at 10 gestational weeks) with 85% of marks the end of the embryonic period.
fetuses exhibiting predominance of right-hand Initially, the ectoderm gives rise to the earli-
movements and right thumb sucking in 90% at 13 est neural tissue, the neural plate, which folds
weeks (Hepper et al., 1990). Importantly, follow-up into the neural tube, a symmetric midline struc-
studies show a positive correlation between early ture. The rostral end of the neural tube grows
thumb sucking preference and later handedness ventrally, leading to the formation of the prosen-
(Hepper et al., 2005). Given that the motor cortex is cephalon, mesencephalon, and rhombencephalon
functionally linked to the spinal cord only after 15 by the sixth gestational week. The prosencepha-
weeks gestational age, it remains unlikely that the lon, in turn, gives rise to the cerebral hemispheres
cortex is the only driver of this form of lateraliza- by eight gestational weeks, and structural brain
tion (ten Donkelaar et al., 2014). Instead, it remains hemispheric asymmetries emerge after the begin-
likely that lateralization begins in ontogenesis at ning of the fetal period at 11 gestational weeks
several independent induction nodes throughout (Schmitz et al., 2019). Between 11 and 13 gesta-
the nervous system and probably under the effect tional weeks, the earliest structural hemispheric
of locally acting genes (Ocklenburg et al., 2017). asymmetry is noted: Enlargement of the left cho-
This proposed multiplex origin of lateralization can roid plexus. This asymmetry in length, area, and
increase the variability of outcome and diminish the circumference has been suggested to be a precur-
odds that lateralization is a simple binary system of sor of other hemispheric asymmetries (Corballis,
left- and right-handedness. Ocklenburg et al. (2017) 2013; Schmitz et al., 2019). In addition, at 20 ges-
proposed that some time before 10 weeks post-con- tational weeks, a leftward asymmetry emerges in
ception comprises the critical period of handedness the volume of the occipital lobes, as does for the
determination; throughout this period, asymmetrical entire left hemisphere (Schmitz et al., 2019). The
DNA methylation and posttranscriptional regulation asymmetry of the sylvian fissures becomes visible
leads to a spike in RNA expression asymmetries even before the fissure is fully closed, soon after
in the spinal segments that innervate the arms and the twentieth week of gestation, and finally, the
hands, which in turn is capable of influencing neural leftward planum temporale asymmetry, percepti-
circuit development throughout the nervous system, ble at 31 gestational weeks, is claimed to result
provoking behavioral asymmetries after 15 weeks from interhemispheric differences in cerebrospi-
when motor cortex and spinal cord are function- nal fluid production and accompanying asym-
ally connected (ten Donkelaar et al., 2014). In this metries in cytokines, growth factors, and peptides
scenario, the cerebral cortex is at least partly slav- (Schmitz et al., 2019).
ish to influences coming up from below. This is akin
to saying that cortical cognitive systems develop at
least partially in relation to inputs from the sensory
organs in a bottom-up fashion. Ciliopathies
Cilia consist of hundreds of proteins organized
FACTORS ASSOCIATED WITH BRAIN around a microtubular scaffold and have key roles
in cell locomotion, fluid movement, including the
ASYMMETRIES propulsion of seminal fluids containing sperm, and
sensory functions (Ferkol and Leigh, 2012). Cilia
Body Asymmetries are involved in the development of the entire human
organism and play an essential role in the develop-
The human body is seen to be symmetric during ing central nervous system. Notably, cilia function
early embryonic development (Figure 17.2). determines visceral asymmetry in vertebrates
Cerebral Lateralization and Hemispheric Specialization 265

Figure 17.2 Formation of body asymmetries during embryonic development.


Body symmetry is first disrupted at the heart during embryonic development, and brain
asymmetries become observable at 11 weeks.

(Trulioff et al., 2017). During embryonic develop- more detail is needed. Cilia malfunctioning can
ment, clockwise rotation of cilia creates transient induce abnormalities in left-right patterning, infer-
leftward fluid flows that contribute to left-right pat- tility, and the clearance of secretions from the lung,
terning. These flows are known to guide the asym- resulting in severe medical conditions known as
metric activation of the Nodal (TGFβ) signaling ciliopathies (Table 17.2).
pathway transforming growth factor-β (TGFβ), The Bardet-Biedl syndrome, Meckel syn-
which directs asymmetric morphogenesis in devel- drome, Joubert’s syndrome, Senior-Loken syn-
oping organs (Dasgupta and Amack, 2016). Two drome, Jeune’s syndrome, Siewert’s/Kartagener’s
hypotheses have been proposed for how nodal flow syndrome, and Alstrom syndrome are examples
is perceived by nodal and perinodal cells. The che- of ciliopathies, where genetic mutations result
mosensory hypothesis suggests that the nodal flow in abnormally formed or malfunctioning cilia
produces a gradient of left node vesicular parcels (Waters and Beales, 2011). Ciliopathies may
with hedgehog proteins, resulting in a downstream include, to a varying extent, heterotaxy, which
signaling cascade of nodal in the left-side perinodal is the abnormal arrangement of the thoracoab-
cells (Tanaka et al., 2005). In contrast, the mecha- dominal organs across the left–right axis of the
nosensory hypothesis proposes that the left nodal human body (Kim, 2011). Cilia disruption, both
flow leads to the asymmetrical increase influx of anatomical and functional, has also been seen in
Ca2+ ions in the sensory cilia cells, which in turn is association with the developmental dyslexia can-
associated with the nodal activation in the left-side didate gene DYX1C1, when its homolog, dyx1c1,
perinodal cells (McGrath et al., 2003; Tanaka et al., is deleted in the mouse, where developmental dys-
2005). Both proposals can explain asymmetric gene lexia itself is associated with brain asymmetry var-
expression and asymmetric organogenesis, although iants (Galaburda et al., 1985; Tarkar et al., 2013).
266 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Table 17.2 Ciliopathies overview

Importance Cilia plays an essential role in the central nervous system development and determines
visceral asymmetry in vertebrates (Trulioff et al., 2017).
Related growth factors The nodal (TGFβ) influences asymmetric morphogenesis in developing organs (Dasgupta
and Amack, 2016).
Hypotheses Chemosensory Hypothesis Mechanosensory Hypothesis
The nodal flow produces a gradient of left The left nodal flow leads to the asymmetrical
node vesicular parcels with hedgehog increase influx of Ca2+ ions in the sensory
proteins, resulting in a downstream cilia cells, which in turn is associated
signaling cascade of nodal in the left- with the nodal activation in the left-side
side perinodal cells (Tanaka et al., 2005). perinodal cells (McGrath et al., 2003;
Tanaka et al., 2005).
Malfunctioning • Abnormalities in left-right patterning
implications • Infertility
• Lung secretions clearance
Examples of ciliopathies • Bardet-Biedl syndrome
• Meckel syndrome
• Joubert’s syndrome
• Senior-Loken syndrome
• Jeune’s syndrome
• Siewert’s/Kartagener’s syndrome
• Alstrom syndrome
• Heterotaxy

Although cilia are distributed widely in the CNS the genetic variant is associated with brain
and characterize the morphology of fluid propel- asymmetries.
ling ependymal cells lining the ventricles, a role of GWAS have identified 27 independent lead
cilia in the formation of brain asymmetry remains SNPs across 21 genomic loci associated with
to be confirmed. different aspects of brain asymmetry (Sha et al.,
2021). These lead SNPs were also associated with
cortical surface area, cortical thickness, or sub-
Genetics and Heritability cortical volumes (Sha et al., 2021). Furthermore,
the loci associated with brain asymmetries were
Brain asymmetry is heritable, but the genetic mapped to several genes, including those respon-
foundations of human brain asymmetry remained sible for encoding proteins involved in the reor-
largely unknown until recently. Genome-wide ganization of the actin cytoskeleton and the
association studies (GWAS) have helped eluci- stabilization of microtubules (Sha et al., 2021).
date whether genomic variation in large popula- Similarly, some of these loci were mapped to
tion datasets relates to variation in adult brain genes related to brain phenotypes or develop-
asymmetry using MRI scans and genetic data. ment. For instance, ZIC4 is involved in the devel-
Although these are correlational data, they repre- opment of visual and auditory pathways, and
sent a good step toward finding the biological NR2F1 is associated with neural activity during
cause or causes of brain asymmetry. A GWAS is cortical patterning, BBS9 causes Bardet–Biedl
performed to identify regions of the genome asso- syndrome when mutated, and COL18A1 plays a
ciated with a given phenotype, in this case, brain major role in neural tube closure and is mutated in
asymmetries. Thousands of individuals with and Knobloch syndrome, potentially resulting in skull
without specific brain asymmetries are recruited abnormalities (Sha et al., 2021). The assays that
and classified as cases and controls. Then, their disclose SNP variants are, of course, hypothesis-
DNA is genotyped and analyzed for the frequency free, so it is rewarding to see the emergence of
of genetic variants in the form of single nucleo- brain-related genes in the analyses. However, as
tide polymorphisms (SNPs) across the entire can be seen from the examples listed above, there
genome. If a genetic variant is statistically more is no obvious connection between what is known
common among individuals who show brain about the functions of these genes and the emer-
asymmetry than for those who do not, taking into gence of brain asymmetry. One exception may be
account multiple comparisons, then that suggests the association with genes involved with the actin
Cerebral Lateralization and Hemispheric Specialization 267

cytoskeleton, as the latter is crucial for cilia func- genetic component, they are also considered plas-
tion, and cilia are implicated in asymmetry forma- tic since they can be reversed early in life. If
tion (see above). Additional help is obtained by functional lateralization can be switched early,
analyzing the expression patterns of these candi- what does it say about the relationship between
date genes, both spatially and temporally. In this functional lateralization and anatomical asymme-
way, for instance, genes expressed, say, only after try, which does not switch? Some growth of the
puberty could be excluded from further study. smaller side is possible from use, as suggested by
Thus, it is informative to note that by using GWAS Merzenich et al. (1996). Alternatively, the quality
results, scientists found higher mRNA expression of the recovered skill may not be as high as if
of genes related to brain asymmetry during early- there had not been any injury causing a switch, or
and mid-prenatal brain development, ranging the switch may be only partial (Buetefisch, 2015).
from 9 to 24 post-conceptional weeks (Sha et al., On the other hand, there has been disagreement
2021), a timeframe overlaping with the emergence about whether anatomical asymmetries predict
of detectable gross anatomical asymmetries in functional lateralization strongly and consistently
the human brain. That said, it would be reward- (Beaton, 1997; Foundas et al., 1994).
ing to observe that some of the asymmetry can- Twin and family studies have estimated that
didate genes are also expressed in somatic organs shared environmental factors account for up to
that also develop asymmetrically (e.g., the liver, 36% of the variance in brain asymmetry in specific
the spleen, the heart, although the mechanisms for regions (Yoon et al., 2010). What does appear to
body and brain asymmetry may be different). One be the case is that, even though anatomical asym-
reason for stating this is that, although brain areas metries are present early, it may take language
are asymmetrical in size, they are present on both experience, especially written language learning,
sides of the brain, whereas, for instance, there is to help express this laterality at the functional
no tiny right-sided spleen or a small mirror image level (Dehaene et al., 2010). Another example of
liver on the left. a putative environmental effect may be the season
Single nucleotide polymorphism (SNP) herit- of conception. For instance, more left-handers
ability describes the extent to which variance in have been found among males born during winter
brain asymmetry is linked to common genetic (Duboc et al., 2015). This phenomenon could be
variation across the entire genome (Kong et al., explained by the seasonal variation in the mother’s
2021; Sha et al., 2021). In genetic epidemiological hormones. Higher levels of androgens during fetal
studies, brain asymmetry heritability has ranged development in spring could contribute, along
from 2.2% to 24% in specific brain structures with genetic factors, to a left shift in handedness
(Kong et al., 2018, 2021; Sha et al., 2021), while (Duboc et al., 2015). However, cultural pressure
twin and family studies have estimated heritabil- has also been shown to push a shift from left to
ity for brain asymmetry measurements up to 37% right-handedness during early life (Fagard, 2013),
(Jahanshad et al., 2010), suggesting that genetic but to our knowledge, there are no examples of any
variation is partly responsible for global brain particular culture pressing a large-scale change of
asymmetry. Similarly, for velvet monkeys, global hand preference from right to left.
brain asymmetry has been reported between 10%
and 30% (Fears et al., 2011). In contrast, estimates
for the heritability of asymmetry in specific brain Sex Hormones
structures range from 21% to 32% for chimpan-
zees, with a total brain asymmetry heritability of Testosterone has been a focal point of interest
41% (Gómez-Robles et al., 2016). These findings when studying brain lateralization because it may
suggest a genetic origin behind the level of brain play an important role in handedness among
plasticity within higher primates and monkeys, males (Galaburda et al., 1985; Hampson and
which, in turn, shows that brain asymmetries are, Sankar, 2012). However, while some have impli-
at least in part, heritable, and that they did not arise cated testosterone in the shift to left-handedness
de novo with humans and are instead part of a phy- (Galaburda et al., 1985), others have observed that
logenetic history. left-handed males have lower bioavailable testos-
terone levels than right-handed males (Hampson
and Sankar, 2012). Notably, males with mixed
handedness have weaker androgen receptors;
Experiential and Environmental however, this does not seem to impact testosterone
Factors levels compared to right-handers (Hampson and
Sankar, 2012). One of the complex issues to
Although lateralization of handedness and lan- resolve relates to the exact timing during develop-
guage have been demonstrated to have a heritable ment when steroids may modulate laterality
268 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

effects and the distinction between absolute tes- adulthood. ADHD symptoms include lack of
tosterone levels and variable testosterone sensitiv- attention, concentration, focus, disorganization,
ity to a given testosterone level, which is also and sometimes hyperactivity (Magnus et al.,
controlled by the genes. 2022). A case-controlled study claimed that an
anatomical asymmetry of the caudate nucleus
might play a role in developing ADHD (Dang
BRAIN ASYMMETRIES IN BRAIN et al., 2016). Another correlational study, in an
effort to shed light on the relationship between the
DISORDERS brain’s white matter and ADHD, used diffusion
tensor imaging data to estimate the mean frac-
Neurodevelopmental Disorders tional anisotropy (FA) of 40 brain regions (Wu
et al., 2020). Among the findings, an increase in
Dyslexia leftward lateralization of the posterior thalamic
Developmental language disorders and learning radiation was associated with ADHD in children
disabilities such as dyslexia have been linked to (Wu et al., 2020). Similarly, a magnetic resonance
anatomical disruptions of brain asymmetry imaging study demonstrated that individuals with
(Altarelli et al., 2014; Galaburda et al., 1985; ADHD show increased leftward lateralization in
Leonard and Eckert, 2008). The planum temporale, the inferior orbitofrontal cortex and decreased
a region of interest in dyslexia asymmetry studies, rightward lateralization in the medial orbitofrontal
is found directly posterior to Heschl’s gyrus, which, cortex and rectus gyrus (Zou and Yang, 2021). It
in turn, contains the primary auditory cortex is not clear why these areas are implicated, as the
(Galaburda et al., 1985). The planum temporale physiology of this disorder points instead to the
overlaps, at least in part, with Wernicke’s temporo- dorsomedial, dopaminergic cortex (Brown et al.,
parietal language area. In particular, it contains the 2010). Furthermore, these same authors found dif-
auditory association cortex, which has a role in ferent asymmetry manifestations between men
representing and processing a necessary skill for and women, where the clinical manifestations also
fluent reading (Leonard and Eckert, 2008). differ.
Therefore, a structural anomaly in this region may
be expected to alter efficient phonological repre-
sentation and processing and, consequently, fluent, Autism Spectrum Disorder
efficient treading. Anatomically, the planum tem-
porale is typically larger in the left hemisphere than Disruption of standard anatomical brain asym-
the right hemisphere, and this manifestation of metries has been suggested to be one of the factors
asymmetry is altered in individuals with develop- influencing the development of autism spectrum
mental dyslexia (Altarelli et al., 2014; Galaburda disorder (ASD) (Postema et al., 2019). ASD is
et al., 1985). During the 1980s, Galaburda acquired characterized by impaired communication,
a series of postmortem brains from individuals with impaired reciprocal social interactions, and
dyslexia and reported an enlargement of the planum restricted and repetitive behaviors or interests
temporale in the right hemisphere, which abolished (Faras et al., 2010). Postema et al. (2019) mapped
the typical asymmetry of this region (Galaburda differences in brain asymmetry between individu-
et al., 1985; Leonard and Eckert, 2008). This was als with ASD and controls. They found altered
taken as an example of larger not being better, and asymmetries in seven regional cortical thickness
anti-phrenological bend. Later studies using mag- measurements among individuals with ASD, pre-
netic resonance imaging have replicated these find- dominantly involving medial frontal, orbitofron-
ings in males but not in females (Altarelli et al., tal, inferior temporal, and cingulate regions
2014). Developmental dyslexia is less common in (Postema et al., 2019), areas implicated in theory
women than in men, and when it occurs, the pathol- of mind, risk assessment and reward, and sociali-
ogy may, in fact, be different (Galaburda et al., zation. In particular, the rightward asymmetry of
1985; Humphreys et al., 1990). the medial orbitofrontal surface area was
decreased in individuals with ASD, as was the
leftward asymmetry of the lateral orbitofrontal
surface area. Also, individuals with ASD showed
Attention Deficit-hyperactivity increased leftward asymmetry of putaminal
Disorder volume (Postema et al., 2019). Understanding the
neuroanatomical biology of ASD, including brain
Attention deficit-hyperactivity disorder (ADHD) asymmetries, could potentially lead to earlier
is the most commonly reported neurodevelopmen- diagnosis, clinical subgrouping, and the develop-
tal disorder in children, which may extend into ment of individually targeted treatments.
Cerebral Lateralization and Hemispheric Specialization 269

Neurodegenerative Disorders the excess or deficiency of neurotransmitters


such as dopamine, serotonin, and glutamate
Corticobasal degeneration (Patel et al., 2014). Notably, other theories sug-
Corticobasal neurodegeneration or corticobasal syn- gest that hemispheric laterality plays a signifi-
drome, one of the degenerative brain disorders cant role in the pathophysiology of schizophrenia.
known as tauopathies, was initially considered to be For instance, it has been argued that schizophre-
strictly a motor disorder; however, it is currently nia may result from a genetically determined
categorized as a complex condition that includes failure of normal cerebral lateralization (Oertel-
sensorimotor and cognitive features (Parmera et al., Knöchel and Linden, 2011). Thus, the authors
2016). Symptoms may include language dysfunc- observed that individuals with schizophrenia are
tion, left-sided apraxia, sensory and visual neglect left-handed more often than the base rate in the
affecting the left side of space, apathy, disinhibition, general population. Magnetic resonance imaging
extrapyramidal stiffness, myoclonus, difficulty and postmortem studies of gray matter volume
swallowing, poor balance, and coordination asymmetry indicate that reduced or reversed
(Parmera et al., 2016). Corticobasal degeneration is asymmetry of the planum temporale is part of
widely known for presenting asymmetrical cortical the anatomical phenotype of schizophrenia, as is
atrophy in magnetic resonance studies, including the reduction or absence of normal right frontal
diffusion-tensor imaging, being more pronounced and left occipital petalias (Oertel-Knöchel and
in the right hemisphere (Constantinides et al., 2019). Linden, 2011). In addition, studies have reported
Volumetric studies have shown that cortical atrophy that people with schizophrenia have a higher
is primarily observed in the pericentral region, likelihood of being mixed-handed, also a devia-
including the pre- and post-central gyri and poste- tion from the standard pattern of lateralization
rior portions of the frontal lobe and anterior portions (Sommer et al., 2001). Schizophrenic individu-
of the parietal lobe (Constantinides et al., 2019). als actively experiencing hallucinations do not
show a right ear advantage to external auditory
stimuli, suggesting that auditory hallucinations
could be arising in the left hemisphere (Hugdahl
Pick’s Disease et al., 2008).
Pick’s disease, one type of frontotemporal lobar
degeneration or frontotemporal dementia, is
another example of tauopathy. It is characterized
by behavioral changes and language dysfunction Bipolar Affective Disorder
owing to frontal and temporal cortical neurode- Bipolar affective disorder is a psychiatric disorder
generation (Pippin and Gupta, 2023). The pathol- characterized by severe mood swings, including
ogy of Pick’s disease mimics to a fair degree the manic, hypomanic, and depressive episodes (Leow
clinical symptomatology, whereby when language et al., 2013). Convergent evidence suggests that
is particularly strongly affected, the left temporal abnormalities in the structural network organiza-
lobe shows greater pathology, and when behavior tion have a role in bipolar disorder. For instance,
aberrations are a prominent manifestation, the brain white matter analysis has revealed lower
pathology is worse on the right side. While the integrity in the corpus callosum among bipolar
disease is often asymmetric in presentation, there individuals (Leow et al., 2013). Similarly, the left
is much individual variability (Irwin et al., 2018). hippocampus, the left lateral orbitofrontal cortex,
and the bilateral cingulate isthmus, which are part
of the limbic system, display abnormalities in
nodal network measures in bipolar subjects (Leow
PSYCHIATRIC DISORDERS et al., 2013).
Magnetic resonance imaging studies show
Schizophrenia asymmetries of the right and left cerebral hemi-
spheres in individuals with bipolar disorder, and,
Schizophrenia is a chronic psychiatric disorder specifically, the disturbance of the right hemi-
characterized by delusions, hallucinations, dis- sphere has been mainly associated with mood
organized thought and behavior, and impaired regulation. In contrast, simultaneous disturbances
cognitive ability (Kahn et al., 2015; Patel et al., of the right and left hemispheres are most likely
2014). Most theories around the pathophysiol- related to mania, which in turn explains the coex-
ogy of schizophrenia point to abnormalities in istence of psychotic and affective symptoms in
neurotransmission, paying particular attention to bipolar disorder (Caligiuri et al., 2004).
270 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Major Depressive Disorder puzzling is why the two sides of the brain are not
simply mirror images of one another, both struc-
Major depressive disorder (MDD) is commonly turally and functionally. One of the most widely
known for persistent low mood and anhedonia, accepted theories is that our brain is asymmetric
fatigue, and sleep disturbances (APA, 2013). to conserve energy. Repetition or redundancy
Electroencephalogram measures of hemispheric across the hemispheres can be costly in an organ
asymmetry have shown abnormalities among indi- that already consumes 20% of the cardiac output.
viduals with MDD (Bruder et al., 2016). For Furthermore, more efficient packing permits a
instance, individuals at a higher risk for MDD greater supply of skills. One could also argue that
have a hemispheric asymmetry for electroen- callosal transmission is a challenging function,
cephalogram measures at rest (Bruder et al., fraught with potential errors, and should be kept at
2005). Studies have demonstrated that MDD is a minimum, thus the packing of networks as much
associated with right asymmetry in the frontal as possible within a hemisphere, but why so often
brain activity (Bruder et al., 1997). Robinson and within the same hemisphere in the species?
colleagues brought attention to the laterality of Much work has been done to document ana-
strokes and their effects on mood, with the left tomical and functional instances of normal brain/
inferior prefrontal region and right posterior mind asymmetry. Given this asymmetry, injury to
regions being particularly relevant in depression the left or right side of the brain results in differ-
causing lesions (Robinson et al., 1984; Robinson ent patterns of functional loss. Even though much
and Starkstein, 1990). of this knowledge has been acquired over the past
nearly 200 years, the fundamental underpinnings
of brain/mind asymmetry are unknown. With the
advent of newer and more powerful tools, some
Obsessive-compulsive Disorder clues are emerging as to how asymmetry origi-
Obsessive-compulsive disorder (OCD) is a psy- nates during development, but why there is a need
chiatric disorder known for the presence of repeti- for asymmetry in the first place remains unknown.
tive obsessions and persistent thoughts or urges One could argue that to build perfectly symmetri-
that are intrusive and related to anxiety (Stein cal brain regions would require additional machin-
et al., 2019). Functional laterality research for ery for comparing and controlling left and right
OCD has suggested psychometric deficits in the growth, and it is unclear what that would accom-
visual-spatial domain (Maril et al., 2007) and plish, at least for cognitive functions. Thus, sensa-
altered emotional processing (Schienle et al., tion must be roughly equal from the two sides of
2005). Both right- and left-sided dysfunction have the body, lest one side becomes more vulnerable
been observed among individuals with OCD. For to injury, but this is less clear for functions such as
instance, studies have reported greater impairment language. So, instead of precisely controlling for
in the visuospatial memory than verbal memory in symmetry, development consists of launching the
OCD (right-sided dysfunction). In contrast, the growth of areas on both sides and allowing the two
detection of left-sided dysfunction is based mainly sides to progress stochastically at their own speed
on neuropsychological and neuroimaging obser- to whatever degree. However, this would not
vations (Kong et al., 2020). explain why asymmetries have a preferred direc-
Recent work performed by the Enhancing tionality at the population level. Thus, it is not
Neuro-Imaging Genetics through Meta-Analysis asymmetry that needs an explanation, but rather
(ENIGMA) consortium used magnetic resonance the preferential asymmetry seen in the population
imaging scans to investigate alterations of brain (e.g., most people are right-handed).
asymmetry in individuals with OCD (Kong et al., It may be the case that asymmetry is inevita-
2020). Their findings highlight volume asymme- ble at the population level because chemical and
try of the thalamus and pallidum influencing OCD physical systems are inherently asymmetric in
(Kong et al., 2020). this region of the universe. Thus, electromagnetic
fields have directionality, and all planets in the
solar system move in the same direction and by
and large rotate in the same direction too; biologi-
cal molecules are of only one enantiomeric kind;
SUMMARY AND CONCLUSIONS for instance, levorotatory sugars and dextro-rota-
tory amino acids are left out of biological systems,
The fact that our species has two hemispheres is and when introduced artificially, they are inactive;
not surprising, given that all vertebrates and many cilia move in only one direction and can set up
invertebrates consist of two paired, joined struc- molecular gradients of only one asymmetric type;
tures across a vertical meridian. What is more and so on.
Cerebral Lateralization and Hemispheric Specialization 271

Perhaps we should be less interested in why handedness excludes simple genetic models.
brain systems are asymmetric and more inter- Heredity, 112(3), 221–225.
ested in those examples that appear less asym- Arning, L., Ocklenburg, S., Schulz, S., Ness, V., Gerd-
metric, such as seen in individuals with bilateral ing, W. M., Hengstler, J. G., … Beste, C. (2015).
brain representation for language and bimanual Handedness and the X chromosome: The role of
“dexterity.” It also remains of great interest as to androgen receptor CAG-repeat length. Scientific
why some patterns, including a more symmetric or Reports, 5(1). doi: 10.1038/srep08325
reverse asymmetric organization, are often associ- Bear, M. F., Connors, B. W., & Paradiso, M. A. (1996).
ated with negative functional consequences, such Neuroscience: Exploring the brain. Baltimore, MD:
as a tendency to developmental cognitive-percep- Williams & Wilkins.
tual disorders. Yet, this may be a false impres- Beaton, A. A. (1997). The relation of planum tempo-
sion. For instance, adding say five left-handers rale asymmetry and morphology of the corpus
with language problems to the whole population callosum to handedness, gender, and dyslexia: A
of left-handers is more noticeable than adding review of the evidence. Brain and Language,
five right-handers with language problems to the 60(2), 255–322.
whole population of right-handers, given that Becker, Y., Phelipon, R., Sein, J., Velly, L., Renaud, L.,
the former is a much smaller population than the & Meguerditchian, A. (2022). Planum temporale
latter, giving the impression of pathological left- grey matter volume asymmetries in newborn mon-
handedness when there is, in fact, an equal number keys (Papio anubis). Brain Structure & Function,
of pathological right-handedness. So, studies of 227(2), 463–468.
pathological association with one or another pat- Behrens, T., & Johansen-Berg, H. (2009). Preface. In
tern of laterality are confounded by the absence H. Johansen-Berg, & T. E. J. Behrens (Eds.), Diffu-
of standard and variant laterality markers. An sion MRI: From quantitative measurement to in
important step, we believe, will be to learn how vivo neuroanatomy (p. xi). Amsterdam: Elsevier/
to identify pathological left- and right-handers Academic.
and predict their deficits at an early age, better to Brown, A. B., Biederman, J., Valera, E. M., Doyle, A.
test them and implement preventive and treatment E., Bush, G., Spencer, T., … Seidman, L. J. (2010).
modalities. Effect of dopamine transporter gene (SLC6A3)
variation on dorsal anterior cingulate function in
attention-deficit/hyperactivity disorder. American
Journal of Medical Genetics: Neuropsychiatric
Genetics, 153B(2), 365–375.
REFERENCES Brown, T. T., Lugar, H. M., Coalson, R. S., Miezin, F.
M., Petersen, S. E., and Schlaggar, B. L. (2005).
Allen, H. N., Bobnar, H. J., & Kolber, B. J. (2021). Left Developmental changes in human cerebral func-
and right hemispheric lateralization of the amyg- tional organization for word generation. Cerebral
dala in pain. Progress in Neurobiology, 196. doi: Cortex, 15(3), 275–290.
10.1016/j.pneurobio.2020.101891 Bruder, G. E., Alvarenga, J., Abraham, K., Skipper, J.,
Altarelli, I., Leroy, F., Monzalvo, K., Fluss, J., Billard, Warner, V., Voyer, D., Peterson, B. S., & Weissman,
C., Dehaene-Lambertz, G., Galaburda, A. M., & M. M. (2016). Brain laterality, depression and anxi-
Ramus, F. (2014). Planum temporale asymmetry in ety disorders: New findings for emotional and
developmental dyslexia: Revisiting an old question. verbal dichotic listening in individuals at risk for
Human Brain Mapping, 35(12), 5717–5735. depression. Laterality, 21(4-6), 525–548.
Amunts, K., Schlaug, G., Schleicher, A., Steinmetz, Bruder, G. E., Fong, R., Tenke, C. E., Leite, P., Towey,
H., Dabringhaus, A., Roland, P. E., & Zilles, K. J. P., Stewart, J. E., McGrath, P. J., & Quitkin, F. M.
(1996). Asymmetry in the human motor cortex (1997). Regional brain asymmetries in major
and handedness. NeuroImage, 4(3), 216–222. depression with or without an anxiety disorder: A
Annett, M. (1998). Handedness and cerebral domi- quantitative electroencephalographic study. Bio-
nance. Journal of Neuropsychiatry and Clinical logical Psychiatry, 41(9), 939–948.
Neurosciences, 10(4), 459–469. Bruder, G. E., Tenke, C. E., Warner, V., Nomura, Y.,
Annett, M. (2013). Handedness and brain asymmetry: Grillon, C., Hille, J., Leite, P., & Weissman, M. M.
The right shift theory. London: Psychology Press. (2005). Electroencephalographic measures of
APA. (2013). Diagnostic and statistical manual of regional hemispheric activity in offspring at risk for
mental disorders (5th ed.). Washington, DC: depressive disorders. Biological Psychiatry, 57(4),
American Psychiatric Association. 328–335.
Armour, J. A. L., Davison, A., & McManus, I. C. Buetefisch, C. M. (2015). Role of the contralesional
(2014). Genome-wide association study of hemisphere in post-stroke recovery of upper
272 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

extremity motor function. Frontiers in Neurology, Faraone, S. V., Asherson, P., Banaschewski, T., Bieder-
6. doi: 10.3389/fneur.2015.00214 man, J., Buitelaar, J. K., Ramos-Quiroga, J. A., …
Caligiuri, M. P., Brown, G. G., Meloy, M. J., Eyler, L. Franke, B. (2015). Attention-deficit/hyperactivity
T., Kindermann, S. S., Eberson, S., Frank, L. R., & disorder. Nature Reviews: Disease Primers, 1. doi:
Lohr, J. B. (2004). A functional magnetic reso- 10.1038/nrdp.2015.20
nance imaging study of cortical asymmetry in Faras, H., Al Ateeqi, N., & Tidmarsh, L. (2010).
bipolar disorder. Bipolar Disorders, 6(3), 183–196. Autism spectrum disorders. Annals of Saudi Medi-
Cantalupo, C., & Hopkins, W. D. (2001). Asymmetric cine, 30(4), 295–300.
Broca’s area in great apes. Nature, 414. doi: Fears, S. C., Scheibel, K., Abaryan, Z., Lee, C., Ser-
10.1038/35107134 vice, S. K., Jorgensen, M. J., … Woods, R. P.
Clarke, E., & O’Malley, C. D. (1996). The human (2011). Anatomic brain asymmetry in vervet mon-
brain and spinal cord: A historical study illustrated keys. PloS One, 6(12). doi: 10.1371/journal.
by writings from antiquity to the twentieth cen- pone.0028243
tury. San Francisco: Norman. Ferkol, T. W., & Leigh, M. W. (2012). Ciliopathies: The
Constantinides, V. C., Paraskevas, G. P., Paraskevas, P. central role of cilia in a spectrum of pediatric dis-
G., Stefanis, L., & Kapaki, E. (2019). Corticobasal orders. Journal of Pediatrics, 160(3), 366–371.
degeneration and corticobasal syndrome: A Finger, S. (2012). Cerebral dominance. In R. W.
review. Clinical Parkinsonism & Related Disorders, Rieber (Ed.), Encyclopedia of the history of psycho-
1, 66–71. logical theories. New York: Springer.
Corballis, M. C. (2013). Early signs of brain asymme- Foundas, A. L., Leonard, C. M., Gilmore, R., Fennell,
try. Trends in Cognitive Sciences, 17(11), E., & Heilman, K. M. (1994). Planum temporale
554–555. asymmetry and language dominance. Neuropsy-
Corballis, M. C. (2014). Left brain, right brain: Facts chologia, 32(10), 1225–1231.
and fantasies. PLoS Biology, 12(1). doi: 10.1371/ Fox, M. D. (2018). Mapping symptoms to brain net-
journal.pbio.1001767 works with the human connectome. New England
Dang, L. C., Samanez-Larkin, G. R., Young, J. S., Journal of Medicine, 379(23), 2237–2245.
Cowan, R. L., Kessler, R. M., & Zald, D. H. (2016). Francks, C., Maegawa, S., Laurén, J., Abrahams, B.
Caudate asymmetry is related to attentional S., Velayos-Baeza, A., Medland, S. E., … Monaco,
impulsivity and an objective measure of ADHD-like A. P. (2007). LRRTM1 on chromosome 2p12 is a
attentional problems in healthy adults. Brain Struc- maternally suppressed gene that is associated
ture & Function, 221(1), 277–286. paternally with handedness and schizophrenia.
Dasgupta, A., & Amack, J. D. (2016). Cilia in verte- Molecular Psychiatry, 12(12), 1129–1139.
brate left-right patterning. Philosophical Transac- Frasnelli, E., Vallortigara, G., & Rogers, L. J. (2012).
tions of the Royal Society of London. Series B, Left-right asymmetries of behaviour and nervous
Biological Sciences, 371. doi: 10.1098/rstb. system in invertebrates. Neuroscience and Biobe-
2015.0410 havioral Reviews, 36(4), 1273–1291.
Dehaene, S., Pegado, F., Braga, L. W., Ventura, P., Frayer, D. W., Clarke, R. J., Fiore, I., Blumenschine, R.
Nunes Filho, G., Jobert, A., … Cohen, L. (2010). J., Pérez-Pérez, A., Martinez, L. M., … Bondioli, L.
How learning to read changes the cortical net- (2016). OH-65: The earliest evidence for right-
works for vision and language. Science, 330, handedness in the fossil record. Journal of Human
1359–1364. Evolution, 100, 65–72.
Denenberg, V. H., Rosen, G. D., Hofmann, M., Gall, Gainotti, G. (2019). The role of the right hemisphere
J., Stockler, J., & Yutzey, D. A. (1981). Neonatal in emotional and behavioral disorders of patients
postural asymmetry and sex differences in the rat. with frontotemporal lobar degeneration: An
Brain Research, 254(3), 417–419. updated review. Frontiers in Aging Neuroscience,
Dorsaint-Pierre, R., Penhune, V. B., Watkins, K. E., 11. doi: 10.3389/fnagi.2019.00055
Neelin, P., Lerch, J. P., Bouffard, M., & Zatorre, R. J. Galaburda, A. M., Sherman, G. F., Rosen, G. D.,
(2006). Asymmetries of the planum temporale and Aboitiz, F., & Geschwind, N. (1985). Developmen-
Heschl’s gyrus: Relationship to language lateraliza- tal dyslexia: Four consecutive patients with cortical
tion. Brain: Journal of Neurology, 129(5), anomalies. Annals of Neurology, 18(2), 222–233.
1164–1176. Geschwind, N., & Galaburda, A. M. (1985). Cerebral
Duboc, V., Dufourcq, P., Blader, P., & Roussigné, M. lateralization: Biological mechanisms, associations,
(2015). Asymmetry of the brain: Development and and pathology. III: A hypothesis and a program for
implications. Annual Review of Genetics, 49, research. Archives of Neurology, 42(7), 634–654.
647–672. Giljov, A., Karenina, K., Ingram, J., & Malashichev, Y.
Fagard, J. (2013). The nature and nurture of human (2015). Parallel emergence of true handedness in
infant hand preference. Annals of the New York the evolution of marsupials and placentals. Cur-
Academy of Sciences, 1288, 114–123. rent Biology, 25(14), 1878–1884.
Cerebral Lateralization and Hemispheric Specialization 273

Gómez-Robles, A., Hopkins, W. D., Schapiro, S. J., & listening study. Cognitive Neuropsychiatry, 13(2),
Sherwood, C. C. (2016). The heritability of chim- 166–179.
panzee and human brain asymmetry. Proceedings Humphreys, P., Kaufmann, W. E., & Galaburda, A. M.
of the Royal Society B: Biological Sciences, 283. (1990). Developmental dyslexia in women: neuro-
doi: 10.1098/rspb.2016.1319 pathological findings in three patients. Annals of
Guadagno, A., Verlezza, S., Long, H., Wong, T. P., & Neurology, 28(6), 727–738.
Walker, C.-D. (2020). It is all in the right amygdala: Irwin, D. J., McMillan, C. T., Xie, S. X., Rascovsky, K.,
Increased synaptic plasticity and perineuronal nets Van Deerlin, V. M., Coslett, H. B., … Grossman, M.
in male, but not female, juvenile rat pups after (2018). Asymmetry of post-mortem neuropathol-
exposure to early-life stress. Journal of Neurosci- ogy in behavioural-variant frontotemporal demen-
ence, 40(43), 8276–8291. tia. Brain: Journal of Neurology, 141(1), 288–301.
Hampson, E., & Sankar, J. S. (2012). Hand preference Jahanshad, N., Lee, A. D., Barysheva, M., McMahon,
in humans is associated with testosterone levels K. L., de Zubicaray, G. I., Martin, N. G., … Thomp-
and androgen receptor gene polymorphism. Neu- son, P. M. (2010). Genetic influences on brain
ropsychologia, 50(8), 2018–2025. asymmetry: A DTI study of 374 twins and siblings.
Harciarek, M., & Mańkowska, A. (2021). Hemispheric NeuroImage, 52(2), 455–469.
stroke: Mood disorders. Handbook of Clinical Neu- Josse, G., & Tzourio-Mazoyer, N. (2004). Hemispheric
rology, 183, 155–167. specialization for language. Brain Research
Harris, L. J. (1984). Louis Pierre Gratiolet, Paul Broca, Reviews, 44(1), 1–12.
et al. on the question of a maturational left–right Kahn, R. S., Sommer, I. E., Murray, R. M., Meyer-
gradient: Some forerunners of current-day models. Lindenberg, A., Weinberger, D. R., Cannon, T. D.,
Behavioral and Brain Sciences, 7(4), 730–731. … Insel, T. R. (2015). Schizophrenia. Nature
Heimer, L. (2005). Gross dissection of the human Reviews: Disease Primers, 1(1). doi: 10.1038/
brain. Neurosurgical Focus, 18(6), 1–2. nrdp.2015.67
Heimer, L., & Robards, M. J. (1981). Epilogue. In L. Killgore, W. D. S., & Yurgelun-Todd, D. A. (2007). The
Heimer & M. J. Robards (Eds.), Neuroanatomical right-hemisphere and valence hypotheses: Could
tract-tracing methods (pp. 539–540). Boston, MA: they both be right (and sometimes left)? Social Cog-
Springer. nitive and Affective Neuroscience, 2(3), 240–250.
Heine, B., Kuteva, T., and Kaltenböck, G. (2014). Kertesz, A., Black, S. E., Polk, M., & Howell, J. (1986).
Discourse grammar, the dual process model, and Cerebral asymmetries on magnetic resonance
brain lateralization: Some correlations. Language imaging. Cortex, 22(1), 117–127.
and Cognition, 6(1), 146–180. Kim, S.-J. (2011). Heterotaxy syndrome. Korean Cir-
Hepper, P. G., Shahidullah, S., & White, R. (1990). culation Journal, 41(5), 227–232.
Origins of fetal handedness. Nature, 347. doi: Knecht, S., Deppe, M., Dräger, B., Bobe, L., Lohm-
10.1038/347431b0 ann, H., Ringelstein, E., & Henningsen, H. (2000).
Hepper, P. G., Wells, D. L., & Lynch, C. (2005). Prena- Language lateralization in healthy right-handers.
tal thumb sucking is related to postnatal handed- Brain: Journal of Neurology, 123(1), 74–81.
ness. Neuropsychologia, 43(3), 313–315. Kong, X.-Z., Boedhoe, P. S. W., Abe, Y., Alonso, P.,
Hickok, G., & Poeppel, D. (2007). The cortical organi- Ameis, S. H., Arnold, P. D., … Francks, C. (2020).
zation of speech processing. Nature Reviews: Mapping cortical and subcortical asymmetry in
Neuroscience, 8(5), 393–402. obsessive-compulsive disorder: Findings from the
Hillis, A. E. (2014). Inability to empathize: brain ENIGMA consortium. Biological Psychiatry, 87(12),
lesions that disrupt sharing and understanding 1022–1034.
another’s emotions. Brain: Journal of Neurology, Kong, X.-Z., Mathias, S. R., Guadalupe, T., ENIGMA
137(4), 981–997. Laterality Working Group, Glahn, D. C., Franke, B.,
Holder, M. K. (2001). Why are more people right- … Francks, C. (2018). Mapping cortical brain
handed? Scientific American. http://www.scienti asymmetry in 17,141 healthy individuals world-
ficamerican.com/article/why-are-more-people- wide via the ENIGMA consortium. Proceedings of
right/ (retrieved Nov. 11, 2021) the National Academy of Sciences of the United
Holland, S. K., Plante, E., Weber Byars, A., Straws- States of America, 115(22), E5154–E5163.
burg, R. H., Schmithorst, V. J., & Ball, W. S., Jr. Kong, X.-Z., Postema, M., Schijven, D., Castillo, A.
(2001). Normal fMRI brain activation patterns in C., Pepe, A., Crivello, F., … Francks, C. (2021).
children performing a verb generation task. Neu- Large-scale phenomic and genomic analysis of
roImage, 14(4), 837–843. brain asymmetrical skew. Cerebral Cortex, 31(9),
Hugdahl, K., Løberg, E.-M., Jørgensen, H. A., Lun- 4151–4161.
dervold, A., Lund, A., Green, M. F., & Rund, B. Kulynych, J. J., Vladar, K., Jones, D. W., & Wein-
(2008). Left hemisphere lateralisation of auditory berger, D. R. (1993). Three-dimensional surface
hallucinations in schizophrenia: A dichotic rendering in MRI morphometry: A study of the
274 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

planum temporale. Journal of Computer Assisted McManus, I. C. (1985). Handedness, language domi-
Tomography, 17(4), 529–535. nance and aphasia: A genetic model. Psychologi-
Lanciego, J. L., & Wouterlood, F. G. (2020). Neuro- cal Medicine: Monograph Supplement, 8, 3–40.
anatomical tract-tracing techniques that did go Merzenich, M., Wright, B., Jenkins, W., Xerri, C., Byl,
viral. Brain Structure & Function, 225(4), N., Miller, S., & Tallal, P. (1996). Cortical plasticity
1193–1224. underlying perceptual, motor, and cognitive skill
Lecours, A. R., Mehler, J., Parente, M. A., Beltrami, M. development: Implications for neurorehabilitation.
C., Canossa de Tolipan, L., Cary, L., … Dehaut, F. Cold Spring Harbor Symposia on Quantitative Biol-
(1988). Illiteracy and brain damage. 3: A contribu- ogy, 61, 1–8.
tion to the study of speech and language disorders Ocklenburg, S., Hirnstein, M., Beste, C., & Güntürkün,
in illiterates with unilateral brain damage (initial O. (2014). Lateralization and cognitive systems. Fron-
testing). Neuropsychologia, 26(4), 575–589. tiers in Psychology, 5. doi: 10.3389/fpsyg.2014.01143
Leonard, C. M., & Eckert, M. A. (2008). Asymmetry Ocklenburg, S., Schmitz, J., Moinfar, Z., Moser, D.,
and dyslexia. Developmental Neuropsychology, Klose, R., Lor, S., … Güntürkün, O. (2017). Epige-
33(6), 663–681. netic regulation of lateralized fetal spinal gene
Leow, A., Ajilore, O., Zhan, L., Arienzo, D., GadElka- expression underlies hemispheric asymmetries.
rim, J., Zhang, A., … Altshuler, L. (2013). Impaired eLife, 6. doi: 10.7554/eLife.22784
inter-hemispheric integration in bipolar disorder Ocklenburg, S., Ströckens, F., & Güntürkün, O.
revealed using brain network analyses. Biological (2013). Lateralisation of conspecific vocalisation in
Psychiatry, 73(2), 183–193. non-human vertebrates. Laterality, 18(1), 1–31.
Lindell, A. K. (2013). Continuities in emotion laterali- Oertel-Knöchel, V., & Linden, D. E. J. (2011). Cerebral
zation in human and non-human primates. Fron- asymmetry in schizophrenia. Neuroscientist, 17(5),
tiers in Human Neuroscience, 7. doi: 10.3389/ 456–467.
fnhum.2013.00464 Parmera, J. B., Rodriguez, R. D., Studart Neto, A.,
Liu, H., Stufflebeam, S. M., Sepulcre, J., Hedden, T., Nitrini, R., & Brucki, S. M. D. (2016). Corticobasal
& Buckner, R. L. (2009). Evidence from intrinsic syndrome: A diagnostic conundrum. Dementia &
activity that asymmetry of the human brain is con- Neuropsychologia, 10(4), 267–275.
trolled by multiple factors. Proceedings of the Patel, K. R., Cherian, J., Gohil, K., & Atkinson, D.
National Academy of Sciences of the United States (2014). Schizophrenia: Overview and treatment
of America, 106(48), 20499–20503. options. Pharmacy & Therapeutics, 39(9),
Llorente, M., Riba, D., Palou, L., Carrasco, L., Mos- 638–645.
quera, M., Colell, M., & Feliu, O. (2011). Popula- Pippin, M. M., & Gupta, V. (2023). Pick disease. In
tion-level right-handedness for a coordinated StatPearls [Internet]. Treasure Island (FL):
bimanual task in naturalistic housed chimpanzees: StatPearls.
Replication and extension in 114 animals from Postema, M. C., van Rooij, D., Anagnostou, E.,
Zambia and Spain. American Journal of Primatol- Arango, C., Auzias, G., Behrmann, M., … Francks,
ogy, 73(3), 281–290. C. (2019). Altered structural brain asymmetry in
Lonsdorf, E. V., & Hopkins, W. D. (2005). Wild chim- autism spectrum disorder in a study of 54 data-
panzees show population-level handedness for sets. Nature Communications, 10(1). doi: 10.1038/
tool use. Proceedings of the National Academy of s41467-019-13005-8
Sciences of the United States of America, 102(35), Robinson, R. G., Kubos, K. L., Starr, L. B., Rao, K., &
12634–12638. Price, T. R. (1984). Mood disorders in stroke
Lozano, M., Mosquera, M., de Castro, J. M. B., patients. Importance of location of lesion. Brain:
Arsuaga, J. L., & Carbonell, E. (2009). Right hand- Journal of Neurology, 107(1), 81–93.
edness of Homo Heidelbergensis from Sima de los Robinson, R. G., & Starkstein, S. E. (1990). Current
Huesos (Atapuerca, Spain) 500,000 years ago. research in affective disorders following stroke.
Evolution and Human Behavior, 30(5), 369–376. Journal of Neuropsychiatry and Clinical Neuro-
Magnus, W., Nazir, S., Anilkumar, A. C., & Shaban, K. sciences, 2(1), 1–14.
(2022). Attention deficit hyperactivity disorder. In Rogers, L. J., Vallortigara, G., & Andrew, R. J. (2013).
StatPearls [Internet]. Treasure Island (FL): StatPearls. Divided brains: The biology and behaviour of brain
Maril, S., Hermesh, H., Gross-Isseroff, R., & Tomer, R. asymmetries. West Nyack, NY: Cambridge Univer-
(2007). Spatial attention and neural asymmetry in sity Press.
obsessive-compulsive disorder. Psychiatry Ross, E. D. (2010). Cerebral localization of functions
Research, 153(2), 189–193. and the neurology of language: Fact versus fiction
McGrath, J., Somlo, S., Makova, S., Tian, X., & or is it something else? Neuroscientist, 16(3),
Brueckner, M. (2003). Two populations of node 222–243.
monocilia initiate left-right asymmetry in the Ross, E. D. (2021). Differential hemispheric lateraliza-
mouse. Cell, 114(1), 61–73. tion of emotions and related display behaviors:
Cerebral Lateralization and Hemispheric Specialization 275

Emotion-type hypothesis. Brain Sciences, 11(8). Carson (Eds.), Handbook of clinical neurology
doi: 10.3390/brainsci11081034 (Vol. 139, pp. 213-228). Amsterdam: Elsevier.
Scerri, T. S., Brandler, W. M., Paracchini, S., Morris, A. Talamonti, G., D’Aliberti, G., Debernardi, A., & Inno-
P., Ring, S. M., Richardson, A. J., … Monaco, A. P. centi, G. (2013). Vittorio Marchi, MD (1851-1908).
(2011). PCSK6 is associated with handedness in Neurosurgery, 73(5), 887–893.
individuals with dyslexia. Human Molecular Genet- Tanaka, Y., Okada, Y., & Hirokawa, N. (2005). FGF-
ics, 20(3), 608–614. induced vesicular release of sonic hedgehog
Schachter, S. C. (2000). The quantification and defi- and retinoic acid in leftward nodal flow is criti-
nition of handedness: Implications for handedness cal for left-right determination. Nature, 435,
research. In M. K. Mandal, M. B. Bulman-Fleming 172–177.
& G. Tiwari (Eds.), Side bias: A neuropsychological Tarkar, A., Loges, N. T., Slagle, C. E., Francis, R.,
perspective (pp. 155–174). New York: Kluwer/ Dougherty, G. W., Tamayo, J. V., … Omran, H.
Academic. (2013). DYX1C1 is required for axonemal dynein
Schienle, A., Schäfer, A., Stark, R., Walter, B., & Vaitl, assembly and ciliary motility. Nature Genetics,
D. (2005). Neural responses of OCD patients 45(9), 995–1003.
towards disorder-relevant, generally disgust-induc- ten Donkelaar, H. J., Lammens, M., & Hori, A. (2014).
ing and fear-inducing pictures. International Jour- Clinical neuroembryology: Development and
nal of Psychophysiology, 57(1), 69–77. developmental disorders of the human central
Schmitz, J., Güntürkün, O., & Ocklenburg, S. (2019). nervous system (2nd ed.). Berlin: Springer-Verlag.
Building an asymmetrical brain: The molecular Trulioff, A., Ermakov, A., & Malashichev, Y. (2017).
perspective. Frontiers in Psychology, 10. doi: Primary cilia as a possible link between left-right
10.3389/fpsyg.2019.00982 asymmetry and neurodevelopmental diseases.
Sha, Z., Schijven, D., Carrion-Castillo, A., Joliot, M., Genes, 8. doi: 10.3390/genes8020048
Mazoyer, B., Fisher, S. E., Crivello, F., & Francks, C. Tzourio-Mazoyer, N., Josse, G., Crivello, F., &
(2021). The genetic architecture of structural left– Mazoyer, B. (2004). Interindividual variability in the
right asymmetry of the human brain. Nature hemispheric organization for speech. NeuroImage,
Human Behaviour, 5(9), 1226–1239. 21(1), 422–435.
Sharma, D., Gulati, R., & Misra, I. (2020). Exploring Vanderah, T. W., & Gould, D. J. (2020). Nolte’s the
consistency in right hemispheric hypothesis and human brain: An introduction to its functional
valence hypothesis for perception of emotions in anatomy (8th ed.). New York: Elsevier.
brain. Psychological Studies, 65(3), 318–326. Waters, A. M., & Beales, P. L. (2011). Ciliopathies: An
Sherman, G. F., & Galaburda, A. M. (1985). Asym- expanding disease spectrum. Pediatric Nephrol-
metries in anatomy and pathology in the rodent ogy, 26(7), 1039–1056.
brain. In S. Glick (Ed.), Cerebral lateralization in non- White, L. E., Lucas, G., Richards, A., & Purves, D.
human species (pp. 89–107). New York: Academic. (1994). Cerebral asymmetry and handedness.
Siniscalchi, M., d’Ingeo, S., Fornelli, S., & Quaranta, Nature 368, 197–198.
A. (2018). Lateralized behavior and cardiac activity Wood, A. G., Harvey, A. S., Wellard, R. M., Abbott,
of dogs in response to human emotional vocaliza- D. F., Anderson, V., Kean, M., Saling, M. M., &
tions. Scientific Reports, 8(1). doi: 10.1038/ Jackson, G. D. (2004). Language cortex activation
s41598-017-18417-4 in normal children. Neurology, 63(6), 1035–1044.
Sommer, I., Ramsey, N., Kahn, R., Aleman, A., & Wu, Z.-M., Wang, P., Yang, L., Liu, L., Sun, L., An, L.,
Bouma, A. (2001). Handedness, language laterali- … Wang, Y.-F. (2020). Altered brain white matter
sation and anatomical asymmetry in schizophre- microstructural asymmetry in children with ADHD.
nia: Meta-analysis. British Journal of Psychiatry, Psychiatry Research, 285. doi: 10.1016/j.
178(4), 344–351. psychres.2020.112817
Stein, D. J., Costa, D. L. C., Lochner, C., Miguel, E. C., Yoon, U., Fahim, C., Perusse, D., & Evans, A. C.
Janardhan Reddy, Y. C., Shavitt, R. G., van den (2010). Lateralized genetic and environmental
Heuvel, O. A., & Blair Simpson, H. (2019). Obses- influences on human brain morphology of 8-year-
sive–compulsive disorder. Nature Reviews: Disease old twins. NeuroImage, 53(3), 1117–1125.
Primers, 5(1). doi: 10.1038/s41572-019-0102-3 Zou, H., & Yang, J. (2021). Temporal variability-based
Stone, J., & Aybek, S. (2016). Functional limb weak- functional brain lateralization study in ADHD.
ness and paralysis. In M. Hallett, J. Stone & A. Journal of Attention Disorders, 25(6), 839–847.
This page intentionally left blank
PART IV
Neural Dynamics and Processes
This page intentionally left blank
18
Mirror Mechanism in Cognition
Giacomo Rizzolatti and Leonardo Fogassi

INTRODUCTION centers involved in producing motor and visceral


responses characteristic of specific emotions such
as disgust, fear, and hilarity.
The main objective of this chapter is to show how
The mirror mechanism is also underlying our
the mirror mechanism represents a basic mecha-
capacity of recognizing the forms of actions.
nism endowed with unique functional characteris-
Indeed, an action can be performed in a gentle
tics. Essentially, the mirror mechanism transforms
or in a brusque way; one can show an irrepress-
sensory representations of others’ behaviors into
ible or a slight sense of disgust; the anger can be
motor representations that the observers would
explosive or repressed. The recognition of these
use if they were to make the same behavior. As the “vitality forms,” as named by Stern, is at the basis
anatomical and functional properties of the areas of social life.
endowed with mirror mechanism can differ sig- Finally, we will expound the theory that mirror
nificantly one from another, the incoming sensory responses play a distinctive role in understanding
information and the outgoing motor representa- actions, vitality forms and emotions we observe
tions may vary. Responses recorded from the in others. In developing this idea, we will put for-
premotor areas are different from, for example, ward the notion of understanding from the inside
those of the insula or the amygdala. In spite of (Rizzolatti and Sinigaglia, 2008). This way of
these differences, all centers endowed with the understanding others is in contrast with the classi-
mirror mechanism use the motor representations cal logical-inferential way, which is used too, but
involved in a given behavior to process informa- only for actions for which there is no motor repre-
tion about similar behavior observed in other sentation (e.g., dog barking, bird flights).
people. This transformation is not related to the
merely visual aspects of actions being observed
nor to a specific sensory modality, but rather
depends primarily on the capacity of the observer
to motorically represent possible action goals. HISTORICAL PREMISE
One important discovery concerning the mir-
ror mechanism has been that it is not a preroga- The discovery of mirror neurons was published
tive of the motor areas, but is also present in for the first time in 1992 (Di Pellegrino et al.,
280 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

1992). Mirror neurons are motor neurons that areas 4 and 6, the anatomical picture of this corti-
activate both during execution of a motor act and cal sector has considerably changed. Although in
during observation of the same motor act per- the twentieth century several neuroanatomists had
formed by another individual. This discovery did already shown that the agranular frontal cortex
not come out from a completely unexplored back- can be further subdivided into a higher number of
ground but was prepared by a previous, long cytoarchitectonic areas, more recently, the com-
investigation on the cognitive properties of the bined use of architectonic and neurochemical
cortical motor system, based on neuroanatomical techniques has allowed a better definition of the
and single neuron data. For this reason, it is neces- architectonic borders between distinct areas
sary, before describing in depth the mirror neuron (Belmalih et al., 2007; Geyer et al., 2000). Here,
mechanism, to frame it within the properties of we will use the parcellation introduced by our
the motor system. neuroanatomical group, that subdivided the motor
cortex into seven different areas, named with an
“F” followed by progressive numbers (Rizzolatti
et al., 1998; Figure 18.1). In this classification,
area F1 corresponds to the primary motor cortex
CORTICAL MOTOR SYSTEM AND ITS (M1), the area from which it is possible to elicit
COGNITIVE PROPERTIES movements of all body parts with electrical micro-
stimulation at low intensity thresholds (Porter and
Since the first parcellation by Brodmann (1909) of Lemon 1995). Areas from F2 to F7 correspond to
the agranular frontal cortex of the macaque the traditional premotor cortex. Electrical micro-
monkey, who described two cytoarchitectonic stimulation shows that these areas are completely

Figure 18.1 Lateral view of the monkey brain showing the subdivisions of the agranular
frontal and posterior parietal cortices. Intraparietal and arcuate sulci are shown unfolded.
Agranular frontal areas are defined according to Matelli et al. (1985, 1991) and Belmalih
et al. (2009), frontal eye fields (FEF) according to Bruce and Goldberg (1985), and areas of
posterior parietal convexity according to Pandya and Seltzer (1982) and Gregoriou et al.
(2006). The areas buried inside the intraparietal sulcus and V6A are defined on the basis of
functional properties (see Rizzolatti et al. 1998).
AI, inferior arcuate sulcus; AIP, anterior intraparietal area; AS, superior arcuate sulcus; C, cen-
tral sulcus; DLPF, dorsolateral prefrontal cortex; IO, inferior occipital sulcus; IP, intraparietal
sulcus; L, lateral fissure; LIP, lateral intraparietal area; Lu, lunate sulcus; MIP, medial intrapari-
etal area; P, principal sulcus; ST, superior temporal sulcus; VIP, ventral intraparietal area; VLPF,
ventrolateral prefrontal cortex.
Mirror Mechanism in Cognition 281

or partially excitable. Of them, caudal premotor field (SEF) (Gerbella et al., 2010; Lu et al., 1994;
areas (F2, F3, F4, and F5) have strong anatomical Luppino et al., 1993). Concerning the connec-
connections with F1, while the rostral ones (F6 tions with posterior parietal cortex, dorsolateral
and F7) are not connected with F1, but show prefrontal cortex is connected with area V6A and
stronger connections with the other premotor PGm (Borra et al., 2019), while ventrolateral pre-
areas and with prefrontal cortex (Rizzolatti and frontal cortex is connected with inferior parietal
Luppino 2001; Rizzolatti et al., 1998). areas AIP, PFG and PG, plus second somatosen-
Similar to the agranular frontal cortex, sory area (SII) (Borra et al., 2011, 2019; Gerbella
the posterior parietal cortex, from its first et al., 2013). Altogether, these connections allow a
Brodmann’s subdivision into areas 5 and 7, also high-level control exerted by prefrontal cortex on
has been later parcellated into several cytoarchi- the parieto-premotor circuits related to reaching in
tectonic areas forming the inferior (IPL) and the space and object grasping.
superior (SPL) parietal lobules. The relevance of
these areas for the motor system derives from
their strong reciprocal connections with motor
areas. Although neurophysiological data showed
the presence, in these parietal areas, of many CODING THE GOAL OF MOTOR ACTS
sensory, unimodal, and polymodal features,
that traditionally led to the concept of “associa- One of the main functional properties demon-
tive cortex” (Critchley), the evidence of the last strated in the last decades in the premotor cortex
three decades showed that these sensory aspects is that its neurons do not mainly code movement
are mostly related to the motor control of dif- parameters, as maintained in the traditional view,
ferent effectors (Rizzolatti and Luppino 2001; but rather the goal of motor acts.1 For example,
Rizzolatti et al., 1998). Neuroanatomical find- neurons of area F5 code hand related motor acts,
ings show that each posterior parietal area has such as grasping, manipulating, tearing, holding,
a main reciprocal connection with a specific while neurons of area F4 code proximal and axial
motor area, forming a dedicated circuit; thus, motor acts such as reaching or orienting (Rizzolatti
several dedicated parieto-premotor circuits et al., 1988). This means that premotor areas store
have been described (Rizzolatti and Luppino several types of motor goals that can be then
2001; Rizzolatti et al., 1998; Rozzi et al., 2006). implemented, depending on the context, through
Broadly speaking, areas of SPL are connected their connections with F1 or through direct
with dorsal premotor cortex, while areas of IPL ­cortico-spinal or cortico-brainstem descending
are linked with ventral premotor cortex. Single pathways.
neuron studies demonstrated that in the two As already suggested by the above described
nodes of each parieto-premotor circuit there parieto-premotor connections, similar properties
are neurons sharing similar sensory and motor have also been demonstrated in the inferior pari-
properties that are exploited for the organization etal cortex, leading to a change of interpretation
of motor acts/actions performed with different of the role of this cortical sector, now to be con-
effectors such as the arm, hand and eyes. These sidered as part of the motor system (Rozzi et al.,
studies revealed two main functions of these 2008).
neuroanatomical circuits: the capacity to operate Based on these concepts and on a series of neu-
several types of sensory-motor transformations rophysiological investigations, it has been possi-
and that of coding motor cognitive properties. ble to study in depth the properties and the role of
These two functions are always strictly linked two circuits, one linking ventral intraparietal area
within the same circuit, as it will be exemplified (VIP) with premotor area F4, the other linking
below. In conclusion, these parieto-premotor cir- anterior intraparietal area (AIP) with area F5. The
cuits can be considered as the functional bases former circuit is involved in the transformation of
of the cortical motor system (Rizzolatti and the spatial location of objects into the appropri-
Luppino 2001; Rizzolatti et al., 1998). ate arm movements necessary to reach objects.
The parieto-premotor circuits have also strong Neurons of this circuit code space in a motor for-
connections with both dorsolateral and ventrolat- mat (Fogassi et al., 1996; Gentilucci et al., 1988).
eral sectors of prefrontal cortex. Generally speak- The second circuit is involved in the transfor-
ing, the connections of dorsolateral prefrontal mation of the physical properties of objects into
cortex with the agranular frontal cortex are mainly the appropriate hand motor acts, and it has been
with area F7, while those of ventrolateral pre- called the grasping circuit (Jeannerod et al., 1995;
frontal cortex are with area F5, and both prefron- Murata et al., 1987, 2000). Neurons of this circuit
tal sectors project to F6 and supplementary eye provide a “pragmatic” description of objects.
282 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

MIRROR NEURONS IN THE MONKEY motor acts having the same goal (e.g., grasping
with the hand or the mouth) (Ferrari et al., 2003).
Mirror neurons were first described in area F5 of Among the acts coded by mirror neurons, grasping
the macaque monkey (Di Pellegrino et al., 1992; is the most represented. Although most mirror
Gallese et al., 1996; Rizzolatti et al., 1996) and neurons are invariant with respect to many details
then in the inferior parietal lobule (area PFG) of observed motor acts, the original observations
(Fogassi et al., 2005; Gallese et al., 2002, Rozzi showed that some mirror neurons can be modu-
et al., 2008). The motor properties of these neu- lated by the space in which the motor act is per-
rons are not distinguishable from those of F5 formed, the type of grip used to grasp an object,
hand-related motor neurons (Rizzolatti et al., the hand of the agent (left or right) performing the
1988), thus they also code the goal of motor acts. motor act, or the direction (contra-ipsi or ipsi-
However, they also show a visual response when contra) in which the motor act is performed
the monkey observes a motor act (e.g., grasping) (Gallese et al., 1996; Rizzolatti et al., 1996).
performed by another individual (another monkey Since its first description, based also on their
or a human being) similar to that coded, motori- motor properties, it has been suggested that the
cally, by the same neuron (Figure 18.2). The visual response of mirror neurons code the goal of
observed hand motor acts more effective in pro- another’s motor act. However, it was important to
ducing mirror neurons response are grasping, assess whether this type of coding strongly depends
manipulating, holding, tearing, hands interactions. on the complete pictorial description of the motor
In later studies it has been shown that, in a sector act performed by the observed individual, as it is
of F5 lateral to that initially explored, there are for most visual neurons of inferotemporal cortex,
mirror neurons responding during execution and or whether it can also occur when the observed
observation of mouth motor acts, such as grasping, act is only partially visible. In order to directly
licking, chewing, or even of both hand-and-mouth test this experimental issue, Umiltà et al. (2001)

Figure 18.2 Example of mirror neuron discharging when the monkey grasps a piece of food
(B) and when it observes the experimenter performing the same grasping act (A). On the
right, the neuronal discharge in five trials for each condition is shown. The arrows indicate
grasping onset, on which the neuronal discharge is aligned.
Source: From Di Pellegrino et al. 1992.
Mirror Mechanism in Cognition 283

compared two conditions, one in which the mon- the congruence is based on both the goal of the
key could directly see the grasping act, the other motor act and the means used to achieve it (“strictly
in which part of grasping (hand-target interaction) congruent” mirror neurons) or mainly on the goal
was hidden behind a screen. The results showed (“broadly congruent” neurons), in both cases these
that mirror neurons show the same discharge in neurons represent a mechanism matching observed
both conditions. Interestingly, in the control con- and executed acts, since the visual (or auditory)
ditions, in which grasping was pantomimed, both description of a motor act can access the motor rep-
in full vision or hidden behind the screen, the same resentation of that motor act. This mechanism would
neurons did not discharge. This demonstrates that allow the observing individual to understand oth-
mirror neurons can code the goal of motor act even ers’ motor acts. In other words, when an individual
when part of it is not visible, likely based on the observes others’ behaviors similar to those s/he is
stored motor representation, and on the available able to produce, thanks to the “mirror mechanism”
contextual information (initial vision of the hand s/he automatically understands this behavior (see
and memory of the object presence). section on the Mirror System in humans).
A further confirmation that the main property of One could think that, during observation, the
mirror neurons is that of coding the goal of others’ retrieval of the motor act representation could lead
acts came from the study of Kohler et al. (2002), to the overt execution of that motor act, since the
showing that many mirror neurons can respond not output of these neurons could activate other neu-
only to observation of motor acts, but also to the rons directly involved in driving movement execu-
sound/noise produced by them (for example pea- tion (Mazurek and Schieber, 2019). This does not
nut breaking or noisy object manipulation). This happen, likely because of an internal mechanism
category of mirror neurons also responds to the inhibiting this automatic motor reproduction. A
sound alone. This finding confirms that through possible neural basis for this inhibitory mecha-
mirror neurons it is possible to access the meaning nism has been described by Kraskov et al. (2009),
of a motor act by using different sensory modali- who showed that half of F5 mirror neurons pro-
ties in which this act can be presented. jecting to the spinal cord had a strongly excitatory
Although the early studies carried out on mirror response during grasping execution, but an inhibi-
neurons were focused on their motor responses and tory response during grasping observation. They
on visual responses to motor acts performed with speculated that during observation of several types
biological effectors (hand, mouth), more recently of motor acts this suppression of activity could
two studies showed that these neurons can also be cause a “disfacilitation” of motor neurons.
sensitive to motor acts done with artificial effec- Recent studies showed that mirror neurons are
tors (e.g., tools). Umiltà et al. (2008) described F5 also capable to encode specific visual features of
single-neuron activity in monkeys trained to grasp the observed motor acts. A first study (Caggiano
objects by means of two different types of pliers, et al., 2009) was aimed at assessing whether the
direct and reverse. The important feature was that visual discharge of mirror neurons is influenced
the two types of pliers required an opposite pat- by the space (near or far) where the motor act is
tern of finger movements (opening-closure or vice executed by the observed agent. It has been found
versa) in order to grasp a piece of food. Very inter- that half of F5 mirror neurons had a different dis-
estingly, when tested while the monkey grasped charge depending on whether a grasping act was
food with the hand or with the pliers, motor neu- performed by an experimenter in the monkey
rons in area F5 discharged during the attainment reaching space (peripersonal space) or outside it
of the goal (grasping the object) independently (extrapersonal space), while the other half were
of the used effectors (either the hand or the pli- not modulated by the distance. Interestingly, by
ers) and of the pattern of finger movements used inserting a transparent barrier between the mon-
to achieve it (closure or opening of the fingers) key and the observed motor act, “extrapersonal”
(Figure 18.3). The same type of response was mirror neurons began to discharge also within the
also found during the observation of motor acts peripersonal space, while “peripersonal” neurons
performed with the hand and both types of pliers stopped their response, suggesting that space-
(Rochat et al., 2010). Altogether, these findings induced modulation is also plastically related to
demonstrate that after motor learning mirror neu- the operational monkey space. As a whole, these
rons can generalize their visual response, likely data suggest that the modulation of mirror neu-
relying on the training-induced expansion of the rons activity due to the distance of the observed
motor repertoire. act could be attributed to the possibility of the
The “key” property of mirror neurons is the observing individual to socially interact with the
congruence between the effective observed and the performing agent, for example to cooperate or to
effective executed motor act. Independent of whether compete.
284 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 18.3 Example of an F5 neuron active when a monkey executes a grasping act with
normal (A) and reverse (B) pliers. Rasters and histograms are aligned with the end of the
grasping closure phase of the motor act (asterisks). The curves below each histogram indi-
cate the hand position as a function of the distance between the pliers handles. When the
curve goes down, fingers close; when it goes up, fingers open. Vertical axes indicate the
measure of finger closure/opening, performed with a potentiometer. Neuron activity is
expressed in spikes per second.
Source: From Umiltà et al. 2008.

A second study (Caggiano et al., 2011) allowed, conclusion of this study, it has been proposed that
first of all, to show that F5 mirror neurons can also view-dependent neurons could either constitute an
respond to motor acts presented by means of vide- intermediate step in the formation of invariant neu-
oclips, although in a lower percentage with respect rons or could participate in encoding particular per-
to those presented in a naturalistic setting. The spectives through the feedback interaction with high
presence of a response to videoclips allowed, in a level visual areas (see below), in which pictorial
subsequent experiment, to present to the monkey descriptions of these perspectives are represented.
videos of grasping motor acts seen from different
perspectives (frontal, lateral, subjective), with the
advantage that in all trials related to a specific per-
spective the video was identical, thus avoiding the
variability intrinsic to naturalistic presentation. PARIETO-FRONTAL MIRROR NEURON
The results showed that three-quarter of mir- CIRCUIT
ror neurons were modulated by visual perspective
and that the three different perspectives were rep- Both premotor area F5 and inferior parietal area
resented, in terms of preferential discharge, by not PFG contain mirror neurons, that is neurons in
statistically different percentages of neurons. As a which both visual and motor responses are present.
Mirror Mechanism in Cognition 285

However, their visual input must be provided by and the inferior parietal area PFG are the main
high order visual areas. The main candidate for this node of the mirror neuron system. These two
role is the anterior region of the superior temporal anatomical locations are also in line with the idea
sulcus (STSa), that contains neurons responding to that the mirror mechanism is based on a motor
the presentation of biological stimuli in motion, scaffold formed by neurons coding the goal of
such as walking, head rotation, forelimb movements motor acts, well demonstrated both in F5 and
(Barraclough et al., 2006; Perrett et al., 1989). PFG. However, in the recent years, neuroana-
Among them there are also neurons discharging tomical and neurophysiological evidence showed
during the observation of goal-directed hand move- that the “grasping network,” involving neurons
ments similar to those activating mirror neurons, but related to grasping execution, is larger than previ-
devoid of any motor-related discharge. Likely, this ously thought, including further areas of premo-
visual input is sent to inferior parietal cortex, to tor and parietal cortex (Borra et al., 2017).
which the inferotemporal region is strongly con- Similarly, single neurons studies showed that
nected (Borra et al., 2008; Rozzi et al., 2006). other cortical areas contain neurons with “mirror”
The presence of mirror neuron circuits start- properties.
ing from temporal visual areas is also confirmed
by functional magnetic resonance (fMRI) studies
in monkeys. In fact, Nelissen et al., (2005; 2011)
recorded the BOLD activation of the brain of
awake monkeys trained to observe movies, show- FRONTAL CORTEX
ing grasping actions, where either the hand or the
entire agent could be seen. They found that the Two studies showed neurons that discharged
convexity of F5 (F5c) (Figure 18.1), where mirror during observation of a video, although the motor
neurons are usually recorded, was more activated act was not directly visible. Cisek and Kalaska
by the observation of the entire agent performing (2004) have described neurons of dorsal premotor
the grasping act, while areas F5a and F5p (Figure cortex (PMd) that activated both when a trained
18.1), buried in the depth of the arcuate sulcus, monkey moved a cursor to reach a target on a
activated stronger during the observation of a screen and when observed the cursor, moved by
grasping hand. Furthermore, they demonstrated the experimenter (not seen from the monkey),
that, in the inferior parietal cortex, the activation achieving the same target. They proposed that this
was stronger in area PFG during observation of visual discharge could represent a rehearsal of the
the whole person grasping an object, while area related motor act.
AIP was more activated by the view of a grasp- In another study, Tkach et al. (2007) described
ing hand. Finally, during grasping observation, neurons of MI and PMd that activated when
several areas of STS, including both upper and the monkey used a cursor to reach a target on
lower banks, were active. Thus, this study revealed a screen and when it observed the replay of the
that also AIP, an area having a strong role in the same movement. If only the cursor movement
grasping circuit, is part of the mirror system. More without the target was presented, the response
generally, these findings show that the visual input was absent. This neuronal behavior resembles the
about the observed action originating from STS differential discharge recorded in mirror neurons
can follow two different anatomo-functional path- when comparing the goal related motor act with
ways, one linking the upper lip of STS with area its pantomime.
PFG that in turn would project to area F5c, the More recently, a very original study was car-
other linking the lower bank of the STS with area ried out by Yoshida et al. (2011). They used a
AIP, then projecting to areas F5a and F5p. The first task in which two monkeys, positioned one in
pathway would be more related to the observed front of the other, alternated their action in a
agent, while the second would allow to understand common task. Single neurons recordings from
observed acts exploiting the details on the type of pre-supplementary and cingulate cortex showed
hand grip and on object meaning (Figure 18.4). the presence of neurons activated only during
own execution (“self”), during observation of
other’s movement (“other”) or in both cases.
This latter category resembles mirror neurons.
They suggested that this part of the mesial cor-
OTHER CORTICAL AREAS SHOWING tex is important for self-other distinction. A
MIRROR PROPERTIES recent study (Livi et al., 2019) carried out in
area F6 (pre-supplementary cortex), using a go/
Since their first discovery in area F5, many stud- nogo grasping execution and observation task,
ies consolidated the notion that this latter area besides showing neurons categories similar
286 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 18.4 Grasping observation circuits in the monkey brain. A. Lateral view of the
monkey brain indicating the sulci shown unfolded in (B). Black and gray arrows link areas
belonging to the STPm–PFG–F5c circuit and the LB2–AIP–F5a/p circuit. Parietal areas PF, PFG,
PG, and Opt are defined according to the parcellation of Pandya and Seltzer (1982) and
Gregoriou et al. (2006), prefrontal areas 45a and 45b according to Gerbella et al. 2010.
AIP, anterior intraparietal area; FEF, Frontal eye fields; F5c, F5 convexity; F5p, F5 (bank)
posterior; F5a, F5 (bank) anterior; FST, lower superior temporal area; IAS, inferior arcuate
sulcus; IPS/IPL, intraparietal sulcus/inferior parietal lobule; LIPa, anterior part of the lateral
intraparietal area; MSTd, middle superior temporal area, dorsal part; MT, middle temporal
area; MTp, middle temporal area, peripheral part; STPm, middle superior temporal polysen-
sory area; STS, superior temporal sulcus. LB1, LB2, UB1, UB2 are defined according to Nelissen
et al. (2011).
Source: From Nelissen et al. 2011.

to those reported by Yoshida et al., found that PARIETAL CORTEX


the “mirror-type” neurons responded to object
presentation when this latter was the target of The confirmation of the presence of neurons with
another’s actions. mirror properties in AIP came from some single
Mirror Mechanism in Cognition 287

neurons studies. Maeda et al. (2015) recorded mirror neuron mechanism. In humans, it is very
neurons from area AIP responding to videoclips of difficult, for ethical reasons, to provide a direct
the monkey own hand action and of the experi- demonstration of this mechanism at the single
menter’s action. They proposed that the activity of neuron level. The possibility to directly perform
both categories of neurons can be important for intracortical recording is permitted only in par-
monitoring the hand position based on visual ticular types of patients, such as epileptic
feedback. Lanzilotto et al. (2016) confirmed that patients, with the aim to localize the epileptic
neurons in the posterior part of AIP respond to locus. However, also in this case, the implanted
visual presentation of several types of static and probes usually allow to record electroencephalo-
dynamic hand acts. Among them, grasping is the graphic (EEG) data from a small population of
most represented, but also other acts, such as neurons close to the recording electrode. Only in
dragging, dropping, or pushing are coded. a few cases, it has been possible to record single
A different consideration must be made relative neuron activity from a brush of electrodes at the
to possible mirror properties in prefrontal cortex. tip of the probe (see below) (Mukamel et al.,
It is well known that visual responses in prefrontal 2011).
cortex are often recorded when they are linked to The above considerations explain why the mir-
a subsequent behavioral reaction, although there ror mechanism in humans has been studied and
are also neurons activated by passive visual stimu- demonstrated using electrophysiological (TMS,
lation, such as presentation of faces (Romanski EEG, MEG) and neuroimaging (PET, fMRI) tech-
and Diehl, 2011; Scalaidhe et al., 1997). Simone niques that reveal the activity of populations of
et al. (2017) recording from ventrolateral prefron- neurons.
tal cortex, a cortical sector strongly connected The first publication demonstrating that the
with inferior parietal areas PFG and AIP and with motor cortex activates during observation of motor
infero-temporal, found neurons specific for the acts was provided by a transcranial magnetic stim-
presentation of videos of goal-related motor acts ulation (TMS) study by Fadiga et al. (1995). They
performed by another monkey or a human agent, applied single pulse magnetic stimulation at low
as compared to non goal-related biological move- threshold to the hand and arm representation of
ment or object motion. Interestingly, these neu- the motor cortex of participants, who were asked
rons were also activated when part of the video to attentively observe an experimenter grasping an
was obscured, suggesting a high order represen- object or, as control, the same experimenter per-
tation of the observed action. Although many forming meaningless arm movements in the air.
of these neurons did not show a corresponding The results showed a specific enhancement, dur-
motor response, they have been found in sectors in ing grasping, with respect to control conditions,
which neurons activated during action execution of the EMG activity (motor evoked potentials,
(Simone et al., 2015). Thus, it is possible that the MEPs) of extrinsic and intrinsic hand muscles
response to observation of goal-related motor acts normally used to execute the observed motor act.
is exploited for the planning of behavioral reac- Furthermore, it was also observed an enhanced
tions in a social context. activity during observation of meaningless arm
Altogether, all these new data on frontal and movements. The same pattern of activation was
parietal areas suggest the existence of a network, found when the observing participants executed
more extended than the core circuit of the classi- the same hand and arm movements. These find-
cal mirror neuron system, composed by areas that ings suggest that the motor representations of
can exploit their response to obervation of motor hand motor acts are activated in the observer
acts/actions for several roles, from those contrib- during observation of the same acts, thus indi-
uting the details necessary for the recognition of cating the presence, also in humans, of a mirror
the acting hand to those allowing the distinction mechanism. Furthermore, the activation of motor
between the self and the other or that exploit social cortex during observation of meaningless move-
information for planning appropriate responsive ments suggested that in humans, differently from
behavior. monkeys, also non-goal directed movements can
be recognised through a similar matching mecha-
nism. In a subsequent TMS study, Gangitano et al.
(2004) stimulated the hand motor field at different
time points of the observed movements (e.g., hand
HUMAN MIRROR SYSTEM starting movement, finger opening, finger closure,
etc.), showing that the pattern of cortical activa-
Single neurons recording studies in monkeys tion was congruent with the timing of the observed
demonstrated the existence of the parieto-frontal finger movements.
288 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

A further TMS study by Cattaneo et al. (2009) three nodes are homologues of the monkey areas
compared the MEPs produced in participants responding to observed motor acts. Specifically,
observing grasping an object or closure-opening the STS sector corresponds to the monkey area
without object, executed by means of normal containing neurons responding during observa-
and reversed pliers (cf. monkey study by Umiltà tion of biological movements but devoid of motor
et al., 2008). The TMS pulse was given above properties (Perrett et al., 1989). The IPL sectors
the field of representation of the opponens pol- correspond to area PFG, while the frontal nodes
licis. The results showed that during observation correspond to F5 region. These studies provided
of goal-directed grasping, MEPs were enhanced a good confirmation of the phylogenetic origin of
when both types of pliers took possession of the the human mirror neuron system from the monkey
object, independently of the movement performed one. However, most of them were limited to the
(extension with the inverted pliers, flexion with observation of hand and mouth motor acts. On the
the normal pliers). Yet, during observation of non- other hand, the mirror neurons described in mon-
goal directed movements, MEP activation always keys are mainly related to these latter effectors. In
occurred during observation of finger closure, most recent years the investigation also extended
independently of the type of pliers. Thus, the for- to other effectors.
mer activation was due to the goal of the observed
movements.
Although the TMS technique clearly reveals
the presence of the mirror mechanism likely
underpinned by mirror neurons, it does not pro- SOMATOTOPY OF OBSERVED ACTS
vide the precise anatomical location of the motor
sectors that are directly activated by observation. One of the first studies trying to assess which
Since 1996, several positron emission tomography motor sectors are activated by the observation of
(PET) and fMRI studies showed that, during obser- acts performed with different effectors is that of
vation of goal-directed motor acts there is always Buccino et al. (2001). Participants to this study
an activation of three main cortical nodes: a sec- had to observe goal-related motor acts performed
tor around the STS, the supramarginal gyrus (part with the mouth (e.g., biting an apple), the hand
of IPL), and the ventral premotor cortex/posterior (e.g., grasping a glass), and the leg (e.g., kicking a
sector of the inferior frontal gyrus (IFG) (Frey ball). The results showed a somatotopic activation
and Gerry, 2006; Grafton et al., 1996; Iacoboni of premotor and parietal cortices. Observation of
et al., 1999; Koski et al., 2003; cf. Caspers et al., mouth motor acts activated IFG and the lateral-
2010; Rizzolatti et al., 2014; Figure 18.5). These most part of ventral premotor cortex, that of hand

Figure 18.5 Lateral view of the two hemispheres of the human brain, in which the areas
usually activated by action observation, as a result of a metanalysis, are indicated. The main
activations are in a sector of the superior temporal sulcus (pMTG-V5), in the inferior parietal
lobule (hIP3 and PFt) plus a small part of the superior parietal lobule (7A); in the ventral pre-
motor cortex (BA6), plus the posterior part of the inferior frontal gyrus (BA44-45).
Source: From Caspers et al. (2010).
Mirror Mechanism in Cognition 289

motor acts activated a most medial sector of ven- system in both conditions, in spite of the differ-
tral premotor cortex, while observation of feet ence in shape and kinematics of the two effectors,
motor acts activated dorsal premotor cortex. This suggesting that this system decodes more strongly
activation resembled the Penfield’s motor homun- the achieved aim of the action rather than the
culus, indicating that motor cortex mirrors the details of the effector. Additional evidence in
observed motor acts also in terms of effectors. A favour of goal coding is provided by an experi-
somatotopic activation was also observed in the ment carried out on two aplasic patients and
posterior parietal cortex, following a rostro-caudal healthy control subjects who observed videoclips
direction (mouth represented more rostrally, fol- showing hand grasping actions. The two aplasic
lowed by hand and foot), but in this case a higher individuals also made actions with their feet and
overlap between fields of adjacent effectors was mouth. It was found that in the aplasic individuals
observed. the parieto-frontal mirror circuit activated during
Other studies, focusing on reaching movements observation of hand grasping acts. These activa-
or on movements of several body parts, including tions overlapped with the premotor and the pari-
the trunk and the leg, substantially confirm the etal sectors active during the execution of foot and
somatotopic concept derived from Buccino et al. mouth movements. Since these patients had never
(2001). Filimon et al. (2007) asked participants executed the observed hand actions, the matching
to execute, imagine, and observe reaching move- between observation and execution was not based
ments. Except for the areas that are only typical of on the effector, but on the common action goal
a specific condition (e.g., the primary motor cortex (Gazzola et al., 2007).
in the execution condition) in all conditions there
was an activation of a dorsal cortical network,
including dorsal premotor cortex and superior
parietal lobule. Calvo-Merino et al. (2005) asked
participants expert in two categories of dance ELECTROPHYSIOLOGICAL STUDIES
(classic ballet or capoeira) to observe videoclips
showing steps of the two types of dances. Naïve A further interesting issue concerns the time
subjects were the control group. The results show course of cortical activation during action obser-
that, during observation, all groups of subjects vation. This issue can be better addressed using
presented an activation of a temporo-parieto- EEG and magnetoencephalographic (MEG) tech-
frontal circuit, but the activation was higher in niques. Using these techniques, independently,
dancers (expertise effect). In particular, the activa- Altschuler et al. (1997) and Cochin et al. (1999)
tion included ventral and dorsal premotor cortex, confirmed that during both action observation and
inferior and superior parietal lobule and posterior action execution there is an activation of motor
STS. It is very likely that the activation of different cortex, as shown by a desynchronization of the μ
premotor and parietal areas reflected the observa- rhythm, while Nishitani and Hari (2000) showed
tion of movements of different effectors. A further that during action observation the activation of the
study supporting the concept of somatotopy is that IFG followed that of visual cortex, and preceded
of Abdollahi et al. (2013), who asked subjects to that of primary motor cortex.
observe manipulation, locomotion, and climbing. In humans, ethical reasons do not allow a direct
They found that while the regions of overlapping demonstration of the existence of mirror neurons
activation are restricted, observation of manipula- at the single neuron level. Some studies, however,
tion tended to activate more area AIP, while obser- verified the overlap between activations occurring
vation of climbing activated more the rostral part during observation and execution, by applying
of the dorsal superior parietal lobule. single-voxel analysis, demonstrating the activa-
tion of the same voxels in the two conditions in
single participants (Gazzola et al., 2006).
Despite the difficulty to record single neurons
in humans, Mukamel et al. (2010), using a special
CODING OF THE GOAL probe, were able to record single neurons from
epileptic patients required to observe and execute
Several evidence point to the most relevant role of reaching-grasping motor acts and facial expres-
the human mirror system, as that of the monkey, sions. The recorded areas were mesial cortical
in representing the action goal. Firstly, in the structures, namely pre-supplementary (pre-SMA)
study by Gazzola et al. (2007), participants were and supplementary (SMA) motor cortex, anterior
presented with videos of grasping executed by a cingulate cortex, hippocampus, parahippocam-
human or a robot hand. The data showed a very pal cortex, and entorhinal cortex. They found,
similar activation of the parieto-frontal mirror in SMA and hippocampus, neurons responding
290 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

during both observation and execution, some of Associative sensorimotor learning is consid-
them being excited during execution and inhibited ered, by some researchers (Heyes et al., 2010), to
during observation. Areas on the medial wall such be fundamental for the creation of mirror neurons,
as SMA seem relevant to movement initiation and although they suggest that it is not necessary to
movement sequences, while neurons recorded in postulate that the mirror mechanism is genetically
hippocampus could be involved in the memory of coded. Another hypothesis, the epigenetic one
the action formed during action execution. (Ferrari et al., 2013), proposes that the develop-
ment of mirror neurons may involve both genetic
and epigenetic factors. In this hypothesis, visual
stimuli provided by the behavior of others, i.e.
social stimuli, would activate a specific population
PHYLOGENESIS AND DEVELOPMENT OF of visuomotor neurons. This specificity would
THE MIRROR MECHANISM have been mediated by epigenetic mechanisms
giving advantages to the individuals possessing
The existence of a mirror mechanism has also them, and natural selection would have then led to
been demonstrated, with electrophysiological and their stabilization.
neuroimaging techniques, in children (Lepage & Independently of which is the contribution of
Theoret, 2007; Nystrom, 2008; Nystrom et al., phylogenetic and ontogenetic mechanism, it is
2011; Marshall et al., 2011) as early as six months very clear, from both monkeys and humans stud-
old (Shimada and Iraki, 2006). Thus, it is very ies, that the mirror neuron system, although keep-
likely that this mechanism, as a product of primate ing its main properties, can be plastically modified
evolution, is already present at birth. An indirect by experience, that allows an extension of the
confirmation of this idea is provided by neonatal individual’s motor repertoire (Calvo-Merino et al.,
imitation, a phenomenon initially demonstrated by 2005; Umiltà et al., 2008).
Meltzoff and Moore (1989) in human newborns a
few hours after birth and then found also in apes
(Myowa, 1996; Myowa-Yamakoshi et al., 2004) ROLE OF SOMATOSENSORY CORTICES IN
and monkeys (Ferrari et al., 2006 ). It consists in
the replication, by the newborn, of the facial ges- MIRRORING SOMATOSENSATION
tures – mostly communicative – performed by an
adult in front of her/him. This phenomenon is very Beyond the classical activation of mirror neurons
likely related to a hard-wired mirror mechanism, during observation of others’ goal-directed motor
matching the observed gestures on their motor acts, there is also evidence of mirror activity when
representations. Interestingly, by recording EEG individuals observe movements that produce, on
activity from 1 to 7 day old infant macaques others, tactile sensation. On the side of single
during imitation of facial gestures performed by neuron studies, Ishida et al. (2010), recording
the experimenter, Ferrari et al. (2012) could dem- from VIP, an area receiving both visual and soma-
onstrate a desynchronization of the alpha rhythm tosensory input, recorded neurons responding
from central electrodes. Note that this desynchro- when a monkey was touched (e.g., on the arm)
nization typically occurs in adults and children and when it could see an experimenter stroking
during action observation (Muthukumaraswamy the corresponding body part. They suggested that
et al., 2004; Lepage and Theoret, 2007). these neurons could contribute to the perception
However, these data do not provide direct infor- of others’ body parts using their own body as
mation on the formation of the mirror mechanism reference.
and whether this latter is influenced by experi- In human studies, there is evidence of activa-
ence. Some theoretical models tried to address tion of somatosensory cortices during observation
these issues. According to Keysers et al. (2014), of touch. For example, in an fMRI experiment
hebbian learning would allow, first of all, to cre- (Keysers et al., 2004), participants received a tac-
ate, in the baby, a coupling between premotor and tile stimulus on the legs and observed the tactile
STS neurons, thanks to the temporal contingency stimulation of others’ legs or, as control, of an
between own action execution and the visual or object. The tactile condition produced activation
acoustic re-afference of the executed action. Then, of SI and SII, while the observation of touch pro-
by means of generalization, the same visual/acous- duced activation of SII. In another experiment
tic afference produced by another’s action would (Ebish et al., 2008), observations of intentional
activate the same STS-premotor potentiated con- and accidental touch were compared. The study
nection, thus originating the mirror function (cf. confirmed an overlapping activation between the
Casile et al., 2011). experience of being touched and that of observing
Mirror Mechanism in Cognition 291

others being touched, in bilateral SII. A difference participants were asked to observe and execute
of activation between the intentional and acciden- manipulative actions, generally confirming the
tal condition was found in SI, suggesting a higher subcortical activation reported in the literature.
resonance of this latter cortical sector in the pres- The results of conjunction analysis between obser-
ence of an intentional agent. In order to determine vation and execution conditions showed bilateral
the differential processing during visual observa- shared activations in cerebellar lobules V, VI, crus
tion of tactile stimuli, Bolognini et al. (2011) used I, VIIIa, and VIIIb. These sectors are compatible
repetitive TMS applied to SI or SII while partici- with those classically considered as part of the
pants looked a finger touching a hand, or a finger motor loop. A shared activation was also observed
moving in the same direction without touching. in the dentate nucleus, that constitutes the main
The results showed that only the inactivation of SI output channel of the cerebellum to the motor
influences the subject’s interpretation of a tactile thalamus (Strick, 1985).
event in the observed stimulus. Altogether, these data indicate that a mir-
The differential contribution of somatosensory ror mechanism is present also in the cerebel-
cortices was also shown, in some studies, dur- lar circuitry. What is its possible role within the
ing action observation. In particular, area 2 of SI mirror system? Relevant theories proposed that
appears more activated than SII during observation cerebellum, during movement execution, could
of manipulation, suggesting that beyond the motor be involved in both forward and inverse models
resonance, there is also an activation of the hap- (Caligiore et al., 2013; Callan et al., 2013; Frith,
tic somatosensory input associated to the action 2007; Ito, 2008; Kilner et al., 2003; Weeks et al.,
(Schaefer et al., 2009; cf. Keysers et al., 2010). 1999). Since during action observation cerebel-
lar activation appears to reproduce the processing
occurring during execution, this suggests that in
the former condition the motor system performs
an internal simulation of the action. The role of
SUBCORTICAL STRUCTURES ACTIVATED cerebellum could be related, in particular, to pro-
DURING ACTION OBSERVATION cessing of sequential and temporal dynamics of
the action (Bijsterbosch et al., 2011; Caligiore
In monkeys, there is no evidence, up to now, of et al., 2013; de Solages et al., 2008).
neurons with mirror properties in subcortical Concerning the activation of basal ganglia dur-
structures. The two major candidates for the pos- ing action observation, the only electrophysiologi-
sibility to find neurons with these properties are cal study (Alegre et al., 2010) during movement
the basal ganglia and cerebellum, due to their observation and execution showed, in the subtha-
strong involvement in neuroanatomical loops with lamic nucleus (STN), an EEG beta-reduction. This
agranular frontal and posterior parietal cortices. effect was stronger during the execution condition.
Anatomical data show, in particular, that frontal Ge and colleagues (2018), with fMRI, showed a
and parietal areas containing mirror neurons pro- putamen activation during observation of grasp-
ject to specific sectors of the putamen, different ing performed in first- or third-person perspective.
from the sector specifically connected with MI They suggested that, during reinforcement learn-
(Gerbella et al., 2016). ing or imitation, this activation could be combined
In humans, a few studies reported the activa- with internal simulation of the same actions.
tion of cerebellum during action observation, but In the above reported study of Errante et al.
this structure was not the main focus of the inves- (2020) on observation/execution of object manip-
tigation (Calvo-Merino et al., 2005, 2006; Errante ulation, there was an activation of GP and STN.
and Fogassi, 2020; Frey and Gerry, 2006; Gazzola The results of conjunction analysis between
and Keysers, 2009). A recent study (Abdelgabar observation and execution conditions showed sig-
et al., 2019), in which participants had to observe nificant shared activations in both GP and STN,
reaching-grasping actions, showed specific acti- while there was no shared activation of the motor
vation of cerebellar lobules VI, VIIb and VIIIa in sector of the putamen. This could be explained by
both hemispheres. a possible enhancement of activity of the so-called
The existence of the mirror mechanism in cer- hyper-direct pathway (linking directly motor cor-
ebellum was reported by a study (Gazzola and tex with the STN) (Caligiore et al., 2013), instead
Keysers, 2009) showing bilateral shared voxels of the direct and indirect pathways, usually
for observation and execution of hand, mouth, and involved during movement execution. This would
foot actions, with a higher percentage of hand- lead to an inhibition of the thalamic motor nuclei,
related voxels, but the exact localization was not that could be explained by the need to block move-
reported. In a recent study by Errante et al. (2020), ment during observation (Bonini, 2017).
292 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

MIRROR NEURONS AND INTENTION of PFG and F5 grasping neurons discharged dif-
UNDERSTANDING ferently according to the final goal of the action
in which the grasping act was embedded, some
The data discussed up to now, in both monkeys discharging stronger during grasping-to-eat, oth-
and humans, mostly demonstrate that the mirror ers being more activated by grasping-to-place
mechanism allows the individual to automatically (Figure 18.6). This modulation indicates that these
understand the “what” of the action. However, it is neurons are able to reflect the motor intention of
not clear whether this mechanism enables observ- the agent. From these results, it has been proposed
ers to also decode others’ motor intentions, mean- that action organization is achieved through dif-
ing with this term the final goal of an action. This ferent chains of neurons, each chain encoding a
latter represents the behavioral outcome of an different intention (for example reach-grasp-bring
action, for example grasping a cup of coffee and to the mouth-eat) (Fogassi et al., 2005; cf. Chersi
bringing it to the mouth to drink from it. The issue et al., 2011).
of intention decoding is very important, because it Subsequently, mirror neurons were recorded
is a basis of social interaction. from PFG and F5 while the monkey observed the
As a premise, we have widely described that experimenter performing the same two actions.
most neurons belonging to the parieto-premotor Then, the monkey performed the same motor
system code the goal of motor acts. Is this type task as described before. The results showed that,
of coding modulated by the intention underlying during grasping observation, the visual discharge
the action? First of all, we carried out a series of of mirror neurons presents the same modulation
experiments in monkeys to assess the response of observed in grasping neurons during the motor
grasping neurons during execution of two types of task (Figure 18.6). This means that during obser-
actions- grasping for eating a piece of food and vation of the grasping act performed by the exper-
grasping to place an object in a container – both imenter, mirror neurons allow the observer to
involving a grasping act (Bonini et al., 2010, 2011; predict agent’s intention. It has been hypothesized
Fogassi et al., 2005). Grasping neurons were that, in a given context, the observation of the
recorded from areas PFG and F5, while a mon- grasping acts retrieves the motor chain associated
key performed the two actions. The results showed to a specific intention, thus enabling the observer
that, although the grasping act was identical in the to automatically understand the intention of the
eating and placing conditions, a high percentage observed agent. It is possible that this function,

Figure 18.6 Congruence of intentional coding between the visual and the motor response
of a PFG and an F5 mirror neuron. Rasters and histograms showing the motor and visual
responses are aligned with the moment in which the hand of the monkey or of the experi-
menter touched the target object.
Mirror Mechanism in Cognition 293

demonstrated in monkeys, constitutes the basic DISGUST AND THE ANTERIOR INSULA
mechanism from which other more sophisticated
mechanisms of mindreading could have evolved Convincing evidence that disgust is encoded in the
(Fogassi et al., 2005). anterior insula was provided at the end of nineties
The presence of such a mechanism has also by Phillips (1997, 1998) and Sprengelmeyer
been studied in humans (Iacoboni et al., 2005). (1998) and their coworkers. Both these groups
In this study participants had to observe three demonstrated, using fMRI, a selective activation
different types of videoclips, corresponding to of the anterior insular (AI) cortex to presentation
three conditions: a video representing either a of faces showing disgust relative to that elicited by
breakfast to be started or ended (condition “con- other emotional facial expressions.
text”), a video showing a hand grasping a cup in An experiment that strongly contributed to prove
an empty background (condition “action”) and a the existence of the mirror mechanism in AI was
hand grasping a cup in one of the contexts of the carried out by (Wicker et al., 2003) in collabora-
first condition (condition “intention”). The results tion with Keysers, Gallese, and Rizzolatti. This
showed that the “intention” condition, as com- study consisted of two distinct sessions: one olfac-
pared to the sum of the other two conditions, pro- tory and one visual. In the former, participants were
duced a stronger activation in the right IFG. This administered pleasant (rose scent) and unpleasant
effect was independent of whether the participants (rotten eggs) odorants. In the second one, partici-
were asked to purely observe the three types of pants observed videos where individuals sniffed at
videos or to observe them in order to explicitly glasses containing an unpleasant odorant, a pleas-
understand the intention. antly perfumed liquid, and a liquid with no smell
whatsoever, and reacted with the appropriate facial
expression. The results showed that, as expected,
disgusting odorants activated the AI, but, most
MIRROR MECHANISMS AND EMOTIONS interestingly, this activation coincided, although
partially, with the insula activation induced by
The first evidence suggesting the existence of the personal experience of disgusting odorants (olfac-
mirror mechanism for emotions has been pro- tory condition). This experiment for the first time
vided by Dimberg et al. (1982). He investigated showed an overlapping activation in the AI, induced
whether the typical face and body configurations by first-hand experience of disgust and by observ-
of basic emotions that appear on the person expe- ing grimaces of disgust on the faces of other people.
riencing them were also present on an individual Besides fMRI studies, electrical stimulation
observing that person. He recorded the EMG experiments in monkeys and humans established
activity of two facial muscles, the zygomaticus a fundamental role of the AI in encoding dis-
major and upper corrugator, in volunteers observ- gust. This had been shown already during the last
ing static faces expressing happiness or anger. century by using macrostimulation. Much more
The results showed that observation of happy recently, researchers of the University of Parma
faces determined activation of the zygomaticus carried out an extensive exploration of the insula
major, while observing angry faces caused using intracortical microstimulation (Caruana
increase of the EMG activity in the upper corru- et al., 2011; Jezzini et al., 2012). They found that,
gator. In other words, the EMG pattern in the in macaques, stimulation of the ventral sector of
observer mirrored that of the individual express- the AI elicited disgust, sometimes accompanied
ing a given emotion. by bouts of retching.
An early interpretation of these data proposed Results congruent with those observed in the
that the mirror mechanism was located in the cor- monkey were found in drug-resistant epileptic
tical motor system and that the activation of the patients electrically stimulated during surgical
emotional centers was a secondary consequence excision of an epileptic focus. Reaction of disgust
of the motor activations. Although this is a pos- was reported in early studies as well as in more
sibility, there is convincing evidence that the emo- recent research based on the stereo EEG technique.
tional responses during observation of emotion in Particularly interesting is the study by Krolak-
others are mostly due to a direct activation of cent- Salmon et al. (2003), who found ERP responses
ers encoding that emotion. in the ventral insula while the person was observ-
Strong evidence that emotional centers are ing faces expressing disgust. The same authors
endowed with a mirror mechanism came from stimulated the active sites electrically, showing
fMRI and stereo-EEG studies. In the next sections that almost all patients complained of an unpleas-
we will discuss the data for anterior insula, the ant disgust-like sensation. Thus, both vision of a
amygdala and the anterior cingulate cortex. disgusted face and electrical stimulation of the
294 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

same site show that detection of disgust in others more recent cingulate parcellation, which will be
and feeling disgust oneself are mediated by the adopted in this chapter, subdivides the cingulate
same part of the insula, indicating that this region cortex into three major sectors: the anterior cingu-
is endowed with the mirror mechanism. late (subgenual, pregenual, and postgenual), the
middle cingulate (anterior middle and posterior
middle, both, in turn, subdivided into a dorsal and
ventral part) and the posterior cingulate
FEAR AND THE AMYGDALA (Palomero-Gallagher, 2009, 2015; Vogt and Vogt,
2003; Vogt et al., 2005) (Figure 18.7).
Although amygdala is a complex subcortical ACC, unlike its caudal sectors, plays a role
structure formed by about 12 different nuclei, for in affect, with the pACC playing a role in posi-
the purpose of the present chapter we will describe tive affect and the subgenual ACC (sACC) being
it as a single entity and will consider its main involved in the negative ones (Vogt et al., 2005).
function: encoding fear. In fact, albeit there is The most evident expression of positive affect
little doubt that some of its nuclei may also have is laughter. Yet, until recently, a locus from
other functions, the most consistent evidence on which laughter could be consistently elicited
its function concerns fear. was controversial, being laughter occasionally
A selective activation of the amygdala for sight evoked by several cortical sites, among which
of facial expressions of fear, relative to faces the region that lies between the superior frontal
showing happiness, was demonstrated in fMRI gyrus and the supplementary motor area (Fried
studies carried out in the 1990s (Morris et al., et al., 1998; Krolak-Salmon et al., 2006; Schmitt
1996; Phillips et al., 1997). Subsequently, it was et al., 2006).
shown that amygdala is selectively activated by A fundamental step in discovering the main
vocal manifestation of fear (Phillips et al., 1998; center for laughing was recently completed by
cf. Dolan et al., 2001). The relation between fear researchers at the University of Parma and the
and activation of amygdala was confirmed by Sato Epilepsy Surgery Center of Milan’s Niguarda
(2004). Hadjikhani and de Gelder (2003) provided Hospital (Caruana et al., 2015). By using intrac-
evidence, also in an fMRI experiment, that the ranial electric stimulation, they examined the
amygdala was activated when participants saw responses of over fifty drug-resistant epileptic
bodily postures communicating a sense of fear. patients. They found that the stimulation of pACC
Whereas these brain imaging data demonstrate induced laughter and hilarity.
a selective activation of amygdala to frightening Gibson et al. (2016) confirmed these results
stimuli, there is now evidence that this structure using a more complex technique. These authors
becomes also active when individuals experience combined deep brain stimulation of the internal
fear themselves. Meletti et al. (2006), in collabo- capsule and ventral striatum with fMRI. There
ration with the surgeons of Niguarda Hospital in was previous evidence that this deep stimulation
Milan, stimulated the temporal lobe region, includ- procedure can modify the mood of patients with
ing the amygdala, in over 70 patients with drug- compulsive-obsessive disorders, making them
resistant epilepsy. The most important result was the more cheerful and eliciting smiles and even burst
high frequency of responses with emotional content of laughing. Gibson et al. (2016) found that this
almost all consisting in manifestation of fear as deep brain stimulation activates the pACC and,
well as in verbal report of fear and anxiety. Similar most interestingly, pACC activation was associ-
results were found by Lanteaume et al. (2007). ated with laughter and hilarity. Along the same
Taken together, these findings indicate that the lines, Matsunaga et al. (2016) reported that the
amygdala is endowed with the mirror mechanism activation of the rostral cingulate cortex increases
for those processes that involve fear. As in the AI with variations in the positive state of mind.
for disgust, in the amygdala the mirror mechanism Taken together, these data support the notion
transforms visual and auditory frightening stimuli in that pACC controls positive affect. Most impor-
fearful experiences as those we experience ourselves. tantly for the aim of this chapter, convincing
evidence shows that pACC is endowed with the
mirror mechanism. A meta-analysis by Fuser-
Poli et al. (2009) on the perception of emotional
faces confirmed this conclusion, showing that the
LAUGHING AND THE PREGENUAL observation of faces expressing happiness, relative
ANTERIOR CINGULATE CORTEX (pACC) to faces with neutral expressions, activates selec-
tively the pACC.
Classically, the cingulate cortex has been subdi- A strong proof that the pACC is endowed
vided into the anterior and posterior cingulate. A with the mirror mechanism has been recently
Mirror Mechanism in Cognition 295

Figure 18.7 Subdivision of human cingulate cortex. Top: mesial view showing eight cin-
gulate sectors. Bottom: Same subdivisions, shown in representative coronal sections. The
sectors are defined as follows: subgenual sector (sACC), that includes areas 25, s24, and s32
(Palomero-Gallagher et al. 2015); pregenual sector (pACC) and pregenual area 32 (Palomero-
Gallagher et al. 2008); rostrocaudal subdivisions of MCC (aMCC and pMCC) (Palomero-
Gallagher et al. 2009; Vogt and Vogt 2003; Vogt et al. 2005); PCC (Leech and Sharp 2014; Vogt
et al. 2003;). aMCC and pMCC were further subdivided in dorsal (aMCCd and pMCCd) and
ventral (aMCCv and pMCCv) (Palomero-Gallagher et al. 2009).
Source: From Caruana et al. 2018.

provided by Caruana et al. (2017) combining was looking at the video-clips of an actor crying or
intracranial electrical stimulation and intracranial showing a neutral face.
EEG recordings in a patient with drug-resistant Summing up, these data show that during the
focal epilepsy. The authors presented the patient observation of smiling faces, pACC is activated,
with videos showing an actor laughing, crying or and triggers a complex pattern of motor processes
expressing no emotion whatsoever. As expected, characterizing laughter and contributing to form-
smile was elicited when the pACC was stimulated. ing the feeling of hilarity. In fact, the sensory
The most interesting result was, however, that the information arriving principally from the temporal
sites whose stimulation evoked laughter, showed regions are transformed, in the pACC, into motor
an increase of gamma activity (50–100 Hz). This representations, allowing the individual to recog-
occurred exclusively while the patient was look- nize other people’s expressions of laughter by acti-
ing at the video-clip showing laughing. No sig- vating the same cortical and subcortical circuits
nificant activation was observed when the patient activated by stimuli that induce laughter.
296 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

MOTOR REACTION AND THE ANTERIOR motor behavior that can subserve defensive or
MIDDLE CINGULATE CORTEX (aMCC) aversive reactions.

The first evidence of the existence of the mirror


mechanism in human cingulate cortex was pro-
vided by Hutchinson et al. (1999). The activity of MIRROR MECHANISMS AND VITALITY
single neurons was recorded from aMCC in FORMS
patients undergoing bilateral cingulotomy. Before
proceeding with the ablation, the surgeons tested In all actions there are two components: the action
the responsiveness of aMCC neurons to different content (i.e., the action goal) and the action form
types of nociceptive stimuli as well as to simple (i.e., how the goal is achieved). Whatever its con-
touch. They found that a set of neurons that tent, actions may be energetic, gentle, abrupt, but
responded selectively when a patient was pricked also hesitant, or effortless. Specifications of how
with a needle, also activated when the patient saw an action is performed do not refer only to “cold”
the experimenter being pricked with a needle. actions (i.e., those devoid of an emotional con-
Some years later, an apparent confirmation that tent), but also to “hot” actions involving emotions.
aMCC is a pain center and convincing evidence Thus, an individual can experience an irrepressi-
that aMCC is endowed with mirror properties was ble sense of disgust, or a spurt of explosive anger
provided by Singer et al. (2004). that had been suppressed.
A clear proof that aMCC in not a pain center Stern called the forms of an action “vitality
was subsequently provided by an fMRI study by forms.” Vitality forms express the agent’s attitudes
Iannetti and coworkers (Mouraux et al., 2011), towards others. They describe the “How” of an
who compared the activation of aMCC elicited action, not its “What” or “Why” (Stern, 2010).
by nociceptive somatosensory, non-nociceptive An important point worth mentioning (Stern,
somatosensory, auditory, and visual stimuli. The 1985; cf. Braten, 1998; Trevarthen, 1998) is that
results showed similar aMCC activations elicited “sharing vitality forms” is the “oldest, most direct
by all four types of stimuli. In addition, the acti- and immediate” way of interpersonal relations so
vations were stronger when the participants felt “deeply rooted” and “natural” as to pervade every
the stimuli as salient. The authors concluded that social interaction (Stern, 2010).
activation of the aMCC reflects processes that In the last few years, a series of experiments
are essential to orientate to salient stimuli, and to tried to establish the brain region responsible for
assess the potential threat they pose and, where vitality forms. The first experiment was an fMRI
necessary, to trigger the appropriate response. study performed in Parma in collaboration with
Intracortical brain stimulation studies fully Daniel Stern (Di Cesare et al., 2014). In this exper-
confirmed this interpretation. In the early 1970s, iment, participants were presented with video-
neurosurgeon Jean Talairach had observed how clips showing transitive actions (e.g., giving an
electric stimulation of aMCC sites produced object) and four intransitive actions (e.g., stopping
complex motor behavior. These motor responses gesture). All actions were performed with two dif-
could affect the whole body or the upper limbs. ferent vitality forms (gentle or abrupt). The stimuli
Most importantly, none of the aMCC stimulation were presented in pairs of consecutive video-clips,
produced pain or any form of emotional behav- where the “what” of the observed action, and the
ior. The findings of Talairach were recently con- “how” of it could be the same or change between
firmed in a study conducted in collaboration with the video-clips pairs. There were two tasks (what
the Epilepsy Surgery Center of Milan’s Niguarda and how). In the what task, the participants were
Hospital (Caruana et al., 2018). They found that required to pay attention to the goal of the actions
stimulation of the aMCC elicited complex motor observed in the two consecutive videos and decide
behaviors, but no actual feeling of pain. For whether the two actions were the same or differ-
example, when stimulated, patients made postural ent, regardless of their vitality form. In the how
adjustments that clearly indicated an impellent task the participants were required to focus their
necessity to get out from the bed and even from attention on the vitality form of the two actions
the room. and decide whether the vitality form was the same
If we consider the data from the fMRI study or different in the two consecutive videos, in spite
conducted by Iannetti (Mouraux et al., 2011) of the fact the observed actions were different.
together with the intracortical stimulation studies, The results showed, in both tasks, activa-
it emerges that aMCC plays a distinct role in trans- tions of the parieto-frontal network known to
forming a great variety of stimuli into specific be involved in action execution and observation.
Mirror Mechanism in Cognition 297

Most importantly, the contrast “how” vs. “what” in the DCI transforms the visual representation of
revealed a specific activation in the right dorso- vitality forms into their motor representation.
central insula (DCI), providing evidence that
central insula plays a specific role in processing
vitality forms.
In a subsequent fMRI study, Di Cesare et al.
(2015) tested whether DCI was also active dur- SUMMARY AND CONCLUSIONS
ing the execution of actions endowed with vitality
forms (Figure 18.8. The experiment was carried Mirror neurons are a specific class of neurons that
out on healthy right-handed individuals, asked to activate during both execution and observation of
perform three different tasks: observation (OBS), the same motor acts. This mechanism enables the
imagination (IMA) and execution (EXE). In the observer to achieve an automatic understanding of
OBS task, the participants were shown video- others’ behavior. This suggests a fundamental role
clips in which an actor was passing an object of this neural system in social cognition. The dis-
to another one, either in a gentle or a rude way covery of mirror neurons was preceded by pro-
(vitality condition) or in which an actor was longed anatomical and functional investigation of
placing a small ball in a box (control condition). premotor and parietal areas, demonstrating the
In the EXE task, the participants were asked to cognitive function of these areas. The mirror
move an object in a rude or gentle way (execu- neuron mechanism has been shown to be present
tion task) or to place a small ball in a box in the also in the parietal and frontal cortex of the human
most neutral way as possible. Finally, in the IMA brain, and subsequently has also been demon-
task, participants were asked to imagine them- strated in the basal ganglia and cerebellum. Recent
selves passing an object towards the actor located investigations have increased knowledge of the
in front of them in a gentle or in a rude way or functional role of the mirror mechanism, includ-
to imagine themselves placing a small ball in the ing the decoding of others’ motor intentions, emo-
box without any specific vitality form (control tions, and vitality forms.
imagination task). The empirical evidence reviewed above indi-
In all three tasks there was a bilateral activation cates that the DCI is a brain center controlling the
of the premotor and parietal cortices, and a strong vitality forms for movements of the forelimb and
activation of the left somatosensory cortex, motor is endowed with the mirror mechanism for these
cortex, and of the cerebellum. Importantly, there actions. We postulated that vitality forms exist
was also a strong activation of the DCI in all three also for emotions. However, at present, there is no
tasks showing that the mirror mechanism present evidence to localize the brain centers responsible

Figure 18.8 Cortical areas active during three different tasks (observation, imagination, and
execution) in the two experimental conditions (rude and gentle vs. control). The parasagittal
sections show the insular activations in two hemispheres.
LH, left hemisphere; RH, right hemisphere.
Source: From Di Cesare et al. 2015.
298 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

for giving form to the expression of emotion and Bijsterbosch, J. D., Lee, K.H., Hunter, M. D., Tsoi, D. T.,
further studies are necessary to clarify this. Lankappa, S., Wilkinson, I. D., … Woodruff P. V. R.
(2011). The role of the cerebellum in sub- and
supraliminal error correction during sensorimotor
synchronization: Evidence from fMRI and TMS. Jour-
Note nal of Cognitive Neuroscience, 23, 1100–1112.
Bolognini, N., Rossetti, A., Maravita, A., & Miniussi,
1  In this chapter we use the term “motor act” to C. (2011). Seeing touch in the somatosensory
indicate a synergy of movements aimed to attain cortex: A TMS study of the visual perception of
a goal (e.g., grasping an object), and the term touch. Human Brain Mapping, 32, 2104–2114.
“action” to indicate a series of motor acts that Bonini, L., Rozzi, S., Ugolotti Serventi, F., Simone, L.,
allow attaining a behavioral goal (e.g., grasping- Ferrari, P. F., & Fogassi L. (2010). Ventral premotor
to-eat). and inferior parietal cortices make distinct contri-
bution to action organization and intention under-
standing. Cerebral Cortex, 20, 1372–1385.
Bonini, L., Serventi, F. U., Simone, L., Rozzi, S., Ferrari,
REFERENCES P. F., & Fogassi L. (2011). Grasping neurons of
monkey parietal and premotor cortices encode
action goals at distinct levels of abstraction during
Abdelgabar, A. R., Suttrup, J., Broersen, R., complex action sequences. Journal of Neurosci-
Bhandari, R., Picard, S., Keysers, C., De Zeeuw, C. ence, 31(15), 5876–5886.
I., & Gazzola, V. (2019). Action perception Bonini, L. (2017). The extended mirror neuron net-
recruits the cerebellum and is impaired in patients work. Neuroscientist, 23, 56–67.
with spinocerebellar ataxia. Brain, 142(12), Borra, E., Belmalih, A., Calzavara, R., Gerbella, M.,
3791–3805. Murata, A., Rozzi, S., & Luppino G. (2008). Corti-
Abdollahi, R. O., Jastorff, J., & Orban, G. A. (2013). cal connections of the macaque anterior intrapari-
Common and segregated processing of observed etal (AIP) area. Cerebral Cortex, 18, 1094–1111.
actions in human SPL. Cerebral Cortex, 23(11), Borra, E., Gerbella, M., Rozzi, S., & Luppino, G.
2734–2753. (2011). Anatomical evidence for the involvement
Alegre, M., Rodríguez-Oroz M. C., Valencia, M., of the macaque ventrolateral prefrontal area 12r in
Pérez-Alcázar, M., Guridi, J., Iriarte, J., … Artieda, controlling goal-directed actions. Journal of Neu-
J. (2010). Changes in subthalamic activity during roscience, 31, 12351–12363.
movement observation in Parkinson’s disease: Is Borra, E., Gerbella, M., Rozzi, S., & Luppino, G.
the mirror system mirrored in the basal ganglia? (2017). The macaque lateral grasping network: A
Clinical Neurophysiology, 121(3), 414–425. neural substrate for generating purposeful hand
Altschuler, E. L., Vankov, A., Wang, V., Ramachan- actions. Neuroscience and Biobehavioral Reviews,
dran, V. S., & Pineda, J. A. (1997). Person see, 75, 65–90.
person do: Human cortical electrophysiological Borra, E., Ferroni, C. G., Gerbella, M., Giorgetti, V.,
correlates of monkey see monkey do cells. Poster Mangiaracina, C., Rozzi, S., & Luppino G. (2019).
presented at the 27th annual meeting of the Soci- Rostro-caudal connectional heterogeneity of the
ety for Neuroscience, New Orleans, Louisiana. dorsal part of the Macaque prefrontal area 46.
Barraclough, N. E., Xiao, D., Oram, M. W., & Perrett, Cerebral Cortex, 29(2), 485–504.
D.I. (2006). The sensitivity of primate STS neurons Bråten, S. (1998). (Ed.), Intersubjective communica-
to walking sequences and to the degree of articu- tion and emotion in early ontogeny. Cambridge,
lation in static images. Progress in Brain Research, UK: Cambridge University Press.
154, 135–148. Bruce, C. J., & Goldberg, M. E. (1985). Primate fron-
Belmalih, A., Borra, E., Contini, M., Gerbella, M., tal eye fields. I. Single neurons discharging before
Rozzi, S., & Luppino G. (2007). A multiarchitec- saccades. Journal of Neurophysiology, 53(3),
tonic approach for the definition of functionally 603–635.
distinct areas and domains in the monkey frontal Buccino, G., Binkofski, F., Fink, G. R., Fadiga, L.,
lobe. Journal of Anatomy, 211, 199–211. Fogassi, L., Gallese, V., … Freund H. J. (2001).
Belmalih, A., Borra, E., Contini, M., Gerbella, M., Action observation activates premotor and parietal
Rozzi, S., & Luppino, G. (2009). Multimodal archi- areas in a somatotopic manner: An fMRI study.
tectonic subdivision of the rostral part (area F5) of European Journal of Neuroscience, 13, 400–404.
the macaque ventral premotor cortex. Journal of Caggiano, V., Fogassi, L., Rizzolatti, G., Thier, P., &
Comparative Neurology, 512(2), 183–217. Casile, A. (2009). Mirror neurons differentially
Mirror Mechanism in Cognition 299

encode the peripersonal and extrapersonal space Chersi, F., Ferrari, P. F., & Fogassi, L. (2011). Neuronal
of monkeys. Science, 324, 403–406. chains for actions in the parietal lobe: A computa-
Caggiano, V., Fogassi, L., Rizzolatti, G., Pomper, J. K., tional model. PLoS One, 6(11). doi: 10.1371/jour-
Thier, P., Giese, M. A., & Casile A. (2011). View- nal.pone.0027652
based encoding of actions in mirror neurons of Cisek, P., & Kalaska, J. F. (2004). Neural correlates of
area F5 in macaque premotor cortex. Current Biol- mental rehearsal in dorsal premotor cortex.
ogy, 21, 144–148. Nature, 431, 993–996.
Caligiore, D., Pezzulo, G., Miall, R. C., & Baldassarre, Cochin, S., Barthelemy, C., Roux, S., & Martineau, J.
G. (2013). The contribution of brain sub-cortical (1999). Observation and execution of movement:
loops in the expression and acquisition of action similarities demonstrated by quantified electroen-
understanding abilities. Neuroscience and Biobe- cephalography. European Journal of Neurosci-
havioral Reviews, 37, 2504–2515. ence, 11, 1839–1842.
Callan, D. E. Terzibas, C., Cassel, D. B., Callan, A., de Solages, C. Szapiro, G., Brunel, N., Hakim, V.,
Kawato, M., & Sato, M. A. (2013). Differential Isope, P., Buisseret, P., … Léna C. (2008). High-
activation of brain regions involved with error- frequency organization and synchrony of activity
feedback and imitation based motor simulation in the Purkinje cell layer of the cerebellum.
when observing self and an expert’s actions in Neuron, 58, 775–788.
pilots and non-pilots on a complex glider landing Di Cesare, G., Di Dio, C., Rochat, M. J., Sinigaglia, C.,
task. NeuroImage, 72, 55–68. Bruschweiler-Stern, N., Stern, D. N., & Rizzolatti G.
Calvo-Merino, B., Glaser, D. E., Grezes, J., Passing- (2014). The neural correlates of ‘vitality form’ rec-
ham, R. E., & Haggard, P. (2005). Action observa- ognition: An fMRI study. Social Cognitive and
tion and acquired motor skills: An FMRI study with Affective Neuroscience, 9(7), 951–960.
expert dancers. Cerebral Cortex, 15, 1243–1249. Di Cesare. G., Di Dio. C., Marchi, M., & Rizzolatti, G.
Calvo-Merino, B., Grezes, J., Glaser, D. E., Passing- (2015). Expressing our internal states and under-
ham, R. E., & Haggard, P. (2006) Seeing or doing? standing those of others. Proceedings of the
Influence of visual and motor familiarity in action National Academy of Sciences of the United States
observation. Current Biology, 16, 1905–1910. of America, 112(33), 10331–10335.
Caruana, F., Jezzini, A., Sbriscia-Fioretti, B., Rizzolatti, G., Dimberg, U. (1982). Facial reactions to facial expres-
& Gallese, V. (2011). Emotional and social behaviors sions. Psychophysiology, 19(6), 643–647.
elicited by electrical stimulation of the insula in the Di Pellegrino, G., Fadiga, L., Fogassi, L., Gallese, V., &
macaque monkey. Current Biology, 21, 195–199. Rizzolatti, G. (1992). Understanding motor events:
Caruana, F., Avanzini, P., Gozzo, F., Francione, S., Car- A neurophysiological study. Experimental Brain
dinale, F., & Rizzolatti, G. (2015). Mirth and laugh- Research, 91, 176–180.
ter elicited by electrical stimulation of the human Dolan, R.J., Morris, J.S., & de Gelder, B. (2001). Cross-
anterior cingulate cortex. Cortex, 71, 323–331. modal binding of fear in voice and face. Proceed-
Caruana, F., Avanzini, P., Gozzo, F., Pelliccia, V., ings of the National Academy of Sciences of the
Casaceli, G., & Rizzolatti, G. (2017). A mirror United States of America, 98, 10006–10010.
mechanism for smiling in the anterior cingulate Ebisch, S. J. H., Perrucci, M. G., Ferretti, A., Del
cortex. Emotion, 17(2), 187–190. Gratta, C., Romani, G. L., Gallese, V. (2008). The
Caruana, F., Gerbella, M., Avanzini, P., Gozzo, F., Pel- sense of touch: Embodied simulation in a visuotac-
liccia, V., Mai, R., … Rizzolatti, G. (2018). Motor tile mirroring mechanism for observed animate or
and emotional behaviours elicited by electrical inanimate touch. Journal of Cognitive Neurosci-
stimulation of the human cingulate cortex. Brain, ence 20, 1611–1623.
141(10), 3035–3051. Errante, A., & Fogassi, L. (2020). Activation of cerebel-
Casile, A., Caggiano, V., & Ferrari, P. F. (2011) The lum and basal ganglia during the observation and
mirror neuron system: A fresh view. Neuroscien- execution of manipulative actions. Scientific
tist, 17(5), 524–538. Reports, 10(1). doi: 10.1038/s41598-020-68928-w
Caspers, S., Zilles, K., Laird, A. R., & Eickhoff, S. B. Fadiga, L., Fogassi, L., Pavesi, G. & Rizzolatti, G.
(2010). ALE meta-analysis of action observation (1995). Motor facilitation during action observa-
and imitation in the human brain. NeuroImage, tion: A magnetic stimulation study. Journal of
50, 1148–1167. Neurophysiology, 73(6), 2608–2611.
Cattaneo, L., Caruana, F., Jezzini, A., & Rizzolatti, G. Ferrari, P.F., Gallese, V., Rizzolatti, G., & Fogassi, L.
(2009). Representation of goal and movements (2003). Mirror neurons responding to the observa-
without overt motor behavior in the human motor tion of ingestive and communicative mouth actions
cortex: A transcranial magnetic stimulation study. in the monkey ventral premotor cortex. European
Journal of Neuroscience, 29, 11134–11138. Journal of Neuroscience, 17(8), 1703–1714.
300 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Ferrari, P. F., Visalberghi, E., Paukner, A., Fogassi, L., neuron system responds to human and robotic
Ruggiero, A., & Suomi, S. J. (2006) Neonatal imita- actions. NeuroImage, 35, 1674–1684.
tion in rhesus macaques. PLoS Biology, 4(9). doi: Gazzola, V., van der Worp, H., Mulder, T., Wicker, B.,
10.1371/journal.pbio.0040302 Rizzolatti, G., & Keysers, C. (2007). Aplasics born
Ferrari, P. F., Vanderwert, R. E., Paukner, A., Bower, S., without hands mirror the goal of hand actions
Suomi, S. J., & Fox, N. A. (2012) Distinct EEG with their feet. Current Biology, 17, 1235–1240.
amplitude suppression to facial gestures as evi- Gazzola, V., & Keysers, C. (2009). The observation
dence for a mirror mechanism in newborn mon- and execution of actions share motor and soma-
keys. Journal of Cognitive Neuroscience, 24(5), tosensory voxels in all tested subjects: single-sub-
1165–1172. ject analyses of unsmoothed fMRI data. Cerebral
Ferrari, P. F., Tramacere, A., Simpson, E. A., & Iriki, A. Cortex, 19, 1239–1255.
(2013). Mirror neurons through the lens of epigenet- Ge, S., Liu, H., Lin, P., Gao, J., Xiao, C., & Li, Z.
ics. Trends in Cognitive Sciences, 17(9), 450–457. (2018). Neural basis of action observation and
Filimon, F., Nelson, J. D., Hagler, D. J., & Sereno, M. understanding from first- and third-person per-
I. (2007). Human cortical representations for spectives: An fMRI study. Frontiers in Behavioral
reaching: Mirror neurons for execution, observa- Neuroscience, 12. doi: 10.3389/fnbeh.2018.00283
tion, and imagery. NeuroImage, 37, 1315–1328. Gentilucci, M., Fogassi, L., Luppino, G., Matelli, M.,
Fogassi, L., Gallese, V., Fadiga, L., Luppino, G., Camarda, R., & Rizzolatti, G. (1988). Functional
Matelli, M., & Rizzolatti, G. (1996). Coding of organization of inferior area 6 in the macaque
peripersonal space in inferior premotor cortex (area monkey. I. Somatotopy and control of proximal
F4). Journal of Neurophysiology, 76, 141–157. movements. Experimental Brain Research, 71,
Fogassi, L., Ferrari, P. F., Gesierich, B., Rozzi, S., 475–490.
Chersi, F., & Rizzolatti, G. (2005). Parietal lobe: Gerbella, M., Belmalih, A., Borra, E., Rozzi, S., & Lup-
from action organization to intention understand- pino, G. (2010). Cortical connections of the
ing. Science, 308, 662–667. macaque caudal ventrolateral prefrontal areas 45A
Frey, S. H., & Gerry, V. E. (2006). Modulation of and 45B. Cerebral Cortex, 20(1), 141–168.
neural activity during observational learning of Gerbella, M., Borra, E., Rozzi, S., Tonelli, S., & Lup-
actions and their sequential orders. Journal of pino, G. (2013). Connectional heterogeneity of
Neuroscience, 26, 13194–13201. the ventral part of the macaque area 46. Cerebral
Fried, I., Wilson, C. L., MacDonald, K. A., & Behnke, Cortex, 23(4), 967–987.
E. J. (1998). Electric current stimulates laughter. Gerbella, M., Borra, E., Mangiaracina, C., Rozzi, S., &
Nature, 391. doi: 10.1038/35536 Luppino, G. (2016). Corticostriate projections from
Fusar-Poli, P., Placentino, A., Carletti, F., Landi, P., areas of the “Lateral grasping network” evidence
Allen, P., Surguladze, S., … Politi, P. (2009). Func- for multiple hand-related input channels. Cerebral
tional atlas of emotional faces processing: A voxel- Cortex, 26, 3096–3115.
based meta-analysis of 105 functional magnetic Geyer, S., Matelli, M., Luppino, G., & Zilles, K. (2000).
resonance imaging studies. Journal of Psychiatry & Functional neuroanatomy of the primate isocorti-
Neuroscience, 34, 418–432. cal motor system. Anatomy and Embryology, 202,
Gallese V., Fadiga L., Fogassi L., & Rizzolatti G. 443–474.
(1996). Action recognition in the premotor cortex. Gibson, W. S., Cho, S., Abulseoud, O. A., Gorny, K.
Brain, 119(2), 593–609. R., Felmlee, J. P., Welker, K. M., … Lee K. H.
Gallese, V., Fadiga, L., Fogassi, L., & Rizzolatti, G. (2016). The impact of mirth-inducing ventral stri-
(2002). Action representation and the inferior atal deep brain stimulation on functional and
parietal lobule. In W. Prinz & B. Hommel (Eds.), effective connectivity. Cerebral Cortex, 27,
Common mechanisms in perception and action: 2183–2194.
Attention and performance (Vol. 19, pp. 334– Grafton, S. T., Arbib, M. A., Fadiga, L., & Rizzolatti,
355). Oxford, UK: Oxford University Press. G. (1996). Localization of grasp representations in
Gangitano, M., Mottaghy, F. M., & Pascual-Leone, A. humans by positron emission tomography. Experi-
(2004). Modulation of premotor mirror neuron mental Brain Research, 112(1), 103–111.
activity during observation of unpredictable grasp- Gregoriou, G. G., Borra, E., Matelli, M., & Luppino,
ing movements. European Journal of Neurosci- G. (2006). Architectonic organization of the infe-
ence, 20(8), 2193–2202. rior parietal convexity of the macaque monkey.
Gazzola, V, Aziz-Zadeh, L, & Keysers, C. (2006). Empa- Journal of Comparative Neurology, 496(3),
thy and the somatotopic auditory mirror system in 422–451.
humans. Current Biology, 16, 1824–1829. Hadjikhani, N., & de Gelder, B. (2003). Seeing fearful
Gazzola, V., Rizzolatti, G., Wicker, B., & Keysers, C. body expressions activates the fusiform cortex and
(2007). The anthropomorphic brain: The mirror amygdala. Current Biology, 13, 2201–2205.
Mirror Mechanism in Cognition 301

Heyes, C. (2010). Where do mirror neurons come Kraskov, A., Dancause, N., Quallo, M.M., Shepherd,
from? Neuroscience and Biobehavioral Reviews, S., & Lemon, R. N. (2009). Corticospinal neurons
34(4), 575–583. in macaque ventral premotor cortex with mirror
Hutchison, W. D., Davis, K. D., Lozano, A. M., Tasker, properties: A potential mechanism for action sup-
R. R., & Dostrovsky, J. O. (1999). Pain-related neu- pression? Neuron, 64, 922–930.
rons in the human cingulate cortex. Nature Neuro- Krolak-Salmon, P., Hénaff, M.-A., Isnard, J., Tallon-
science, 2(5), 403–405. Baudry, C., Guénot, M., Vighetto, A., … Mau-
Iacoboni, M., Woods, R. P., Brass, M., Bekkering, H., guière, F. (2003). An attention modulated response
Mazziotta, J. C., & Rizzolatti G. (1999). Cortical to disgust in human ventral anterior insula. Annals
mechanisms of human imitation. Science, 286, of Neurology, 53, 446–453.
2526–2528. Krolak-Salmon, P., Hénaff, M.-A., Vighetto, A., Bau-
Iacoboni, M., Molnar-Szakacs, I., Gallese, V., Buccino, chet, F., Bertrand,, O., Mauguière, F., & Isnard, J.
G., Mazziotta, J. C., & Rizzolatti, G. (2005). Grasp- (2006). Experiencing and detecting happiness in
ing the intentions of others with one’s own mirror humans: The role of the supplementary motor
neuron system. PLoS Biology, 3(3). doi: 10.1371/ area. Annals of Neurology, 59, 196–199.
journal.pbio.0030079. Lanteaume, L., Khalfa, S., Regis, J., Marquis, P., Chau-
Ishida, H., Nakajima, K., Inase, M., & Murata, A. vel, P., & Bartolomei, F. (2007). Emotion induction
(2010). Shared mapping of own and others’ after direct intracerebral stimulations of human
bodies in visuotactile bimodal area of monkey amygdala. Cerebral Cortex, 17, 1307–1313.
parietal cortex. Journal of Cognitive Neuroscience Lanzilotto, M., Livi, A., Maranesi, M., Gerbella, M.,
22(1), 83–96. Barz, F., Ruther, P., … Bonini, L. (2016). Extending
Ito, M. (2008). Control of mental activities by internal the cortical grasping network: Pre-supplementary
models in the cerebellum. Nature Reviews: Neuro- motor neuron activity during vision and grasping
science, 9, 304–313. of objects. Cerebral Cortex, 26(12), 4435–4449.
Jeannerod, M., Arbib, M.A., Rizzolatti, G., & Sakata, Lanzilotto, M., Ferroni, C. G., Livi, A., Gerbella, M.,
H. (1995). Grasping objects: The cortical mecha- Maranesi, M., Borra, E., … Orban, G. A. (2019).
nisms of visuomotor transformation. Trends in Anterior intraparietal area: A hub in the observed
Neuroscience, 18(7), 314–320. manipulative action network. Cerebral Cortex, 29,
Jezzini, A., Caruana, F., Stoianov, I., Gallese, V., & 1816–1833.
Rizzolatti, G. (2012). The functional organization Leech, R., & Sharp, D.J. (2014). The role of the pos-
of the insula and of inner perisylvian regions: An terior cingulate cortex in cognition and disease.
intracortical microstimulation study. Proceedings Brain, 137(1), 12–32.
of the National Academy of Sciences of the United Lepage, J. F., & Théoret, H. (2006). EEG evidence for
States of America, 109, 10077–10082. the presence of an action observation-execution
Keysers, C., Wicker, B., Gazzola, V., Anton, J.L., matching system in children. European Journal of
Fogassi, L., & Gallese, V. (2004). A touching sight: Neuroscience, 23(9), 2505–2510.
SII/PV activation during the observation and expe- Livi, A., Lanzilotto, M., Maranesi, M., Fogassi, L., Riz-
rience of touch. Neuron, 42(2), 335–346. zolatti, G., & Bonini, L. (2019). Agent-based repre-
Keysers, C., Kaas, J.H., & Gazzola, V. (2010) Soma- sentations of objects and actions in the monkey
tosensation in social perception. Nature Reviews: pre-supplementary motor area. Proceedings of the
Neuroscience, 11(6), 417–428. National Academy of Sciences of the United States
Keysers, C., & Gazzola, V. (2014). Hebbian learning of America, 116(7), 2691–2700.
and predictive mirror neurons for actions, sensa- Lu, M. T., Preston, J. B., & Strick, P. L. (1994) Intercon-
tions and emotions. Philosophical Transactions of nections between the prefrontal cortex and the
the Royal Society B, 369. doi: 10.1098/ premotor areas in the frontal lobe. Journal of
rstb.2013.0175 Comparative Neurology, 341, 375–392.
Kilner, J. M., Friston, K. J., & Frith, C. D. (2007). Pre- Luppino, G., Matelli, M., Camarda, R., & Rizzolatti,
dictive coding: An account of the mirror neuron G. (1993). Corticocortical connections of area F3
system. Cognitive Processing, 8, 159–166. (SMA-proper) and area F6 (pre-SMA) in the
Kohler, E., Keysers, C., Umiltà, M. A., Fogassi, L., Gal- macaque monkey. Journal of Comparative Neurol-
lese, V., & Rizzolatti G. (2002). Hearing sounds, ogy, 338, 114–140.
understanding actions: Action representation in Maeda, K., Ishida, H., Nakajima, K., Inase, M., &
mirror neurons. Science, 297, 846–848. Murata, A. (2015). Functional properties of pari-
Koski, L., Iacoboni, M., Dubeau, M. C., Woods, R. P., etal hand manipulation-related neurons and
& Mazziotta, J. C. (2003). Modulation of cortical mirror neurons responding to vision of own hand
activity during different imitative behaviors. Jour- action. Journal of Cognitive Neuroscience, 27(3),
nal of Neurophysiology, 89, 460–471. 560–572.
302 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Marshall, P. J., Young, T., & Meltzoff, A. N. (2011). Brain Research: Cognitive Brain Research, 19(2),
Neural correlates of action observation and execu- 195–201.
tion in 14-month-old infants: An event-related Myowa, M. (1996). Imitation of facial gestures by an
EEG desynchronization study. Developmental Sci- infant chimpanzee. Primates, 37, 207–213.
ence,14(3), 474–480. Myowa-Yamakoshi, M., Tomonaga, M., Tanaka, M.,
Matelli, M., Luppino, G., & Rizzolatti, G. (1985). Pat- & Matsuzawa, T. (2004). Imitation in neonatal
terns of cytochrome oxidase activity in the frontal chimpanzees (Pan troglodytes). Developmental
agranular cortex of the macaque monkey. Behav- Science, 7(4), 437–442.
ioral Brain Research, 18(2), 125–136. Nelissen, K., Luppino, G., Vanduffel, W., Rizzolatti,
Matelli, M., Luppino, G., & Rizzolatti, G. (1991) G., & Orban, G. A. (2005). Observing others: Mul-
Architecture of superior and mesial area 6 and the tiple action representation in the frontal lobe. Sci-
adjacent cingulate cortex in the macaque monkey. ence, 310, 332–336.
Journal of Comparative Neurology, 311(4), Nelissen, K., Borra, E., Gerbella, M., Rozzi, S., Lup-
445–462. pino, G., Vanduffel, W., … Orban, G. A. (2011).
Matsunaga, M., Kawamichi, H., Koike, T., Yoshihara, Action observation circuits in the macaque monkey
K., Yoshida, Y., Takahashi, H. K., … Sadato, N. cortex. Journal of Neuroscience, 31, 3743–3756.
(2016). Structural and functional associations of Nishitani, N., & Hari, R. (2000). Temporal dynamics of
the rostral anterior cingulate cortex with subjective cortical representation for action. Proceedings of
happiness. NeuroImage, 134, 132–141. the National Academy of Sciences of the United
Mazurek, K. A., & Schieber, M. H. (2019) Mirror neu- States of America, 97(2), 913–918.
rons precede non-mirror neurons during action Nystrom, P. (2008). The infant mirror neuron system
execution. Journal of Neurophysiology, 122(6), studied with high density EEG. Social Neurosci-
2630–2635. ence, 3, 334–347.
Meletti, S., Tassi, L., Mai, R., Fini, N., Tassinari, C. A., Nyström, P., Ljunghammar, T., Rosander, K., von Hof-
& Russo, G. L. (2006). Emotions induced by intrac- sten, C. (2011). Using mu rhythm desynchroniza-
erebral electrical stimulation of the temporal lobe. tion to measure mirror neuron activity in infants.
Epilepsia, 47, 47–51. Developmental Science 14(2), 327–335.
Meltzoff, A. N., & Moore, M. K. (1989). Imitation in O’Scalaidhe, S. P., Wilson, F. A. W., & Goldman-Rakic, P.
newborn infants: Exploring the range of gestures S. (1997). Areal segregation of face-processing neu-
imitated and the underlying mechanisms. Devel- rons in prefrontal cortex. Science, 278, 1135–1138.
opmental Psychology, 25(6), 954–962. Palomero-Gallagher, N., Mohlberg, H., Zilles, K., &
Miall, R. C. (2003). Connecting mirror neurons and Vogt, B. (2008). Cytology and receptor architec-
forward models. NeuroReport, 14, 2135–2137. ture of human anterior cingulate cortex. Journal of
Morris, J. S., Frith, C. D., Perrett, D. I., Rowland, D., Comparative Neurology, 508(6), 906–926.
Young, A. W., … Dolan, R. J. (1996). A differential Palomero-Gallagher, N., Vogt, B. A., Schleicher, A.,
neural response in the human amygdala to fearful Mayberg, H. S., & Zilles, K. (2009). Receptor archi-
and happy facial expressions. Nature, 383, tecture of human cingulate cortex: Evaluation of
812–815. the four-region neurobiological model. Human
Mouraux, A., Diukova, A., Lee, M.C., Wise, R.G., & Brain Mapping, 30(8), 2336–2355.
Iannetti, G.D. (2011). A multisensory investigation Palomero-Gallagher, N., Eickhoff, S.B., Hoffstaedter,
of the functional significance of the “pain matrix.” F., Schleicher, A., Mohlberg, H., Vogt, B. A., …
NeuroImage, 54(3), 2237–2249. Zilles, K. (2015). Functional organization of human
Mukamel, R., Ekstrom, A. D., Kaplan, J., Iacoboni, subgenual cortical areas: Relationship between
M., & Fried I. (2010). Single-neuron responses in architectonical segregation and connectional het-
humans during execution and observation of erogeneity. NeuroImage, 115, 177–190.
actions. Current Biology, 20, 750–756. Pandya, D. N., & Seltzer, B. (1982). Intrinsic connec-
Murata, A., Fadiga, L., Fogassi, L., Gallese, V., Raos, V., tions and architectonics of posterior parietal cortex
& Rizzolatti, G. (1997). Object representation in the in the rhesus monkey. Journal of Comparative
ventral premotor cortex (area F5) of the monkey. Neurology, 204(2), 196–210.
Journal of Neurophysiology, 78, 2226–2230. Perrett, D. I., Harries, M. H., Bevan, R., Thomas, S.,
Murata, A., Gallese, V., Luppino, G., Kaseda, M., & Benson, P. J., Mistlin, A. J., … Ortega, J. E. (1989).
Sakata, H. (2000). Selectivity for the shape, size, Frameworks of analysis for the neural representa-
and orientation of objects for grasping in neurons tion of animate objects and actions. Journal of
of monkey parietal area AIP. Journal of Neuro- Experimental Biology, 146, 87–113.
physiology, 83(5), 2580–2601. Phillips, M.L., Young, A. W., Senior, C., Brammer, M.,
Muthukumaraswamy, S. D., Johnson, B. W., & Andrew, C., Calder, A. J., … David, A. S. (1997). A
McNair, N. A. (2004). Mu rhythm modulation specific neural substrate for perceiving facial
during observation of an object-directed grasp. expressions of disgust. Nature, 389, 495–498.
Mirror Mechanism in Cognition 303

Phillips, M. L., Young, A. W., Scott, S. K., Calder, A. somatosensory activations during observation of
J., Andrew, C., Giampietro, V., … Gray, J. A. touch. Human Brain Mapping, 30(9), 2722–2730.
(1998). Neural responses to facial and vocal Schmitt, J. J., Janszky, J., Woermann, F., Tuxhorn, I.,
expressions of fear and disgust. Proceedings of the & Ebner, A. (2006). Laughter and the mesial and
Royal Society London B, 265, 1809–1817. lateral premotor cortex. Epilepsy Behavior, 8(4),
Porter, R., & Lemon, R. (1995). Corticospinal function 773–775.
and voluntary movement. Oxford, UK: Oxford Shimada, S., & Hiraki, K. (2006). Infant’s brain
University Press. responses to live and televised action. NeuroIm-
Rizzolatti, G., Camarda, R., Fogassi, L., Gentilucci, M., age, 32(2), 930–939.
Luppino, G., & Matelli, M. (1988). Functional Simone, L., Rozzi, S., Bimbi, M., & Fogassi, L. (2015).
organization of inferior area 6 in the macaque Movement-related activity during goal-directed
monkey. II. Area F5 and the control of distal move- hand actions in the monkey ventrolateral prefron-
ments. Experimental Brain Research, 71, 491–507. tal cortex. European Journal of Neuroscience,
Rizzolatti, G., Fadiga, L., Gallese, V., & Fogassi, L. 42(11), 2882–2894.
(1996). Premotor cortex and the recognition of Simone, L., Bimbi, M., Rodà, F., Fogassi, L., & Rozzi,
motor actions. Cognitive Brain Research, 3, S. (2017). Action observation activates neurons of
131–141. the monkey ventrolateral prefrontal cortex. Scien-
Rizzolatti, G., Luppino, G., & Matelli, M. (1998). The tific Reports, 7. doi: 10.1038/srep44378
organization of the cortical motor system: New Singer, T., Seymour, B., O’Doherty, J., Kaube, H.,
concepts. Electroencephalography and Clinical Dolan, R.J., & Frith, C.D. (2004). Empathy for pain
Neurophysiology, 106(4), 283–296. involves the affective but not sensory components
Rizzolatti, G., & Luppino, G. (2001). The cortical of pain. Science, 303, 1157–1162.
motor system. Neuron, 31, 889–901. Sprengelmeyer, R., Rausch, M., Eysel, U. T., &
Rizzolatti, G., & Sinigaglia, C. (2008). Mirrors in the Przuntek, H. (1998). Neural structures associated
brain. Oxford, UK: Oxford University Press. with recognition of facial expressions of basic
Rizzolatti, G., Cattaneo, L., Fabbri-Destro, M., Rozzi, emotions. Proceedings: Biological Sciences, 265,
S. (2014). Cortical mechanisms underlying the 1927–1931.
organization of goal-directed actions and mirror Stern, D.N. (1985). The interpersonal world of the
neuron-based action understanding. Physiological infant. New York: Basic Books.
Reviews, 94(2), 655–706. Stern, D.N. (2010). Forms of vitality exploring
Rochat, M. J., Caruana, F., Jezzini, A., Escola, L., dynamic experience in psychology, arts, psycho-
Intskirveli, I., Grammont, F., … Umiltà, M.A. therapy, and development. Oxford, UK: Oxford
(2010). Responses of mirror neurons in area F5 to University Press.
hand and tool grasping observation. Experimental Strick, P. L. (1985). How do the basal ganglia and
Brain Research, 204, 605–616. cerebellum gain access to the cortical motor
Romanski, L. M., & Diehl, M. M. (2011). Neurons areas? Behavioral Brain Research, 18, 107–123.
responsive to face-view in the primate ventrolateral Tkach, D., Reimer, J., & Hatsopoulos, N. G. (2007).
prefrontal cortex. Neuroscience, 189, 223–235. Congruent activity during action and action obser-
Rozzi, S., Calzavara, R., Belmalih, A., Borra, E., Gre- vation in motor cortex. Journal of Neuroscience,
goriou, G. G., Matelli, M., & Luppino, G. (2006). 27, 13241–13250.
Cortical connections of the inferior parietal cortical Trevarthen, C. (1998). The concept and foundations of
convexity of the macaque monkey. Cerebral infant intersubjectivity. In S. Braten (Ed.), Intersub-
Cortex 16, 1389–1417. jective communication and emotion in early ontog-
Rozzi, S., Ferrari, P.F., Bonini, L., Rizzolatti, G., & eny. Cambridge, UK: Cambridge University Press.
Fogassi, L. (2008). Functional organization of infe- Umiltà, M.A., Kohler, E., Gallese, V., Fogassi, L.,
rior parietal lobule convexity in the macaque Fadiga, L., Keyzers, C., & Rizzolatti, G. (2001). I
monkey: Electrophysiological characterization of know what you are doing: A neurophysiological
motor, sensory and mirror responses and their cor- study. Neuron, 31, 155–165.
relation with cytoarchitectonic areas. European Umiltà, M. A., Escola, L., Intskirveli, I., Grammont, F,
Journal of Neuroscience, 28, 1569–1588. Rochat, M., Caruana, F., … Rizzolatti, G. (2008).
Sato, W., Yoshikawa, S., Kochiyama, T., & Mat- When pliers become fingers in the monkey motor
sumura, M. (2004). The amygdala processes the system. Proceedings of the National Academy of Sci-
emotional significance of facial expressions: An ences of the United States of America, 105,
fMRI investigation using the interaction between 2209–2213.
expression and face direction. NeuroImage, 22(2), Vogt, B.A., & Vogt, L. (2003). Cytology of human
1006–1013. dorsal midcingulate and supplementary motor
Schaefer, M., Xu, B., Flor, H., & Cohen, L. G. (2009). cortices. Journal of Chemical Neuroanatomy,
Effects of different viewing perspectives on 26(4), 301–309.
304 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Vogt, B.A., Vogt, L., Farber, N.B., & Bush, G. (2005). Wicker, B, Keysers, C, Plailly, J, Royet, JP, Gallese, V,
Architecture and neurocytology of monkey cingu- & Rizzolatti, G. (2003). Both of us disgusted in my
late gyrus. Journal of Comparative Neurology, insula: The common neural basis of seeing and
485(3), 218–239. feeling disgust. Neuron, 40(3), 655–664.
Weeks, R. A., Gerloff, C., Honda, M., Dalakas, M. C., Yoshida, K, Saito, N, Iriki, A, & Isoda, M. (2011).
& Hallett, M. (1999). Movement-related cerebellar Representation of others’ action by neurons in
activation in the absence of sensory input. Journal monkey medial frontal cortex. Current Biology, 21,
of Neurophysiology, 82, 484–488. 249–253.
19
Spatio-temporal Dynamics of
Brain Rhythms
M a r t i n S e e b e r, T h o m a s K o e n i g , a n d
Christoph M. Michel

INTRODUCTION processes has become an intense research topic in


human neuroscience (for a comprehensive review
It is commonly understood that neuronal oscilla- see (Lopes da Silva, 2013).
tions are not epiphenomena but are basic neuro- EEG oscillations occur at different frequen-
physiological mechanisms that are essential for cies. Traditionally, EEG signals are divided into
information transmission and processing in the frequency bands from infraslow (<0.2 Hz), Delta
brain (Buzsaki, 2006). Studies of local field (from 0.2 to 3.5 Hz), Theta (from 4 to 7.5 Hz),
potentials (LFP) in animals and in the human Alpha (from 8 to 13 Hz), Beta (from 14 to 30 Hz),
brain have repeatedly demonstrated that LFP Gamma (from 30 to 90 Hz), to high-frequency
oscillations contribute to information coding in oscillations (HFO; >90 Hz). While the limits of
the brain by adjusting the timing of neuronal these bands are rather artificially defined and
firing to the phase of a certain oscillation (Fries, the neurophysiological mechanisms underlying
2005; Jensen and Lisman, 2000). Neurons thereby these different rhythms is not yet well understood
form dynamical assemblies that work in syn- (Buzsaki, 2006), it is generally agreed that slow
chrony (Lopes da Silva, 1991), and multiple such oscillations tend to spread across large cortical
neuronal assemblies are usually simultaneously areas, while higher frequency oscillations are
active and constitute synchronized functional net- spatially more restricted. Low-frequency activ-
works (Dehaene et al., 1998). Synchronization of ity is related to neural mechanisms of inhibition
large-scale brain networks is thus a fundamental (Lundstrom, 2015). In non-REM sleep, low-
mechanism in the coding of information with frequency synchronization with frequent appear-
respect to perception, memory, attention, cogni- ance of large oscillations (slow waves) reflects the
tion, and action (Fries, 2015). switch between up- and down-states, a bi-stability
Given that EEG signals reflect LFPs that are that causes an impairment of causal interac-
transferred to the scalp by volume conduction, tions among brain networks and loss of neuronal
they also convey information about neuronal syn- integration and communication (Pigorini et al.,
chronization in the brain. It is therefore not sur- 2015; Sanchez-Vives and McCormick, 2000).
prising that investigations of oscillations of the Oscillations in the theta and alpha range regu-
spontaneous EEG and their relation to cognitive late information transfer between cortical areas
306 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

such as the hippocampus and the cortex for theta assumption is not correct: the EEG exhibits not
(Mizuseki et al., 2009) and the thalamus and the only spectral, but also spatial signatures which
cortex for alpha (Lopes da Silva et al., 1980). High change dynamically over time in a sub-second
frequency oscillations allow for binding informa- range (Michel, 2019). A simple examination of the
tion among neuronal assemblies (Gray et al., spati-temporal dynamic of a filtered EEG clearly
1989). A comprehensive discussion of the func- shows that the spatial configuration of the scalp
tional connotation of the different frequency bands electric field is not stable over time. It changes
can be found in (Lopes da Silva, 2013). dynamically in the sub-second range (Figure 19.1).
Since different map configurations are by physical
laws due to differences in the underlying genera-
tors (Michel and Murray, 2012), the assumption of
spatial stationarity of the generators producing a
SPATIO-TEMPORAL DYNAMICS OF certain oscillation over longer time periods is inva-
BRAIN RHYTHMS lid. Therefore, changes of frequency power at one
specific electrode could be due to changes of the
The traditional analysis of oscillations in the oscillation of one specific brain network, or due to
ongoing spontaneous EEG is the spectral analyses changes of the network configuration that leads to
of the signal at a given electrode using Fast a different projection of the activity at the given
Fourier Transform and calculating the power of electrode.
the different frequency bands (Lopes da Silva and However, assuming that there is a limited set
Mars, 1987). In multichannel recordings, the of brain functions that explain some interesting
quantitative EEG (qEEG) approach consists of psycho-physiological correlates and assuming
calculating the power spectrum for each channel that these functions materialize as spatially and
and then constructing power maps averaged temporally well-defined networks of brain electric
within a given frequency band and comparing activity, it follows from the physics of EEG and
them topographically between experimental or volume conduction that there must be an equally
pathological conditions (Duffy, 1985; John and limited set of spatially defined EEG components
Prichep, 2006). While this analysis has undoubt- that correspond, in their dynamics and in their
edly led to a series of clinically relevant findings, spatial distribution, to those functions and net-
it contains numerous pitfalls that prevent non- works. Unfortunately, this inverse problem (i.e.,
ambiguous conclusions about the neurophysiolog- decomposing the spatio-temporal components
ical mechanisms underlying observed spectral of the EEG) generally has no unique solution.
power changes. Accordingly, much of the efforts in EEG analysis
First, the observation of oscillatory power is has gone in the development and empirical evalu-
dependent on the reference electrode because ation of priors that allow identifying these compo-
the recorded voltages in EEG are always differ- nents (Blankertz et al., 2007; Makeig et al., 1996;
ences between two leads. Brain signals originat- Parra et al., 2005; Wu et al., 2020).
ing in specific locations are propagated to all scalp Many of the methods developed to tackle this
electrodes mostly dependent on their distance and problem of meaningfully decompose an EEG sig-
source orientation because of volume conduction nal can be framed as the identification and appli-
(Fender, 1987). Given the reference dependence cation of particular spatial filters (Michel and
and volume conduction, the spatial association of Pascual-Leone, 2020): Such spatial filters are ide-
oscillatory power at single electrodes is thus not ally designed to suitably compute signal dynamics
straightforward and experimental effects observed associated with some functional network. These
at a single electrode may eventually have very dif- signal dynamics are computed as weighted sum of
ferent, and spatially remote origins in brain space the recorded voltages at a given time sample. The
(Michel et al., 2009). This implies that the spatial spatial filters are composed of the weights for each
interpretation of oscillations (e.g., by frequency recording electrode. In other words, weights are
analyses) should be handled with great care. used to transform the data from recording space
Second, the spectral analysis of single chan- into component space. However, weights of spatial
nel EEG implicitly assumes that the generators filters should not be physiologically interpreted.
of the oscillations remain the same across time For example, large positive and negative weights
and between conditions and thus that differences at certain electrodes could only serve the cancel-
in spectral power between conditions are due to lation or suppression of unwanted signals such as
differences in the oscillations of the same gen- noise. The recorded signals can be described by
erators in the brain. This assumption also under- the summation of the signal components multi-
lies the qEEG approach that considers the power plied with their spatial patterns. Spatial patterns
map averaged over time as spatially stable. This (Figure 19.2) which are applied to back-project
Spatio-temporal Dynamics of Brain Rhythms 307

Figure 19.1 Spatial non-stationarity of Alpha-EEG. Conventional frequency power analy-


sis assumes that the generators of the oscillatory activity remain the same over time. This
assumption is not valid as illustrated here: The top shows the spectral power of a 30-sec,
eyes-closed EEG calculated by a wavelet transformation and averaged over all channels. It
shows typical Alpha bursts at different time periods. Potential maps at some of the peaks of
Alpha activity are plotted below. Obviously, the topography of the maps at different time
points are very different, which signifies different networks generating the Alpha activity
at these time points. Even within a short period of 1-sec EEG, the topographies vary as illus-
trated on the bottom where all maps during a period of one Alpha burst are shown (high-
lighted in the time-frequency plot and on the EEG of one electrode during this period). This
example illustrates that even during an alpha burst, map topographies change, indicating
changes in the underlying generators.
Source: From Michel, 2019.

signal components into recording space can be appropriate as their optimization criterion is con-
physiologically meaningful (Blankertz et al., sidering the experimental design and related data.
2007; Cohen, 2021; Haufe et al., 2014; Parra et al., Two prominent families of spatial decomposi-
2005; Wu et al., 2020). tions are Independent Component Analysis (ICA)
Yet, there are plural ways to determine spatial and Generalized Eigenvalue Decompositions
filters and hence decompose recorded signals into (GED). ICA aims for statistical independence
specific components. Any decomposition is based between the resulting components and therefore
on assumptions of the data and resulting decom- is data-driven. ICA decomposition typically yields
posed components. Given the limited dimension- to dipolar scalp maps and are thought to isolate
ality of the data there is no unique solution without single brain sources while minimizing volume
introducing additional priors. Different decompo- conduction effects (Delorme et al., 2012).
sition methods have different optimization criteria GED is hypothesis-driven and optimizes signal
determining the spatial filters and resulting sig- differences between separate sets of data (e.g.,
nal components. Therefore, each decomposition experimental conditions; Cohen, 2021; Parra et al.,
method and associated spatial filters are only as 2005). So, the spatial patterns resulting from GED
308 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 19.2 Illustration of the spatial filtering of the EEG. The EEG is filtered in the Alpha
Band and the EEG microstates are defined using k-means cluster analysis. The Alpha-filtered
EEG is then spatially filtered for each microstate map. As can be seen, the Alpha power as
well as the temporal dynamics are distinctly different for the five spatially filtered signals.
Source localization of these spatially filtered signals allows to determine the networks
underlying these different oscillations.
Source: Adapted from Michel and Pascual-Leone, 2020.

can signify both single and distributed underlying a milliseconds level (e.g., by their instantaneous
sources. In the case where multiple well-separate amplitude or phase – see Figure 19.3).
cortical patches could be identified as sources of Within this ongoing debate on suitable priors
GED patterns, that would indicate a distributed for the decomposition of EEG signal, the con-
functional network oscillating synchronously at cept of so-called microstates has become increas-
zero-phase lags. ingly successful over the last two decades of EEG
In summary, diverse spatial decomposition research (Michel and Koenig, 2018). In brief, this
approaches have different inherent properties that concept is driven by the point that at the level
should be considered while interpreting their sig- where scalp EEG is recorded, the driving pro-
nal components and spatial patterns (Parra et al., cesses that determine most of the data are func-
2005). The main advantage of spatial filtering is tional state dynamics that affect the brain on its
that resulting signal components are “unmixed” upmost, and therefore global scale. This implies
following the criterion of the particular decompo- that any (sufficiently large) change in the overall
sition method. Moreover, the spatial configuration spatial configuration of brain electric activity and
of these signals are well-defined and spatial pat- thus any (sufficiently large) change in scalp field
terns can be used to estimate the underlying brain configuration implies a change in the global func-
structures generating these patterns and associated tional state of the brain. Framed otherwise, the
signal components. Importantly, all of these spa- microstate approach postulates that there is, for
tial filter approaches imply that the sources of the conceptual reasons, no overlap among the suitably
putatively isolated processes are either spatially identifiable components of an EEG.
compact (i.e. close to dipoles) or share a highly Starting from this a-priori rationale, it turns
correlated or anticorrelated time course (Michel out that empirically, these changes in global
and Koenig, 2018). Thus, while recordings at functional state are discontinuous and separated
single electrodes contain the summation of many by transient periods of quasi stability (Lehmann
signals with distinct spatial origin, frequency anal- et al., 1987), which led to the term “microstates,”
yses that are performed typically on time windows and that in addition, there seems to be a relatively
of seconds could “wash out” the spatio-temporal low number of prototypical microstate configura-
dynamics on the milliseconds level. However, suit- tions that can be efficiently identified using spa-
ably unmixed signal components are associated tial clustering algorithms (Pascual-Marqui et al.,
with well-defined spatial configurations and there- 1995). The approach to describe spontaneous EEG
fore are more appropriate to study oscillations on as series of short-lasting periods of quasi-stable
Spatio-temporal Dynamics of Brain Rhythms 309

Figure 19.3 Decomposition of two epochs of 30-sec multichannel EEG during eyes-closed
wakeful resting and during sleep onset into oscillatory events that are characterized as a
network of synchronously active sources (components) that produce prototypical scalp maps
(left row) and that are separated among themselves by time, frequency, or phase. The second
column shows the energy distribution of these classes of oscillatory EEG events as a function
of time and frequency. The third row shows the reconstructed time-course of these compo-
nents, and the last row their frequency distribution.

field configuration that can be assigned to one of and mental disorders and various cognitive states
class of a low number of prototypical configu- (Bréchet et al., 2019; da Cruz et al., 2020; Khanna
rations has, over the last two decades, lead to a et al., 2015; Michel and Koenig, 2018; Zanesco
rapidly growing empirical body of findings on et al., 2020).
the functional role of these prototypical states in On the other side, the microstate approach to
the context of brain development, neurological the EEG decomposition problem, as it is currently
310 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

implemented, can be criticized as it has been multiple recording sites allows to study the relation
applied almost exclusively to the time domain. between them. Therefore, it is possible to investi-
This implies that at any given moment in time, gate the connectivity between sites in addition to
there is only one, eventually broad frequency- the local, frequency-specific power activity.
spectrum event that ought to account for the given Despite these mere methodological considerations,
data. This clearly runs against the at least equally on a more conceptual level, connectivity metrics
successful strategy in EEG research to decom- are relevant to assess synchrony between sites,
pose the data into a temporally overlapping set which is thought as key-mechanism for grouping
of processes that are separated by frequency, as neuronal populations into assemblies.
it is frequently and very successfully done e.g. in Gray et al. (1989) showed that neurons across
sleep research (Borbély, 1982), pharmaco-EEG different columns of the cat visual cortex syn-
(Herrmann et al., 1989; Jobert et al., 2012), or chronized their oscillatory responses specifically
when analyzing event related spectral changes of to visual stimuli with matching features (i.e., ori-
EEG (Makeig, 2002; Pfurtscheller and Lopes da entation and movement direction). In a follow-up
Silva, 1999). In other words, the assumption that study, Engel et al. (1991) provided evidence that
for the processes to be identified as not overlap- neurons in the left and right hemisphere syn-
ping, the claim that they must not overlap in time chronized and therefore bind features within and
may turn out to be overly strong and force what a between visual hemifields. Importantly, inter-
microstate must represent to include a package of hemispheric synchronization was dependent on
processes that certainly commonly form an overall corticocortical connections, because it became
global state, but that may nevertheless also func- absent after dissection of the corpus callosum. In
tionally segregate in interesting ways, and that both of these seminal studies, synchronization was
may be separable by their spectral distribution. present in the Gamma range with zero phase dif-
Fortunately, this apparent dilemma between ferences between distant neuronal populations on
two very successful, but seemingly incompatible average.
approaches to the problem of decomposing the More recently, oscillatory synchrony in the Beta
EEG can be avoided by relaxing the “no overlap” range differed for specific rules in the prefrontal
priority of microstate analysis from the traditional cortex in non-human primates (Buschman et al.,
“no overlap in time of momentary maps”, to “no 2012). Furthermore, spike-field synchrony, a met-
overlap in time, frequency and phase of transient ric for LFP phase-locked spiking of neurons for
oscillations” (Koenig et al., 2001; Studer et al., a given frequency, revealed that Beta oscillations
2006). As a result, this time-frequency approach selected neurons specifically for distinct rules.
decomposes an ongoing EEG into a set of tran- Therefore, Beta oscillatory synchrony was sug-
sient oscillations that are each uniquely defined by gested to form rule-specific neural ensembles dur-
their joint distribution in time and frequency, and ing decision making. These results are moreover
that again assumingly join all the EEG correlates in favor for the idea that oscillations contribute to
of the different elements of the brain networks that the grouping of spatially separated neurons into
produce each of these oscillations into a common ensembles selectively for distinct representations.
event (Figure 19.3). Combining resting state EEG Given these seminal invasive studies in ani-
and fMRI data, Schwab et al could demonstrate mals showing oscillatory synchrony between dis-
that the spontaneous fluctuations such synchro- tant neurons and LFP oscillations contributing to
nized cortical oscillations seem to be associated establish synchrony, it is of relevance to translate
with the activity of particular thalamic subsystems these findings to the human brain. For example,
(Schwab et al., 2015). Beta band synchronization between distributed
cortical regions predicted the perception in sub-
jects responding to ambiguous audiovisual stimuli
in and MEG experiment (Hipp et al., 2011) and
attention modulated interregional synchronization
CONNECTIVITY MEDIATED BY in a frequency-specific manner along the visual
BRAIN RHYTHMS pathway (Siegel et al., 2008).
Interregional co-activity was consistently
In the last decades, invasive multi-site recordings reported by another line of research studying
are increasingly available along with non-invasive blood oxygen level dependent (BOLD) signals on
high-density EEG. These developments open up a much slower time scale in humans during rest,
novel possibilities in studying long-range interac- periods of minimal sensory input without specific
tions between different sites of the brain. Instead of task instructions (Biswal et al., 1995; Raichle,
determining the frequency and power of a certain 2010; Smith et al., 2009). Intrinsic fluctuations
brain rhythm at a single recording electrode, form meaningful, so-called resting-state networks
Spatio-temporal Dynamics of Brain Rhythms 311

(RSN), between distant but functionally coupled the scalp. To distinguish saccadic artifacts from
brain regions. genuine gamma band responses approaches such
To investigate the electrophysiological basis of as topographic and source analyses or ICA were
RSN, several groups investigating the large-scale suggested (Carl et al., 2012; Keren et al., 2010).
networks derived either from EEG or MEG using Indeed, Hassler et al. (2011) showed induced
source reconstruction (Hipp et al., 2012; Mantini gamma band responses related to object recogni-
et al., 2007; Samogin et al., 2019) or directly from tion after controlling for microsaccadic artifacts.
intracranial recordings (Hacker et al., 2017; He Therefore, high-frequency oscillations in human
et al., 2008; Kucyi et al., 2018). EEG/MEG source EEG are suitable for the investigation of cogni-
reconstruction provides full coverage of the brain, tive processes after appropriate control for artifact
but the spatial resolution of these techniques is contamination.
moderate. Invasive recordings provide more direct
measures of local brain fluctuations but are limited
to the electrodes implantation sites.
These studies either confirmed or showed par-
allels between RSN and electrophysiological field ZERO-PHASE SYNCHRONIZATION
potentials. Based on the much slower time scale of BEYOND VOLUME CONDUCTION
BOLD fluctuation, most studies either focused on
infraslow fluctuation or frequency-specific enve- Studies based on invasive recordings in humans
lopes to investigate the electrophysiological basis provided substantial insight into large-scale cou-
of RSN. Although envelope correlation is capable pling related to RSN recordings (Hacker et al.,
to quantify inter-regional functional coupling, 2017; He et al., 2008; Kucyi et al., 2018).
it does not necessarily express oscillatory phase However, these studies investigated infraslow
synchrony between these regions. Taken together, dynamics or amplitude coupling. Interregional
phase synchrony and envelope correlation are two phase synchrony is less studied in humans, mostly
distinct coupling modes in terms of their time because of spurious correlations confounding
scale, neural mechanisms and putative functions genuine zero-phase relations in non-invasive
(Engel et al., 2013). EEG/MEG recordings.
Motivated by invasive studies in animals show- While volume conduction is generally the rea-
ing quasi zero-phase relations (Andreas et al., son why we can measure brain activities using
1991; Campo et al., 2019; Roelfsema et al., 1997) scalp EEG it inherently leads to spurious corre-
between distant regions several groups investi- lations between scalp recordings. These spuri-
gated synchrony in humans related to stimulus ous correlations impact scalp-based connectivity
perception and feature binding (Rodriguez et al., estimates considerably (Brunner et al., 2016; Van
1999; Srinivasan et al., 1999; Tallon-Baudry et al., de Steen et al., 2017). Consequently, connectiv-
1996). ity analyses following source estimation from
As with previous animal studies, increased multi-channel EEG/MEG were proposed to pro-
gamma coherence and power recorded by scalp vide more meaningful and spatially interpretable
EEG was suggested to visual feature binding results (He et al., 2019; Michel and He, 2018).
and perception (Rodriguez et al., 1999; Tallon- Although to a lesser extent, spurious correla-
Baudry et al., 1996). However, Yuval-Greenberg tions are still present in source reconstructed sig-
and Deouell (2009), as well as Yuval-Greenberg nals, because of the limited spatial resolution of
et al. (2008), raised some awareness that minia- EEG/MEG recording. When the number of sources
ture saccadic eye movements result in transient being estimated extents the number of recording
gamma band activity that overlap with genuine, sensors, redundancy in source reconstructions is
narrow-banded gamma responses in the scalp inevitable. One approach to correct for spurious
EEG. Consequently, electrical activity produced correlation confounding connectivity estimates is
by microsaccades confound gamma band activity. eliminating instantaneous phase relations between
Following these concerns, (micro)saccadic signals investigated. Because volume conduction
eye artifacts were thoroughly characterized. is instantaneous, only signal components orthogo-
The appearance of microsaccadic artifacts at nal to each other could be investigated to dimin-
parietal and occipital electrodes can be caused ish spurious effects (Colclough et al., 2015; Hipp
by an unfortunate choice of the reference elec- et al., 2012; Nolte et al., 2004).
trode at the nose or, more generally, electrodes However, this approach comes with the cost
close to the eyes. Second, volume conduction of ignoring genuine zero-phase relations, which
of the head’s biological tissues propagates elec- previously were proposed as key-mechanism to
trical signals to some extent to all electrodes on group distant neuronal populations. Phase-lagged
312 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

relations can be investigated after orthogonaliza- By investigating the instantaneous phase rela-
tion while diminishing spurious relations. Yet, tion between the network nodes, we quantified the
phase-lagged metrics are sensitive to investigate time course of synchronization. These analyses
directional interactions (e.g., feedforward and suggest that the manifestation of simultaneous
feedback interactions between different regions synchronous networks is briefly stable, not persis-
along the visual hierarchy following a stimu- tently. Given the flexible spatial reconfiguration of
lus). Meaning phase-lagged connectivity metrics distinct networks, simultaneous synchronization
capture rather the transmission of information might be instrumental to the establishment of par-
between different processing stages than the dis- ticular, re-occurring states. If a particular source
tributed coherent representation of information network configuration maintains quasi-zero phase
and global states of the brain (Ju and Bassett, relations for a certain period, that necessarily
2020). leads to a stable topography of the scalp potential
Moreover, if all zero-phase relations that are field. EEG microstates are defined as topographies
present in multichannel recordings would result being briefly stable in time. Therefore, the brief
from volume conduction, that would imply that establishment of specific quasi zero-lag-related
only one active source in the brain is active, which network patterns can be seen as source dynam-
is highly implausible from a physiological point ics underlying the scalp-level observation of
of view. microstates.
In addition to invasive animal literature, recent Based on simulated and real data, Sjogard et al.
studies indeed demonstrate inter-regional zero- (2019) demonstrated the relevance of quasi zero-
phase relation that cannot be explained by volume lag synchronization between posterior midline cor-
conduction (O’Reilly and Elsabbagh, 2021; Seeber tices of the default-mode network. Interestingly,
and Michel, 2021). O’Reilly and Elsabbagh (2021) posterior midline synchrony was only present
used intracranial recordings to explicitly investi- when using distributed source reconstruction, but
gate zero-lag connectivity between distant brain absent when using beamformers. Beamforming
regions. They found zero-phase relations between minimizes linear correlation between source
homotopic brain areas but not between heterotopic reconstructions (Darvas et al., 2004; Van Veen
areas. Furthermore, zero-lag functional connec- et al., 1997), which leads to underestimating of
tion were found between distant regions that are the well-established coupling of posterior midline
>100 mm apart. In intracranial recordings, volume areas. Furthermore, while comparing functional
conduction does not confound signals on such MRI with source reconstructed EEG networks,
large distances. So, it is highly unlikely that these Rizkallah et al. (2020) showed that orthogonaliza-
results are an effect of residual volume conduc- tion is considerably reducing correlation between
tion. In addition, recent findings combining EEG these different modalities (Figure 19.4).
and PET data in patients with dementia clearly Taken together, zero-phase relations are physi-
point at the beneficial functional role of zero- ological meaningful and should not be ignored.
phase, as opposed to lagged interactions among To date, to the best of our knowledge no appro-
brain regions (Smailovic et al., 2020). priate leakage correction algorithm is existing.
Taken another angle on the same matter, we sys- Orthogonalization approaches do have limita-
tematically compared simultaneous (i.e., zero-lag, tions (Palva et al., 2018) and do ignore all zero-
synchronization with the residual bias from spatial phase relations, which is not desirable for many
smoothness of EEG source estimates). Based on studies as outlined above. Therefore, we propose
these analyses we indeed report simultaneous syn- either experimental or statistical assessment of
chronization exceeding confounds resulting from zero-phase interaction being beyond the level of
volume conduction (Seeber and Michel, 2021). In volume conduction in order to distinguish genuine
this study, we focused on the investigation of two from spurious effects.
different networks spanning over frontal versus
parietal brain areas. We found simultaneous syn-
chronization between homologous brain areas of
the two different hemispheres. These functional
networks were not limited to two notes, but typi- SUMMARY AND CONCLUSIONS
cally included at least one medial node (Figure
19.4). The finding of quasi zero-lags between Multichannel scalp EEG recordings allow to
distant areas could signify the functional conver- investigate neuronal oscillations at the whole brain
gence of involved regions during rest, where little level and thus provide a window into the basic
sensory input is perturbing intrinsic oscillations neurophysiological mechanisms underlying com-
minimally. munication between remote brain areas. EEG
provides temporal, spectral and spatial information
Spatio-temporal Dynamics of Brain Rhythms 313

Figure 19.4 Zero-phase synchronization beyond volume conduction. A: Simultaneous, i.e.,


zero-lag synchronization, between distant brain regions exceeds the residual bias from spa-
tial smoothness of EEG source estimates. B: Coupling of posterior midline areas that are pre-
sent in distributed source solutions (MNE) but are underestimated by beamformers (LCMV)
that minimize linear correlation between source reconstructions. C: Zero-phase relations are
present in intracranial recordings in homotopic, distant regions, as shown by introducing
an offset of 1 sample before computing phase lag index (PLI), which is insensitive to zero-
phase relations otherwise. D: Ignoring zero-phase relations significantly reduces the correla-
tion of functional networks derived from EEG and functional MRI. Functional EEG networks
were estimated using the Phase Locking Value (PLV), PLI, and multivariate orthogonalization
(PLVCol) and then compared to fMRI networks. Correlation between different modalities
(EEG and fMRI) is higher for PLV measures that are sensitive to zero-lag relations compared
to metrics ignoring zero-phase relations (i.e., PLI and PLVCol).
Source: Panels A–D adapted from (Seeber and Michel, 2021a), (Sjogard et al., 2019), (O’Reilly and Elsabbagh, 2021) and
(Rizkallah et al., 2020) respectively.

about the functioning of large-scale brain net- A second important point to consider is volume
works. While this makes EEG a potentially power- conduction. A common pitfall in the interpreta-
ful technique to study brain neuronal dynamics, tion of the results of scalp EEG analysis is the
the different dimensions lead to complex signals assumption that the activity recorded at a certain
on the scalp that need to be properly understood, electrode is generated by the neuronal population
analyzed, and interpreted. underlying this electrode. Due to volume conduc-
A first important fact to consider is that brain tion neuronal activity spreads to the scalp surface
network dynamics is extremely fast and network in all directions and, consequently, activity of a
configurations change in the sub-second time given brain area is captured by all electrodes to a
scale. Assuming stationarity in time and in space, certain extent, depending on distance and orien-
as conventional frequency analysis methods do, tation. Interpretation of brain areas generating a
is not justified and leads to erroneous interpreta- given scalp recording is only possible by solving
tion. Decomposing the signals using spatial filters the so-called inverse problem (i.e., by converting
followed by time-resolved frequency analysis can the signals from the sensor to the source-space).
overcome this problem. Powerful distributed inverse solutions based on
314 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

realistic head-models provide reliable estimations spatiotemporal dynamics of self-generated, task-


of the brain sources leading to the scalp recordings. initiated thoughts with EEG and fMRI. NeuroIm-
A third aspect concerns the study of functional age, 194, 82–92.
connectivity between brain areas using scalp Brunner, C., Billinger, M., Seeber, M., Mullen, T. R., &
EEG measures. Obviously, due to the spread of Makeig, S. (2016). Volume conduction influences
the neuronal activity to all scalp electrodes and scalp-based connectivity estimates. Frontiers in Com-
the reference-dependency, functional connectiv- putational Neuroscience, 10. doi: 10.3389/fncom.
ity analysis on the scalp-level is difficult to inter- 2016.00121
pret. After converting the data to the source-space, Buschman, T. J., Denovellis, E. L., Diogo, C., Bullock,
interpretation becomes more meaningful. Still, D., & Miller, E. K. (2012). Synchronous oscillatory
volume conduction leads to spatial leakage. One neural ensembles for rules in the prefrontal cortex.
way to overcome this problem is to ignore simul- Neuron, 76(4), 838–846.
taneous (zero-phase) synchronization when ana- Buzsaki, G. (2006). Rhythms of the brain. Oxford,
lyzing functional connectivity between different UK: Oxford University Press.
nodes. This, however, ignores the well-known fact Campo, A. T., Vázquez, Y., Álvarez, M., Zainos, A.,
that genuine zero-phase synchronization plays an Rossi-Pool, R., Deco, G., & Romo, R. (2019). Feed-
important functional role in information repre- forward information and zero-lag synchronization
sentation, maintenance, and processing. Methods in the sensory thalamocortical circuit are modu-
that control for leakage are needed in order to lated during stimulus perception. Proceedings of
disentangle genuine from spurious simultaneous the National Academy of Sciences of the United
synchronizations. States of America, 116(15), 7513–7522.
A fourth aspect concerns temporal coding of Carl, C., Açık, A., König, P., Engel, A. K., & Hipp, J. F.
information beyond frequency synchronization. (2012). The saccadic spike artifact in MEG. Neuro-
An interesting observation in broad-band EEG is Image, 59(2), 1657–1667.
the fact that the spatial configuration of the scalp Cohen, M. X. (2021). A tutorial on generalized
potential fields remains stable for a few tenths eigendecomposition for denoising, contrast
of milliseconds (80–120 ms) and then quickly enhancement, and dimension reduction in multi-
changes to a new configuration in which it remains channel electrophysiology. NeuroImage, 247. doi:
stable again. This empirical observation led to 10.1016/j.neuroimage.2021.118809
the concept of EEG microstates as basic build- Colclough, G. L., Brookes, M. J., Smith, S. M., &
ing blocks of conscious mentation. An increasing Woolrich, M. W. (2015). A symmetric multivariate
number of studies have demonstrated the sensitiv- leakage correction for MEG connectomes. Neuro-
ity of the temporal dynamics of specific micro- Image, 117, 439–448.
states to the functional state of the brain, to the da Cruz, J. R., Favrod, O., Roinishvili, M., Chkonia, E.,
content of the ongoing mental process, to the level Brand, A., Mohr, C., … Herzog, M. H. (2020). EEG
of consciousness, and, most importantly, to differ- microstates are a candidate endophenotype for
ent neuropsychiatric pathologies. EEG microstates schizophrenia. Nature Communications, 11(1).
offer to study the syntax of flow of mental pro- doi: 10.1038/s41467-020-16914-1
cesses that is not necessarily controlled by syn- Darvas, F., Pantazis, D., Kucukaltun-Yildirim, E., &
chronization in specific frequencies. Leahy, R. (2004). Mapping human brain function
with MEG and EEG: methods and validation. Neu-
roImage, 23, S289–S299.
Dehaene, S., Kerszberg, M., & Changeux, J. P.
(1998). A neuronal model of a global workspace
REFERENCES in effortful cognitive tasks. Proceedings of the
National Academy of Sciences of the United States
Biswal, B., Yetkin, F. Z., Haughton, V. M., & Hyde, J. S. of America, 95(24), 14529–14534.
(1995). Functional connectivity in the motor cortex Delorme, A., Palmer, J., Onton, J., Oostenveld, R., &
of resting human brain using echo-planar MRI. Makeig, S. (2012). Independent EEG sources are
Magnetic Resonance in Medicine, 34(4), 537–541. dipolar. PLoS One, 7(2). doi:10.1371/journal.pone.
Blankertz, B., Tomioka, R., Lemm, S., Kawanabe, M., 0030135
& Muller, K.-R. (2007). Optimizing spatial filters for Duffy, F. H. (1985). The BEAM method for neuro-
robust EEG single-trial analysis. IEEE Signal Pro- physiological diagnosis. Annals of the New York
cessing Magazine, 25(1), 41–56. Academy of Sciences, 457, 19–34.
Borbély, A. A. (1982). A two process-model of sleep Engel, A. K., Gerloff, C., Hilgetag, C. C., & Nolte, G.
regulation. Human Neurobiology, 1(3), 195–204. (2013). Intrinsic coupling modes: multiscale inter-
Bréchet, L., Brunet, D., Birot, G., Gruetter, R., actions in ongoing brain activity. Neuron, 80(4),
Michel, C. M., & Jorge, J. (2019). Capturing the 867–886.
Spatio-temporal Dynamics of Brain Rhythms 315

Engel, A. K., König, P., Kreiter, A. K., & Singer, W. Jobert, M., Wilson, F. J., Ruigt, G. S., Brunovsky, M.,
(1991). Interhemispheric synchronization of oscil- Prichep, L. S., Drinkenburg, W. H., … IPEG Phar-
latory neuronal responses in cat visual cortex. Sci- maco-EEG Guidelines Committee (2012). Guidelines
ence, 252, 1177–1179. for the recording and evaluation of pharmaco-EEG
Fender, D. (1987). Source localization of brain electri- data in man: The International Pharmaco-EEG Soci-
cal activity. In A. S. G. A. Remond (Ed.), Methods ety (IPEG). Neuropsychobiology, 66(4), 201–220.
of analysis of brain electrical and magnetic signals John, E. R., & Prichep, L. S. (2006). The relevance of
(Vol. 1). Amsterdam: Elsevier. QEEG to the evaluation of behavioral disorders
Fries, P. (2005). A mechanism for cognitive dynamics: and pharmacological interventions. Clinical EEG
Neuronal communication through neuronal coher- and Neuroscience, 37(2), 135–143.
ence. Trends in Cognitive Science, 9(10), 474–480. Ju, H., & Bassett, D. S. (2020). Dynamic representa-
Fries, P. (2015). Rhythms for cognition: Communica- tions in networked neural systems. Nature Neuro-
tion through coherence. Neuron, 88(1), 220–235. science, 23(8), 908–917.
Gray, C. M., Konig, P., Engel, A. K., & Singer, W. Keren, A. S., Yuval-Greenberg, S., & Deouell, L. Y.
(1989). Oscillatory responses in cat visual cortex (2010). Saccadic spike potentials in gamma-band
exhibit inter-columnar synchronization which reflects EEG: Characterization, detection and suppression.
global stimulus properties. Nature, 338, 334–337. NeuroImage, 49(3), 2248-2263.
Hacker, C. D., Snyder, A. Z., Pahwa, M., Corbetta, Khanna, A., Pascual-Leone, A., Michel, C. M., &
M., & Leuthardt, E. C. (2017). Frequency-specific Farzan, F. (2015). Microstates in resting-state EEG:
electrophysiologic correlates of resting state fMRI current status and future directions. Neuroscience
networks. NeuroImage, 149, 446–457. and Biobehavioral Reviews, 49, 105–113.
Hassler, U., Barreto, N. T., & Gruber, T. (2011). Koenig, T., Marti-Lopez, F., & Valdes-Sosa, P. (2001).
Induced gamma band responses in human EEG Topographic time-frequency decomposition of the
after the control of miniature saccadic artifacts. EEG. NeuroImage, 14(2), 383–390.
NeuroImage, 57(4), 1411–1421. Kucyi, A., Schrouff, J., Bickel, S., Foster, B. L., Shine,
Haufe, S., Meinecke, F., Görgen, K., Dähne, S., J. M., & Parvizi, J. (2018). Intracranial electrophysi-
Haynes, J.-D., Blankertz, B., & Bießmann, F. (2014). ology reveals reproducible intrinsic functional con-
On the interpretation of weight vectors of linear nectivity within human brain networks. Journal of
models in multivariate neuroimaging. NeuroIm- Neuroscience, 38(17), 4230–4242.
age, 87, 96–110. Lehmann, D., Ozaki, H., & Pal, I. (1987). EEG alpha
He, B., Astolfi, L., Valdes-Sosa, P. A., Marinazzo, D., map series: brain micro-states by space-oriented
Palva, S., Benar, C. G., … Koenig, T. (2019). Elec- adaptive segmentation. Electroencephalography
trophysiological brain connectivity: Theory and and Clinical Neurophysiology, 67, 271–288.
implementation. IEEE Transactions on Biomedical Lopes da Silva, F. H. (1991). Neural mechanisms
Engineering. doi: 10.1109/TBME.2019.2913928 underlying brain waves: From neural membranes
He, B. J., Snyder, A. Z., Zempel, J. M., Smyth, M. D., to networks. Electroencephalography and Clinical
& Raichle, M. E. (2008). Electrophysiological cor- Neurophysiology, 79(2), 81–93.
relates of the brain’s intrinsic large-scale functional Lopes da Silva, F. H. (2013). EEG and MEG: relevance
architecture. Proceedings of the National Academy to neuroscience. Neuron, 80(5), 1112–1128.
of Sciences of the United States of America, Lopes da Silva, F. H., & Mars, N. J. I. (1987). Paramet-
105(41), 16039–16044. ric methods in EEG analysis. In A. S. Gevins & A.
Herrmann, W. M., Abt, K., Coppola, R., Etevenon, E. Rémond (Eds.), Methods of analysis of brain elec-
T., Ferber, G., Fink, M., … Scheuler, W. (1989). trical and magnetic signals (pp. 243–260). Amster-
International Pharmaco-EEG Group (IPEG). Recom- dam: Elsevier.
mendation for EEG and evoked potential map- Lopes da Silva, F. H., Vos, J. E., Mooibroek, J., & Van
ping. Neuropsychobiology, 22, 170–176. Rotterdam, A. (1980). Relative contributions of
Hipp, J. F., Engel, A. K., & Siegel, M. (2011). Oscillatory intracortical and thalamo-cortical processes in the
synchronization in large-scale cortical networks generation of alpha rhythms, revealed by partial
predicts perception. Neuron, 69(2), 387–396. coherence analysis. Electroencephalography and
Hipp, J. F., Hawellek, D. J., Corbetta, M., Siegel, M., Clinical Neurophysiology, 50(5–6), 449–456.
& Engel, A. K. (2012). Large-scale cortical correla- Lundstrom, B. N. (2015). Modeling multiple time
tion structure of spontaneous oscillatory activity. scale firing rate adaptation in a neural network of
Nature Neuroscience, 15(6), 884–890. local field potentials. Journal of Computational
Jensen, O., & Lisman, J. E. (2000). Position recon- Neuroscience, 38(1), 189–202.
struction from an ensemble of hippocampal place Makeig, S. (2002). Response: Event-related brain
cells: contribution of theta phase coding. Journal dynamics: Unifying brain electrophysiology. Trends
of Neurophysiology, 83(5), 2602–2609. in Neurosciences, 25(8), 390.
316 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Makeig, S., Bell, A., Jung, T.-P., & Sejnowski, T. J. Pfurtscheller, G., & Lopes da Silva, F. H. (1999). Event-
(1996). Independent component analysis of elec- related EEG/MEG synchronization and desynchro-
troencephalographic data. Advances in neural nization: basic principles. Clinical Neurophysiology,
information processing systems, 8, 145–151. 110(11), 1842–1857.
Mantini, D., Perrucci, M. G., Del Gratta, C., Romani, Pigorini, A., Sarasso, S., Proserpio, P., Szymanski, C.,
G. L., & Corbetta, M. (2007). Electrophysiological Arnulfo, G., Casarotto, S., … Massimini, M.
signatures of resting state networks in the human (2015). Bistability breaks-off deterministic
brain. Proceedings of the National Academy of responses to intracortical stimulation during non-
Sciences of the United States of America, 104(32), REM sleep. NeuroImage, 112, 105–113.
13170–13175. Raichle, M. E. (2010). Two views of brain function.
Michel, C. M. (2019). High-resolution EEG. Hand- Trends in Cognitive Sciences, 14(4), 180–190.
book of Clinical Neurology, 160, 185–201. Rizkallah, J., Amoud, H., Fraschini, M., Wendling, F.,
Michel, C. M., & He, B. (2018). EEG Mapping and & Hassan, M. (2020). Exploring the correlation
Source Imaging. In D. L. Schomer & F. H. Lopes da between M/EEG source-space and fMRI networks
Silva (Eds.), Niedermeyer’s electroencephalography at rest. Brain Topography, 33(2), 151–160.
(7 ed., pp. 1135–1156). New York: Oxford Univer- Rodriguez, E., George, N., Lachaux, J.-P., Martinerie,
sity Press. J., Renault, B., & Varela, F. J. (1999). Perception’s
Michel, C. M., & Koenig, T. (2018). EEG microstates shadow: Long-distance synchronization of human
as a tool for studying the temporal dynamics of brain activity. Nature, 397, 430–433.
whole-brain neuronal networks: A review. Neuro- Roelfsema, P. R., Engel, A. K., König, P., & Singer, W.
Image, 180(B), 577–593. (1997). Visuomotor integration is associated with
Michel, C. M., Koenig, T., Brandeis, D., Gianotti, L. R. zero time-lag synchronization among cortical
R., & Wackermann, J. (Eds.). (2009). Electrical neu- areas. Nature, 385, 157.
roimaging. Cambridge, UK: Cambridge University Samogin, J., Liu, Q., Marino, M., Wenderoth, N., &
Press. Mantini, D. (2019). Shared and connection-spe-
Michel, C. M., & Murray, M. M. (2012). Towards the cific intrinsic interactions in the default mode net-
utilization of EEG as a brain imaging tool. Neuro- work. NeuroImage, 200, 474–481.
Image, 61(2), 371–385. Sanchez-Vives, M. V., & McCormick, D. A. (2000).
Michel, C. M., & Pascual-Leone, A. (2020). Predicting Cellular and network mechanisms of rhythmic
antidepressant response by electroencephalogra- recurrent activity in neocortex. Nature Neurosci-
phy. Nature Biotechnology, 38(4), 417–419. ence, 3(10), 1027–1034.
Mizuseki, K., Sirota, A., Pastalkova, E., & Buzsaki, G. Schwab, S., Koenig, T., Morishima, Y., Dierks, T.,
(2009). Theta oscillations provide temporal win- Federspiel, A., & Jann, K. (2015). Discovering fre-
dows for local circuit computation in the entorhi- quency sensitive thalamic nuclei from EEG micro-
nal-hippocampal loop. Neuron, 64(2), 267–280. state informed resting state fMRI. NeuroImage,
Nolte, G., Bai, O., Wheaton, L., Mari, Z., Vorbach, S., 118, 368–375.
& Hallett, M. (2004). Identifying true brain interac- Seeber, M., & Michel, C. M. (2021). Synchronous
tion from EEG data using the imaginary part of brain dynamics establish brief states of communal-
coherency. Clinical Neurophysiology, 115(10), ity in distant neuronal populations. eNeuro, 8(3).
2292–2307. doi: 10.1523/ENEURO.0005-21.2021
O’Reilly, C., & Elsabbagh, M. (2021). Intracranial Siegel, M., Donner, T. H., Oostenveld, R., Fries, P., &
recordings reveal ubiquitous in-phase and in- Engel, A. K. (2008). Neuronal synchronization
antiphase functional connectivity between homo- along the dorsal visual pathway reflects the focus
topic brain regions in humans. Journal of of spatial attention. Neuron, 60(4), 709–719.
Neuroscience Research, 99(3), 887–897. Sjogard, M., De Tiege, X., Mary, A., Peigneux, P.,
Palva, J. M., Wang, S. H., encePalva, S., Zhigalov, A., Goldman, S., Nagels, G., … Wens, V. (2019). Do
Monto, S., Brookes, M. J., … Jerbi, K. (2018). the posterior midline cortices belong to the elec-
Ghost interactions in MEG/EEG source space: A trophysiological default-mode network? NeuroIm-
note of caution on inter-areal coupling measures. age, 200, 221–230.
NeuroImage, 173, 632–643. Smailovic, U., Koenig, T., Savitcheva, I., Chiotis, K.,
Parra, L. C., Spence, C. D., Gerson, A. D., & Sajda, P. Nordberg, A., Blennow, K., … Jelic, V. (2020).
(2005). Recipes for the linear analysis of EEG. Neu- Regional disconnection in Alzheimer dementia
roImage, 28(2), 326–341. and amyloid-positive mild cognitive impairment:
Pascual-Marqui, R. D., Michel, C. M., & Lehmann, D. Association between EEG functional connectivity
(1995). Segmentation of brain electrical activity and brain glucose metabolism. Brain Connectivity,
into microstates: Model estimation and validation. 10(10), 555–565.
IEEE Transactions on Biomedical Engineering, 42, Smith, S. M., Fox, P. T., Miller, K. L., Glahn, D. C., Fox,
658–665. P. M., Mackay, C. E., … Beckmann, C. F. (2009).
Spatio-temporal Dynamics of Brain Rhythms 317

Correspondence of the brain’s functional architec- activity via linearly constrained minimum variance
ture during activation and rest. Proceedings of the spatial filtering. IEEE Transactions on Biomedical
National Academy of Sciences of the United States Engineering, 44(9), 867–880.
of America, 106(31), 13040–13045. Wu, W., Zhang, Y., Jiang, J., Lucas, M. V., Fonzo, G.
Srinivasan, R., Russell, D. P., Edelman, G. M., & Tononi, A., Rolle, C. E., … Etkin, A. (2020). An electroen-
G. (1999). Increased synchronization of neuromag- cephalographic signature predicts antidepressant
netic responses during conscious perception. Jour- response in major depression. Nature Biotechnol-
nal of Neuroscience, 19(13), 5435–5448. ogy, 38(4), 439–447.
Studer, D., Hoffmann, U., & Koenig, T. (2006). From Yuval-Greenberg, S., & Deouell, L. Y. (2009). The
EEG dependency multichannel matching pursuit broadband-transient induced gamma-band response
to sparse topographic EEG decomposition. Journal in scalp EEG reflects the execution of saccades. Brain
of Neuroscience Methods, 153(2), 261–275. Topography, 22(1), 3–6.
Tallon-Baudry, C., Bertrand, O., Delpuech, C., & Pernier, Yuval-Greenberg, S., Tomer, O., Keren, A. S.,
J. (1996). Stimulus specificity of phase-locked and Nelken, I., & Deouell, L. Y. (2008). Transient
non-phase-locked 40 Hz visual responses in human. induced gamma-band response in EEG as a mani-
Journal of Neuroscience, 16(13), 4240–4249. festation of miniature saccades. Neuron, 58(3),
Van de Steen, F., Faes, L., Karahan, E., Songsiri, J., 429–441.
Valdes-Sosa, P. A., & Marinazzo, D. (2017). Critical Zanesco, A. P., King, B. G., Skwara, A. C., & Saron,
comments on EEG sensor space dynamical connec- C. D. (2020). Within and between-person corre-
tivity analysis. Brain Topography, 32, 643–654. lates of the temporal dynamics of resting EEG
Van Veen, B. D., Van Drongelen, W., Yuchtman, M., microstates. NeuroImage, 211. doi: 10.1016/
& Suzuki, A. (1997). Localization of brain electrical j.neuroimage.2020.116631
20
Sleep and Circadian Rhythms:
Alpha Rhythm and
Alertness/Sleepiness
Arcady A. Putilov

INTRODUCTION: SLEEP, ALERTNESS, AND night-time sleep. Throughout each 24-hour period,
CIRCADIAN RHYTHMS alertness–sleepiness level dips and rises, impact-
ing the amount of sleepy and alert substates of the
wakefulness state.
This chapter briefly describes the regulating pro-
The basic properties of any biological time-
cesses underlying two major human behaviour
measuring system, including the circadian clocks,
rhythms, the 24-hour sleep–wake cycle, and cor-
have easily lent themselves to mathematical mod-
responding alertness–sleepiness rhythm. In their
eling. Better understanding the rhythmic human
scientific elaboration, the chapter also provides an
illustration of the important role of brain waves, behaviours is provided by such modeling enriched
including the alpha rhythm. by model-based simulations of empirical data. The
Circadian (∼one day) rhythms in human behav- best example is the two-process model of sleep–
iour and physiology are regulated by a light sensi- wake regulation (Daan et al., 1984) that, for more
tive circadian pacemaker (i.e., master body clocks) than three decades, has become the major con-
localized in the suprachiasmatic nuclei of the tributor to the current understanding of the mecha-
hypothalamus (Pittendrigh and Daan, 1976). As nisms underlying the human 24-hour sleep–wake
predicted by the general theory of oscillation and cycle (Borbély et al., 2016). In accord with this
confirmed by the empirical findings, the 24-hour model (Daan et al., 1984), two body processes
light-dark cycle is able to entrain this circadian control sleep and wake states. These are the cir-
pacemaker. The entrainment results in matching a cadian process and sleep homeostatic process.
period of the pacemaker to that of the environmen- The circadian process represents the influence
tal cycle, i.e., 24 hours. The circadian pacemaker, of the circadian clocks on sleep timing, and the
in turn, entrains the behavioural, physiological sleep homeostatic process adjusts sleep intensity
and hormonal rhythms, including the sleep–wake and duration as a function of the duration of prior
cycle and daily fluctuations of alertness–sleepi- wakefulness (Borbély et al., 2016; Daan et al.,
ness level (Pittendrigh and Daan, 1976). Sleep– 1984).
wake cycle refers to the 24-hour daily sleep To examine human sleep and alertness/sleepi-
pattern which usually consists of approximately ness, electrodes are attached to the scalp and face
16 hours of daytime wakefulness and 8 hours of to allow measurement of electroencephalographic
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 319

(EEG) signal, eye movements, and muscle tone. SWA(t ) = [ XSWA l + C (t )] −


EEG recordings of brain waves derived from elec-
{SWA d − [SWA l + C (t )]}* e − ( t − t 2) /[ Td − k *C ( t )])   (20.1b)
trical activity in the brain are analysed for their
distribution and classified in accord with their
frequency, in cycles per second, or hertz (Hz). whereby C ( t ) = A * sin ( 2π * t / τ + )   (20.2)
0
Frequencies range from very slow to very fast
(labeled using Greek letters “delta” and “gamma,”
is a periodic function with a period τ assigned to
respectively). The frequency ranges are: delta (1
24 hours (i.e., this term represents the modulating
Hz–4 Hz), theta (5 Hz–8 Hz), beta (13 Hz–30 Hz),
action of the circadian process – body clocks – on
and gamma bandwaves (>30 Hz), respectively
the parameters of sleep homeostatic process in this
(Buzsaki, 2006). In addition, the sigma bandwave
version of two-process model).
(12–16 Hz) is especially important for sleep stud-
Figure 20.1a illustrates the results of apply-
ies due to the overlap with spindle activity (Holz
ing this model to simulate sleep times for several
et al., 2012). The two-process model of sleep–
days with the model’s parameters listed in Table
wake regulation (Daan et al., 1984) proposes that
20.1. Throughout a week, wake state is spontane-
sleep homeostatic process can be traced in experi-
ously terminated by this sleep–wake regulating
mental research by measuring brain activity thus
mechanism irrespective of whether sleep state was
allowing the simulations of the sleep–wake cycle
also spontaneously terminated (at free days) or
in terms of this model. From one approximately
terminated a bit earlier with alarm clocks on five
90-minute sleep cycle to the next, the amplitude
weekdays. An index of SWA is building up and
of waves in very slow (delta) range gradually
decaying during wake and sleep states within its
decreases exponentially. Delta activity is also
upper and lower thresholds, respectively.
found to be higher after sleep restriction/depriva-
Figure 20.1a also illustrates that, if wake state
tion, or lower after extended sleep duration or an
was not spontaneously terminated in the evening
afternoon nap. This pattern allows the utilization
(see the end of free day 1), “sleep debt” is accumu-
of the indexes of EEG Slow Wave Activity (SWA)
lated in the course of further (extended) wakeful-
as the conventional markers of the homeostatic
ness as indicated by a further buildup of the SWA
process (Borbély et al., 2016). SWA typically cor-
index above the upper threshold (Figure 20.1a).
responds to the signal power in the delta frequency
Consequently, this “debt” is expected to be “paid”
range (cf. Zielinski et al., 2019).
back during the following more intensive (recov-
In the Daan et al. (1984) model, SWA has been
ery) sleep.
proposed to decrease exponentially during the
One of the examples of application of the two-
sleep phase of the sleep–wake cycle, while its
process models is the result of evaluation of the
compensating rise during the wakefulness phase is
role of sleep homeostatic process in the well-
approximated by an inverse exponential function
documented ontogenetic changes in sleep timing
(it cannot be measured directly via EEG record-
and duration. The model-based simulations of
ing). The circadian process in the Daan et al.
experimental and epidemiological data pointed
(1984) model determines the circadian times for
at the kinetics of sleep homeostatic process as a
the spontaneous switches from the first to the sec-
cause of differences between mature and prepu-
ond phase (sleep offset) and back from the second
bescent adolescents in sleep timing and duration
to the first phase (sleep onset).
(Crowley et al., 2014; Hagenauer and Lee, 2013;
Putilov and Verevkin, 2018). Moreover, Skeldon
et al. (2016) demonstrated that changes in sleep
timing and duration occurring from adolescence
MODELING AND SIMULATION OF THE to old ages can be understood as a consequence of
SLEEP–WAKE CYCLE changes in the kinetics of this process. In contrast,
this process seems not to be involved in determi-
nation of the inter-individual differences in sleep
If t1 and t2 are the initial times for the buildup and associated with the phase of sleep–wake cycle.
decay phases of the 24-h sleep–wake cycle (i.e., the These differences exist in people who do not dif-
spontaneous sleep off- and on-set, respectively), the fer in sleep duration but differ in this phase (e.g.,
process of sleep–wake regulation, SWA(t), can be the so-called larks and owls with earlier and later
simulated (Figure 20.1a and Table 20.1) using the sleep timing, respectively).
following equations (Putilov, 1995): The predictive power of this version of two
process model (Putilov, 1995) is demonstrated by
SWA ( t ) = [SWA u + C (t )] − the experimental evidence supporting predictions
{[SWA u + C (t )] − SWA b }* e − ( t − t1) /[ Tb − k *C ( t )])    (20.1a) by the simulations’ modifying influence of the
320 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 20.1 Simulations of sleep times and sleepiness.


(a) Simulation of 10-day time course of SWA(t) associated with the 24-hour alternations of
wakefulness and sleep; throughout these days, the range of build-up and decay of SWA (1)
always remains to be limited by the upper and lower thresholds (SWAd and SWAb, respec-
tively); SD: Further build-up of SWA above the upper threshold, SWAd, is expected due to
prolongation of wakefulness beyond its normal everyday interval (i.e., sleep deprivation
associated with accumulation of “sleep debt”). (b) Simulation of 2.5-day time course of
sleepiness (3) indexed by the inverted score on the nine-step Karolinska Sleepiness Scale
(Åkerstedt and Gillberg, 1990), KSS(t) = 9-KSS score (see more details in Table 20.1).
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 321

Table 20.1 Model’s parameters in simulations of sleep times and sleepiness levels
Simulated sleep-wake cycle and alertness-sleepiness rhythm SWA Inverted KSS

Sine wave-form circadian modulation of the parameters of homeostatic processes (2):


A (circadian amplitude) 0.50 0.30
ΦMax (circadian phase for maximal rise of sine function), hours 15.0 5.0
τ (entrained circadian period), hours 24.0 24.0
Exponential buildup/decay of alertness/sleepiness, X(t) (1):
Xl(t) (lower asymptote for decay phase), see below
Xb (lowest decay of alertness during normal waking) 4.75
Xd (highest buildup of alertness during normal waking) 5.73
Xu(t) (upper asymptote for buildup phase), see below
Td (decay phase constant of the rhythm), hours 12.71
Tb (buildup phase constant of the rhythm), hours 19.23
Exponential buildup/decay of sleep/wakefulness that modulates SWAl(t) and SWAu(t) (3):
SWAl0 (lower asymptote for decay phase of the cycle) 0.70 4.50
SWAb0 (lowest decay during normal waking phase of the cycle) 0.76 5.93
SWAd0 (highest buildup during normal sleep phase of the 3.00 7.54
cycle)
SWAu0 (upper asymptote for buildup phase of the cycle) 5.50 7.60
Td (decay phase constant of the cycle), hours 26.63 20.57
Tb (buildup phase constant of the cycle), hours 2.75 2.24
Initial times for buildup/decay of sleep/wakefulness and alertness/sleepiness:
t2 (free day switch from buildup to decay phase), clock hours 23.4 21.0
t1 (free day switch from decay to buildup phase), clock hours 8.5 7.3

Note. For simulation of sleep process indexed by Slow-Wave Activity (SWA), SWA in the baseline night episode was set to 1.0
(see the simulation of the time courses of SWA in Figure 20.1a). For simulation of subjective alertness–sleepiness measure
(9-KSS score), sleep histories in the pre-experimental day self-reported by 48 study participants (Figure 20.2b) were used to
set mean initial times for going to bed (12.30 am) and waking up (8.48 am) in Figure 20.1b.

circadian process on the parameters of SWA with pandemic. This “lockdown” provided a possibility
the peak of such influence in the afternoon hours to demonstrate the predictive power of simulations
(Lazar et al., 2015). based on sleep–wake regulating models and their
The utility of mathematical modeling pertains ability to predict the counterintuitive findings.
to its ability to enable novel insights into human Model-based predictions (Putilov and Verevkin,
rhythmic behaviour that are not simply intuitive. 2018) suggested that weekend sleep durations
Simulations of weekday and weekend sleep times reported before, during and after “lockdown”
predict that sleep missed on weekdays are com- would be practically identical despite a significant
pensated for by prolongation of weekend sleep increase in weekday sleep duration during “lock-
(Putilov and Verevkin, 2018). The simulations down” as compared with sleep duration before or
suggested that, in any five work/school days, the after “lockdown.” This counterintuitive predic-
pressure for sleep (i.e., SWA level) builds up until tion was supported by empirical evidence based
its expected upper threshold only (Figure 20.1a). on the analysis of sleep times before and during
Indeed, when individuals try to sleep for longer “lockdown” (Putilov, 2021). In fact, it was demon-
periods of time on weekends to make up for sleep strated that weekend times in bed before and dur-
lost during the week, they fail to prolong their ing “lockdown” were practically identical despite
sleep beyond its normal duration. By April 2020, the lockdown-associated shift of weekday wake-
about half of the world’s population was under ups on later hours that led to a longer weekday
some form of “lockdown” due to the COVID-19 time in bed (Putilov, 2021).
322 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

MODELING AND SIMULATION OF THE the conceptualization of sleep–wake cycle in the


ALERTNESS–SLEEPINESS RHYTHM framework of so-called opponent process model
of sleep–wake regulation that postulates two
antagonistic drives – for wake and for sleep – that
Despite a common-sense view that daily variation oppose one another to regulate the cycle (Dijk and
in alertness–sleepiness level is simply a reflection Czeisler, 1995; Edgar et al., 1993).
of human sleep–wake cycle, this variation can be Figure 20.1b illustrates the results of simula-
actually regarded as an example of a “strong” tion with this model the time course of subjectively
(rigid) circadian rhythm, such as the daily varia- assessed sleepiness (KSS) during two-day sleep
tion in core body temperature and melatonin deprivation experiment (see more details on the
secretion. A “strong” circadian rhythm is charac- model’s parameters in Table 20.1). Throughout
terized by a narrow range of entrainment that is this experiment, a step-like, not gradual change in
the range of synchronizer’s periods to which a sleepiness level occurred. It reflects the modulation
given rhythm is able to entrain. Unlike a weaker influence of the circadian clocks and sleep homeo-
(lax) rhythm, such as the sleep–wake cycle, a static process on the wake homeostatic process.
“strong” rhythm cannot be entrained by a time This model of alertness–sleepiness rhythm was,
giver with a much longer or much shorter period in particular, applied for simulating age-associated
(Pittendrigh and Daan, 1976; Granada et al., changes in the 24-hour fluctuations of subjective
2013). Experimental results indicate that the range sleepiness. These simulations suggested that the
of entrainment of alertness–sleepiness rhythm is age-associated changes in the parameters of pro-
even narrower than that of the core body tempera- cesses regulating the fluctuations of sleepiness
ture rhythm (Folkard et al., 1985). Therefore, the seem to be rather small compared to the age-­
simulations of the sleep–wake cycle and the simu- associated changes in the parameters of the processes
lations of alertness–sleepiness rhythm can require regulating the sleep–wake cycle (Putilov, 2016).
somewhat different models. At least one addi- In sum, we have described the regulatory pro-
tional process is usually included in the model of cesses underlying the 24-hour sleep–wake cycle
alertness–sleepiness fluctuations (Akerstedt and and alertness–sleepiness rhythm and the role of
Folkard, 1997). brain waves in scientific research into the sleep–
Since in the course of prolonged wakeful- wake cycle. Changes in EEG waves during sleep
ness, alertness demonstrates a gradual declining and changes in subjective sleepiness during sleep
trend from one day to another, this trend might be deprivation were found to relate to the compli-
accounted by postulating that the upper asymptote, mentary effect of the circadian, and homeostatic
Xu, is not, as it is suggested in equation (20.1a), regulatory processes impacting on the drive for
a constant throughout the buildup (wake) phase sleep. The results point to the prospect of using
(Putilov et al., 2015). On the contrary, it can be changes in SWA and weekday–weekend shifts of
proposed to exponentially decline (as in equation sleep times as markers of the mechanisms regulat-
20.1b) to reflect the influence of the correspond- ing rhythmicity of sleep and wakefulness.
ing phase of another homeostatic process, Xu(t):

X u (t ) = [ X lu + C (t )] −
{X u1 − [ X lu + C (t )]* e − ( t − t1) /[ Tdu − k *C ( t )]}   (20.3),
ALPHA RHYTHM, SLEEPINESS, AND
where Xu1 is a value at the initial time point t1, ALERTNESS
whereas Xlu and Tdu are an asymptote and a time
constant, respectively, of the declining upper Unfortunately, the changes in SWA levels fail to
asymptote Xu(t). reflect the changes in homeostatic sleep process
In such a bit more complicated variant of the during wakefulness, but, fortunately, the changes
model (Putilov et al., 2014, 2015, 2019), the in some other frequency ranges of the EEG spec-
process X(t) can be interpreted as the homeo- trum might help to differentiate between alert and
static process representing a wake drive (i.e., a sleepy substates of wakefulness state. Most reports
wake-promoting process), whereas the process have suggested the possibility of using changes in
Xu(t) can be suggested to be an opposing sleep oscillations in theta and alpha frequency ranges as
drive (i.e., a sleep-promoting process associated the spectral EEG indicators of sleepiness level
with accumulation of “sleep debt” during wake- (Leproult et al., 2003; Lorenzo et al., 1995;
fulness in the initial version of the two-process Marzano et al., 2007; Putilov and Donskaya, 2014;
model). This interpretation seems to resemble Strijkstra et al., 2003).
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 323

Alpha waves fit in the middle of the brain daily variation in the levels of performance, atten-
wave spectrum, between theta and beta frequency tion, perception, etc. For instance, Aeschbach et al.
ranges, 8 Hz–12 Hz. Despite falling in the mid- (1999) found that the circadian rhythm in alpha fre-
dle of the EEG spectrum, the first letter of the quency band exhibited a minimum near the body
Greek alphabet was used for labelling alpha waves temperature minimum. Moreover, alpha activity
because they were the first of the discovered pat- has a potential as an index of homeostatically regu-
terns of the EEG signal. In the early twentieth cen- lated alternations between alertness and sleepiness
tury, Berger (1929) pioneered in observing these substates of wakefulness state. At least, it was
waves by recording electrical activity from the previously shown that this activity is attenuating
human scalp. This observation was the first of the with increasing duration of wakefulness beyond
most important discoveries in the almost 100-year its normal interval in the 24-hour sleep–wake cycle
history of the EEG research. (Putilov and Donskaya, 2014; Putilov et al., 2021).
In healthy, awake adults, alpha waves occur Thus, since their discovery (Berger, 1929),
while resting with the eyes closed. They disappear and until recently, alpha waves have remained the
during sleep and vanish when there is concentra- most salient EEG observation during wakefulness.
tion on a specific task or when eyes are opening They may be fundamental for various cognitive
(Klimesch, 2012). Alpha waves are maximal over processes, might be associated with individual dif-
the occipital (back) region of the brain (Barzegaran ferences in these processes, and have a potential
et al., 2017; Hughes and Crunelli, 2005). Alpha as a marker of regulatory mechanisms responsible
frequency and amplitude of alpha waves are for the diurnal alternations between alertness and
known to change systematically over the lifespan sleepiness substates of wakefulness.
(Anderson and Perone, 2018; Bell et al., 2012;
Bernhard and Skoglund, 1939; Campus et al.,
2021; Cellier et al., 2021; Chiang et al., 2011;
Clarke et al., 2001; Marshall et al., 2002). ALPHA WAVES AS AN OBJECTIVE
Alpha waves were found to be the most salient
EEG event during wakefulness and may be funda-
MARKER OF SLEEPINESS: DEPRIVATION
mental for various cognitive processes (Ito et al., STUDIES
2005; Lustenberger et al., 2015; Samuel et al.,
2018; Von Stein and Sarnthein, 2000) includ- The studies of functional role of alpha oscillations
ing attention (Boudewyn and Carter, 2018; Foxe have usually focused on their relation with atten-
and Snyder, 2011; Saalmann et al., 2021), per- tion and perception. A less explored area for
ception (Arnal and Giraud, 2012; Capilla et al., application of the concept and measurement of
2014; Samaha et al., 2015), working memory alpha waves is an objective evaluation of changes
(Hanslmayr et al., 2012; Jensen, 2002; Riddle in vigilant state. Below such kind of evaluation is
et al., 2020), inter alia. exemplified by the results of two sleep deprivation
The current EEG research suggested that brain experiments in which alpha activity was utilized
oscillations in the alpha frequency band not only as an objective index of sleepiness level.
reflect an “idle” state of cortical activity, but also Subjective sensation of sleepiness prior to
take a more active role in the generation of com- habitual bedtime might have evolved to motivate
plex cognitive functions (Anokhin and Vogel, humans to switch from any kind of current day-
1996; Grandy et al., 2013; Prent and Smit, 2020; time activities to sleep-preparatory behaviours
Sadaghiani and Kleinschmidt, 2016; Stevens and (Axelsson et al., 2020; Shochat et al., 2021).
Zabelina, 2019). For instance, more than 60% of Such conceptualization of sleepiness motivated to
the observed inter-subject variability in perceptual address the question of whether people, indepen-
learning can be ascribed to ongoing alpha activity dently of their age, are equipped with the ability
(Freyer et al., 2013). to become more and more sleepy due to prolonga-
The circadian rhythm (period length around tion of wakefulness beyond habitual bedtime hour
24 hours) exerts the strongest influence on almost (i.e., as indicated by the time courses of both sub-
all investigated characteristics of the EEG signal jective and objective measures of sleepiness). To
(Croce et al., 2018; Dijk and Duffy, 1999; Gundel address this question, indexes of alpha and theta
et al., 1983; Lehnertz et al., 2021) and changes activities were utilized for objective measurement
in EEG waves are related to diurnal variation in of sleepiness. It was hypothesized that an ability to
vigilance states and substates. In particular, alpha perceive sleepiness is preserved in people of older
waves can serve as indicators of the processes reg- age and that feeling sleepy is associated with the
ulating circadian rhythmicity of sleep and wakeful- specific pattern of spectral EEG signal (Putilov
ness states, substates of alertness and sleepiness, and Donskaya, 2020).
324 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Methods of Deprivation Studies Subjective sleepiness was self-scored in each


EEG recording session using the nine-step KSS
The protocols of two experimental sleep depriva- (Åkerstedt and Gillberg, 1990). Each individual
tion studies were approved by the ethics commit- KSS score obtained in any of the EEG recording
tee of the Institute for Molecular Biology and sessions was expressed as a deviation from the
Biophysics of the Federal Research Centre for initial score, at 19:00 hours for the two-day sleep
Fundamental and Translational Medicine deprivation study, and at 21:00 hours for the one-
(Novosibirsk, Russia). The experiments were per- night sleep deprivation study.
formed in accordance with the ethical standards Two electrodes, frontal (Fz–A2) and occipi-
laid down in the Declaration of Helsinki. Informed tal (either Oz-A2 or O2-A2), were used for all
written consents were obtained from all partici- EEG recordings in accord with the International
pants studied as paid volunteers. They were 10–20 system of electrode placement. For fixing
recruited from the staff of the conglomeration of the electrodes, Ten20 conductive paste (Nicolet
medical research institutes and from the surround- Biomedical, Madison, WI, USA) was utilized. The
ing community. The participants did not undergo electrodes were removed after each EEG measure-
a clinical evaluation, but, during a pre-experimen- ment. Their exact positions were recorded using
tal interview, they denied current health problems, a permanent ink marker. A 16-channel electroen-
history of psychiatric or sleep disorders, and shift cephalograph (Neuron-Spectrum-2, Neurosoft,
work or trans-meridian flights in the preceding Ivanovo, Russia) was used to record EEG signals,
month. For a week prior to the experiment, they conditioned by the high-pass, low-pass and notch
were asked to keep regular sleep–wake schedule filters (0.5 Hz, 35 Hz, and 50 Hz, respectively),
and to report history of their sleep for each of sampled and stored on a hard disc with a fre-
these seven days. quency of 200 Hz. The EEG records were visually
Previously, the two analysed here EEG datasets inspected on two-second epochs for removing all
were used for introducing a single spectral EEG epochs containing artifacts from further analy-
measure of sleepiness calculated by summing 16 sis. The FFTW (Fastest Fourier Transform in the
weighted single-Hz power densities of the EEG West) package (Frigo and Johnson, 2005) was
spectrum (Putilov et al., 2019). The main EEG applied to compute power spectra densities for the
dataset was collected from 48 participants (27 artifact-free epochs (see www.fftw.org for more
females) in two-day sleep deprivation experi- detail). Rectangular window taper was used on
ments. Ages ranged between 15 and 67 years with one-second epochs without overlap for calculating
mean age ± standard deviation of 36.6 ± 13.1 absolute spectral power densities (μV2) for each
years. The participants were grouped into three of 16 first single-Hz frequency bandwidths (i.e.,
age groups of 15–18 participants each (younger: 0.50–1.49 Hz, 1.50–2.49 Hz, 2.50–3.49 Hz, etc.).
15–26 years, intermediate: 30–40 years, and older: These single-Hz power densities were aver-
46–67 years). They arrived in the research unit on aged across each of the 1-minute intervals of the
Friday before 18:30 hours and left on Sunday at EEG record and ln-transformed. These power
19:30 hours. It was expected that they would be densities were further averaged over derivations,
able to complete 25 or fewer sessions of resting and further averaging was performed over 2-min-
EEG recording separated by two-hour intervals ute intervals with eyes open and then closed, and
(2- and 5 minutes with eyes open and then closed, across 5-minute intervals with eyes closed, for
respectively). both the 4 4-Hz frequency range and the alpha
The additional EEG dataset from the experi- range (9 Hz-12 Hz). Each such value obtained in
ment on a shorter (one-night) sleep deprivation the 13–25 EEG recording sessions of each of 48
was collected from a bigger number of volunteers participants during the two-day deprivation study
aged between 15 and 67 years with mean age ± was subtracted from the first (evening) value
SD = 29.4 (± 12.2) years. The sample of 130 par- obtained at 19:00 hours. In the additional dataset
ticipants (76 females) was divided into four age with 130 participants, each value was subtracted
groups (youngest: 15–21 years, younger: 22-24 from the value obtained at 21:00 hours on the
years, intermediate: 25–39 years, and older: 40–67 evening preceding the experimental prolongation
years). They arrived in the research unit between of wakefulness.
8:00 and 8:30 pm to leave it at 9:30 am next day Participants were engaged in performance
after completing nine sessions of resting EEG measurements and questionnaire assessments.
recordings separated by 3-hour intervals (one During their free time they can surf the Internet,
minute with open eyes of a study participant and watch TV, read, write, listen to music, play board,
another with his/her eyes closed). and computer games. Participants can freely
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 325

move between several rooms of the research unit. study, F3,128 = 11.74, p < 0.001 in one-night
Throughout the experiment, they consumed at study). However, there were not any significant
self-chosen times light snacks and drinks (with age-associated differences in self-reported nap
exception of alcohol and caffeinated beverages). frequency and duration, night sleep onset
Taking any medications and heavy meals, smok- latency, and night sleep satisfaction. Moreover,
ing, vigorous physical activity, and exposure to a significant association with age was found for
light >500 lux were not allowed. neither subjective nor objective evening sleepi-
All calculations and all tests of significance ness measures prior to the experimental prolon-
of the effects of age on subjective and objective gation of wakefulness (i.e., at 19:00 hours and
measures of sleepiness were performed using 21:00 hours in the first and second studies,
the SPSS23.0 statistical software package (IBM, respectively).
Armonk, New York, USA). Pearson’s correlation During the first day of sleep deprivation,
coefficient was calculated for the associations power in the alpha range (9 Hz–12 Hz) drastically
between time courses of objective and subjective decreased in the young study participants, while
sleepiness on the interval of 25 or 9 EEG recording in the old participants they remained almost unaf-
sessions and in order to relate age of participants fected by the moderate sleep loss (Figure 20.2).
to sleepiness measures obtained for each EEG However, the expected attenuation of the alpha
recording session. Correlation analyses and one- rhythm was found within any age group after fur-
way ANOVAs with the independent factor “Age” ther extension of wakefulness to the second day
were performed to examine age-associated differ- (Figure 20.2). The difference between age groups
ences in pre-experimental sleep times and baseline during the first day of sleep deprivation was even
sleepiness measurements. In two-way ANOVAs, more profound in the response to sleep loss of the
the additional independent factor “Sex” was slower frequency ranges including theta range (5
added. Moreover, significance of main effect of Hz-8 Hz). Power density increased in this range
independent factor “Age” on a sequence of subjec- in older participants but decreased in younger par-
tive and objective measurements of sleepiness was ticipants (Figure 20.2a and 20.2b). In contrast, the
tested with two-way repeated measure ANOVAs theta power only increased after prolongation of
(rANOVAs). The repeated measure was “Clock wakefulness into the second day (Figure 20.2).
hour” on day 1 and/or day 2 (between 21:00 hours The significance of such differences between
Friday or Saturday and 19:00 hours Saturday or the young and old participants in changes of
Sunday) and on night 1 (between 0:00 hours and power densities throughout the first stage of
9:00 hours) in a two-day and one-night sleep dep- sleep deprivation was evident for the triple inter-
rivation study, respectively. The independent fac- action between the independent factor “Age” and
tor “Sex” was added to “Age” “Clock hour” in repeated measures, “Clock hour” and “Single-Hz”
three-way rANOVAs. Finally, “Clock hour” and (F330,7425 = 1.55, p = 0.043, and F315,11340 = 1.27,
“Single-Hz” powers from 1 Hz to 16 Hz, were p = 0.049 in two-day and one-night experimental
included in three-way rANOVAs to test signifi- studies, respectively). This interaction became
cance of interaction of these two repeated meas- non-significant during the second day indicating
ures with the independent factor “Age.” Mauchly’s the increase of similarity of ages in their response
test was conducted to assess the sphericity and, if to further prolongation of wakefulness (F270,3630 =
necessary, the Greenhouse–Geiser correction was 1.16, p = 0.298, in two-day experimental study).
used to adjust the degrees of freedom, but the The dual interactions of “Age” with “Single-Hz”
original degrees of freedom are reported in Results (Figure 20.2a) was significant in each of two
and Tables 20.2 and 20.3. days (F30,675 = 1.85, p = 0.007, and F30,330 = 3.03,
p = 0.006).
As illustrated in Figure 20.2b, the correlation
coefficient between time course of KSS score and
Results of Deprivation Studies 6-Hz power (i.e., within theta frequency range)
attained the value of 0.96 for the older partici-
During a week preceding the experimental dep- pants on the first day of the two-day experiment,
rivation studies, older study participants while the correlation for this day was much lower
reported earlier risetime than did younger par- for younger aged participants, 0.56. The second
ticipants (F2,47 = 4.44, p = 0.017, and F3,128 = day correlations were 0.24 and 0.52, respectively
14.27, p < 0.001 in two-day and one-night (Figure 20.2b). In contrast, the first day correla-
experimental studies, respectively). Significantly tions between time courses of KSS score and
earlier bedtime was found in one of the studies 10-Hz power (within alpha frequency range) were
(although F2,47 = 2.38, p = 0.104 in a two-day weaker for the older participants, –0.67, and much
326 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

stronger for younger participants, –0.93 (Figure single-Hz powers (6 Hz and 10 Hz) related to the
20.2b). The coefficients were relatively similar initial score and initial powers at 19:00 hours; they
during the second day, –0.80 and –0.81 (first- and again were calculated separately on two 24-hour
second-day coefficients were –0.98 and –0.82 in intervals of two-day deprivation study (N = 48
the whole sample). and 25). (c) Two-day time courses of KSS score
Although the strong correlations with subjec- and the example of time courses of Alpha power
tive sleepiness were not uncommon for both 6-Hz (frequency range from 9 Hz to 12Hz) expressed
and 10-Hz powers and for both younger and older relative to the initial score and power at 19:00
study participants, the patterns and strength of hours; (d) One-night time courses of KSS score
correlation varied depending upon amount of and Alpha power related to the score and power at
accumulated sleep debt, frequency, and age 21:00 hours (N = 130).
(Figure 20.2). Alpha power was found to correlate signifi-
For example, as suggested by results on the first cantly with age in almost all EEG recording ses-
day of sleep deprivation in the two-day depriva- sions (Table 20.2). A weaker attenuation of alpha
tion study (Figure 20.2b) and by results of the one- rhythm in older participants was confirmed by sig-
night study, the changes in theta frequency range nificant main effect of independent factor “Age”
were strong predictors of the change in subjective in two-way and three-way rANOVAs (Table 20.3).
sleepiness score in older age participants while Thus, the associations with alpha range power
the changes in alpha range were strong predictors were the most reliable and persistent (over the
of self-reported sleepiness in the participants of time of experiment) compared to all other possi-
younger age. As illustrated in Figure 20.2a, dur- ble objective sleepiness markers of sleepiness. The
ing the first day spectral powers in theta range association with sleepiness and age was the same
did not increase significantly in younger partici- directional (Figure 20.2). In contrast, KSS score
pants while spectral powers in alpha range did did not show consistent significant relationship
not decrease significantly in older participants, with age in any of the studies (Figure 20.2b–20.2d
but more resembling one another changes in the and Tables 20.2 and 20.3).
expected direction were shown by these ages dur-
ing the second day (Figure 20.2a, 20.2b).
On the two-day interval of sleep deprivation
study, the time course of KSS score strongly cor- Discussion of Deprivation Studies
related with the majority of the 16 timecourses
of single-Hz spectral powers, but the strongest Analysis of the self-ratings of sleepiness failed to
(below –0.98) coefficient was shown by 10-Hz reveal significant differences between ages in
power at the second minute with eyes closed. strength of their ability to feel sleepy during sleep
Correlation coefficients of a similar strength were deprivation. In contrast, the results also sug-
also obtained after averaging over the first two or gested that, objectively, older adults had a
all five minutes with eyes closed and over frequen- reduced vulnerability to sleep loss as indicated by
cies in alpha frequency range, 9 Hz–12 Hz (Figure the decrease of alpha activity. However, the
20.2b and 20.2c). Therefore, alpha power for the changes of activity in lower frequency band asso-
first two minutes and for the first minute of the ciated with increase of sleepiness appeared ear-
eyes closed section of the EEG record was chosen lier in the course of prolongation of wakefulness
for quantifying the strength of association between by older adults. Consequently, the results revealed
objective sleepiness and age of participants of the that motivational action of subjective sleepiness
two-day and one-night study, respectively (Figure seemed to be age-independent. To some extent
20.2, and Tables 20.2 and 20.3). this result contradicts with the age-associated
Data for the first two minutes with eyes closed decline of objective sleepiness seen in the results
(a–c) and for one minute with eyes closed (d) in on responsiveness to sleep deprivation evaluated
two-day and one-night sleep deprivation study, with alpha waves or with theta waves. Alpha
respectively, for the whole sample (left graphs) waves showed low responsiveness in older adults,
and separately for three or four ages (on the right and theta waves showed lower responsiveness in
from these left graphs). (a) Power (density) spectra younger adults. Therefore, the answer to such a
on the interval of frequencies from 1 Hz to 16 Hz question as are younger adults become objec-
expressed as deviation from the initial spectrum tively sleepier than older adults after sleep loss
at 19:00 hours; they were calculated separately depends upon the chosen measure (whether
for the first and second day of two-day study (N = sleepiness was evaluated using the alpha index or
48 and 25–42, respectively). (b) Time courses of with the lower frequency index or by
KSS score and two examples of time courses of self-scoring).
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 327

Figure 20.2 Power spectra and time courses of sleepiness.


328 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Table 20.2 Objective and subjective sleepiness measures as the correlates of age
h n Whole n Male n Female n Whole n Male n Female

Alpha power, day 1 Alpha power, day 2


21 48 .37* 21 .28 27 .41* 42 .49*** 19 .53* 23 .47*
23 48 .28 21 .15 27 .35 42 .56*** 19 .57* 23 .61**
1 48 .34* 21 .32 27 .35 41 .42** 19 .54* 22 .35
3 48 .40** 21 .34 27 .47* 39 .43** 18 .52* 21 .38
5 48 .51*** 21 .48* 27 .57** 37 .35* 17 .45 20 .30
7 48 .39** 21 .37 27 .43* 36 .49** 17 .60* 19 .41
9 48 .33* 21 .32 27 .35 33 .36* 16 .50* 17 .25
11 48 .36* 21 .33 27 .39* 29 .50** 14 .63* 15 .42
13 48 .38** 21 .38 27 .39* 27 .49** 12 .52 15 .48
15 48 .32* 21 .35 27 .32 27 .39* 12 .35 15 .43
17 48 .31* 21 .31 27 .33 25 .22 11 .11 14 .29
19 48 .44** 21 .36 27 .49** 25 .48* 11 .43 14 .53
Alpha Rhythm power, night 1 KSS score, night 1
0 130 .03 54 –.12 76 .11 130 .07 54 .27* 76 –.08
3 130 .20* 54 .16 76 .20 130 –.15 54 –.16 76 -.18
6 130 .25** 54 .31* 76 .20 130 –.12 54 –.19 76 –.12
***
9 130 .32 54 .32* 76 .33** 130 –.19* 54 –.06 76 –.24*

Notes. Data on two first minutes with eyes closed in two-day sleep deprivation study and on one minute with eyes closed in
one-night sleep deprivation study. Pearson correlation coefficient between age of study participant and sleepiness measure.
The measure is either objective or subjective: either alpha power and KSS score related to 21:00 hours in one-night sleep
deprivation study or only alpha power related to 19:00 hours in two-day sleep deprivation study (for KSS score of two-day
study, none of the coefficients was found to be significant). Whole, Male and Female: Whole sample of study participants,
Male and Female study participants; h: Clock Hour; n: Number of study participants (it is gradually decreasing on day 2 of
two-day study due to dropouts occurring when desire to sleep becomes irresistible). Level of significance for Pearson correla-
tion coefficient: * p < 0.05, ** p < 0.01, ***p < 0.001.

Table 20.3 Main effects of the independent factors “Age” and “Sex” in rANOVAs
rANOVA Two-way Three-way
df Age df Age df Sex df Interaction

Day 1
Alpha power 2/45 5.32** 2/42 5.74** 1/42 2.34 2/42 0.16
KSS score 2/45 0.48 2/42 0.48 1/42 0.00 2/42 0.52
Day 2
Alpha power 2/22 4.96* 2/19 4.41* 1/19 0.24 2/19 0.41
KSS score 2/22 0.53 2/19 0.43 1/19 0.74 2/19 0.16
Night 1
Alpha power 3/126 3.15* 3/122 2.48 1/122 0.39 3/122 0.21
KSS score 3/126 0.77 3/122 1.11 1/122 4.60* 3/122 1.07

Notes. Data on the first two minutes with eyes closed collected during the first and second days of two-day sleep depriva-
tion study and data on one minute with eyes closed collected in one-night sleep deprivation study. “Clock hour” was the
repeated measure and either “Age” or “Age” and “Sex” were independent factors in two- and three-way rANOVAs, respec-
tively; objective and subjective sleepiness measures were alpha power (9 Hz–12 Hz) and KSS score, respectively. Interaction:
Interaction between independent factors. Estimates of sleepiness were related to either 19:00 hours or 21:00 hours in either
two-day or one-night sleep deprivation study, respectively. See Methods of deprivation studies and Figure 20.2c and 20.2d
for the division into either three or four age groups. Level of significance for F-ratio: * p < 0.05, ** p < 0.01.
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 329

Sleep deprivation experiments consistently other spectral EEG indexes of sleepiness applica-
point at younger rather than older people as most ble to old volunteers. Moreover, the results sug-
susceptible to impairment of alertness and perfor- gesting the age independent response of subjective
mance during extension of wakefulness beyond its sleepiness to sleep loss do not provide evidence
normal duration (Brendel et al., 1990; Cajochen for such hypothetical age-associated sleep pathol-
et al., 2006; Dijk et al., 2010; Duffy et al., 2009; ogies as a reduction in the neural mechanisms for
Landolt et al., 2012; Philip et al., 2004; Putilov regulating sleep need and a desensitization to the
and Donskaya, 2016; Silva et al., 2010; Smulders homeostatic sleep drive (Mander et al., 2017).
et al., 1997; Zitting et al., 2018). The results on Thus, the diurnal variation in vigilance states
alpha index supported these findings. The fact and sub-states can be linked to changes in EEG
that lower frequency indexes responded earlier waves in the eyes closed condition. Consequently,
to sleep deprivation in older adults might explain these waves might serve as objective markers of
identity of response of subjective sleepiness in the mechanisms regulating the alertness–sleepi-
younger and older adults. Subjective sleepiness ness rhythm. One such marker (alpha waves in
might reflect an integrative response of different the eyes closed condition), might provide a quan-
brain functions to sleep loss, some of them decline titative description of the processes regulating
earlier in younger adults while other decline ear- alertness levels during extended periods of wake-
lier in older adults, and the sum of these responses fulness. These prospects have been illustrated by
perceived as feeling sleepy might be identical in the results on evaluation of alpha waves as objec-
younger and older people. tive indexes of sleepiness in sleep deprivation
The results on the EEG response to sleep loss experiments. The tracing changes in these waves
are in agreement with the previous reports indi- in 130 and 48 participants of one- and two-day
cating that, in eyes closed condition, sleepiness sleep deprivation experiments, respectively, has
is associated with a decrease of spectral power suggested that, similar to subjective measures
density in the fast frequency rage and a change of sleepiness, alpha waves in the eyes closed
in the opposite direction in slow frequency condition can quantitatively measure processes
range (i.e., alpha rhythm attenuates while theta regulating the alertness–sleepiness rhythm. This
and delta waves increase in amplitude; Leproult objective index might be used instead of a subjec-
et al., 2003; Lorenzo et al., 1995; Marzano et al., tive index (Figure 20.1b) in simulations of time
2007; Putilov and Donskaya, 2014; Strijkstra course of sleepiness in the two-day sleep depriva-
et al., 2003). As previously proposed (Putilov tion experiment.
and Donskaya, 2016), the brain response to sleep
loss might affect a wide range of frequencies of
the EEG signal rather than just one of the tradi-
tionally recognized frequency ranges. Such EEG
changes might represent the underlying influ- SUMMARY AND CONCLUSIONS
ence of, at least, two antagonistic sleep–wake
regulating processes (i.e., the drives for wake The processes underlying two major human
and sleep). The contribution of each of single-Hz behaviour rhythms, the 24-hour sleep–wake cycle,
powers and each of the opposing drives might be and corresponding alertness–sleepiness rhythms
modified by age, duration of permanent wakeful- and the role of brain waves including the alpha
ness, and so on (Dorokhov et al., 2021; Putilov rhythm in their scientific research, have been
and Donskaya, 2016; Putilov et al., 2021). described. It has been demonstrated that the
Olbrich et al. (2009) proposed to determine the changes in electroencephalographic waves can be
1st of two stages of drowsiness as a change from related to the diurnal variation in vigilance states
predominant frontal alpha activity to low volt- and substates. The results point to the prospect of
age EEG without this activity, while an increase using these waves as markers of the mechanisms
of delta and theta power they recommended as regulating rhythmicity of sleep and wakefulness
a marker of the next drowsiness stage preced- states, alertness and sleepiness substates, levels of
ing sleep onset. However, the present results performance, attention, perception, etc. One such
suggested that such sequence of two-step spec- marker, slow waves during sleep episodes, tradi-
tral EEG changes during transition from alert to tionally have been utilized for simulation of the
very sleep substate of wake state is typical only kinetics of sleep homeostasis, and another marker,
for samples consisting of rather young study par- alpha waves in an eyes closed condition, might
ticipants (i.e., 15 people aged 19–35 years in the serve as a quantitative measure of the processes
report by Olbrich et al., 2009). The significant age regulating alertness level during extended wake-
difference in the spectral EEG response to moder- fulness. The feasibility of such descriptors has
ate sleep loss votes for the necessity to identify been illustrated by sleep deprivation experiments
330 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

evaluating alpha waves as objective indexes of attention to spoken language. Neuropsychologia,


sleepiness. In addition to subjective measures of 117, 148–155.
sleepiness, alpha waves in an eyes closed condi- Borbély, A. A., Daan, S., Wirz-Justice, A., & Deboer,
tion can provide a quantitative description and T. (2016). The two-process model of sleep regula-
simulation of the processes regulating the alert- tion: A reappraisal. Journal of Sleep Research, 25,
ness–sleepiness rhythm. 131–143.
Brendel, D. H., Reynolds, C. F. III, Jennings, J. R.,
Hoch, C. C., Monk, T. H., Berman, S. R., … Kupfer,
D. J. (1990). Sleep stage physiology, mood, and
vigilance responses to total sleep deprivation in
REFERENCES healthy 80-year-olds and 20-year-olds. Psycho-
physiology, 27, 677–685.
Aeschbach, D., Matthews, J. R., Postolache, T. T., Buzsaki, G. (2006). Rhythms of the brain. New York:
Jackson, M. A., Giesen, H. A., & Wehr, T. A. Oxford University Press.
(1999). Two circadian rhythms in the human elec- Cajochen, C., Münch, M., Knoblauch, V., Blatter, K.,
troencephalogram during wakefulness. American & Wirz-Justice, A. (2006). Age-related changes in
Journal of Physiology: Regulatory, Integrative and the circadian and homeostatic regulation of human
Comparative Physiology, 46, R1771–R1779. sleep. Chronobiology International, 23, 461–474.
Akerstedt, T., & Folkard S. (1997). The three-process Campus, C., Signorini, S., Vitali, H., De Giorgis, V.,
model of alertness and its extension to perfor- Papalia, G., Morelli, F., & Gori, M. (2021). Sensitive
mance, sleep latency, and sleep length. Chronobi- period for the plasticity of alpha activity in humans.
ology International, 14, 115-123. Developmental Cognitive Neuroscience, 49. doi:
Åkerstedt, T., & Gillberg, M. (1990). Subjective and 10.1016/j.dcn.2021.100965
objective sleepiness in the active individual. Inter- Capilla, A., Schoffelen, J.M., Paterson, G., Thut, G.,
national Journal of Neuroscience, 52, 29–37. & Gross, J. (2014). Dissociated alpha-band modu-
Anderson, A. J., & Perone, S. (2018). Developmental lations in the dorsal and ventral visual pathways in
change in the resting state electroencephalogram: visuospatial attention and perception. Cerebral
Insights into cognition and the brain. Brain and Cortex, 24, 550–561.
Cognition, 126, 40–52. Chiang, A., Rennie, C., Robinson, P., Van Albada, S.,
Anokhin, A., & Vogel, F. (1996). EEG alpha rhythm & Kerr, C. (2011). Age trends and sex differences
frequency and intelligence in normal adults. Intel- of alpha rhythms including split alpha peaks. Clini-
ligence, 23(1), 1–14. cal Neurophysiology, 122, 1505–1517.
Arnal, L. H., & Giraud, A.-L. (2012). Cortical oscilla- Cellier, D., Riddle, J., Petersen, I., & Hwang, K.
tions and sensory predictions. Trends in Cognitive (2021). The development of theta and alpha
Sciences, 16(7), 390–398. neural oscillations from ages 3 to 24 years. Devel-
Axelsson, J., Ingre, M., Kecklund, G., Lekander, M., opmental Cognitive Neuroscience, 50. doi:
Wright, K. P., & Sundelin, T. (2019). Sleepiness as 10.1016/j.dcn.2021.100969
motivation: A potential mechanism for how sleep Clarke, A. R., Barry, R. J., McCarthy, R., & Selikowitz,
deprivation affects behavior. Sleep, 43. doi: M. (2001). Age and sex effects in the EEG: Devel-
10.1093/sleep/zsz291 opment of the normal child. Clinical Neurophysiol-
Bell, M. A., & Cuevas, K. (2012). Using EEG to study ogy, 112, 806–814.
cognitive development: issues and practices. Jour- Croce, P., Quercia, A., Costa, S., & Zappasodi, F.
nal of Cognitive Development, 13, 281–294. (2018). Circadian rhythms in fractal features of
Berger, H. (1929). Ueber das elektrenkephalogramm EEG signals. Frontiers of Physiology, 9. doi:
des menschen. Archives für Psychiatrie und Ner- 10.3389/fphys.2018.01567
venkrankheiten, 87, 527–570. Crowley, S. J., Van Reen, E., LeBourgeois, M. K.,
Bernhard, C.G., & Skoglund, C.R. (1939). On the Acebo, C., Tarokh, L., Seifer, R., Barker, D. H., &
alpha frequency of human brain potentials as a Carskadon, M. A. (2014). A longitudinal assess-
function of age. Skandinavisches Archives für ment of sleep timing, circadian phase, and
Physiologie, 82(2), 178–184. phase angle of entrainment across human ado-
Barzegaran, E., Vildavski, V. Y., & Knyazeva, M. G. lescence. PLoS One, 9(11). doi: 10.1371/journal.
(2017). Fine structure of posterior alpha rhythm in pone.0112199
human EEG: Frequency components: Their cortical Daan, S., Beersma, D. G. M, & Borbély, A. A. (1984).
sources, and temporal behavior. Scientific Reports, Timing of human sleep: recovery process gated by
7. doi: 10.1038/s41598-017-08421-z a circadian pacemaker. American Journal of Physi-
Boudewyn, M. A., & Carter, C. S. (2018). I must have ology: Regulatory, Integrative and Comparative
missed that: Alpha-band oscillations track Physiology, 246, R161–R178.
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 331

Dijk, D. J., & Czeisler, C. A. (1995). Contribution of memory: The information via desynchronization
the circadian pacemaker and the sleep homeostat hypothesis. Frontiers in Human Neuroscience, 6.
to sleep propensity, sleep structure, electroen- doi: 10.3389/fnhum.2012.00074
cephalographic slow waves, and sleep spindle Holz, J., Piosczyk, H., Feige, B., Spiegelhalder, K.,
activity in humans. Journal of Neuroscience, 15, Baglioni, C., Riemann, D., & Nissen, C. (2012).
3526–3538. EEG Σ and slow-wave activity during NREM sleep
Dijk, D.-J., & Duffy, J. F. (1999). Circadian regulation correlate with overnight declarative and proce-
of human sleep and age-related changes in its dural memory consolidation. Journal of Sleep
timing, consolidation and EEG characteristics. Research, 21(6), 612–619.
Annals of Medicine, 31, 130–140. Hughes, S. W., & Crunelli, V. (2005). Thalamic mech-
Dijk, D. J., Groeger, J. A., Stanley, N., & Deacon, S. anisms of EEG alpha rhythms and their pathologi-
(2010). Age-related reduction in daytime sleep cal implications. Neuroscientist, 11, 357–372.
propensity and nocturnal slow wave sleep. Sleep, Ito, J., Nikolaev, A. R., & Van Leeuwen, C. (2005).
33, 211–223. Spatial and temporal structure of phase synchroni-
Dorokhov, V. B., Sveshnikov, D. S., Puchkova, A. N., zation of spontaneous alpha EEG activity. Biologi-
Taranov, A. I., Sakharov, D. S., Gruzdeva, S. S., … cal Cybernetics, 92, 54–60.
Putilov A. A. (2021). Differential relationship of two Jensen, O. (2002). Oscillations in the alpha band
measures of sleepiness with the drives for sleep and (9-12 Hz) increase with memory load during reten-
wake. Sleep & Breathing, 25, 2179–2187. tion in a short-term memory task. Cerebral Cortex,
Duffy, J. F., Willson, H. J., Wang, W., & Czeisler, C. A. 12, 877–882.
(2009). Healthy older adults better tolerate sleep Klimesch, W. (2012). Alpha-band oscillations, atten-
deprivation than young adults. Journal of the tion, and controlled access to stored information.
American Geriatrics Society, 57, 1245–1251. Trends in Cognitive Sciences, 16, 606–617.
Granada, A. E., Bordyugov, G., Kramer, A., & Herzel, Landolt, H. P., Rétey, J. V., & Adam, M. (2012).
H. (2013). Human chronotypes from a theoretical Reduced neurobehavioral impairment from sleep
perspective. PLoS One, 8(3). doi: 10.1371/journal. deprivation in older adults: Contribution of aden-
pone.0059464 osinergic mechanisms. Frontiers in Neurology, 3.
Grandy, T. H., Werkle-Bergner, M., Chicherio, C., doi: 10.3389/fneur.2012.00062
Lövdén, M., Schmiedek, F., & Lindenberger, U. Lazar, A. S., Lazar, Z. I., & Dijk, D.-J. (2015). Circadian
(2013). Individual alpha peak frequency is related regulation of slow waves in human sleep: Topo-
to latent factors of general cognitive abilities. graphical aspects. NeuroImage, 116, 123–134.
NeuroImage, 79, 10–18. Lehnertz, K., Rings, T., & Bröhl, T. (2021). Time in brain:
Gundel, A., & Witthöft, H. (1983). Circadian Rhythm How biological rhythms impact on EEG signals and
in the EEG of Man. International Journal of Neuro- on EEG-derived brain networks. Frontiers in Net-
science, 19, 287–292. work Physiology, 1. doi: 10.3389/fnetp.2021.755016
Edgar, D. M., Dement, W. C. & Fuller, C. A. (1993). Leproult, R., Colecchia, E. F., Berardi, A. M., Stickgold,
Effect of SCN lesions on sleep in squirrel monkeys: R., Kosslyn, S. M., & Van Cauter, E. (2003). Individual
Evidence for opponent processes in sleep–wake reg- differences in subjective and objective alertness
ulation. Journal of Neuroscience, 13, 1065–1079. during sleep deprivation are stable and unrelated.
Folkard, S., Hume, K. I., Minors, D. S., Waterhouse, J. American Journal of Physiology: Regulatory, Integra-
M., & Watson F. L. (1985). Independence of the tive and Comparative Physiology, 284, R280–R290.
circadian rhythm in alertness from the sleep/wake Lorenzo, I., Ramos, J., Arce, C., Guevara, M. A., &
cycle. Nature, 313, 678–679. Corsi-Cabrera, M. (1995). Effect of total sleep
Foxe, J. J., & Snyder, A. C. (2011). The role of alpha- deprivation on reaction time and waking EEG
band brain oscillations as a sensory suppression activity in man. Sleep, 18, 346–354.
mechanism during selective attention. Frontiers of Lustenberger, C., Boyle, M. R., Foulser, A. A., Mellin,
Psychology, 2. doi: 10.3389/fpsyg.2011.00154 J. M., & Fröhlich, F. (2015). Functional role of frontal
Freyer, F., Becker, R., Dinse, H. R., & Ritter, P. (2013). alpha oscillations in creativity. Cortex, 67, 74–82.
State-dependent perceptual learning. Journal of Mander, B. A., Winer, J. R., & Walker, M. P. (2017).
Neuroscience, 33, 2900–2907. Sleep and human aging. Neuron, 94, 19–36.
Frigo, M., & Johnson, S. G. (2005). The design and Marshall, P. J., Bar-Haim, Y., & Fox, N. A. (2002). Devel-
implementation of FFTW3. Proceedings of the opment of the EEG from 5 months to 4 years of
IEEE, 93, 216–231. age. Clinical Neurophysiology, 113, 1199–1208.
Hagenauer, M. H., & Lee, T. M. (2013). Adolescent Marzano, C., Fratello, F., Moroni, F., Pellicciari, M. C.,
sleep patterns in humans and laboratory animals. Curcio, G., Ferrara, M., Ferlazzo, F., & De Gennaro,
Hormones and Behavior, 64, 270–279. L. (2007). Slow eye movements and subjective
Hanslmayr, S., Staudigl, T., & Fellner, M. C. (2012). estimates of sleepiness predict EEG power changes
Oscillatory power decreases and long-term during sleep deprivation. Sleep, 30, 610–616.
332 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Olbrich, S., Mulert, C., Karch, S., Trenner, M., Leicht, Putilov, A. A., Donskaya, O. G., & Verevkin, E. G.
G., Pogarell, O., & Hegerl, U. (2009). EEG-vigilance (2019). Can we feel like being neither alert nor
and BOLD effect during simultaneous EEG/fMRI sleepy? The electroencephalographic signature of
measurement. NeuroImage, 45, 319–332. this subjective sub-state of wake state yields an
Pittendrigh, C., & Daan, S. (1976). The entrainment accurate measure of objective sleepiness level. Inter-
of circadian pacemakers in nocturnal rodents. IV. national Journal of Psychophysiology, 135, 33–43.
Entrainment: pacemaker as clock. Journal of Com- Putilov, A. A., & Verevkin, E. G. (2018). Simulation of
parative Physiology A, 106, 291–331. the ontogeny of social jet lag: A shift in just one of
Prent, N., & Smit, D. J. A. (2020). The dynamics of the parameters of a model of sleep–wake regulat-
resting-state alpha oscillations predict individual ing process accounts for the delay of sleep phase
differences in creativity. Neuropsychologia, 142. across adolescence. Frontiers of Physiology, 9. doi:
doi: 10.1016/j.neuropsychologia.2020.107456 10.3389/fphys.2018.01529
Putilov, A. A. (1995). The timing of sleep modelling: Riddle, J., Scimeca, J. M., Cellier, D., Dhanani, S., &
Circadian modulation of the homeostatic process. D’Esposito, M. (2020). Causal evidence for a role
Biological Rhythm Research, 26. doi: 10.1080/ of theta and alpha oscillations in the control
09291019509360320 of working memory. Current Biology, 30,
Putilov, A. A. (2016). Age-associated advance of 1748–1754.
sleep times relative to the circadian phase of alert- Saalmann, Y. B., Pinsk, M. A., Wang, L., Li, X., &
ness–sleepiness rhythm: Can it be explained by Kastner, S. (2012). The pulvinar regulates informa-
changes in general features of the underlying tion transmission between cortical areas based on
oscillatory processes? Current Aging Science, 9, attention demands. Science, 337, 753–756.
44–56. Sadaghiani, S., & Kleinschmidt, A. (2016). Brain net-
Putilov, A. A. (2021). Sleep during “lockdown” high- works and α-oscillations: Structural and functional
lighted the need to rethink the concept of week- foundations of cognitive control. Trends in Cogni-
end catch-up sleep. Sleep & Breathing. doi: tive Science, 20, 805–817.
10.1007/s11325-021-02492-z Samaha, J., & Postle, B. R. (2015). The speed of
Putilov, A. A., & Donskaya, O. G. (2014). Alpha alpha-band oscillations predicts the temporal reso-
attenuation soon after closing the eyes as an lution of visual perception. Current Biology, 25,
objective indicator of sleepiness. Clinical and 2985–2990.
Experimental Pharmacology and Physiology, 41, Samuel, I. B. H., Wang, C., Hu, Z., Ding, M. (2018).
956–964. The frequency of alpha oscillations: Task-depend-
Putilov, A. A., & Donskaya, O. G. (2016), Evidence for ent modulation and its functional significance.
age-associated disinhibition of the wake drive pro- NeuroImage, 183, 897–906.
vided by scoring principal components of the rest- Shochat, T., Santhi, N., Herer, P., Dijk, D. J., & Skel-
ing EEG spectrum in sleep provoking conditions. don, A. C. (2021). Sleepiness is a signal to go to
Chronobiology International, 33, 995–1008. bed: Data and model simulations. Sleep, 11,
Putilov, A. A., & Donskaya, O. G. (2020). Are younger 44(10), doi: 10.1093/sleep/zsab123
people sleepier than older people after missing Skeldon, A. C., Derks, G., & Dijk, D. J. (2016). Model-
bedtime and night sleep? It depends…The 12th ling changes in sleep timing and duration across
international multiconference: Bioinformatics of the lifespan: Changes in circadian rhythmicity or
genome regulation and structure\systems biology sleep homeostasis? Sleep Medicine Reviews, 28,
(pp. 669–670). Institute of Cytology and Genetics, 96–107.
Siberian Branch of Russian Academy of Sciences, Smulders, F. T., Kenemans, J. L., Jonkman, L. M., &
Novosibirsk State University. Kok, A. (1997). The effects of sleep loss on task
Putilov, A. A., Donskaya, O. G., Poluektov, M. G., & performance and the electroencephalogram in
Dorokhov, V. B. (2021). Age- and gender-associ- young and elderly subjects. Biological Psychology,
ated differences in the sleepy brain’s electroen- 45, 217–239.
cephalogram. Physiological Measurement, 42. doi: Stevens, C. E., & Zabelina, D. L. (2019). Creativity
10.1088/1361-6579/abcdf3 comes in waves: An EEG-focused exploration of
Putilov, A. A., Donskaya, O. G., & Verevkin, E. G. the creative brain. Current Opinion in Behavioral
(2014). Phase difference between chronotypes in Sciences, 27, 154–162.
self-reported maximum of alertness rhythm: An Strijkstra, A. M., Beersma, D. G., Drayer, B., Hal-
EEG predictor and a model-based explanation. besma, N., & Daan, S. (2003). Subjective sleepi-
Journal of Psychophysiology, 28, 242–256. ness correlates negatively with global alpha (8-12
Putilov, A. A., Donskaya, O. G., & Verevkin, E. G. Hz) and positively with central frontal theta
(2015). How many diurnal types are there? A (4-8 Hz) frequencies in the human resting awake
search for two further “bird species.” Personality electroencephalogram. Neuroscience Letters, 340,
and Individual Differences, 72, 12–15. 17–20.
Sleep and Circadian Rhythms: Alpha Rhythm and Alertness/Sleepiness 333

Von Stein, A., & Sarnthein, J. (2000). Different fre- activity and cortical-dependent recognition
quencies for different scales of cortical integration: memory. Sleep, 42(10). doi: 10.1093/sleep/
From local gamma to long range alpha/theta syn- zsz143
chronization. International Journal of Psychophysi- Zitting, K. M., Münch, M. Y., Cain, S. W., Wang, W.,
ology, 38, 301–313. Wong, A., Ronda, J. M., … Duffy, J. F. (2018).
Zielinski, M. R., Atochin, D. N., McNally, J. M., McK- Young adults are more vulnerable to chronic sleep
enna, J. T., Huang, P. L., Strecker, R. E., & Gerash- deficiency and recurrent circadian disruption than
chenko, D. (2019). Somatostatin+ /nNOS+ neurons older adults. Scientific Reports, 8. doi: 10.1038/
are involved in delta electroencephalogram s41598-018-29358-x
This page intentionally left blank
PART V
Sensory-Perceptual
Systems and Cognition
This page intentionally left blank
21
Auditory Processing
Erich Schröger and Alexandra Bendixen

INTRODUCTION introduce some important subfunctions that aid


our auditory system in this challenging task and
Auditory information processing is in the service some models explaining how these functions are
of gaining information about the physical, psy- achieved.
chological, and social world provided by the In addition to seeing, hearing is of high impor-
acoustic environment. One important task is to tance for human communication and other cog-
establish neural or mental representations of nitive functions. Analogous to electromagnetic
sound sources (e.g., a particular speaker) and of waves in vision, vibrations of a certain wavelength
the auditory stimulus events they generate (e.g., a are picked up in hearing and converted into sen-
spoken word). These events (i.e., discrete sounds) sations. In the context of auditory processing, the
are consciously perceived as auditory objects and oscillations are air pressure fluctuations caused
can be used by other processing modules of the by the sound source, that is, the sound-producing
brain or mind. Following the definition of Bizley entity such as a musical instrument, the human
and Cohen (2013), auditory objects are regarded vocal apparatus, or the collision of two enti-
as perceptual constructs which correspond to ties. Often, the physical input parameters and
sound events that can be assigned to a particular their psychophysical transformation are equated
source. In order to accomplish this task, the sen- between hearing and seeing: the amplitude of the
sory stimulation caused by the arriving sound vibration is perceived as intensity (i.e., loudness
waves has to be analyzed and related to our exist- or brightness), the wavelength (frequency) of the
ing knowledge about the world. This includes vibration corresponds psychologically to the pitch
many subfunctions such as the identification of a or the experienced hue. But even for this simple
particular sound source (e.g., a human speaker), analogy one can already find counter-arguments.
the identification of the message conveyed by the One is that pitch is perceptually represented in
activity of that sound source (e.g., the understand- one dimension (i.e., all pitches can be arranged
ing of a spoken sentence), the separation of con- next to each other on a scale), while colour space
currently active sound sources (also known as is typically perceived as multidimensional – it is
cocktail-party problem; Cherry, 1953; Holmes not possible to sort all colours in order (Kavšek,
et al., 2021) and many others. This chapter will 1996). Another counter-argument is the difference
338 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

in naming performance: while naming colours is was intentionally created by the listener of the
not a problem for most people, correctly naming sound or by somebody else (i.e., impact of action).
the pitch of a tone on a musical scale is a com- And vice versa, a loud sound in the back could make
paratively rare gift – so-called absolute pitch is us turn (i.e., impact on motor behaviour / action)
possessed by few people only (e.g., Elmer et al., and visually analyze the sound source (i.e., impact
2015). Such differences suggest that auditory pro- on visual attention).
cessing is organized by (partly) different princi-
ples than visual processing.
Possibly the most striking difference between
vision and audition is that the objects of the
respective sensory system are quite different with PHYSICAL, PHYSIOLOGICAL AND
respect to their ontological state. The objects of PSYCHOPHYSICAL PRINCIPLES OF
seeing usually exist in ways that one can approach HEARING
them, can touch them, can determine their weight
and size; they are usually stable, that is, they are
there before one has seen them and continue to be Mapping Physical to Perceptual
there after one has seen them. The objects of hear- Dimensions (Psychophysics)
ing (the sounds) do not exist in the same sense.
Sounds are volatile, they often fade within a frac- Pitch
tion of a second, one usually cannot reinspect Auditory stimulus events are (discrete) sounds
them in the same way as a visual object. In other generated by an active sound source, that is, a
words, sounds do not materialize similarly to the sound-producing entity such as a musical instru-
objects of visual perception. Although generated ment, the vocal apparatus of a bird, or the colli-
by an existing entity (i.e., the sound source such as sion of a spoon with a plate. The sound-producing
a human speaker), the perceived sound is phenom- entity moves quickly back and forth (i.e., vibrates)
enologically not equivalent to this entity. Unlike so that fluctuations in air pressure are generated,
in vision, where the seen person is the person, that is, alternating zones of compressed and
the heard word uttered by the speaker is not the decompressed air, which propagate as longitudi-
speaker. There are similar, but exceptional cases nal waves. That is, in everyday life, acoustic infor-
in vision – for example, warning or signal lights mation is transmitted by changes in air pressure.
are usually not taken for the lamp or flashlight that These changes (travelling sound waves) can often
sends them, but for the message the signal com- be described as a superposition of different sinu-
municates (e.g., SOS). soidal oscillations with a defined frequency. For
The information in the acoustic signal is distrib- example, a human speaker emits signals of a cer-
uted in time; for example, on a microscale the air tain fundamental frequency with several harmonic
pressure fluctuations occurring a thousand times overtones (integer multiples of the fundamental
per second at the eardrum results in the percep- frequency). The fundamental frequency depends
tion of a high-pitched tone; on a macroscale the primarily on the speaker’s current vocal pitch, the
sequential presentation of tones with different composition of the overtones on his breathing
pitch may result in the perception of a familiar technique as well as on anatomical parameters of
melody. The exploitation of information contained his vocal apparatus.
in sequences on different time scales and the con- The frequency of a tone is expressed in Hertz
catenation of this information seems to be a char- (Hz; number of oscillation cycles per second). Fast
acteristic processing principle in audition. This vibrations are perceived as high-pitched tones,
ability to recognize sequences and “sequences slow vibrations as low-pitched tones. Humans can
of sequences” (Kiebel et al., 2009) is essential in hear frequencies with a lower limit of about 16 Hz
auditory information processing. and an upper limit of about 20 kHz. This frequency
The auditory processing system interacts with range declines significantly with age, especially
other information processing systems (e.g., vision, the upper limit. For that reason, elderly listen-
action, memory, intention, emotion) as it provides ers show difficulties in hearing high-­frequency
and receives information to and from these other sounds, which impedes their ability to differen-
systems. For example, the perceived location of a tiate certain speech sounds. Yet sounds with fre-
sound may be affected by the visual information quencies outside the audible frequency range can,
about the sound source (i.e., impact of vision for to some extent, still be perceived. Intense low-
audition), the quality of the representation of the frequency infrasound (< 16 Hz) can cause soma-
sound event may depend on the voluntary alloca- tosensory feelings, and high-frequency ultrasound
tion of attention (i.e., impact of attention), the per- (> 20 kHz) can lead to auditory percepts when it
ceived loudness may depend on whether the sound reaches the cochlea via bone conduction.
Auditory Processing 339

Loudness studied for the sound of musical instruments


The second important characteristic of a vibration (McAdams et al., 1995). However, it also plays an
besides frequency is its amplitude. This physical important role in recognizing other sounds, espe-
parameter translates into loudness perception at cially in recognizing speakers by their voice. The
the psychological level (although loudness is also characteristic timbre of a sound source is deter-
influenced by various other factors, such as sound mined by the envelope, especially the time course
duration, complexity and distance of the sound at the onset of a sound (rise time), the temporal
source). Natural sound waves typically do not development of harmonics over time, and the rela-
have a constant amplitude, but show amplitude tive proportions of the energy of odd and even
changes over time. These amplitude changes can overtones in the sound mixture. The importance of
be described by the envelope of the sound. In timing in a sound can easily be demonstrated by
contrast, the temporal fine structure is determined playing a short piano piece backwards, which
by the relative mixture of the frequency compo- sounds as if played by an organ. Some sounds
nents of the sound signal occurring at a particular increase in amplitude only gradually (e.g., an
time. Surprisingly, complex signals such as speech organ sound), while others begin with a very
can be understood quite well based on the enve- sudden increase in amplitude (e.g., a piano tone).
lope alone. However, in noisy environments, fine It is still investigated which other physical proper-
structure becomes more important (Moon et al., ties contribute to the psychological experience of
2014; Moore, 2008). timbre and how timbre can be objectively meas-
The physical quantities of loudness are sound ured (e.g., Rahne et al., 2010).
intensity and sound pressure level (SPL). Sound
intensity is the energy of sound that passes Location
through a given area per second. Sound pressure In addition to pitch, timbre, and loudness, the
level is the more common term for the strength location from which the sensory information
of a sound. It is usually reported as a logarith- originates is of importance, especially in contexts
mic quantity relative to a reference sound pres- in which the auditory system provides a warning
sure of 20Pa (2*10−5 N/m²), which is indicated function for the organism. In order to be able to
by SPL after the sound level value (e.g., 40 dB react quickly to warning signals (the horn of a
SPL in a typical office environment). In terms of vehicle, the roar of a lion), it is indispensable that
audible sound intensities or sound pressures, the our auditory sense not only informs us that there
human auditory system has a wide dynamic range, is danger “somewhere” in the environment, but
extending from the absolute threshold of hearing that it also immediately indicates the location (or
(which is at about 0 dB SPL for a 2000 Hz sound, at least the direction) of the danger. Unlike in
but can be very different for sounds of other fre- vision (or somatosensation), the neural signal
quencies) to the threshold of pain (which is at generated by sound waves in the inner ear does
about 140 dB SPL, but again very much depend- not contain direct information about the sound
ent on the frequency composition). With a senso- location. In vision, the light (emitted or reflected
rineural hearing loss that often comes with age or by the seen entity) hits a restricted area of the
with a long history of noise exposure, the absolute retina, that is, the two-dimensional location infor-
hearing thresholds increase – that is, sounds need mation is present at the respective sensory organ
to be physically more intense for the listener to be and propagated to the corresponding areas in the
able to hear them. This threshold increase happens visual cortex (retinotopy). There is no such direct,
in a frequency-specific manner. At the same time, receptor-encoded location information in audi-
the threshold of pain does not increase, instead it tion. Nevertheless, the location or direction of a
usually decreases with sensorineural hearing loss sound can be indirectly calculated by the auditory
as well. This convergence of the lower and upper system. The lateralization of sound sources is
thresholds of hearing leads to a compression of the largely based on binaural hearing, i.e., on an inte-
dynamic range for loudness perception. gration of information from the two ears. In this
integration process, interaural time differences
Timbre (ITDs) and interaural intensity differences (IIDs,
A perceptually significant aspect of a sound is also: interaural level differences, ILDs) are evalu-
timbre, which enables the listener to distinguish ated, which give information about the location of
similar sounds by the sources that produced them the sound source. A sound signal coming from the
(e.g., a musical note played by two different left reaches the left ear earlier (ITD) and with
instruments, or a word uttered by two different higher intensity (ILD) than the right ear, the latter
speakers). Unlike frequency or amplitude, timbre due to energy losses over time and due to the
is a multidimensional quantity. Timbre is best sound shadow caused by the intervening head.
340 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

ITD and ILD are not directly perceptual properties matching (cross-disparity), and monocular cues
of sound (i.e., one has no conscious access to the (occlusion, motion parallax, and the like). Despite
information that the left ear was stimulated earlier this limitation compared to visual perception, we
or more intensely, respectively), but they are auto- can find our way auditorily quite well in familiar
matically recoded into directional information acoustic environments. Even in a new acoustic
(i.e., the conscious percept is that of a sound environment, we quickly learn to orient ourselves
source located to the left of the listener). The on the basis of acoustic information.
resolvable travel time differences are far below Together, information about azimuth, elevation,
one millisecond – an amazingly precise comput- and distance enables an approximate three-dimen-
ing power of the auditory system. sional perception of a sound’s location. Seemingly
With these binaural cues, mainly the horizon- even more complicated, but ecologically more
tal localization of sound sources (left vs. right, relevant, is the localization of a sound source that
so-called azimuth) can be achieved. Vertical is moving in space (e.g., a vehicle approaching or
localization estimation (above vs. below, so-called moving away from us or the mosquito surround-
elevation) is based on monaural cues (i.e., acces- ing us at night), so that the location of the acoustic
sible on the basis of information from only one stimulation is constantly changing relative to the
ear). These are generated by reflections and dif- listener. Interestingly, this does not seem to pre-
fractions of the sound wave in the outer ear, which sent any major difficulties for the auditory sys-
acts like a filter. Such a filter can be described by tem, since it is very well prepared to receive and
the outer-ear- or head-related transfer function relate sequential input signals (i.e., sound events
(HRTF). However, in order to correctly interpret stretched over time). The underlying ability to pro-
this function, a certain knowledge of the physi- cess sequential information will be discussed later.
cal composition of the original signal is required.
This – as well as the experiential knowledge of the
acoustic properties of one’s own outer ear – must
first be learned in the course of life. Neural Encoding of Sounds
By integrating monaural and binaural cues, (Physiology)
sound sources can be localized reasonably well
in two-dimensional space; nevertheless, some- From sound waves to action potentials
times ambiguities remain – that is, a certain acti- The auditory sensory system converts vibrations
vation pattern cannot be clearly assigned to a within the audible frequency range into neuronal
specific sound direction. In these cases, listener activation patterns. When arriving at the listener,
help themselves – often without noticing it – with sound waves are first bundled and amplified by
small head movements, which slightly change the the outer ear consisting of the pinna, the ear canal,
activation pattern and thus resolve the ambiguity. and the outer part of the eardrum. The sound
This example illustrates that perception and motor waves cause vibrations of the eardrum, which are
activity are closely linked in hearing – not only propagated and amplified by a chain of small
in the sense that perception is the basis for motor bones (hammer/malleus, anvil/incus, stirrup/
activity, but also in the opposite sense: “We must stapes) to the oval window enabling the transmis-
perceive in order to move, but we must also move sion of vibration in air into vibration in liquid. The
in order to perceive” (Gibson, 1979). oval window separates the middle (filled with air)
In addition to the direction of sound (left/right, and the inner ear (filled with fluid). The deflec-
up/down), the distance of the sound source from tions at the oval window move the fluid in the
the listener is also a relevant property in order to inner ear; this is possible as the other window
be able to react adequately to a sensory stimulus separating the middle and the inner ear (the round
(the closer the honking car, the greater the need window) moves jointly with the oval window to
to act). Distance information, in turn, can also dissipate energy.
only be inferred indirectly; it is largely based on The actual translation into neural activity is
the loudness of the signal, which is related to the performed by a sensory organ in the inner ear, the
known or average loudness of the sound source so-called organ of Corti on the basilar membrane
in question – similar to the way the visual sys- inside the cochlea. Here, the receptors of the audi-
tem includes the typical size of entities in the tory system (the hair cells) are located. The cochlea
distance calculation. However, it must be taken is a spiral-shaped cavity that is separated into three
into account that the visual system – in contrast chambers or ducts, the vestibular, the tympanic
to the auditory system – usually has numerous and the cochlear duct. They are filled with two
other cues available for spatial depth estimation, different fluids (perilymph, endolymph) which
for example, signals from the oculomotor system are similar to the fluid in which the brain floats.
(accommodation, convergence angle), binocular The vibrations of a sound lead to corresponding
Auditory Processing 341

movements of the endolymph in the cochlear In this respect, analogies exist between tonotopic
duct. When the basilar membrane vibrates, the coding in the auditory system, retinotopic coding
structures on the basilar membrane are shifted in the visual system (i.e., coding according to the
against each other. This particularly affects the location of activation on the retina), and somato-
tectorial membrane, where the outer hair cells, topic coding in the somatosensory system (i.e.,
topped with hair-like structures called stereocilia, coding according to the location of activation on
are attached. The back and forth of the tectorial the body). It is important to keep in mind that loca-
membrane in rhythm with the vibration bends the tion-based encoding in vision and touch (unlike
stereocilia. When the stereocilia are deflected, a audition) directly reflects the actual location in the
sensing potential is generated at the hair cells that world from which the activity originated because
initiates neural information processing (i.e., action of the physical relationships between sensory
potentials in primary auditory neurons). About stimulus and receptor.
3000 inner hair cells and 13000 outer hair cells
are located in the organ of Corti. Inner and outer
refers to the turn of the cochlea: the inner hair Auditory pathway
cells are arranged in the inner turn. The inner hair Most properties of a sound must be extracted from
cells provide 95% of the output of the cochlea. the neural signals along the auditory pathway
The outer hair cells, instead, mainly receive neural between the cochlea and the primary auditory
input from the brain, which influences their motil- cortex. At the level of the hair cells, the mechani-
ity as part of a special function of the cochlea as cal information is converted into neuronal excita-
a kind of mechanical pre-amplifier. This motility tion at the axons of the auditory nerve. In addition
increases the sensitivity to sounds considerably. to hair cell deficiency, prolonged noise exposure
The motility expresses as evoked emissions, which can also lead to damage in the synapses between
can be measured as an echo when a click is played the hair cells and the auditory nerve, and in conse-
into the ear. The evoked emissions are louder and quence to a degeneration of auditory nerve fibers
longer in a well-functioning inner ear than to be (Kujawa and Liberman, 2009). This so-called
expected from a passive system. Measuring these cochlear neuropathy leads to severe impairments
emissions is an important part of routine diagnosis in understanding speech, but it does not come with
for possible hearing deficits in newborn infants. elevated auditory thresholds because it mainly
Damage or deficiency of hair cells is the core affects supra-threshold sound encoding
reason for elevated auditory thresholds that may (Bharadwaj et al., 2014). The term hidden hearing
come with age or with prolonged noise exposure loss has been coined for this deficit (Liberman,
(sensorineural hearing loss). 2015) since – unlike sensorineural hearing loss –
The location at which the hair cells are bent it is not revealed by conventional auditory thresh-
maximally (i.e., the location at which the move- old testing.
ment of the basilar membrane is largest) directly The auditory processing pathway involves a
maps onto the frequency of the vibration. This number of relay stations (nuclei, assemblies of
correspondence between frequency and coch- neuronal bodies) before the information arrives
lear location (called tonotopy) is realized by the at the cortex: the cochlear nucleus, the trapezoid
mechanical properties of the basilar membrane body, the superior olivary nucleus, the lateral lem-
(leading to different resonance frequencies in niscus, the inferior colliculi, and the medial genic-
different locations) and by the properties of the ulate body. The large number of relay stations
inner ear fluid (leading to different propagation indicates a high degree of pre-processing, since at
depths of sound waves of different frequencies). the corresponding synapses – as everywhere in the
For frequencies below about 5000 Hz, there is nervous system – information is not only passed
an additional time-based coding mechanism via on one to one, but is accentuated and recombined.
phase coupling of the activity of neuronal popu- The routing of signals in the auditory pro-
lations that “resonate” with the acoustic stimula- cessing pathway is hierarchical to a large extent.
tion. However, this mechanism reaches its limits Tonotopy as an organizing principle is maintained
at higher frequencies. in all core areas up to the auditory cortex, that is,
Tonotopy is the most important organizing prin- neighbouring populations of neurons respond to
ciple in the auditory system. The frequency selec- similar frequencies. Unlike, for example, in the
tivity is further enhanced by lateral inhibition, somatosensory system, there is no uniquely lat-
where an activated neuron reduces the activity of eralized (i.e., ipsilateral or contralateral) trans-
its neighbouring neurons. Tonotopy is found in all mission of information in audition; instead,
temporally and anatomically downstream stages processing pathways for signals from the left and
of the auditory processing pathway in the brain. right ears converge in several nuclear areas. This
342 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

allows for an exchange of signals received by both SOUND SOURCE SEPARATION


ears. The first core area for binaural integration is
the superior olive, where interaural time and level
differences (ITD, ILD) are computed as the basis The auditory system has to determine which
of sound localization. sound sources exist and which portions of the
Human auditory cortex is located bilaterally complex sound signal belong to which source.
in the superior temporal plane of the temporal This is not a trivial task considering that the
lobe and is largely “hidden” in the lateral sulcus acoustic waves generated by concurrently active
(Sylvian fissure). The auditory cortex is composed sound sources intermix with each other and with
of the primary auditory cortex, which extends over their echoes (generated when the waves bounce
the medial portion of the transverse temporal gyrus against a barrier, e.g., the walls in a classroom).
(also known as Heschl’s gyrus), and several sec- Moreover, the complex signals generated by a
ondary and association areas located in adjacent sound source (such as a human speaker) vary con-
areas of the anterior and posterior superior tempo- siderably over time. In addition, this extraction of
ral plane. The anatomical structure of the auditory sound sources must happen quickly in order to be
cortex shows major differences both between indi- able to monitor the occurrence of new (or the ter-
viduals and between the left and right hemispheres mination of active) sound sources in the dynamic
of the same individual. Hemispheric differences acoustic world. This process is called auditory
are partly associated with functional specializa- scene analysis (Bregman, 1990). Often, one of
tions related to speech and music processing. these sources is then to be selected for further
In vision, two different processing routes were (cognitive) processing. This is typically a function
distinguished, one mainly involved in the process- of attention. Some theories conceptualize the for-
ing of where information, the other in what infor- mation of sound sources and the selection of one
mation. A similar distinction has been proposed of those sound sources as one and the same pro-
for the auditory system. Starting from the primary cess (e.g., Shamma et al., 2011); others view the
auditory cortex, the ventral pathway, which con- two functions as sequential: Auditory scene analy-
nects the auditory cortex to the frontal lobe via sis comes first, followed by selection by the atten-
the anterior temporal lobe, is assumed to play an tional system (e.g., Bregman, 1990).
essential role in the detection and identification of Bregman (1990) proposed that the auditory
sound signals, whereas the dorsal pathway, which system uses rules that apply in the majority of
connects the auditory cortex and the parietal cases and whose application massively simpli-
lobe, is associated with the localization of sound fies the problem of auditory scene analysis. Such
sources (Wang et al., 2008). Overall, the auditory “simple” rules are called heuristics. One can
cortex has numerous connections to frontal brain distinguish bottom-up and top-down heuristics.
areas (Plakke and Romanski, 2014). Bottom-up heuristics are based on characteristics
It should be noted that at each intermediate pro- of the signal and describe typical signs that sound
cessing level of the auditory system, there are not components belong together (i.e. were emitted by
only ascending bottom-up connections but also the same source). They are better known under
various feedback loops. Anatomically, there are the term auditory Gestalt laws. Top-down heu-
often even more top-down than bottom-up connec- ristics describe prior assumptions of the listener,
tions. The function of the top-down connections for example, regarding the sound sources to be
are manifold. They are involved, for example, in expected in a particular listening environment.
the initiation of reflexive muscle contraction in the The Gestalt laws formulate abstract principles
middle ear and in top-down modulation of neural for whether certain components of the visual or
information. As a response to loud sounds, the auditory input should be integrated (i.e. perceived
stapedius muscle and the tensor tympani muscle as belonging together or assigned to the same
in the middle ear are contracted (acoustic reflex). source) or segregated (i.e. perceived as separate
This reduces transmission of mechanical energy to or assigned to different sources). Both integration
the inner ear and thus protects the inner ear from and segregation can refer to a specific point in
noise-induced damage. Neural processing at lower time (instantaneous auditory scene analysis) or
levels can be modified by top-down effects caused unfold over time (sequential or temporal auditory
by implicit expectations, current perceptual inter- scene analysis). Instantaneous or simultaneous
ests, and action goals of the listener: For exam- integration or segregation helps in determining
ple, certain acoustic information is prioritized in how many sound sources are currently active and
processing to focus on single sound sources in the which signal components belong to which one of
environment (e.g., the interlocutor), while other them. Integration or separation over time informs
sound sources (e.g., street noise) are largely sup- whether the sound just heard belongs to the
pressed from processing (see below). same auditory source, for example, whether the
Auditory Processing 343

same speaker who had already articulated sev- perceptual segregation of sound components and
eral words before continued to speak or whether is generated in the auditory cortex. The processing
another speaker kicked in. of inharmonicity has also been traced further down
the auditory processing hierarchy, at the level of
scalp-recorded brainstem ERPs within tens of mil-
liseconds after the onset of a sound (Bidelman and
Instantaneous Auditory Scene Alain, 2015).
Analysis
The initial decision of whether parts of a signal
belong together is based on cues that are instanta- Sequential Auditory Scene Analysis
neously available (i.e., they do not require taking
stimulus history into account), such as outstand- Unlike in instantaneous grouping, the decision
ing spectral cues, onset relationship, harmonic whether successively (sequentially) occurring
structure or location. The rationale of such an sound parts should be perceptually integrated or
instantaneous grouping rule is, for instance, that segregated requires taking stimulus history into
parts of a sound coming from different directions account. Sequential auditory scene analysis
at the same time could hardly have been produced assesses the similarity of the individual signals
by the same sound source and should therefore be over time: the more different the signals, the less
perceptually segregated. Similarly, natural sound likely it is that they originate from the same sound
sources produce overtones whose frequencies are source. This is consistent with the Gestalt law of
integer multiples of the fundamental frequency. similarity, with similarity referring to a variety of
When a single sound component falls out of such dimensions such as frequency, location, loudness,
a harmonic mixture, it is perceptually segregated or timbre. The rationale of the law of similarity is
and assigned to a different source; this is called that a sound source can produce sounds only
inharmonicity-based sound source segregation. within a well-defined range of characteristics
Conversely, there is no good reason to assume that (e.g., timbre and frequency range of an instru-
a mixture of sounds in perfect harmonic relation ment) or that its location and loudness does not
to each other happened to be produced by differ- vary too much within a short period of time. As a
ent sound sources; perceptual integration there- sound source changes its characteristics after a
fore takes place. The fundamental frequency may pause during which the sound source was inactive
even be missing in the signal and still be heard or not audible, perception assesses the physical
because it can be inferred from the other compo- similarity of the signals before and after the pause.
nents of the mixture. This so-called “missing The longer the pause, the more dissimilar the sig-
fundamental” effect is already found in four- nals can be in order to still be assigned to the same
month-old infants (He and Trainor, 2009) as dem- sound source. With this temporal notion, the
onstrated by recordings of their electrical brain Gestalt law of similarity changes into the Gestalt
activity. law of good continuation: The smaller the extent
The recognition of sounds in a mixture of dif- of feature value change per unit time, the greater
ferent sounds, that is, within a sound texture is the tendency to perceptually integrate the signals.
based on computations performed on the output of The law of good continuation creates a strong
the peripheral auditory system, which delivers the bias for a continuity of already known sound
sub-bands of the input signal and their envelopes. sources. However, when plausible cues for contin-
These computations, as shown by McDermott and uation are missing, the emergence of new sources is
Simoncelli (2011), yield the statistical moments considered. This so-called old-plus-new heuristic
of the compressed envelopes and cross-band cor- (Bregman, 1990) can result in continuity illusions.
relations. Experiments with human participants For example, if one intersperses a sound with regu-
listening to synthetic sounds constructed accord- lar gaps, that sound is correctly perceived as inter-
ing to the model suggest that the auditory system rupted; but if one fills the gaps with loud noise,
represents textures similarly to the principles of the sound is suddenly perceived as continuous
this model. (Dannenbring, 1976; cf. http://webpages.mcgill.
The segregation of sources based on instanta- ca/staff/Group2/abregm1/web/downloadstoc.
neous cues such as harmonic structure has been htm#29). Thus, the auditory system performs
investigated with an event-related brain poten- sound source separation by segregating out of
tial (ERP) component called the Object-Related the noise a sound that is not actually present but
Negativity (ORN) (Alain et al., 2001; Kocsis could be contained in terms of physical inten-
et al., 2016). The ORN occurs about 150–200 ms sity ratios. This phenomenon is called conti-
after the onset of an instantaneous cue indicating nuity illusion. Interestingly, this also works
344 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

when speech is masked with noise at regular et al., 2003a). It should be noticed that the elicita-
intervals (Warren, 1970). One can follow the tion of MMN not only indicates the brain’s detec-
speech signal interspersed with noise bursts bet- tion of a violation of an auditory regularity, but
ter than if it were interspersed with silent gaps also that the regularity must have been extracted
(for an auditory illustration of this picket-fence beforehand. Thus, MMN is a tool that taps into
effect with speech see http://webpages.mcgill. auditory representations and expectations.
ca/staff/Group2/abregm1/web/downloadstoc. The auditory scene analysis mechanisms
htm#31). At first glance, this is surprising. One described above happen to a large extent automati-
might think that the additional noise would rather cally. That is, the listener does not need to intend
confuse the listener, because the acoustic signal to segment the acoustic input – scene analysis
contains the same linguistic information in both takes place during passive listening without the
situations. On closer examination, such examples need to allocate voluntary attention. This is evi-
impressively illustrate how auditory perception denced by studies showing that ORN (Alain et al.,
uses heuristics and draws interpretations during 2002) and MMN (Symonds et al., 2020; Winkler
signal analysis. et al., 2003b) can be elicited when participants
Further grouping phenomena can be described do not actively listen to the sounds. However,
with the Gestalt law of common fate. In vision, this does not imply that ORN and MMN (and the
objects that move together are interpreted as corresponding scene analysis processes) cannot
belonging together. In audition, the joint common be modulated by attention. In fact, it seems that
rise in amplitude at sound onset and the common attention can improve the establishment of percep-
amplitude modulations (comodulation) while the tual representations underlying ORN (Zobel et al.,
sound is played biases perception toward a com- 2015) and MMN (Sussman, 2017). With age and
mon sound source (Bizley and Cohen, 2013). sensorineural hearing loss, the principal function
Based on this principle, the influential theory of of auditory scene analysis should remain intact,
temporal coherence has been formulated (Shamma but its outcome suffers from reductions in fre-
et al., 2011). Coherent changes can be found not quency resolution and in temporal precision that
only in the amplitude but also in the frequency of are part of the many changes the auditory system
the signal components: As the frequency of the undergoes. Hence many elderly listeners experi-
fundamental changes (e.g., intonation at the end of ence difficulties in listening in noisy environ-
an interrogative sentence), so do all the overtones, ments, part of which are probably due to reduced
which in turn provides evidence for the common precision of auditory scene analysis, while other
origin of the signal components. The temporal parts are due to changes of the attentional system
coherence of amplitude and frequency changes is with age (e.g., Weeks and Hasher, 2018). Attention
found on both short and longer time scales, plac- plays a crucial role when trying to extract meaning
ing it at the interface between instantaneous and from one of the sound sources upon segregating
sequential auditory scene analysis. It is possible it out of the mixture. The influence of attention
that this forms the long-neglected link between on the processing of auditory information will be
these two mechanisms of sound source attribution. discussed in the next section.
Sequential auditory scene analysis in humans
cannot be measured as directly from brain record-
ings as instantaneous auditory scene analysis,
since there is no direct electrophysiological marker
like the ORN. Yet auditory stimulus sequences AUDITORY ATTENTION
can be set up to obtain an indirect marker of
sequential auditory scene analysis, namely the Voluntary attention denotes the intended selection
Mismatch Negativity (MMN) component of the of sounds or sound sources in order to focus
ERP (Näätänen et al., 1978; Sussman et al., 2014). resources on relevant information – such as listen-
MMN is elicited when a sound is detected as vio- ing to a speaker against a mixture of background
lating a currently active (implicit) regularity of an noises in a cafeteria (or at a cocktail party, as the
auditory stream. When using MMN as a measure early literature on auditory scene analysis and
of sequential auditory scene analysis, one must attention liked to emphasize). Such resource allo-
embed a regularity into an auditory scene that cation can be achieved by attenuating the process-
can only be extracted if the sound sources in the ing of irrelevant, to-be-ignored information, or by
scene are segregated but not if they are integrated facilitating the processing of relevant information.
(or vice versa; Spielmann et al., 2014). With such Voluntary attention must be distinguished from
specifically tailored paradigms, it has been shown involuntary attention, which comes into play
that newborn infants can already separate sound when something unexpected happens in the audi-
sources in simplified auditory scenes (Winkler tory scene (put more formally, an implicit or
Auditory Processing 345

explicit expectation generated on the basis of the by auditory attention in several auditory and non-
internal model of the auditory environment is auditory cortical regions such as the superior tem-
violated). For example, a wrongly played note in poral, temporo-parietal, superior parietal, inferior
a melody or a mobile phone ringing may call for and middle frontal as well as supplementary motor
an attentional orienting toward the sound and areas (Alho et al., 2015). The attentional selec-
sound source. The sound or sound source can then tion of auditory information can be accomplished
receive additional processing in order to check for within a single dimension such a lateralization of a
the relevance of the “new” information and to pos- sound (e.g., “select left-side sounds, ignore right-
sibly initiate respective actions or to revise the side sounds”) or pitch (e.g., “select high-pitch
model about what to expect in the acoustic envi- sounds, ignore low-pitch sounds”), but also on
ronment. This call for attention by unexpected the basis of sound sources or streams (e.g., “lis-
stimulation may happen at the expense of costs in ten to the female speaker, don’t listen to the male
the processing of information relevant for the task speaker”), to some extent even with feature over-
at hand, and may result in behavioral distraction lap (Woods and McDermott, 2015).
(Parmentier, 2014). Voluntary and involuntary A newly emerging line of research attempts
attention complement each other, and are dis- to decode from listeners’ brain responses which
cussed below. sound source they are paying attention to (Mirkovic
et al., 2016; O’Sullivan et al., 2015). Such attempts
exploit the fact that cortical processing “tracks”
(i.e., temporally aligns with) the envelope of the
Voluntary Auditory Attention sound that is currently being attended (Ahissar
et al., 2001). The attention-decoding research is
Auditory attention research has classically dealt often motivated by the idea of using the informa-
with multi-talker scenarios: How can we recog- tion about the currently attended sound source for
nize what one speaker is saying in the presence of directional steering of a hearing aid, in order to
other active speakers? This amazing capability of facilitate the difficult scene analysis task for those
the auditory attentional system was introduced listeners who are hard of hearing. One challenge
into the literature by Cherry (1953) as the “cock- that remains to be solved is how such an attention-
tail party problem” and is still intensively steered hearing aid could still provide the listener
researched today (Bronkhorst, 2015; Holmes with information about unexpectedly occurring
et al., 2021). Other research is concerned with the sounds in the environment – in other words, how
allocation of attentional resources between modal- to maintain the function of involuntary attention.
ities, e.g., between hearing and vision (Zhang
et al., 2017). Effects of attention on the processing
of sounds can also be compared when attention is
directed to the external versus internal environ- Involuntary Attention
ment (Kam et al., 2021). In all three situations,
voluntary (top-down) attention is supposed to Auditory involuntary attention comes into play
cause a differential processing of attended and when something unexpected happens in the acous-
unattended auditory information, by facilitating tic environment. We orient our attention toward
the processing of to-be-attended information or by the unexpected event, an orienting response may
suppressing the processing of to-be-ignored infor- be elicited (Sokolov, 1990), phenomenologically a
mation (Bidet-Caulet et al., 2010). feeling of surprise or alertness may occur. Two
Attentional modulations along the auditory types of phenomena can trigger involuntary audi-
pathway have been investigated in thousands of tory attention: (1) a change or transient in an oth-
studies in animal and human research. Effects erwise stable acoustic environment, such as a
were reported all along the auditory pathway (Fritz change in the loudness or pitch of a continuous
et al., 2007). Some studies reported effects of tone; (2) a deviation from a regularity in the
attention already at the cochlear level in humans, acoustic environment, such as repetition of a tone
with attention modulating otoacoustic emissions in a series of otherwise alternating tones, or the
(Giard et al., 2000; Walsh et al., 2015; cf., Bell omission of a tone in an otherwise isochronous
and Jedrzejczak, 2021). Animal studies found sequence (note that the fact that attention can be
that spectro-temporal receptive fields (STRFs), triggered by an absence of information alludes to
characterizing the selective responsiveness of a non-trivial underlying mechanism). Two differ-
neurons to sounds, were larger when the sounds ent computational neural mechanisms were pro-
were task-relevant (Kaya and Elhilali, 2017). posed to be involved in these two forms of
Human functional magnetic resonance imaging involuntary attention. One consists in the release
(fMRI) research revealed increases in activation of neural adaptation to a stable acoustic
346 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

environment (May et al., 2015), the other in a duration of about 200 ms, which enables integra-
mismatch between a prediction derived from a tion and persistence phenomena. The other phase
detected regularity in the acoustic environment is the long (synthesized) auditory memory (also
and the actual sensory input (Wacongne et al., echoic memory), whose storage duration is about
2012). Some neurocomputational models attempt 10–30 secs. It represents a more static form of
to integrate these seemingly different mechanisms storage that serves as a kind of database for fur-
into a single mechanism (Chien et al., 2019). ther processing.
In terms of the best possible adaptation to the Psychological concepts of memory trace for-
environment, an appropriate balance between mation and memory fading have greatly influ-
involuntary and voluntary attention must be enced research on auditory perception. Much of
achieved. An imbalance toward voluntary atten- this research has been in the tradition of unidi-
tion can, for example, result in inattentional deaf- rectional bottom-up processing, where sensory
ness, a failure to recognize important auditory information is picked up, passed on, processed
events such as alarms (Scheer et al., 2018). Vice and transformed into mental representations.
versa, an imbalance toward openness for new During the past ~15 years, research on auditory
information can, for example, lead to disruption perception has experienced a paradigm shift from
by unexpected sounds during reading (Vasilev the prevailing framework of unidirectional infor-
et al., 2021). The challenging task of the lis- mation processing toward a framework including
tener lies in paying attention to one sound source feedback loops and exchange of bottom-up infor-
while at the same time monitoring all other sound mation with top-down expectations at all levels of
sources in the auditory scene in order to be open the auditory processing hierarchy. This develop-
to relevant changes in their behaviour. To achieve ment has been strongly driven by the application
this, the listener needs a mental representation of of predictive coding theory to auditory sensory
the whole auditory scene, in which the foreground information processing (Friston, 2005).
representation is processed in detail, and the back-
ground representation is continuously (and more
or less automatically) checked against the incom-
ing sensory information. Such auditory represen- Neural Representation and Predictive
tations have long been postulated in the tradition
of cognitive psychology, and are now increasingly
Coding
being refined in neural terms. The amazing skills of involuntary attention (notic-
ing energy decreases in sound sources that were
previously unattended) and of sequential auditory
scene analysis (assigning incoming signal parts to
AUDITORY REPRESENTATIONS sound sources to which they provide a good con-
tinuation) described above would be very cumber-
some to achieve in a purely bottom-up framework
Auditory Event Representations and of sensory information processing. The notion
Memory that regularities are extracted from the auditory
surroundings and that incoming sounds are
Listeners establish and maintain internal represen- checked against these regularities has been formu-
tations of auditory stimulus events (i.e., of discrete lated in the context of research on irregularity
sounds) that are consciously perceived and that detection (involving the MMN) and involuntary
can be used by other processing modules of the attentional capture. Some early MMN research
brain or mind. One important characteristic of has already expressed the idea that predictions are
auditory event representations is that they usually derived from the regularities (Winkler et al.,
remain active after the respective sound has faded. 1996). This idea has gained momentum when
Hence, they represent a form of memory. Keeping acoustic irregularities were discussed within pre-
information available for some time is important, dictive coding theory, a theoretical framework for
for example, in sentence comprehension or in sensory processing (Friston, 2005). According to
linking acoustic information across interruptions. this view, predictions are derived from the brain’s
In fact, the current concept of auditory event rep- generative model of the environment and com-
resentation (Winkler and Schröger, 2015) corre- pared with the actual sensory input. This dynamic
sponds to the classic concept of auditory memory system of feedback (predictions) and feedforward
from cognitive psychology (Cowan, 1984). For (prediction error) recurrent loops aims at minimiz-
auditory memory, two successive phases of stor- ing the prediction error. At each level of the corti-
age are distinguished. One is the short (pre-per- cal hierarchy, the prediction error is passed on to
ceptual) auditory memory, which has a storage the higher levels. Thereby, the amount of sensory
Auditory Processing 347

data that is fed forward and needs to be processed the interaction of bottom-up processing triggered
further focuses on those parts that are not already by the available sensory data and the top-down
accounted for by the internal model. The predic- processing triggered by our internal models. The
tion error is in turn used to improve the model. respective sensory data are based on excitation
When the prediction error is minimized, that is, patterns in the cochlea. The internal models host
when the prediction conforms to the input, the our beliefs about the world, that is, the representa-
internal model (i.e., the content specified by rep- tions of our mental (memory) system. The result
resentational neurons) is perceived. Predictive of these interactive processing are auditory per-
coding theory has been instantiated in specific ceptual representations of discrete sounds, which
neurocomputational and neurobiological models are linked to and used for various cognitive pro-
that support and refine the mechanistic principles cesses related to perception, attention, discrimina-
underlying the sequential nature of auditory per- tion, evaluation, action planning, and the like.
ception (e.g., Spratling, 2017; Wacongne et al., Predictive coding theory is the currently prevail-
2012). ing account for sequential auditory scene analysis,
Neurophysiological recordings show that irreg- background monitoring and involuntary attention
ularity detection happens at various levels of the shifting. Temporal coherence theory is promising
auditory pathway. Animal single-unit recordings for linking sequential auditory scene analysis with
revealed effects in primary auditory cortex, the instantaneous auditory scene analysis and with
medial geniculate body (MGB), and the inferior mechanisms of voluntary attention. A theoretical
colliculus (IC) (e.g., Valdés-Baizabal et al., 2021). framework joining the underlying principles with
A recent study investigated the functional speciali- one another and with our knowledge on psycho-
zation of auditory areas in the rat for mere change physical and psychophysiological principles will
detection versus irregularity detection (Parras allow us to gain an even better understanding of
et al., 2021). While the primary auditory cortex, the fascinating properties of auditory perception.
the anterior auditory field, the ventral auditory
field, and the suprarhinal auditory field are mainly
engaged in (release from) adaptation, irregularity
detection is processed preferentially in the non-
lemniscal, posterior auditory field. Research using REFERENCES
event-related fMRI revealed auditory irregularity
detection in the auditory cortex, the MGB and Ahissar, E., Nagarajan, S., Ahissar, M., Protopapas, A.,
the IC also in humans (Cacciaglia et al., 2015). Mahncke, H., & Merzenich, M. M. (2001). Speech
Altogether, a picture emerges that predictions are comprehension is correlated with temporal response
locally instantiated and irregularities detected at patterns recorded from auditory cortex. Proceedings
multiple different levels of the auditory pathway of the National Academy of Sciences of the United
(e.g., Tabas and von Kriegstein, 2021). States of America, 98, 13367–13372.
Alain, C., Arnott, S. R., & Picton, T. W. (2001). Bottom-
up and top-down influences on auditory scene
analysis: Evidence from event-related brain poten-
tials. Journal of Experimental Psychology: Human
SUMMARY AND CONCLUSIONS Perception and Performance, 27, 1072–1089.
Alain, C., Schuler, B. M., & McDonald, K. L. (2002).
In this chapter we have discussed principles and Neural activity associated with distinguishing con-
mechanisms of auditory processing from a cogni- current auditory objects. Journal of the Acoustical
tive and neuroscientific view. By means of audi- Society of America, 111, 990–995.
tory processing, we extract information from the Alho, K., Salmi, J., Koistinen, S., Salonen, O., &
acoustic environment about the physical, psycho- Rinne, T. (2015). Top-down controlled and bot-
logical and social world we interact with. This tom-up triggered orienting of auditory attention
information describes the sound sources (e.g., a to pitch activate overlapping brain networks. Brain
particular speaker) and their behavior (e.g., the Research, 1626, 136–145.
message of a spoken sentence). We can, for exam- Bell, A., & Jedrzejczak, W. W. (2021). Muscles in
ple, identify an instrument and enjoy a whole and around the ear as the source of “physiologi-
melody played by it, perceive the trajectory of a cal noise” during auditory selective attention: A
mosquito circling us, detect a familiar voice, rec- review and novel synthesis. European Journal of
ognize the mood of a speaker, understand the Neuroscience, 53, 2726–2739.
content of a spoken message, and notice a car Bharadwaj, H. M., Verhulst, S., Shaheen, L., Liber-
approaching us from behind. This is achieved by man, M. C., & Shinn-Cunningham, B. G. (2014).
348 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Cochlear neuropathy and the coding of supra- Gibson, J. J. (1979). The ecological approach to visual
threshold sound. Frontiers in Systems Neurosci- perception. Hillsdale, NJ: Erlbaum.
ence, 8. doi: 10.3389/fnsys.2014.00026 He, C., & Trainor, L. J. (2009). Finding the pitch of the
Bidelman, G. M., & Alain, C. (2015). Hierarchical missing fundamental in infants. Journal of Neuro-
neurocomputations underlying concurrent sound science, 29, 7718–8822.
segregation: Connecting periphery to percept. Holmes, E., Parr, T., Griffiths, T. D., & Friston, K. J.
Neuropsychologia, 68, 38–50. (2021). Active inference, selective attention, and
Bidet-Caulet, A., Mikyska, C., & Knight, R. T. (2010). the cocktail party problem. Neuroscience & Biobe-
Load effects in auditory selective attention: Evi- havioral Reviews, 131, 1288–1304.
dence for distinct facilitation and inhibition mech- Kam, J. W. Y., Helfrich, R. F., Solbakk, A.-K., Endestad,
anisms. NeuroImage, 50, 277–284. T., Larsson, P. G., Lin, J. J., & Knight, R. T. (2021).
Bizley, J. K., & Cohen, Y. E. (2013). The what, where Top-down attentional modulation in human fron-
and how of auditory-object perception. Nature tal cortex: Differential engagement during external
Reviews Neuroscience, 14, 693–707. and internal attention. Cerebral Cortex, 31,
Bregman, A. S. (1990). Auditory scene analysis: The 873–883.
perceptual organization of sound. Cambridge, Kavšek, M. J. (1996). Multidimensionale Skalierung
MA: MIT Press. von Farbmustern aus der DIN-Farbenkarte [Multi-
Bronkhorst, A. W. (2015). The cocktail-party problem dimensional scaling of color patterns of the DIN
revisited: early processing and selection of multi- color chart]. Zeitschrift für Experimentelle Psy-
talker speech. Attention, Perception, & Psycho- chologie, 43, 547–570.
physics, 77, 1465–1487. Kaya, E. M., & Elhilali, M. (2017). Modelling auditory
Cacciaglia, R., Escera, C., Slabu, L., Grimm, S., San- attention. Philosophical Transactions of the Royal
juán, A., Ventura-Campos, N., & Ávila, C. (2015). Society B: Biological Sciences, 372. doi: 10.1098/
Involvement of the human midbrain and thalamus rstb.2016.0101
in auditory deviance detection. Neuropsychologia, Kiebel, S. J., von Kriegstein, K., Daunizeau, J., & Fris-
68, 51–58. ton, K. J. (2009). Recognizing sequences of
Cherry, E. C. (1953). Some experiments on the rec- sequences. PLOS Computational Biology, 5. doi:
ognition of speech, with one and with two ears. 10.1371/journal.pcbi.1000464
Journal of the Acoustical Society of America, 25, Kocsis, Z., Winkler, I., Bendixen, A., & Alain, C.
975–979. (2016). Promoting the perception of two and
Chien, V. S. C., Maess, B., & Knösche, T. R. (2019). A three concurrent sound objects: An event-related
generic deviance detection principle for cortical potential study. International Journal of Psycho-
On/Off responses, omission response, and mis- physiology, 107, 16–28.
match negativity. Biological Cybernetics, 113, Kujawa, S. G., & Liberman, M. C. (2009). Adding
475–494. Insult to Injury: Cochlear Nerve Degeneration after
Cowan, N. (1984). On short and long auditory stores. “Temporary” Noise-Induced Hearing Loss. Journal
Psychological Bulletin, 96, 341–370. of Neuroscience, 29, 14077–14085.
Dannenbring, G. L. (1976). Perceived auditory conti- Liberman, M. C. (2015). Hidden hearing loss from
nuity with alternately rising and falling frequency everyday noise. Scientific American, 313, 48–53.
transitions. Canadian Journal of Psychology / Revue May, P. J. C., Westö, J., & Tiitinen, H. (2015). Com-
Canadienne de Psychologie, 30, 99–114. putational modelling suggests that temporal inte-
Elmer, S., Rogenmoser, L., Kühnis, J., & Jäncke, L. gration results from synaptic adaptation in auditory
(2015). Bridging the Gap between Perceptual and cortex. European Journal of Neuroscience, 41,
Cognitive Perspectives on Absolute Pitch. Journal 615–630.
of Neuroscience, 35, 366–371. McAdams, S., Winsberg, S., Donnadieu, S., De
Friston, K. (2005). A theory of cortical responses. Soete, G., & Krimphoff, J. (1995). Perceptual scal-
Philosophical Transactions of the Royal Society B: ing of synthesized musical timbres: Common
Biological Sciences, 360, 815–836. dimensions, specificities, and latent subject classes.
Fritz, J. B., Elhilali, M., David, S. V., & Shamma, S. A. Psychological Research, 58, 177–192.
(2007). Auditory attention-focusing the search- McDermott, J. H., & Simoncelli, E. P. (2011). Sound
light on sound. Current Opinion in Neurobiology, texture perception via statistics of the auditory
17, 437–455. periphery: Evidence from sound synthesis. Neuron,
Giard, M. H., Fort, A., Mouchetant-Rostaing, Y., & 71, 926–940.
Pernier, J. (2000). Neurophysiological mechanisms Mirkovic, B., Bleichner, M. G., De Vos, M., & Debener,
of auditory selective attention in humans. Fron- S. (2016). Target speaker detection with concealed
tiers in Bioscience (Landmark Ed), 5. doi: 10.2741/ EEG around the ear. Frontiers in Neuroscience, 10.
giard doi: 10.3389/fnins.2016.00349
Auditory Processing 349

Moon, I. J., Won, J. H., Park, M.-H., Ives, D. T., Nie, Sussman, E. S., Chen, S., Sussman-Fort, J., & Dinces,
K., Heinz, M. G., Lorenzi, C., & Rubinstein, J. T. E. (2014). The five myths of MMN: Redefining
(2014). Optimal combination of neural temporal how to use MMN in basic and clinical research.
envelope and fine structure cues to explain speech Brain Topography, 27, 553–564.
identification in background noise. Journal of Neu- Symonds, R. M., Zhou, J. W., Cole, S. L., Brace, K. M.,
roscience, 34, 12145–12154. & Sussman, E. S. (2020). Cognitive resources are
Moore, B. C. J. (2008). The role of temporal fine distributed among the entire auditory landscape in
structure processing in pitch perception, mask- auditory scene analysis. Psychophysiology, 57. doi:
ing, and speech perception for normal-hearing 10.1111/psyp.13487
and hearing-impaired people. Journal of the Tabas, A., & von Kriegstein, K. (2021). Adjudicating
Association for Research in Otolaryngology, 9, between local and global architectures of predic-
399–406. tive processing in the subcortical auditory path-
Näätänen, R., Gaillard, A. W. K., & Mäntysalo, S. way. Frontiers in Neural Circuits, 15. doi: 10.3389/
(1978). Early selective-attention effect on evoked fncir.2021.644743
potential reinterpreted. Acta Psychologica, 42, Valdés-Baizabal, C., Casado-Román, L., Bartlett, E. L.,
313–329. & Malmierca, M. S. (2021). In vivo whole-cell
O’Sullivan, J. A., Power, A. J., Mesgarani, N., Raja- recordings of stimulus-specific adaptation in the
ram, S., Foxe, J. J., Shinn-Cunningham, B. G., inferior colliculus. Hearing Research, 399. doi:
Slaney, M., Shamma, S. A., & Lalor, E. C. (2015). 10.1016/j.heares.2020.107978
Attentional Selection in a Cocktail Party Environ- Vasilev, M. R., Parmentier, F. B., & Kirkby, J. A. (2021).
ment Can Be Decoded from Single-Trial EEG. Cer- Distraction by auditory novelty during reading:
ebral Cortex, 25, 1697–1706. Evidence for disruption in saccade planning, but
Parmentier, F. B. R. (2014). The cognitive determi- not saccade execution. Quarterly Journal of Experi-
nants of behavioral distraction by deviant auditory mental Psychology, 74, 826–842.
stimuli: a review. Psychological Research, 78, Wacongne, C., Changeux, J.-P., & Dehaene, S.
321–338. (2012). A neuronal model of predictive coding
Parras, G. G., Casado-Román, L., Schröger, E., & accounting for the mismatch negativity. Journal of
Malmierca, M. S. (2021). The posterior auditory Neuroscience, 32, 3665–3678.
field is the chief generator of prediction error sig- Walsh, K. P., Pasanen, E. G., & McFadden, D. (2015).
nals in the auditory cortex. NeuroImage, 242. doi: Changes in otoacoustic emissions during selective
10.1016/j.neuroimage.2021.118446 auditory and visual attention. Journal of the
Plakke, B., & Romanski, L. M. (2014). Auditory con- Acoustical Society of America, 137, 2737–2757.
nections and functions of prefrontal cortex. Fron- Wang, W.-J., Wu, X.-H., & Li, L. (2008). The dual-
tiers in Neuroscience, 8. doi: 10.3389/fnins. pathway model of auditory signal processing.
2014.00199 Neuroscience Bulletin, 24, 173–182.
Rahne, T., Rasinski, C., & Neumann, K. (2010). Meas- Warren, R. M. (1970). Perceptual restoration of miss-
uring timbre discrimination with cross-faded ing speech sounds. Science, 167, 392–393.
synthetic tones. Journal of Neuroscience Methods, Weeks, J. C., & Hasher, L. (2018). Older adults encode
189, 176–179. more, not less: evidence for age-related attentional
Scheer, M., Bülthoff, H. H., & Chuang, L. L. (2018). broadening. Aging, Neuropsychology, and Cogni-
Auditory Task Irrelevance: A Basis for Inattentional tion, 25, 576–587.
Deafness. Human Factors, 60, 428–440. Winkler, I., Karmos, G., & Näätänen, R. (1996). Adaptive
Shamma, S. A., Elhilali, M., & Micheyl, C. (2011). Tem- modeling of the unattended acoustic environment
poral coherence and attention in auditory scene reflected in the mismatch negativity event-related
analysis. Trends in Neurosciences, 34, 114–123. potential. Brain Research, 742, 239–252.
Sokolov, E. N. (1990). The orienting response, and Winkler, I., Kushnerenko, E., Horváth, J., Čeponienė,
future directions of its development. Pavlovian R., Fellman, V., Huotilainen, M., Näätänen, R., &
Journal of Biological Sciences, 25, 142–150. Sussman, E. (2003a). Newborn infants can organ-
Spielmann, M. I., Schröger, E., Kotz, S. A., & Ben- ize the auditory world. Proceedings of the
dixen, A. (2014). Attention effects on auditory National Academy of Sciences of the United
scene analysis: insights from event-related brain States of America, 100, 11812–11815.
potentials. Psychological Research, 78, 361–378. Winkler, I., & Schröger, E. (2015). Auditory perceptual
Spratling, M. W. (2017). A review of predictive coding objects as generative models: Setting the stage for
algorithms. Brain and Cognition, 112, 92–97. communication by sound. Brain and Language,
Sussman, E. S. (2017). Auditory scene analysis: An 148, 1–22.
attention perspective. Journal of Speech, Lan- Winkler, I., Teder-Sälejärvi, W. A., Horváth, J.,
guage, and Hearing Research, 60, 2989–3000. Näätänen, R., & Sussman, E. (2003b). Human
350 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

auditory cortex tracks task-irrelevant sound transient spatial attention using steady-state visual
sources. Neuroreport, 14, 2053–2056. evoked potentials. Experimental Brain Research,
Woods, K. J. P., & McDermott, J. H. (2015). Attentive 235, 1575–1591.
tracking of sound sources. Current Biology, 25, Zobel, B. H., Freyman, R. L., & Sanders, L. D. (2015).
2238–2246. Attention is critical for spatial auditory object
Zhang, D., Hong, B., Gao, S., & Röder, B. (2017). formation. Attention, Perception, & Psychophysics,
Exploring the temporal dynamics of sustained and 77, 1998–2010.
22
Vestibular Processing in Cognition
P a u l C . J . Ta y l o r

INTRODUCTION but rather, detecting how the head is moving. The


structural blueprint of the vertebrate vestibular sys-
Although parts of the vestibular peripheral organs tem has been largely conserved across four hun-
were described in the seventeenth century, Antonio dred million years of evolution (Straka and Baker,
Scarpa provided the first detailed description of 2013). Within each inner ear are three semicircu-
the anatomy of the vestibular organs in 1789. lar canals and two other organs (otoliths), making
Subsequently, in 1842, Marie Jean Pierre Flourens 10 sensory organs, with five on each side. These
lesioned the semicircular canals of pigeons and tiny bodies are all encapsulated within the floor
reported that movements became uncoordinated of the skull (Figure 22.1), reducing any timelag
in particular planes, but concluded that their role between head movement and sensation.
was ultimately in audition. Jan Evangelista The three semicircular canals are loops contain-
Purkinje studied vertigo in 1820, noting like ing fluid, the endolymph, which moves relative to
others before him, that spinning people on rotat- the canal if three conditions are met. Firstly, the
ing chairs lead to systematic eye movements, but head has to rotate. Secondly, this rotation has to
he concluded that it was predominantly cerebellar. involve acceleration or deceleration. Canals are
From the 1870s onward, researchers started to blind to linear, constant velocity motion. Thirdly,
suggest that the vestibular organs play a role in the rotation has to be in the right direction – the
balance. For example, Ernst Mach noticed that the canals are oriented at right angles to each other in
semicircular canals were insensitive to linear three-dimensional space so that all potential head
forces, although he did not believe in a role for the rotations can be detected. The horizontal canals
endolymph’s motion, but rather its pressure. are oriented approximately horizontally, and so
William James noted that out of over 500 deaf are maximally sensitive to shaking the head from
mutes tested, more than one-third seemed immune side to side. Anterior and posterior canals are each
to dizziness after spinning, unlike all but one out tipped by about 45 degrees from the vertical. Each
of 200 of his Harvard academic colleagues. canal’s responses are increased by rotations in
The anatomy of the vestibular sensory organs one direction but inhibited in the opposite direc-
provides important clues and constraints to vestib- tion. The difference between responses between
ular function – core to which is not balance per se, “coplanar” members of each pair carries a rotation
352 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 22.1 The vestibular organs lie within the bone at the base of the skull (left). The
three semicircular canals are oriented at right angles to each other (middle) allowing detec-
tion of all types of head rotation (pitch, yaw, and tilt).

signal. The response of each canal is proportional material: a columnar layer of vertical filaments, a
to the cosine of the angle between the canal plane dense gelatinous layer of randomly arranged fila-
and the plane of motion (e.g., when the angle dif- ments, and the otoconial layer. This upper layer is
ference is low, the response is high). Fluid then a loose fibrous network studded with crystals of
pushes against the end of the canal (the ampulla), calcium carbonate in the form of calcite – a robust
which contains a diaphragm (the cupula), stretch- but inert building material present throughout bio-
ing across the whole extent of the canal, receiv- logical systems, for example in snail shells. If the
ing the full brunt of the shifting fluid. Within the head moves in the right way, specifically a linear
ampulla lie the sensory hair cells which transduce acceleration, this material is moved. As it is denser
mechanical movement into electrical impulses. than the endolymph, it is carried by its own inertia,
Hair cells extend into the cupula such that if the bending the hair cells underneath (in the semicir-
cupula is displaced, the hair bundles are deflected. cular canals, the cupula has a similar specific grav-
If the hair bundle is shifted in one direction, it ity to the surrounding endolymphatic fluid and so
depolarizes, and firing rate increases. A shift in the does not respond to linear forces). If movement
opposite direction leads to hyperpolarization and a orientation is aligned with the hair cells, they bend
reduced response. and trigger the sensory transduction cascade.
Some sources still maintain that the cupula does However, gravity is also a linear acceleration
not reach the ceiling of the ampulla but acts as a (about 9.81 m/s2, or “g”). Otolithic hair cells
swinging gate, because the cupula would retract respond not only if the head moves linearly but
and shrink back when prepared for histology. The also if the head is tipped such that gravity bends
canal response was already well described in 1931 the hair cells, under the weight of the otoconia.
by Wilhelm Steinhausen as an “overdamped tor- It is utterly ambiguous, from otolith responses
sion pendulum” (an analogy being a door handle alone, whether a vestibular stimulus comprises a
with a certain springiness to it, such that it snaps linear acceleration or a tilt with respect to grav-
back when pushed down). The biomechanics of ity. Sudden accelerations during aviation can be
the canal response mean the response varies with mistaken to indicate tilt of the aircraft (somato-
head velocity. Very slow rotations (below 0.1 Hz) gravic illusion) or even of the visual horizon (the
will not be detected because they do not produce oculogravic illusion). Canal input, together with
enough force to lead to relative motion of the visual, somatosensory, proprioceptive, and cogni-
endolymph. For normal head movements, from tive cues, can then resolve this.
about 0.5 Hz up to about 10 Hz (Grossman et al.,
1988), there is no phase difference between head
movement and the bending of the cupula, and so
the fluid viscosity is the deciding factor. The gain Down the Vestibular Nerve
of response reduces as frequency increases.
It is left to the otolith organs (utricle and sac- A vestibular hair cell only needs to be displaced by
cule) to detect linear accelerations. Again, they are a distance of the order of magnitude of a microme-
blind to constant velocity stimulation. The ves- tre (depending on the frequency) to generate a
tibular organs are then inertial sensors. The event depolarizing current through the ion channels
that is detected by the otolithic hair cells is pleas- within microseconds (Holt et al., 1998). These hair
ingly mechanical (Figure 22.2). The hair cells cells synapse onto afferent neural fibres that form
are physically weighed down by three layers of a bundle, the vestibular or vestibulocochlear nerve,
Vestibular Processing in Cognition 353

Figure 22.2 Schematic of otoconial hair cells and the layers of neuroepithelium

a branch of the VIII cranial nerve. Some hair cell four main nuclei and further subgroups, and many
ion channels are open at rest, lending these affer- nuclei have subdivisions and multiple names. The
ents a healthy baseline firing rate (e.g., higher than vestibular nuclei also receive from other brain-
retinal ganglion cells). These vestibular afferents stem structures, cerebellum and cortex.
can then carry information by responding more Additionally, efferent fibres project from the brain
quickly or more slowly, and they have a smooth stem back up to the hair cells, probably modulat-
response profile around the sensory threshold. Due ing hair cell excitability in a general and relatively
to the differing underlying sensory mechanics, diffuse manner (Goldberg and Fernandez, 1980).
afferents from the semicircular canals and otoliths Neurons from this synapse onward show mixed
encode velocity versus linear acceleration, respec- selectivities for velocity, acceleration, jerk, and
tively. Afferent fibres can be classified according to position (Laurens et al., 2017). Responses of the
the regularity of their spontaneous action poten- vestibular sensory organs or the vestibular nerve
tials. Compared with more regular fibres, irregular are independent of whether a head movement is
units are more sensitive, require less sensory input actively generated or passively applied (Cullen
to be activated, and have thicker axons (Goldberg, and Minor, 2002). Yet in one class of vestibular
2000). Irregular afferents can encode not only nuclei neurons of rhesus macaques, firing rates are
acceleration, but also “jerk,” the change in accel- suppressed by more than half if monkeys actively
eration. The two parallel information channels move their head relative to their trunk (Roy and
however do not correspond to different central Cullen, 2001) and the vestibulo-spinal reflexes are
vestibular pathways (Cullen, 2012). suppressed. The cerebellum is thought to predict
the sensory consequences of an action and to send
an (“efference”) copy of the motor command to
sensory areas. The sensation predicted to occur
Into the Subcortex with the action (for example, neck propriocep-
tive input) is then compared with what actually
Only a fraction of the hundreds of human subcor- happens. The neck proprioceptive input differs
tical regions has been mapped with MRI according to whether the head moves on the trunk
(Forstmann et al., 2017), and in general, classical or if the whole animal is moved passively, whereas
connectomics has focused on cortex (Johansen- the vestibular input is the same. If there is no dif-
Berg, 2013). Different nerve fibres project from ference (and no prediction error) this indicates that
the canals, utricle or saccule and lead to intricate everything is going as expected, and the represen-
connectivity patterns within the brain stem. This tation of the head turn in vestibular nuclei neurons
complex of so-called vestibular nuclei consists of is inhibited. This means that the firing of these
354 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

neurons does not indicate head movement per se, movements, and using excitatory or inhibitory neu-
but rather selectively represents unintended, pas- rotransmitters; the oculomotor nuclei also receive
sive head-movements. An expected head motion input from vestibular neurons that lie outside of this
arising from an intended movements does not pro- reflex arc and do not directly receive input from the
duce as much of a signal, and is prevented from vestibular nerve; and in addition to these canal-
causing distraction. driven reflexes, there are also otolithic projec-
tions to oculomotor neurons mediating even more
intricate circuits and other oculomotor reflexes
(Goldberg et al., 2012). In order to move the eyes to
Vestibular Reflexes for Movement shift gaze, the VOR must logically be inhibited so
Control that gaze does not simply stay fixed. Reflex motor
control, although by definition automatic, is subtle
Somatosensory and proprioceptive information and can vary with the motor context (making crisp
also converge on the vestibular nucleus, both from delineations of what is “reflexive” difficult).
the spinal cord, second-order neurons, cerebel- Before we turn to the contribution of vestibu-
lum, and cortex. Some (“vestibular-only”) neu- lar cortical areas, one last impressive property is
rons in the vestibular nuclei project to the spinal known to arise in the subcortex and to contribute
cord to mediate the vestibulo-spinal reflex, but to more abstract spatial cognition. The vestibular
there are also projections to the thalamus and the signals indicating a change in head position have
cerebellar nodulus and uvula, which are important clear relevance for the control of orchestrated
for postural control: vestibular cerebellar patients actions including multiple effectors. Accordingly,
do not show normal galvanically evoked body studies have investigated how the egocentric ref-
sway responses (Kammermeier et al., 2009). erence frame for the head (both vestibular and
The vestibular system plays a crucial role in the visual) is related to an absolute world-based
orienting of the eyes as well as the body. Separate allocentric frame. A key element here is head-­
(“vestibulo-ocular reflex”) neurons are also visu- direction neurons, which fire as a function of
ally sensitive and project to the motor neurons which direction an animal’s head is pointing inde-
for the eye muscles, to control gaze (Cullen and pendently of where the animal is within a given
Taube, 2017). One surprising aspect of subcorti- area (Taube et al., 1990). These neurons func-
cal circuitry is how separate structures are special- tion like little compasses, and have been reported
ized not only for specific types of eye movements throughout the mammal (and insect) brain. The
but also within different planes and sometimes key role of vestibular processing can be demon-
in specific directions (e.g., upward versus down- strated by bilateral peripheral vestibular lesions
ward). Three separate reflex pathways function so in the rat, which greatly disrupt the normal tun-
that the eyes can be kept fixating on an external ing of head direction cells in the anterior thalamus
stimulus even if the head moves. Angular, trans- (Stackman and Taube, 1997). Head direction cells
lational and torsional vestibulo-ocular reflexes form an important component building up to the
(VORs) are each specialized to move the eyes in allocentric medial temporal system culminating
the opposite direction to head movements occur- in hippocampal place cells, originally discovered
ring due to turn, shift, or tilt of the head, respec- in the rat to fire not only when the rat was point-
tively. The direct VOR arc consists of only three ing in a particular direction but also in a particular
neurons after the hair cell: the primary vestibular spatial area in the environment (and, interestingly,
nerve leading away from the labyrinth, the sec- also only if the animal was held down).
ondary neuron leading away from the vestibular
nucleus, and the motor neurons themselves in the
oculomotor nuclei which directly innervate the
muscles around the eyeball. After monkey head- Cortical Processing of Visual and
rotation, some counter-rotation in the eye is appar- Vestibular Heading in the Monkey
ent within about 6ms (Huterer and Cullen, 2002).
Such speed is necessary, because if there were any Given the sophisticated degree of processing
substantial lag between moving one’s head and already occurring in the vestibular nuclei and asso-
the eyes rotating in their socket to compensate ciated subcortical areas, one might be forgiven for
for this, the world would be perceived as moving wondering what more processing could possibly
every time that the head is moved, before the eyes be necessary. A widespread network of cortical
have responded. areas has now been identified to show an associa-
Additional pathways link the semicircular tion with vestibular processing. This evidence
canals to oculomotor neurons, modulating the arises from integrating converging results from
VOR, crossing the midline to co-ordinate binocular different approaches that have used anatomical
Vestibular Processing in Cognition 355

tracing, single-cell recording in monkeys being The same serial order exists for which areas show
moved around on motion platforms, or imaging in the earliest responses to vestibular translational
humans while the vestibular periphery is in some stimulation (Chen et al., 2011). Lesion studies indi-
way stimulated artificially, as well as tasks simulat- cate that the cortical network for heading is distrib-
ing vestibular sensation. uted. Lesions in MSTd (Gu et al., 2012) alone do
Although the thalamus is clearly far from not drastically impair vestibular heading percep-
simply being a relay of information, vestibular tion, nor integrating visual and vestibular cues, but
thalamic neuronal responses often mimic the ves- do impair visual heading. VIP lesions affect nei-
tibular nucleus, and a large thalamo-cortical net- ther visual nor vestibular heading, whereas PIVC
work responds to vestibular stimulation (Lopez lesions affect both (Chen et al., 2016).
and Blanke, 2011). At least three distinct thalam- This corpus of work has also supported more
ocortical pathways (Kotchabhakdi et al., 1980) general models of how information is processed in
innervate the many areas referred to accordingly the brain. Studies of Bayes-optimal cue integration
as “vestibular cortex” (Guldin and Grüsser, 1998). test statistically optimal or ideal observer models:
Unusually, there is no uncontested candidate for a the idea is that if information from one modal-
primary sensory cortex with as much support as ity (say, vestibular) becomes more or less reli-
in other sensory systems, where one cortical area able during an experiment, its signal is weighted
receives by far the most input from the sensory accordingly (Fetsch et al., 2013). Human partici-
thalamus. One promising approach for dissociat- pants weight vestibular information more or less
ing nodes within the vestibular network has been during multisensory judgments depending on the
to measure how neurons respond to vestibular coherence of the visual motion (ter Horst et al.,
stimulation, visual stimulation, and the combina- 2015).
tion of the two. In these experiments, the vestibular
stimulation is generated by actually moving mon-
keys on motion platforms while recording their
brain activity (note that the velocities and accelera- Imaging the Human Cortical
tions that can be used with platforms is well below Vestibular Network During Vestibular
the rapid speeds (up to 8G) at which monkeys can Stimulation
move their heads voluntarily (Carriot et al., 2017)).
The visual stimulation used is optic flow, simulat- The spatial resolution and compatibility with
ing what might be seen when moving through a motion platforms make single-cell recordings
simple visual scene consisting of a cloud of dots. from monkeys an extremely important resource
This reveals a multimodal network of areas which for the student of the human vestibular system.
may function to determine the direction in which Some ingenious experiments have been devised to
motion is toward, referred to as “heading.” Areas be compatible with human participants. Various
PIVC, VIP, and MSTd have received particular methods allow stimulating the vestibular organs
scrutiny, and may follow a serial hierarchical rela- peripherally while people are lying inside an MRI
tionship (Chen et al., 2016). In PIVC, vestibular scanner (Ertl and Boegle, 2019), and the mecha-
neuronal responses are most prominent, and this nism underlying each of these methods also illu-
area has therefore been highlighted as a key hub in minates our understanding of vestibular function.
the monkey: PIVC shows strong connectivity with In caloric irrigation a continuous stream of warm
vestibular subcortical nuclei. VIP shows an inter- or cold water is squirted into the left or right ear
mediate mix of vestibular and visual responses, (and caught below in a dish). This indirectly leads
and a closer association than MSTd with the to unilateral activation of the peripheral vestibular
reported judgement of the monkey: including after system, and enables manipulating the left or right
re-learning of visual-vestibular correspondences, if inner ear independently of the other (unlike during
a constant discrepancy is introduced between the real motion). Robert Bárány in 1906 proposed the
heading conveyed by vestibular and visual stimuli underlying mechanism was that a change in tem-
(Zaidel et al., 2021). MSTd has the greatest degree perature across the labyrinth produced convection
of visual responses (by contrast in MT vestibular currents in the endolymph. However, this cannot
responses are absent). MSTd also shows a particu- explain why caloric irrigation still had effects on
larly high proportion of neurons where the direc- two astronauts under the zero gravity of the
tion tuning for vestibular and visual tuning are European Spacelab 1 mission (in 1983), and so
opposite, thereby representing the combination of other non-convective explanations have been pro-
multimodal stimulation that would occur normally posed, such as changing the endolymph volume or
(e.g., if the head tilts clockwise, the retinal image thermally activating the vestibular nerve (Scherer
tilts counter-clockwise; Takahashi et al., 2007). and Clarke, 1985). In any case, caloric irrigation
356 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

can lead to a vestibular sensation as if rotating similar to the PIVC connectivity reported origi-
(vertigo), and even nausea. Caloric irrigation is nally in the monkey (Guldin and Grüsser, 1998).
also used clinically for testing the VOR which it Secondly, in addition to this region, a widespread
triggers, and the direction of the nystagmus it network is activated, including: on the midline a
causes flips direction depending on both the side medial part of posterior parietal cortex and of the
stimulated (left/ right) and the water temperature anterior cingulate, a ventral part of inferior pari-
(hot/cold). etal cortex and the superior temporal gyrus; and
The ability to use transcranial current stimula- the frontal eye fields – and some of these activa-
tion to stimulate the vestibular system was acci- tions are specific to particular forms of vestibular
dentally discovered (although not understood) at stimulation (Lopez et al., 2012), and this remains
least by the nineteenth century (Purkinje, 1820). a conservative list to be combined with other
Contemporary galvanic vestibular stimulation converging sources of data. In generative mod-
(GVS) in the human passes a current through the els such as predictive coding (Rao and Ballard,
head between an anode and cathode, at least one 1999) higher levels are considered to represent the
of which is placed over a mastoid. The clinical use expected percept, and an error signal is sent up to
to diagnose which side is impaired (using unilat- higher levels only in the event of a breach of that
eral montages) is not routine, due to high interindi- expectation in comparison to the incoming sen-
vidual variability and low selectivity (Dlugaiczyk sory signals from lower levels. According to this
and Straka, 2019). In order to increase the effects, interpretation, the neural activity caused by artifi-
smaller electrodes and higher intensities than in cial vestibular stimulation includes the prediction
conventional tCS are usually used (Nitsche and error caused by the absence of the other expected
Paulus, 2000), with settings of the order of 1–5 mA sensory inputs (Klingner et al., 2016).
(and 9 V). GVS likely directly activates both hair Newer methods for vestibular stimulation are
cells and the vestibular nerve, on the basis of inva- available. Strikingly, it has been demonstrated
sive GVS in the tadpole (Gensberger et al., 2016). that the static magnetic field of an MRI scanner
As with tCS, different temporal profiles of stimu- is capable of stimulating the vestibular system. A
lation can easily be applied, and the precise sen- nystagmus apparently caused by the magnetic field
sation of rotation, and pattern of eye movements is absent in patients with a bilateral labyrinthec-
caused are variable not only with electrode loca- tomy, and beats in different directions depending
tion but also across participants. on whether people are lying in the scanner head
Brief and extremely loud sounds can also stim- first or feet first. The mechanism suggested is via
ulate the vestibular organs, as shown by measur- the static magnetic field interacting with the ion-
ing the reflexive peripheral muscular contractions rich endolymphatic fluid to produce movement of
brought out by the reflex arcs triggered (Halmagyi the fluid inside the canals through ionic currents
et al., 2005). Note then that these three main meth- (“Lorentz forces”) (Roberts et al., 2011).
ods for stimulating the vestibular system during
imaging vary in which of the vestibular organs are
stimulated. Nonetheless two main messages come
from meta-analyses of imaging studies using Vestibular Information for
these methods for vestibular stimulation (Lopez Multisensory Guidance of Action
et al., 2012). Firstly, there is a particularly promi-
nent activation in the deep areas within the lateral Some form of vestibular representation is then
fissure, where the temporal lobe meets the rest combined with information arising from other
of cortex (i.e., the territory of the insula and the senses, often visual, for the control of movement,
parietal and temporal opercula). Although some whether of the eyes, limbs or body. If a constant
descriptions tend to collapse across these, there is velocity rotation is suddenly braked, the mechan-
clear heterogeneity and within these, OP2 may be ics of the semicircular canals mean they continue
a key area (zu Eulenburg et al., 2012). to send a signal indicating self-motion for some
Converging evidence suggests that here or time: this produces an illusory tilt and an altered
within these deep regions may be a human homo- VOR, implying the presence of an internal model
logue of the monkey region PIVC, although con- of gravity (Merfeld et al., 1999). Patients with
sensus is lacking in the field of exactly which vestibular sensory loss show problems with both
brain region in the human corresponds the best postural control and visual stability (Curthoys and
(Pfeiffer et al., 2014). This area shows strong Halmagyi, 1995). Vestibular integration for the
functional connectivity with a core inner circle of sensorimotor control of action depends on the
vestibular regions, including area 3a, other areas context (Medendorp and Selen, 2017). For exam-
around the lateral fissure, and an area near motion ple, to reach toward something while you are
visual cortex (zu Eulenburg et al., 2012): this is moving requires incorporating vestibular signals
Vestibular Processing in Cognition 357

to adjust and adapt to the various forces arising et al., 2018). TMS to the ventral parietal cortex
(Lackner and DiZio, 2005). GVS that simulates increases SVV errors (Fiori et al., 2015), and if the
movement to one side also biases hand trajectories head is tilted in one direction, errors are shifted in
to the other side during reaching (Bresciani et al., the other (Kheradmand et al., 2015). TMS more
2002). dorsally, to the intraparietal sulcus, improves SVV
Visual and vestibular processing interact: per- performance in normals (Willacker et al., 2019).
ception of self-motion relies above all on these Furthermore, through online ERP recording it
senses yet is impossible with either sense alone could be shown that TMS also reduced the normal
(Noel and Angelaki, 2022). Being accelerated to inter-individual variability in frontal neural pro-
the left or right on a platform biases which of two cessing. In contrast, dorsal medial frontal TMS
laterally presented visual stimuli are perceived to reduces the intra-individual variability in the SVV
have been presented first (Rincon-Gonzalez et al., in normals, increasing precision of perceptual
2016). Although people can estimate distance decision making (Willacker et al., 2020). Within-
traveled from optic flow alone, the vestibular (plus subject SVV variability is also increased in
somatosensory) cues from real motion appear patients with somatosensory loss (Barra et al.,
to dominate when both are presented together 2010). The SVV is shifted if a visual background
(Harris et al., 2000). Adaptation to optic flow rotates or if GVS is applied – and the effects of
biases the reported vestibular perception when these two summate linearly (Niehof et al., 2019).
moved on a moving platform afterward, as if ves- The SVV then reflects how verticality per-
tibular processing has been recalibrated (Cuturi ception, and vestibular processing more widely,
and MacNeilage, 2014). employ a wide variety of areas and can use a
Accordingly, acute vestibular loss is one cause range of different modalities and types of infor-
of oscillopsia, where the mechanisms for visual mation. The network includes the “core” regions
stability via the VOR are compromised and so around the insula, but converging data implicates
the visual scene is seen to move when the eyes other areas including members of the dorsal vis-
do (Rinne et al., 1998). Such oscillopsia recovers ual attentional network (Corbetta and Shulman,
over time, and this may reflect central compensa- 2011), such as the intraparietal sulcus, frontal eye
tion: changes in downstream neural excitability in field, and dorsomedial frontal cortex. For example,
such patients are also apparent as the raised thresh- the potential human VIP homologue also shows
olds for inducing phosphenes with TMS (Ahmad responses in fMRI to vision, touch, and audition
et al., 2017). Such patients show decreased rest- (Bremmer et al., 2001). TMS to similar regions
ing-state functional connectivity in the insula/ also affects other types of multimodal judgments
opercular cortex, but interestingly those patients (Konen and Haggard, 2014) and postural control
whose VOR recovered the best showed increased (Kaulmann et al., 2017). Human VIP is activated
connectivity between cerebellum and part of the in tasks requiring dissociating between simulated
posterior parietal cortex around the supramarginal self- and object-motion (Field et al., 2020). More
gyrus (Göttlich et al., 2014). FEF TMS improved generally, parietal cortex can represent posture in
visual acuity in normals in a model of oscillopsia freely foraging rats (Mimica et al., 2018) and may
using optokinetic nystagmus (Mastropasqua et al., integrate self-motion and visual input to form an
2020). The unusual involvement of these more input to the hippocampal formation for naviga-
dorsal areas is a theme that recurs below. tion (Nau et al., 2018). The frontal eye fields also
contain neurons in the macaque with a functional
profile supporting visual-vestibular integration
(Gu et al., 2016). FEF TMS not only affects neu-
Multimodal Representation of ral activity during visual attentional tasks (Taylor
Verticality et al., 2007) but can also elicit neck muscle activa-
tions as if triggering an orienting response via a
A large network of areas has been associated with head movement (Goonetilleke et al., 2011).
pathological tilt when participants are asked to
gauge whether a line presented in front of them is
vertical or tilted to one side. Although areas
around the deep insular/opercular region do show Representation of the Body in Space
a correlation with the subjective visual vertical
(SVV), implicated lesions clearly extend frontally, Vestibular processing then has a role in the transfor-
and parietally all the way up to the intraparietal mation of information between reference frames,
sulcus (Rousseaux et al., 2013). Transcranial the prediction of the sensory consequences of
direct current stimulation to the posterior right movement, and incorporation of intentional signals:
hemisphere shifts the SVV in normals (Santos these are also considered fundamental components
358 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

of attentional selection, spatial cognition, and the “yes” or “no” on the basis of EEG is improved if
control of voluntary movement. Contrary to subjec- those thoughts were previously conditioned with
tive experience, it has been hard to find a consensus GVS (Yoshimura et al., 2021). Establishing how
definition of attention (Nobre and Kastner, 2014): vestibular stimulation, including GVS, may have
although some abandon it as a useful concept effects will require future studies to control for
(Hommel et al., 2019), another approach is to con- the accompanying non-specific sensations arising
sider the prioritization of particular information directly from stimulation (Lopez, 2016) as well as
processing as a function that only seems bafflingly any indirect effects via eye movements or dizziness
general because it can apply to many stages of pro- and, as with tCS, demonstrating statistical differ-
cessing, from perception to action (Luo and ences from direct comparison to identical stimula-
Maunsell, 2019). Disrupting vestibular processing tion of a control site (Parkin et al., 2015).
can affect an egocentric representation used to
guide action. Three neglect patients showed judg-
ments of straight-ahead that were skewed abnor-
mally to the right, consistent with their neglect, but Visually Induced Vestibular Subjective
this was transiently restored after (cold) caloric Experiences
irrigation of the left ear (Karnath, 1994). GVS dis-
torts the reported perception of straight-ahead in It is possible to elicit a strong vestibular percept of
normals, an effect that still occurs with crossed self-motion by looking at a visual stimulus simu-
hands, and could not be attributed to effects of GVS lating what one would see if one was moving, even
on gaze direction (Abekawa et al., 2018). Voxel- with simple rotating patterns (reported by Ernst
based lesion symptom mapping in stroke patients Mach in 1875). Such “vection” is sometimes per-
correlates a deviation in straight-ahead with damage ceived by rail passengers who may feel the illusion
to a large strip of anterior parietal cortex running that their stationary train is moving, if they see a
from dorsal to ventral, whereas postural problems neighbouring train pulling away. In one extreme
were associated with the tissue around the insula case, participants sat still in a small furnished
and parietal operculum (Rousseaux et al., 2013). room, which could rotate fully around them, and
Recent work is exploring the role of vestibular perceived themselves to be performing full 360-
processing in other forms of body representation degree flipping (Allison et al., 1999): vection was
(Ferrè et al., 2013). One patient suffering acute much weaker if the field of view was restricted
vestibular loss, John Crawford (Harvard biolo- (a potential limitation for some fMRI and virtual
gist and physician), wrote an evocative account reality setups). Vection is if anything increased
in 1952 of his own subjective experience. In addi- during microgravity (Cheung et al., 1990) or while
tion to impaired balance, nausea, and oscillopsia, underwater (Fauville et al., 2021), when there are
he reported that if when moving in bed he closed fewer other somatosensory and proprioceptive cues
his eyes (to prevent dizziness), he would have the available to signify that the visual stimulation is
bizarre illusion that the bed itself was flipping incorrect. Vection can arise from other modalities
over on its side around him (cf. Memorial, 2005). and is sensitive to a wide range of stimulus proper-
Caloric irrigation increased the perceived size ties and cognitive factors (Palmisano et al., 2015).
of participants’ own hands, or of objects touch- fMRI studies presenting participants with rela-
ing their hands (Lopez et al., 2012). Deep brain tively large-field optic flow report BOLD changes
stimulation around the ventral part of the pari- in several areas (Berti and Keshavarz, 2020),
etal produced interesting reported experiences in including not only potential human homologues of
a patient: weak stimulation produced vestibular the vestibular areas mentioned above, MST, VIP
sensation (as had been reported in various areas and PIVC, but also V6 and CSv. Neural correlates
by Wilder Penfield in 1957), but high intensity of vection have been reported in the alpha band
stimulation lead to an out-of-body experience with EEG, for example by using sophisticated
where the patient reported seeing part of her body measures such as event-related spectral perturba-
from above (Blanke et al., 2002). Although such tions (Palmisano et al., 2016) or steady-state
experiences also have been reported in the normal evoked potentials (Dowsett et al., 2020). A similar
population, they are nearly three times as likely EEG signature has been found during studies of
in patients suffering from dizziness (Lopez and real motion to reflect the remapping of visual space
Elzière, 2018). (Gutteling et al., 2015), or to be inhibited in
Effects of GVS are increasingly being tested for patients with vestibular loss (Gale et al., 2016).
in other domains. Performance at a visual spatial The alpha band event-related desynchronization
memory task is improved if the targets were previ- evoked by optic flow, and that is related to postural
ously presented during GVS (Smith et al., 2020). sway and vection, can be reduced by top-down
The classification of whether a participant is thinking expectation (Obereisenbuchner et al., 2021).
Vestibular Processing in Cognition 359

Dizziness and sickness can of course be evoked The literature is mixed as to whether there is
by stimulating the vestibular system (e.g. spinning a correlation between these two subjective flow-
with eyes closed). There is also a class of visual induced phenomena, vection, and VIMS (Nooij
stimuli beyond optic flow which make viewers et al., 2017), or not (Golding et al., 2021). This
feel unwell, such as intense dazzling light, or some may depend greatly on factors such as the precise
flicker (Morgan et al., 2020). However, a feeling stimuli used, type of response, and sample size.
of nausea is precipitated if visual and vestibular In one study (Obereisenbuchner et al., 2021),
inputs conflict e.g. visually induced motion sick- the degree of vection correlated with the VIMS.
ness (VIMS). This can of course be one compo- However, there was also clear heterogeneity
nent of the sickness felt during vehicular transport, within the group whereby many people reported
but is also reported during computer games, flight vection without any VIMS, although no-one felt
simulators or virtual reality (“gaming sickness, sick without reporting vection.
“simulator sickness,” “cybersickness”), or many Moving the immediate visual surroundings
of the situations in which vection can be triggered. forward or backward induces postural sway. The
The evolutionary function and neural mechanisms visual stimulation indicates to participants that
underlying motion sickness and VIMS are unclear they are falling over, triggering compensation
(Lackner, 2014). In the study of nausea and vom- for the (erroneously) expected shift in momen-
iting, lesion studies in anaesthetized cats suggest tum. Visually evoked sway can be reduced by
causal roles for the cerebellum and its interac- top-down control, if the optic flow is self-initi-
tions with the vestibular nucleus plus other brain ated (Obereisenbuchner et al., 2021). Vestibular
stem nuclei, the area postrema, lateral tegmental patients are abnormally sensitive to sway-induc-
field, medial medullary reticular formation, and ing optic flow, showing higher magnitude sway
nucleus tractus solitarius (NTS), parabrachial (Redfern and Furman, 1994). Such visual depend-
nuclei, and the ventral respiratory group. Some of ency may lead to visually induced sensations of
these areas project onto respiratory motor neurons sway or dizziness in busy visual environments
(co-contraction of which produces emesis), others after other aspects of the vestibular disorder have
to the hypothalamus and limbic system, others to recovered (visual vertigo), associated with devia-
the cerebellum. Importantly for clinical models, tions of the reported vertical (Guerraz et al., 2001).
the lab rat and mouse cannot vomit, which may A functional (i.e., “psychogenic”) vestibular syn-
be a feature of all rodents, who lack not only some drome is now called persistent postural perceptual
brainstem structures but also bear important dif- dizziness (PPPD), encompassing previously used
ferences in the musculature of the abdomen (Horn terminology (Staab et al., 2017).
et al., 2013). In general, cross-species comparisons Treatments for dizziness depend on the specific
can delineate general principles underlying ves- causal disorder. During episodes of benign parox-
tibular processing, both when convergent mecha- ysmal positioning vertigo (BPPV), for example,
nisms arise, and if they differ due to a species’ particles of otoconia become detached, and floating
particular lifestyle and niche (Straka et al., 2016). freely, lead to vestibular stimulation. Consequent
Although visually induced motion sickness is bouts of dizziness can be exceptionally strong,
reduced in patients with bilateral labyrinthecto- but in some cases are resolved by performing a
mies, it is in some cases not abolished (Johnson sequence of whole-body jerks to physically shake
et al., 1999). Congenitally blind individuals can the otolithic material away (Mandala et al., 2019).
get motion sickness, just as the sighted can. There are several types of physical vestibular reha-
Skylab astronauts in zero gravity no longer felt bilitation which are robust treatments for several
sick during a complex rotation paradigm, as they causes of dizziness. Nevertheless, future strate-
did on Earth. They were tested several days into gies for treating vestibular disorders will need to
the mission, after which any initial space-sick- be geared toward the individual patient (Pavlou,
ness symptoms had improved through adaptation 2017), and there is a need to develop other treat-
or pharmacological treatment. Motion sickness ment approaches involving pharmacology, genetic
is blocked with lesions encompassing both area screening, vestibular implants, and the use of vir-
postrema and NTS (Borison and Borison, 1986). tual reality.
Activity in many of the brain stem nuclei for
vomiting also correlate with motion sickness:
nausea and vomiting may then be functions
of the same pathways, although the role of the SUMMARY AND CONCLUSIONS
many other neural areas responding to vestibular-
vestibular integration in triggering these is under- Within the walls of the skull lie the vestibular
explored (Yates et al., 2014). organs, mechanically tuned to detect specific head
360 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

movements. Although omitted from Aristotle’s list with some highlighting the differences between
of five classic senses, there is a now extensive lit- more reflexive processing versus more abstract
erature on the functions of the vestibular system. spatial cognition (Ferrè and Haggard, 2020), and
This chapter focuses on aspects of vestibular pro- others emphasizing how cognition can modulate
cessing relevant to cognitive neuroscience. Many the earliest stages of processing (Cullen and Wang,
fundamental principles delineating vestibular cog- 2020). Vestibular cognition needs to be considered
nition are inherited from the make-up of sensory conceptually (Garzorz and Deroy, 2020) and in
transduction onward. A full account of the pro- the context of many other systems, functions, and
cesses underlying how vestibular cognition principles mentioned in this book.
occurs, and how this relates to initial vestibular
processing stages, requires synthesizing the key
principles of early vestibular processing. Even the
simplest reflex-like vestibular functions, such as
compensatory eye movements or postural correc- ACKNOWLEDGMENTS
tion, are relevant for cognition. Vestibular sensory
processing is deeply intertwined with the motor The author is grateful to Madeleine Seitz for the
system. Furthermore, throughout the vestibular illustrations.
network and from the earliest stages there is an Funded by the DFG (TA 857/3-2).
extraordinary degree of multisensory interaction
(to the extent that stimuli in other modalities can
elicit vestibular sensations). Incorporating these
three principles of vestibular function – reflex,
motor, multisensory – sets the stage for the contri- REFERENCES
bution of this system to more abstract functions
such as the representation of egocentric space, Abekawa, N., Ferrè, E. R., Gallagher, M., Gomi, H., &
navigation, and the bodily self. In this way Haggard, P. (2018). Disentangling the visual,
research into the role of vestibular processing in motor and representational effects of vestibular
cognition spans fields ranging from sport science input. Cortex, 104, 46–57.
to space science, and from the clinical to the Ahmad, H., Roberts, R. E., Patel, M., Lobo, R., See-
philosophical. mungal, B., Arshad, Q., & Bronstein, A. (2017).
Every sense has its own peculiar proper- Downregulation of early visual cortex excitability
ties and phenomena. What principles dictate the mediates oscillopsia suppression. Neurology,
functioning of the vestibular system? First is the 89(11), 1179–1185.
direct relationship to action, including both rapid Allison, R. S., Howard, I. P., & Zacher, J. E. (1999). Effect
reflexes and voluntary motor control. Second is of field size, head motion, and rotational velocity on
the segregation into four main separable ascend- roll vection and illusory self-tilt in a tumbling room.
ing systems for i) postural stabilization; ii) gaze Perception, 28(3), 299–306.
stabilization; iii) an anterior thalamo-cortical path- Barra, J., Marquer, A., Joassin, R., Reymond, C., Metge,
way leading to the head cell network and medial L., Chauvineau, V., & Pérennou, D. (2010). Humans
temporal structures; and iv) a posterior thalamo- use internal models to construct and update a sense
cortical pathway leading to vestibular cortical of verticality. Brain, 133(12), 3552–3563.
areas and which is itself further divisible (Cullen, Berti, S., & Keshavarz, B. (2020). Neuropsychological
2019; Noel and Angelaki, 2022). Third is the high approaches to visually-induced vection: An over-
and early degree of integration with other senses, view and evaluation of neuroimaging and neuro-
indicating how these different paths may modu- physiological studies. Multisensory Research, 34(2),
late one another and have widespread feedforward 153–186.
feedback effects (Brooks and Cullen, 2019), and Blanke, O., Ortigue, S., Landis, T., & Seeck, M. (2002).
meaning that vestibular perception can be modu- Stimulating illusory own-body perceptions. Nature,
lated or even triggered by stimulation in other 419, 269–270.
modalities. Integration of different types of infor- Borison, H. L., & Borison, R. (1986). Motion sickness
mation beyond the purely vestibular is required to reflex arc bypasses the area postrema in cats. Experi-
appreciate at least some crucial aspects of vestibu- mental Neurology, 92(3), 723–737.
lar function. This leads us to the fourth principle, Bremmer, F., Schlack, A., Shah, N. J., Zafiris, O.,
the profound relationship between supposedly Kubischik, M., Hoffmann, K.-P., …Fink, G. R.
non-cognitive and cognitive processes (Firestone (2001). Polymodal motion processing in posterior
and Scholl, 2016). The best way to frame the dif- parietal and premotor cortex: A human fMRI study
ferent types of more or less cognitive vestibular strongly implies equivalencies between monkeys
processing has also not yet reached a consensus, and humans. Neuron, 29, 287-296.
Vestibular Processing in Cognition 361

Bresciani, J.-P., Blouin, J., Popov, K., Bourdin, C., Sar- Dlugaiczyk, J., Gensberger, K. D., & Straka, H. (2019).
legna, F., Vercher, J.-L., & Gauthier, G. (2002). Galvanic vestibular stimulation: From basic con-
Galvanic vestibular stimulation in humans pro- cepts to clinical applications. Journal of Neuro-
duces online arm movement deviations when physiology, 121(6), 2237–2255.
reaching towards memorized visual targets. Neu- Dowsett, J., Herrmann, C. S., Dieterich, M., & Taylor, P.
roscience Letters, 318(1), 34-38. C. J. (2020). Shift in lateralization during illusory
Brooks, J. X., & Cullen, K. E. (2019). Predictive sens- self-motion: EEG responses to visual flicker at 10 Hz
ing: The role of motor signals in sensory process- and frequency-specific modulation by tACS. Euro-
ing. Biological Psychiatry: Cognitive Neuroscience pean Journal of Neuroscience, 51(7), 1657–1675.
and Neuroimaging, 4(9), 842–850. Ertl, M., & Boegle, R. (2019). Investigating the ves-
Carriot, J., Jamali, M., Chacron, M. J., & Cullen, K. E. tibular system using modern imaging techniques:
(2017). The statistics of the vestibular input experi- A review on the available stimulation and imaging
enced during natural self-motion differ between methods. Journal of Neuroscience Methods, 326.
rodents and primates. Journal of physiology, 595(8), doi: 10.1016/j.jneumeth.2019.108363
2751–2766. Fauville, G., Queiroz, A. C., Woolsey, E. S., Kelly, J.
Chen, A., DeAngelis, G. C., & Angelaki, D. E. (2011). W., & Bailenson, J. N. (2021). The effect of water
A comparison of vestibular spatiotemporal tuning immersion on vection in virtual reality. Scientific
in macaque parietoinsular vestibular cortex, ventral Reports, 11(1). doi: 10.1038/s41598-020-80100-y
intraparietal area, and medial superior temporal Ferrè, E. R., & Haggard, P. (2020). Vestibular cogni-
area. Journal of Neuroscience, 31(8), 3082–3094. tion: State-of-the-art and future directions. Cogni-
Chen, A., Gu, Y., Liu, S., DeAngelis, G. C., & Ange- tive Neuropsychology, 37(7–8), 413–420.
laki, D. E. (2016). Evidence for a causal contribu- Ferrè, E. R., Vagnoni, E., & Haggard, P. (2013). Ves-
tion of macaque vestibular, but not intraparietal, tibular contributions to bodily awareness. Neu-
cortex to heading perception. Journal of Neurosci- ropsychologia, 51(8), 1445–1452.
ence, 36(13), 3789–3798. Fetsch, C. R., DeAngelis, G. C., & Angelaki, D. E.
Cheung, B., Howard, I., & Money, K. (1990). Visually- (2013). Bridging the gap between theories of sen-
induced tilt during parabolic flights. Experimental sory cue integration and the physiology of multi-
Brain Research, 81(2), 391–397. sensory neurons. Nature Reviews: Neuroscience,
Corbetta, M., & Shulman, G. L. (2011). Spatial 14(6), 429–442.
neglect and attention networks. Annual Review of Field, D. T., Biagi, N., & Inman, L. A. (2020). The role of
Neuroscience, 34, 569–599. the ventral intraparietal area (VIP/pVIP) in the per-
Cullen, K. E. (2012). The vestibular system: multi- ception of object-motion and self-motion. NeuroIm-
modal integration and encoding of self-motion age, 213. doi: 10.1016/j.neuroimage.2020.116679
for motor control. Trends in Neurosciences, 35(3), Fiori, F., Candidi, M., Acciarino, A., David, N., & Agli-
185–196. oti, S. M. (2015). The right temporoparietal junc-
Cullen, K. E., & Minor, L. B. (2002). Semicircular canal tion plays a causal role in maintaining the internal
afferents similarly encode active and passive head- representation of verticality. Journal of Neurophysi-
on-body rotations: Implications for the role of ves- ology, 114(5), 2983–2990.
tibular efference. Journal of Neuroscience, 22(11), Firestone, C., & Scholl, B. J. (2016). Cognition does
RC226–RC226. not affect perception: Evaluating the evidence for
Cullen, K. E., & Taube, J. S. (2017). Our sense of “top-down” effects. Behavioral and Brain Sci-
direction: progress, controversies and challenges. ences, 39. doi: 10.1017/S0140525X15000965
Nature Neuroscience, 20(11), 1465–1473. Forstmann, B. U., de Hollander, G., van Maanen, L.,
Cullen K. E. (2019). Vestibular processing during Alkemade, A., & Keuken, M. C. (2017). Towards a
natural self-motion: Implications for perception mechanistic understanding of the human subcor-
and action. Nature Reviews: Neuroscience, 20(6), tex. Nature Reviews: Neuroscience, 18(1), 57–65.
346–363. Gale, S., Prsa, M., Schurger, A., Gay, A., Paillard, A.,
Cullen, K. E., & Wang, L. (2020). Predictive coding in Herbelin, B., … Blanke, O. (2016). Oscillatory
early vestibular pathways: Implications for vestibu- neural responses evoked by natural vestibular
lar cognition. Cognitive Neuropsychology, 37(7–8), stimuli in humans. Journal of Neurophysiology,
423–426. 115(3), 1228–1242.
Curthoys, I. S., & Halmagyi, G. M. (1995). Vestibular Garzorz, I., & Deroy, O. (2020). Why there is a vestibu-
compensation: A review of the oculomotor, neural, lar sense, or how metacognition individuates the
and clinical consequences of unilateral vestibular senses. Multisensory Research, 34(3), 261–280.
loss. Journal of Vestibular Research, 5(2), 67–107. Gensberger, K. D., Kaufmann, A.-K., Dietrich, H.,
Cuturi, L. F., & MacNeilage, P. R. (2014). Optic flow Branoner, F., Banchi, R., Chagnaud, B. P., & Straka,
induces nonvisual self-motion aftereffects. Current H. (2016). Galvanic vestibular stimulation: cellular
Biology, 24(23), 2817–2821. substrates and response patterns of neurons in the
362 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

vestibulo-ocular network. Journal of Neurosci- Holt, J. R., Corey, D. P., & Eatock, R. A. (1998). Mecha-
ence, 36(35), 9097–9110. noelectrical transduction and adaptation in hair cells
Goldberg, J. M. (2000). Afferent diversity and the of the mouse utricle, a low-frequency vestibular
organization of central vestibular pathways. Exper- organ. Journal of Neuroscience, 17(22), 8739–8748.
imental Brain Research, 130(3), 277–297. Hommel, B., Chapman, C. S., Cisek, P., Neyedli, H. F.,
Goldberg, J. M., & Fernandez, C. (1980). Efferent Song, J.-H., & Welsh, T. N. (2019). No one knows
vestibular system in the squirrel monkey: anatomi- what attention is. Attention, Perception, & Psycho-
cal location and influence on afferent activity. physics, 81(7), 2288–2303.
Journal of Neurophysiology, 43(4), 986–1025. Horn, C. C., Kimball, B. A., Wang, H., Kaus, J.,
Goldberg, J. M., Wilson, V. J., Cullen, K. E., Angelaki, Dienel, S., Nagy, A., … Andrews, P. L. R. (2013).
D. E., Broussard, D. M., Buttner-Ennever, J., … Why can’t rodents vomit? A comparative behavio-
Minor, L. B. (2012). The vestibular system: A sixth ral, anatomical, and physiological study. PLoS One,
sense: Oxford, UK: Oxford University Press. 8(4). doi: 10.1371/journal.pone.0060537
Golding, J. F., Rafiq, A., & Keshavarz, B. (2021). Pre- Huterer, M., & Cullen, K. E. (2002). Vestibuloocular
dicting individual susceptibility to visually induced reflex dynamics during high-frequency and high-
motion sickness by questionnaire. Frontiers in Vir- acceleration rotations of the head on body in
tual Reality, 2. doi: 10.3389/frvir.2021.576871 rhesus monkey. Journal of Neurophysiology, 88(1),
Goonetilleke, S. C., Gribble, P. L., Mirsattari, S. M., 13–28.
Doherty, T. J., & Corneil, B. D. (2011). Neck muscle Johansen-Berg, H. (2013). Human connectomics: What
responses evoked by transcranial magnetic stimu- will the future demand? NeuroImage, 80, 541–544.
lation of the human frontal eye fields. European Johnson, W. H., Sunahara, F. A., & Landolt, J. P.
Journal of Neuroscience, 33(11), 2155–2167. (1999). Importance of the vestibular system in visu-
Göttlich, M., Jandl, N. M., Wojak, J. F., Sprenger, A., ally induced nausea and self-vection. Journal of
von der Gablentz, J., Münte, T. F., …Helmchen, C. Vestibular Research, 9(2), 83–87.
(2014). Altered resting-state functional connectiv- Kammermeier, S., Kleine, J., & Buttner, U. (2009). Ves-
ity in patients with chronic bilateral vestibular fail- tibular-neck interaction in cerebellar patients. Annals
ure. NeuroImage: Clinical, 4, 488–499. of the New York Academy of Sciences, 1164(1),
Grossman, G. E., Leigh, R. J., Abel, L. A., Lanska, D. J., 394–399.
& Thurston, S. (1988). Frequency and velocity of Karnath, H.-O. (1994). Subjective body orientation in
rotational head perturbations during locomotion. neglect and the interactive contribution of neck
Experimental Brain Research, 70(3), 470–476. muscle proprioception and vestibular stimulation.
Gu, Y., Cheng, Z., Yang, L., DeAngelis, G. C., & Ange- Brain, 117(5), 1001–1012.
laki, D. E. (2016). Multisensory convergence of Kaulmann, D., Hermsdörfer, J., & Johannsen, L.
visual and vestibular heading cues in the pursuit (2017). Disruption of right posterior parietal cortex
area of the frontal eye field. Cerebral Cortex, 26(9), by continuous theta burst stimulation alters the
3785–3801. control of body balance in quiet stance. European
Gu, Y., DeAngelis, G. C., & Angelaki, D. E. (2012). Journal of Neuroscience, 45(5), 671–678.
Causal links between dorsal medial superior tempo- Kheradmand, A., Lasker, A., & Zee, D. S. (2015).
ral area neurons and multisensory heading percep- Transcranial magnetic stimulation (TMS) of the
tion. Journal of Neuroscience, 32(7), 2299–2313. supramarginal gyrus: A window to perception of
Guerraz, M., Yardley, L., Bertholon, P., Pollak, L., Rudge, upright. Cerebral Cortex, 25(3), 765–771.
P., Gresty, M., & Bronstein, A. M. (2001). Visual ver- Klingner, C. M., Axer, H., Brodoehl, S., & Witte, O. W.
tigo: Symptom assessment, spatial orientation and (2016). Vertigo and the processing of vestibular
postural control. Brain, 124(8), 1646–1656. information: A review in the context of predictive
Guldin, W., & Grüsser, O. (1998). Is there a vestibular coding. Neuroscience & Biobehavioral Reviews, 71,
cortex? Trends in Neurosciences, 21(6), 254–259. 379–387.
Gutteling, T. P., Selen, L. P., & Medendorp, W. P. Konen, C. S., & Haggard, P. (2014). Multisensory
(2015). Parallax-sensitive remapping of visual parietal cortex contributes to visual enhancement
space in occipito-parietal alpha-band activity of touch in humans: A single-pulse TMS study.
during whole-body motion. Journal of Neurophysi- Cerebral Cortex, 24(2), 501–507.
ology, 113(5), 1574–1584. Kotchabhakdi, N., Rinvik, E., Walberg, F., &
Halmagyi, G., Curthoys, I., Colebatch, J., & Aw, S. Yingchareon, K. (1980). The vestibulothalamic
(2005). Vestibular responses to sound. Annals of the projections in the cat studied by retrograde axonal
New York Academy of Sciences, 1039(1), 54–67. transport of horseradish peroxidase. Experimental
Harris, L. R., Jenkin, M., & Zikovitz, D. C. (2000). Brain Research, 40(4), 405–418.
Visual and non-visual cues in the perception of Lackner, J. R. (2014). Motion sickness: More than
linear self motion. Experimental Brain Research, nausea and vomiting. Experimental Brain Research,
135(1), 12–21. 232(8), 2493–2510.
Vestibular Processing in Cognition 363

Lackner, J. R., & DiZio, P. (2005). Motor control and experience. Trends in Cognitive Sciences, 22(9),
learning in altered dynamic environments. Current 810–825.
Opinion in Neurobiology, 15(6), 653–659. Niehof, N., Perdreau, F., Koppen, M., & Medendorp,
Laurens, J., Liu, S., Yu, X.-J., Chan, R., Dickman, D., W. P. (2019). Time course of the subjective visual
DeAngelis, G. C., & Angelaki, D. E. (2017). Trans- vertical during sustained optokinetic and galvanic
formation of spatiotemporal dynamics in the vestibular stimulation. Journal of Neurophysiology,
macaque vestibular system from otolith afferents 122(2), 788–796.
to cortex. Elife, 6. doi: 10.7554/eLife.20787 Nitsche, M. A., & Paulus, W. (2000). Excitability
Lopez, C., & Blanke, O. (2011). The thalamocortical changes induced in the human motor cortex by
vestibular system in animals and humans. Brain weak transcranial direct current stimulation. Jour-
Research Reviews, 67(1–2), 119–146. nal of Physiology, 527(3), 633–639.
Lopez, C., Blanke, O., & Mast, F. (2012). The human Nobre, A. C., & Kastner, S. (2014). Attention: Time
vestibular cortex revealed by coordinate-based capsule 2013. In A. C. Nobre & S. Kastner (Eds.),
activation likelihood estimation meta-analysis. The Oxford handbook of attention: Oxford UK:
Neuroscience, 212, 159–179. Oxford University Press.
Lopez C. (2016). The vestibular system: Balancing Noel, J. P., & Angelaki, D. E. (2022). Cognitive, sys-
more than just the body. Current Opinion in Neu- tems, and computational neurosciences of the self
rology, 29(1), 74–83. in motion. Annual Review of Psychology, 73,
Lopez, C., & Elzière, M. (2018). Out-of-body experi- 103–129.
ence in vestibular disorders: A prospective study of Nooij, S. A., Pretto, P., Oberfeld, D., Hecht, H., & Bül-
210 patients with dizziness. Cortex, 104, 193–206. thoff, H. H. (2017). Vection is the main contributor
Lopez, C., Schreyer, H.-M., Preuss, N., & Mast, F. W. to motion sickness induced by visual yaw rotation:
(2012). Vestibular stimulation modifies the body Implications for conflict and eye movement theo-
schema. Neuropsychologia, 50(8), 1830–1837. ries. PLoS One, 12(4). doi: 10.1371/journal.pone.
Luo, T. Z., & Maunsell, J. H. (2019). Attention can be 0175305
subdivided into neurobiological components cor- Obereisenbuchner, F., Dowsett, J., & Taylor, P. C.
responding to distinct behavioral effects. Proceed- (2021). Self-initiation inhibits the postural and
ings of the National Academy of Sciences of the electrophysiological responses to optic flow and
United States of America, 116(52), 26187–26194. button pressing. Neuroscience, 470, 37–51.
Mandala, M., Salerni, L., & Nuti, D. (2019). Benign Parkin, B. L., Ekhtiari, H., & Walsh, V. F. (2015). Non-
positional paroxysmal vertigo treatment: A practi- invasive human brain stimulation in cognitive
cal update. Current Treatment Options in Neurol- neuroscience: A primer. Neuron, 87(5), 932–945.
ogy, 21(12). doi: 10.1007/s11940-019-0606-x Palmisano, S., Allison, R. S., Schira, M. M., & Barry, R. J.
Mastropasqua, A., Dowsett, J., Dieterich, M., & (2015). Future challenges for vection research:
Taylor, P. C. J. (2020). Right frontal eye field has Definitions, functional significance, measures, and
perceptual and oculomotor functions during opto- neural bases. Frontiers in Psychology, 6. doi:
kinetic stimulation and nystagmus. Journal of 10.3389/fpsyg.2015.00193
Neurophysiology, 123(2), 571–586. Palmisano, S., Barry, R. J., De Blasio, F. M., & Fogarty,
Medendorp, W. P., & Selen, L. J. (2017). Vestibular J. S. (2016). Identifying objective EEG based mark-
contributions to high-level sensorimotor functions. ers of linear vection in depth. Frontiers in Psychol-
Neuropsychologia, 105, 144–152. ogy, 7. doi: 10.3389/fpsyg.2016.01205
Memorial (2005). John Douglas Crawford (Harvard Pavlou, M. (2017). Treatment of the dizzy patient. In
biologist and clinician) https://fa.hms.harvard.edu/ A. Bronstein & T. Lempert (Eds.), Dizziness: A prac-
files/memorialminute_crawford_john_d.pdf tical approach to diagnosis and management. (pp.
(retrieved October 10, 2022) 167–182). Cambridge, UK: Cambridge University
Merfeld, D. M., Zupan, L., & Peterka, R. J. (1999). Press.
Humans use internal models to estimate gravity Pfeiffer, C., Serino, A., & Blanke, O. (2014). The ves-
and linear acceleration. Nature, 398, 615–618. tibular system: A spatial reference for bodily self-
Mimica, B., Dunn, B. A., Tombaz, T., Bojja, V. S., & consciousness. Frontiers in Integrative Neuroscience,
Whitlock, J. R. (2018). Efficient cortical coding of 8. doi: 10.3389/fnint.2014.00031
3D posture in freely behaving rats. Science, 362, Rao, R. P., & Ballard, D. H. (1999). Predictive coding
584–589. in the visual cortex: A functional interpretation of
Morgan, P., Macken, B., Toet, A., Bompas, A., Bray, some extra-classical receptive-field effects. Nature
M., Rushton, S., & Jones, D. (2020). Distraction for Neuroscience, 2(1), 79–87.
the eye and ear. Theoretical Issues in Ergonomics Redfern, M., & Furman, J. (1994). Postural sway of
Science, 21(6), 633–657. patients with vestibular disorders during optic
Nau, M., Julian, J. B., & Doeller, C. F. (2018). How the flow. Journal of Vestibular Research: Equilibrium &
brain’s navigation system shapes our visual Orientation, 4(3), 221–230.
364 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Rincon-Gonzalez, L., Selen, L. P., Halfwerk, K., Straka, H., Zwergal, A., & Cullen, K. E. (2016). Ves-
Koppen, M., Corneil, B. D., & Medendorp, W. P. tibular animal models: Contributions to under-
(2016). Decisions in motion: Vestibular contribu- standing physiology and disease. Journal of
tions to saccadic target selection. Journal of Neu- Neurology, 263(1), 10–23.
rophysiology, 116(3), 977–985. Takahashi, K., Gu, Y., May, P. J., Newlands, S. D.,
Rinne, T., Bronstein, A., Rudge, P., Gresty, M., & DeAngelis, G. C., & Angelaki, D. E. (2007). Multi-
Luxon, L. (1998). Bilateral loss of vestibular func- modal coding of three-dimensional rotation and
tion: Clinical findings in 53 patients. Journal of translation in area MSTd: Comparison of visual
Neurology, 245(6), 314–321. and vestibular selectivity. Journal of Neuroscience,
Roberts, D. C., Marcelli, V., Gillen, J. S., Carey, J. P., 27(36), 9742–9756.
Della Santina, C. C., & Zee, D. S. (2011). MRI mag- Taube, J. S., Muller, R. U., & Ranck, J. B. (1990).
netic field stimulates rotational sensors of the Head-direction cells recorded from the postsubicu-
brain. Current Biology, 21(19), 1635–1640. lum in freely moving rats. I. Description and quan-
Rousseaux, M., Honoré, J., Vuilleumier, P., & Saj, A. titative analysis. Journal of Neuroscience, 10(2),
(2013). Neuroanatomy of space, body, and pos- 420–435.
ture perception in patients with right hemisphere Taylor, P. C., Nobre, A. C., & Rushworth, M. F. (2007).
stroke. Neurology, 81(15), 1291–1297. FEF TMS affects visual cortical activity. Cerebral
Roy, J. E., & Cullen, K. E. (2001). Selective processing Cortex, 17(2), 391–399.
of vestibular reafference during self-generated ter Horst, A. C., Koppen, M., Selen, L. P., & Meden-
head motion. Journal of Neuroscience, 21(6), dorp, W. P. (2015). Reliability-based weighting of
2131–2142. visual and vestibular cues in displacement estima-
Santos, T. E., Favoretto, D. B., Toostani, I. G., Nasci- tion. PLoS One, 10(12). doi: 10.1371/journal.
mento, D. C., Rimoli, B. P., Bergonzoni, E., … pone.0145015
Makkiabadi, B. (2018). Manipulation of human Willacker, L., Dowsett, J., Dieterich, M., & Taylor, P. C. J.
verticality using high-definition transcranial direct (2019). Egocentric processing in the roll plane and
current stimulation. Frontiers in Neurology, 9. doi: dorsal parietal cortex: A TMS-ERP study of the
10.3389/fneur.2018.00825 subjective visual vertical. Neuropsychologia, 127,
Scherer, H., & Clarke, A. (1985). The caloric vestibular 113–122.
reaction in space: Physiological considerations. Willacker, L., Roccato, M., Can, B. N., Dieterich, M.,
Acta Oto-Laryngologica, 100(5–6), 328–336. & Taylor, P. C. (2020). Reducing variability of per-
Smith, L., Gkioka, A., & Wilkinson, D. (2020). Vestib- ceptual decision making with offline theta-burst
ular-guided visual search. Experimental Brain TMS of dorsal medial frontal cortex. Brain Stimula-
Research, 238(3), 689–698. tion, 13(6), 1689–1696.
Staab, J. P., Eckhardt-Henn, A., Horii, A., Jacob, R., Yates, B. J., Catanzaro, M. F., Miller, D. J., & McCall, A. A.
Strupp, M., Brandt, T., & Bronstein, A. (2017). (2014). Integration of vestibular and emetic gastro-
Diagnostic criteria for persistent postural-percep- intestinal signals that produce nausea and vomit-
tual dizziness (PPPD): Consensus document of the ing: Potential contributions to motion sickness.
committee for the classification of vestibular disor- Experimental Brain Research, 232(8), 2455–2469.
ders of the Bárány society. Journal of Vestibular Zaidel, A., Laurens, J., DeAngelis, G. C., & Angelaki,
Research, 27(4), 191–208. D. E. (2021). Supervised multisensory calibration
Stackman, R. W., & Taube, J. S. (1997). Firing proper- signals are evident in VIP but not MSTd. Journal of
ties of head direction cells in the rat anterior tha- Neuroscience, 41(49), 10108–10119.
lamic nucleus: Dependence on vestibular input. zu Eulenburg, P., Caspers, S., Roski, C., & Eickhoff, S.
Journal of Neuroscience, 17(11), 4349–4358. B. (2012). Meta-analytical definition and func-
Straka, H., & Baker, R. (2013). Vestibular blueprint in tional connectivity of the human vestibular cortex.
early vertebrates. Frontiers in Neural Circuits, 7. NeuroImage, 60(1), 162–169.
doi: 10.3389/fncir.2013.00182
23
Somatosensory Processing
J o n H . K a a s , L e a h A . K r u b i t z e r, H u i - X i n Q i ,
and Jamie L. Reed

INTRODUCTION Garey, 1994) and also project to layer 3 of the


hand representation of adjoining Area 1. These
This review focuses on somatosensory processing two cortical areas then contribute tactile infor-
of tactile information from the hand in primates. mation to an array of higher-order somatosen-
Much of this review is based on studies in mon- sory areas, including Brodmann’s Area 2, the
keys, and to some extent humans (Figure 23.1). second somatosensory area (S2), the parietal
Most of the sensory information from the gla- ventral area (PV), and the ventral somatosen-
brous hand, which is an especially important sory areas (VSr and VSc). This network expands
tactile organ, comes from two classes of affer- to include areas of insular cortex, posterior
ents and their receptors, the rapidly adapting parietal cortex, and premotor and motor cortex.
type 1 (RA1) and the slowly adapting type 1 Feedback connections at all levels add to this
(SA1) afferents. Other afferents and their recep- complex network of interconnections.
tors, RA2 and SA2, also subserve the glabrous While tactile afferents from the hand are
hand, and receptors around the base of hairs in very important in humans and other primates
the hairy skin provide additional tactile infor- (Mountcastle, 2005), tactile afferents from other
mation. These afferents from the hand branch as parts of the body form similar projection pat-
they enter the spinal cord to travel in the dorsal terns in parallel with those for the hand to com-
column pathway to the cuneate nucleus or ter- plete the representation of the body surface in
minate on neurons in the dorsal horn of the multiple subcortical and cortical representations
spinal cord that provide second-order input to of receptors from the skin of the contralateral
the cuneate nucleus at the brainstem-spinal cord body. In addition, at least some of these nuclei
junction. Neurons in the cuneate nucleus have and areas contain representations of both the
axons that cross to the contralateral side of the contralateral and ipsilateral tongue and teeth.
brainstem, where they terminate in the ven- The pathways from face and oral structures
troposterior nucleus (VP) of the somatosensory are somewhat different than those described
thalamus. Neurons in VP then project to the for the body. The processed information from
hand representation of the primary somatosen- peripheral receptors allows humans and other
sory cortex (S1, Brodmann’s Area 3b; see primates to identify objects by touch and use
366 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

this information to guide and control sensori- elsewhere. Movement of the skin stretches the
motor behavior. Other mammals have similar corpuscles and activates the SA2 afferents. The
somatosensory systems, but often with some- afferents have large receptive fields with uncer-
what different specializations related to spe- tain borders. SA2 afferents signal the direction of
cific behaviors and peripheral morphologies. In strain and stress of the skin, and likely have a role
addition, the number of cortical areas and the in proprioception and motor control. Contributions
complexity of cortical networks is often less to tactile sensations, if any, are unknown.
elaborate compared to primates. Our review The rapidly adapting type 2 afferents (RA2)
also briefly considers the systems for other innervate Pacinian corpuscles deeper in the skin.
types of receptor-afferent inputs, such as those The layers of the Pacinian corpuscles filter out
for proprioception, pain, and temperature. slow changes in pressure on the corpuscle, while
the corpuscle transmits rapid compressions pro-
duced by high frequency vibrations to the axon in
the core of the corpuscle. The Pacinian corpuscles
are found elsewhere on the body, and in deep tis-
TACTILE RECEPTORS AND AFFERENTS sue. Distant vibrations conducted via a hard sur-
FROM THE GLABROUS SKIN AND face such as a table or floor to the hand or foot can
be detected, and provide useful information. The
HAIRY SKIN receptive fields of Pacinian corpuscle afferents are
hard to define, as vibrations may extend through
For all mammals, sensory inputs from the face, body tissue, and the sources of vibrations may be
lips, tongue, and teeth are highly important as distant from of the body.
most mammals grasp food and prey directly with Other somatosensory afferents stem from the
the mouth and pre-process food by chewing. base of sensory hairs, and from receptors near
Primates also use their hands to explore the envi- hairs (Bolanowski et al., 1994). Merkel cell slowly
ronment, and grasp food and bring it directly to adapting receptors are found both in hair folli-
the mouth. Sensory hairs around the mouth are cles and touch domes on the hairy skin. Rapidly
less important in primates, and missing in humans. adapting afferents also innervate hair follicles
Early studies of afferents from the hand in pri- and adjoining skin. The long sensory vibrissae
mates and humans have identified four types of or whiskers found on the face of mice and rats,
low-threshold mechanosensory receptors and as well as many other mammals, can be actively
afferents (Johansson and Flannagan, 2008; moved to explore and detect objects near the face.
Johnson, 2001). The slowly adapting type I sen- For example, rats can use their extra-large macro
sory afferents (SA1) are activated by a number of vibrissae to discriminate shapes, texture and the
closely spaced Merkel cell receptors located in the locations of objects near their face (Diamond and
superficial skin. These afferents are responsive to Arabzadeh, 2013; Diamond et al., 2008). The pri-
light touch on the skin, and continue to respond mary afferents for touch on the face and mouth
during a maintained contact or skin indentation. form the trigeminal nerve which enters the pons
Merkel cells are most dense in the glabrous skin of the brainstem where the afferents branch and
of the fingertips. Thus, SA1 afferents are impor- terminate in the nuclei of the trigeminal complex.
tant for identifying object form and texture. The principal nucleus of this complex receives
The rapidly adapting type I (RA1) afferents RA1 and SA1 afferent inputs, and neurons in the
rapidly adapt to light touch and skin indentation, principal nucleus project to the contralateral ven-
and are most important for signaling change as a troposterior medial nucleus to join those from the
result of stimulus movement, as well as signaling a dorsal column nuclei from the lower body (Ebner
series of repeated taps in the low frequency range and Kaas, 2015). The neurons in VP project
of flutter. RA1 afferents have the smallest recep- densely to primary somatosensory cortex (S1),
tive fields, and can signal the details of stimulus which corresponds to Area 3b of Brodmann in
form. The receptors of RA1 afferents are Meissner primates (Kaas, 1983). Other terminations of VP
corpuscles, which are most densely packed in the are in the second somatosensory area, S2, of most
skin of the finger tips. Most Meissner corpuscles mammals, and in Area 1 of primates.
are innervated by more than one axon, and each Other afferents that have been related to touch
axon innervates a tight group of corpuscles. The are the wide-dynamic range afferents that respond
SA1 and RA1 afferents make up the vast majority to increasing pressure on the skin into the range
of tactile afferents from the hands. of pain. These afferents terminate on neurons in
The slowly adapting type 2 afferents from the the dorsal horn of the spinal cord and axons from
hand innervate encapsulated Ruffini-like cor- these neurons project contralaterally to form the
puscles that are deeper in the skin, and are also spinothalamic pathway that terminates in the
found in ligaments and tendons of the hand and ventroposterior inferior nucleus (VPI). The VPI
Somatosensory Processing 367

neurons project to the superficial layers of primary less precision. Surprisingly, some of these second-
somatosensory cortex (S1) or Area 3b, and in some ary dorsal horn neurons project to the contralateral
mammals, other cortical areas including S2 and PV. cuneate nucleus (Liao et al., 2015), possibly con-
Proprioceptive afferents from muscle spin- tributing to the inhibitory receptive field sur-
dles, which signal muscle length, provide criti- rounds of relay neurons in the cuneate nucleus.
cally important proprioceptive inputs through the Overall, the contributions of the second-order
dorsal column-medial lemniscal pathway to the afferents to the receptive fields of cuneate neurons
ventroposterior superior nucleus, VPS. Thus, are not well understood, but such inputs are
the ability to perceive joint position survives thought to modulate the responses of cuneate
after joint removal and replacement in humans. relay neurons to the driving inputs of the primary
However, Ruffini type receptors in joints and afferents (Liao et al., 2018).
other deep tissues also contribute to the sense of The main activating inputs to the central core of
joint position. Unmyelinated, slowly conducting the cuneate nucleus in primates are from the RA1
afferents and thinly myelinated, moderately con- and SA1 afferents from the glabrous hand. Recent
ducting afferents signal slow and fast pain, and electrophysiological recordings indicate that the
other afferents signal temperature (Craig, 2015). responding neurons typically receive activating
Thin, slowly conducting afferents from the hairy inputs from both RA1 and SA1 afferents (Suresh
skin appear to signal pleasant (affiliative) touch et al., 2021), although one type or the other may
(Keysers et al., 2010), and project to dorsal horn dominate. The cuneate nucleus also receives
neurons that in turn project to the contralateral feedback connections mainly from the hand rep-
somatosensory thalamus via the spinothalamic resentations in S1 (Area 3b) and other areas of
tract. Thalamic projections are to the insular somatosensory cortex (Liao et al., 2021). These
cortex (Olausson et al., 2002). inputs also seem to be mainly suppressive by acti-
vating inhibitory neurons (Conner et al., 2021).
Tactile afferents from the lower trunk and leg
enter the spinal cord at lower levels, and join the
dorsal column pathway and synapse in the grac-
AFFERENT PATHWAYS AND ile nucleus, medial and adjacent to the cuneate
TARGET NUCLEI nucleus (Qi and Kaas, 2006). These axons also
have a branch that terminates on dorsal horn neu-
Peripheral nerves contain a mixture of several rons that provide a secondary input to the gracile
afferents of different types, and motor axons that nucleus. Primary afferents for touch on the face
travel to the muscles. The cell bodies of sensory travel in the trigeminal nerve to enter the brain-
afferents are in the dorsal root ganglia. The inputs stem and branch to terminate in the principal divi-
to each of the dorsal roots are from receptors in sion of the trigeminal nucleus, analogous to the
strips of skin, the dermatomes, that form the rough gracile and cuneate nuclei, and in the spinal divi-
somatotopic sequence from tail to face (e.g., sion of the trigeminal nucleus, analogous in part to
Pubols and Pubols, 1969). The tactile afferents the dorsal horn of the spinal cord. Relay neurons
from the arm and hand enter at the cervical level in the gracile, cuneate, and principal trigeminal
of the spinal cord and branch to ascend in the nuclei project contralaterally to the ventropos-
dorsal columns to terminate in the cuneate nucleus terior nucleus, which contains a detailed repre-
near the spinal cord – brainstem junction sentation of the contralateral body surface and
(Figure 23.1). The other branches terminate on projects densely to the primary somatosensory
second-order sensory neurons in the dorsal horn area (Area 3b) and less so to Area 1.
of the spinal cord at the same level. The axons Damage to the primary afferents in the cune-
from the second-order tactile neurons of the spinal ate dorsal columns at a high cervical level greatly
cord mainly join the cuneate fasciculus, but some impairs the use of the ipsilateral hand to the extent
travel to the cuneate nucleus via a lateral fascicu- that touch on the hand is not felt. However, if a
lus (Liao et al., 2015). Thus, the cuneate nucleus few primary afferents from the hand survive, hand
gets primary afferents from the arm and hand, and use and the sense of touch on the hand recovers
much of the same information from secondary to the extent that nearly normal hand use returns
neurons in the dorsal horn of the spinal cord. The (Qi et al., 2021).
primary afferents terminate in a very precise Inputs from proprioceptive afferents, such as
somatotopic pattern in the cuneate nucleus so that muscle spindle, tendon, and joint afferents form
the digits and pads of the hand are represented pathways to higher centers that are separate from
separately in somewhat reliable species specific those for tactile afferents. The cuneate dorsal col-
somatotopic patterns (Florence et al., 1989). The umn complex has separate nuclei for muscle spin-
inputs from the dorsal horn neurons appear to dle and other proprioceptive afferents. Those for
conform to this somatotopic pattern, with perhaps the arm terminate in the external cuneate nucleus,
368 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 23.1 Levels of processing in the somatosensory systems of anthropoid primates


Tactile afferents from the hands project to the ipsilateral cuneate nucleus (Cu) at the junction
of the spinal cord and brainstem (arrows). These direct projections are joined by those from
second-order neurons in the dorsal horn of the spinal cord. The cuneate nucleus neurons
then project to the contralateral ventroposterior nucleus (VP) of the thalamus. VP projects to
layer 4 of cortical Area 3b (S1 of somatosensory cortex), and less densely to layer 3 of Area 1.
Area 3b projects to Areas 3a, 1, 2, the second somatosensory area (S2), and the parietal ventral
area PV. These areas provide feedback connections to Area 3b (S1). The hand representation in
Area 3b has few callosal connections, but such connections exist between 3a, 1, 2, S2, and PV.
The cortical connections of areas S2 and PV include posterior parietal cortex and frontal motor
cortex. Somatosensory cortex also projects back to VP and Cu. See text for details.

travel in the gracile dorsal columns at lower lev- (Cusick et al., 1985; Padberg et al., 2009). Area
els, but then leave to terminate in Clarke’s nucleus 3a represents body muscles and other propriocep-
in the intermediate zone of the spinal cord. Many tive inputs in parallel to the tactile representation
of the neurons in Clarke’s nucleus project to the in Area 3b. Area 2 combines tactile and proprio-
cerebellum or project directly to the contralateral ceptive inputs in a representation that is in parallel
thalamus, where they terminate dorsal to the ven- with the Area 1 representation.
troposterior nucleus. While these projections are
often described as in a dorsorostral part of the
ventroposterior nucleus, in primates this target
is widely recognized as a separate ventroposte- SPINOTHALAMIC PATHWAYS
rior superior nucleus (VPS) that caps the tactile
ventroposterior nucleus (VP). VPS projects to Other sensory inputs related to touch, pain, and
Area 3a and Area 2 of the anterior parietal cor- temperature enter the spinal cord or brainstem to
tex, and the somatotopy of VPS mirrors that of VP activate neurons in the dorsal horn or in brainstem
Somatosensory Processing 369

nuclei that have axons that cross to the contralat- histological preparations. Somewhat surprisingly,
eral side to join the lateral spinothalamic tract that the tongue and teeth are represented twice in
terminates in parts of the somatosensory thalamus VPM, one for inputs from the contralateral teeth
near and in the ventroposterior nucleus. In pri- and tongue, as for the rest of the body, and again
mates, one of the targets is identified as the ven- for the ipsilateral teeth and tongue (Rausell and
troposterior inferior nucleus (VPI; Craig, 2006). Jones, 1991). These representations of the ipsilat-
Some neurons in VPI and the septal regions of VP eral mouth depend on ipsilateral projections from
are responsive to “wide-dynamic range,” tactile to the brainstem trigeminal nuclei. Neurons in VP
noxious stimuli. Neurons in VPI project to S1 and largely reflect a mixture of the RA1 and SA1
more densely to higher-order somatosensory areas afferent inputs to the spinal cord and brainstem.
such as S2, PV, and Area 1 (Coq et al., 2004; Individual neurons and clusters of neurons are
Disbrow et al., 2002; Padberg et al., 2009; likely to reflect more or less of these two main
Qi et al., 2002). classes of peripheral afferents (Sur et al., 1984).
In all mammals, VP projects to primary soma-
tosensory cortex (S1 or Area 3b). In most studied
mammals, VP also projects to other areas such as
SOMATOSENSORY MIDBRAIN S2 and PV (Krubitzer and Kaas, 1987). In mon-
keys, and likely humans, VP projections to S2 and
For most vertebrates, the midbrain is the major PV are greatly reduced or absent (Krubitzer and
sensorimotor center for controlling behavior. The Kaas, 1992), while other projections include the
visual midbrain, the superior colliculus (SC) of representations in Area 1, Area 2, and even most of
mammals receives direct input from the retina, Area 5 (Padberg et al., 2009), areas not generally
and the inferior colliculus (IC) is a major auditory recognized in non-primate mammals. VP receives
relay center. Yet, the deeper layers of the SC and feedback connections from primary somatosen-
the external nucleus of the IC receive ascending sory cortex, Area 3b, and from other somatosen-
somatosensory inputs from the trigeminal nuclei, sory areas (Area 1, Area 2, S2, PV).
dorsal column nuclei, and somatosensory cortex Architectonically, the ventroposterior superior
(see Kaas and Huerta (1988)), and these inputs are nucleus, VPS, resembles VP in histological stains
likely important in the multisensory motor func- for neurons, while VPI is quite distinct from VP as
tions of the midbrain. The deeper layers of the SC a cell sparse region that is contiguous with the cell
are divided histologically into modular compart- sparse septa that separate the major body parts in
ments that express acetylcholinesterase (AChE) or VP, which in primates are considered part of VPI,
receive somatosensory inputs and express rather than part of VP. However, both VPI and
parvalbumin (Illing, 1992). VPS are histologically distinct from VP by having
less expression of cytochrome oxidase and more
expression of calbindin (Kaas et al., 2006). Unlike
neurons in VP which have extremely small recep-
SOMATOSENSORY THALAMUS tive fields, neurons in VPS and VPI have much
larger receptive fields and a less precise topography.
The most obvious somatosensory nucleus of the Two other somatosensory nuclei are important
thalamus is the ventroposterior nucleus (VP), parts of the somatosensory thalamus in primates
which traditionally has been divided into the ven- and other mammals. A nucleus on the medial mar-
troposterior lateral sub-nucleus (VPL) represent- gin of VPM, the parvocellular nucleus of VPM
ing the body, and the ventroposterior medial (VPMpc, also called the basal ventral medial
sub-nucleus (VPM) representing the face, mouth, nucleus, VMb), receives taste inputs, as well as
and head (Figure 23.2). Together, they form the tactile inputs from the tongue, and projects to the
ventroposterior nucleus, VP. The activating inputs representation of the tongue in primary soma-
are rapidly adapting and slowly adapting afferents tosensory cortex as well as to gustatory cortex
from the contralateral dorsal column nuclei, and (Iyengar et al., 2007; Kaas et al., 2006). In mon-
the trigeminal principal nucleus, respectively keys, Craig et al. (1984) identified a posterior ven-
(Kaas, 2008). VP represents tactile inputs from tral medial nucleus (VMpo) just caudal to VPMpc
the tail to the tongue in a lateromedial sequence. that receives nociceptive inputs from the spinotha-
Narrow cell poor septal bands separate the more lamic and trigeminothalamic ascending pathways
lateral representation of the foot from that of the (Craig, 2006, 2015). VMpo is not distinct in non-
hand, and narrower septal areas separate represen- primates, and may be included in parts of VPI
tations of the toes and fingers from each other. A and VP. Other relevant nuclei have not been well
wider septal band separates VPL from VPM. established as “somatosensory” nuclei. The ante-
Thus, much of the somatotopy of VP is obvious in rior pulvinar (PLa), proposed for only primates, is
370 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 23.2 The three main somatosensory nuclei of thalamus in primates


The ventroposterior (VP) nucleus, shown here for its two subnuclei, VPL (ventroposterior
lateral) for the limbs, trunk and tail body, and VPM (ventral posterior medial) for the head,
face, and mouth. VP represents tactile inputs relayed from spinal cord and brainstem nuclei.
The ventroposterior inferior nucleus (VPI) receives a mixture of somatosensory inputs
from the second-order neurons in the contralateral spinal cord and brainstem. The ventral
posterior superior nucleus (VPS) receives inputs from contralateral neurons in the spinal
cord and brainstem nuclei that are activated by movement sensitive receptors and afferents
from muscles and joints that provide proprioceptive information. Both brain sections are in
the frontal (coronal) plane and are processed for the glutamate transporter type 2 (VGLUT2),
which is highly expressed in somatosensory nuclei and S1. A brain section is shown for a
squirrel monkey (A) and a macaque monkey (B). Notice that VP is darkly stained for both
primates. Unstained septal regions separate VPL from VPM, and further divide the two
subnuclei. In addition, VPS expresses less VGLUT2 than VP, and VPI expresses much less than
VP. The scale bar = 1 mm. Dorsal (D) is up, and lateral (L) is left. Rt is reticular nucleus of the
ventral thalamus.

a region of the thalamus with broad connections are mainly from layer 6 neurons, but some layer
with areas of somatosensory cortex. Neurons in 5 projections exist (Kaas, 2008). The cortical
the PLa respond to cutaneous stimulation of the projections from layer 6 largely appear to modu-
contralateral body (Padberg et al., 2009; Sherman late thalamic neuron activity by activating local
and Guillery, 2006). The anterior pulvinar projects thalamic inhibitory interneurons (Ergenzinger
to cortical regions of posterior parietal cortex that et al., 1998), while layer 5 inputs are activating
have multisensory or somatosensory functions excitatory neurons (Sherman and Guillery, 2006).
(Gharbawie et al., 2010). In primates, the lateral Cortical areas also project to brainstem and dorsal
posterior nucleus, LP, has connections with pos- column relay nuclei (Liao et al., 2021), where they
terior parietal cortex (Jones, 2007; Padberg et al., appear to activate local inhibitory neurons that
2009), and thus is involved in somatosensory pro- constrain receptive field sizes.
cessing. In non-primate mammals, LP is often
identified as the pulvinar or part of the pulvinar,
confusing comparisons with primates. In some
rodents and other mammals, part of the thalamus
just caudal to VP has been identified as the poste- SOMATOSENSORY CORTEX
rior medial nucleus (POm; cf. Audette et al., 2018;
Ebner and Kaas, 2015), adding confusion by using All mammals have cortex in which neurons
different terminologies for rodents and primates. respond to somatosensory stimuli, and these
Somatosensory areas of cortex also project regions are divided into several cortical areas on
back to somatosensory nuclei. These projections the basis of having separate representations of
Somatosensory Processing 371

tactile and proprioceptive inputs from the contralat- from the glabrous hands. The primary cortical
eral body surface. These separate representations area, Area 3b in primates, is most distinct in archi-
differ in their patterns of connections, and to vary- tecture, including a well-developed layer 4 with
ing extents in their cortical architecture. A primary small, closely packed neurons (Qi et al., 2008).
Area, S1, is basic to all mammals, but has been The S1 representation tends to proceed from tail
modified in separate lines of mammalian evolu- to tongue in a mediolateral sequence across pari-
tion in various ways, most often in relation to etal cortex, with representations of the tongue and
changes in peripheral morphology and sensory teeth located most laterally. Most mammals also
receptor arrays on specialized body parts associ- have two additional somatosensory cortical areas
ated with ethologically relevant behaviors. Inputs that are well developed; the classical second
from the face and mouth are extremely important somatosensory area, S2, and the later discovered
relays in all mammals, and these inputs are repre- parietal ventral area, PV (Krubitzer et al., 1986).
sented by more cortical territory and more neu- Primates have additional areas that are responsive
rons (cortical magnification). In highly dexterous to touch and some may be unique to this order
primates, there is also magnification of the inputs (Figure 23.3).

Figure 23.3 Somatosensory areas of the cortex of an owl monkey


This New World monkey does not have a central sulcus, and thus more somatosensory cortex
is on the exposed cortical surface. Primary somatosensory cortex, Area 3b, and adjoining
Areas 3a, 1, and 2 all contain parallel representations of the contralateral body from foot
to mouth in a mediolateral sequence. Areas 3b, 3a, and 1 have representations of both the
contralateral (C) and ipsilateral (I) teeth and tongue. Smaller somatosensory representations
include the second somatosensory area (S2), the parietal ventral area (PV), the rostroventral
parietal area (PR), which may have gustatory functions (G), and rostral (VSr) and caudal (VSc)
divisions of the ventral somatosensory areas. The lateral sulcus has been opened to show
these areas. Primary motor cortex (M1) has a crude somatotopic representation of move-
ments and somatosensory responses. The supplementary motor area (SMA), dorsal premotor
area (PMd), ventral premotor area (PMv) all have somatosensory inputs. The frontal eye field
(FEF), auditory areas, and visual areas 1 and 2 are shown for reference. The anterior part of
posterior parietal cortex is highly involved in somatosensory-motor functions. Auditory areas
include primary auditory cortex (A1), rostral (R) and rostral temporal (RT) areas, and the
caudal medial (CM) area.
372 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Some of the somatosensory areas in primates suppresses the center response. The response to
correspond to architectonic divisions that were small receptive field centers on the hand in mon-
recognized by Brodmann, and are now known as keys are suppressed by touch on other parts of the
Brodmann’s areas (3b for primary somatosensory hand (Reed et al., 2008, 2010), and even by touch
cortex, Area 3a for a rostrally adjoining strip of on the other hand (Reed et al., 2011), although
cortex, Area 1 for a caudally adjoining strip of this likely requires indirect pathways, as the hand
cortex, and Area 2 for a more caudally adjoining representation of Area 3b has few callosal con-
strip of cortex adjacent to area 1; Figure 23.3). nections (Killackey et al., 1983). Neurons in Area
These areas form adjoining representations of 1 have converging inputs from Area 3b, and thus
the contralateral body from toes most medially to have larger receptive fields. Area 2 has inputs from
tongue and teeth most laterally. These somatosen- 3b and 1, as well as from the thalamic nucleus
sory areas occupy mediolateral strips of cortex in VPS, so Area 2 neurons are activated by proprio-
anterior parietal cortex, including the caudal wall ceptive and tactile inputs. Area 3a neurons are
of the deep central sulcus of those primates where most strongly activated by proprioceptive inputs
such a sulcus is present. Early studies in humans from VPS. Areas S2, PV, and VS are smaller rep-
and monkeys confounded most or all of these four resentations with inputs from Areas 3b, 1, and 2,
representations as parts of S1, as recordings and and have large receptive fields. These higher-order
somatotopic maps were largely based on areas somatosensory areas relate to sensorimotor, action
most accessible on the brain surface, area 1 and specific domains in posterior parietal cortex that
most of Area 2. While the Area 3b representation project to matching action specific domains in pre-
has long been known to correspond to the repre- motor and motor cortex (Kaas et al., 2018). These
sentation of S1 in other mammals (Kaas, 1983), actions are guided by inputs from higher-order
some current researchers still confound the con- somatosensory and visual areas.
cept of S1 by including representations in Areas Early microelectrode recordings from hundreds
3a, 1, and 2. Areas 3a, 3b, 1, and 2 have been of recording sites in anterior parietal cortex in
architectonically compared in macaques, chim- macaque monkeys first established the existence
panzees, and humans (Qi et al., 2008), and have of multiple, separate somatotopic representations
been described in many other primates. corresponding to Brodmann’s Areas 3a, 3b, 1, and
2 (Brodmann, 1909; Kaas, 1983; Krubitzer et al.,
2004). Important features of Area 3b include rep-
resentations of the glabrous surface of digits 1-5
in a lateromedial sequence, with digit tips repre-
PRIMARY SOMATOSENSORY CORTEX, sented rostrally (deep in the central sulcus) and
AREA 3B digit representations separated by narrow, cell
poor septa (Qi and Kaas, 2004). The histologi-
Primary somatosensory cortex, S1 or Area 3b, is cal characteristics of Area 3b are so clear it was
present in the neocortex of all mammals, although possible to relate a complete representation of the
not always identified as Area 3b. The somatotopic contralateral body surface to a single architec-
representations across studied mammals are simi- tonic area in early studies using myelin stained
lar, but differ in the proportional representations or Nissl-stained brain sections. The innovation
as some skin surfaces are variably important of manually flattening the folded cortex after
across taxa (Krubitzer and Stolzenberg, 2014). separating it from the underlying fibers and cut-
Thus, most of S1 and other somatosensory areas ting this flattened cortex parallel to the surface,
represent the highly sensitive bill in the duck- meant that large portions of the Area 3b borders
billed platypus, the hand and mouth in primates, with adjoining Area 3a and Area 1 could be visu-
the glabrous skin of the tail in spider and cebus alized in single sections. This was even easier to
monkeys, the hand of raccoons, the nose of the do in rodents with a smooth cortex without any
star-nosed mole, the wing of bats, and facial fissures, so that histological preparations, includ-
vibrissa of rats and mice (Kaas, 2021). The impor- ing stains such as cytochrome oxidase, allowed all
tant sensory surfaces have more receptors, and of S1 (3b) to be viewed in a single section. These
occupy more of subcortical as well as cortical preparations revealed the somatotopy of the rodent
representations, thus providing proportionally S1 in such detail that it was possible to determine
more neurons for processing sensory information the sex of the animal from the surface view of
at every level. the representation (Lenschow and Brecht, 2018).
Neurons in Area 3b have the smallest recep- Such preparations also reveal the differences in
tive field with excitatory receptive field centers S1 representation across mammalian taxa. For
where light touch evokes a large response, while small mammals with no or few cortical fissures,
also having receptive field surrounds where touch parts of the tactile representations in S1 have large
Somatosensory Processing 373

separations between the representations of the and PV (Burton et al., 1995; Disbrow et al., 2003;
hindlimb, forelimb, lower face, and upper face, Krubitzer and Kaas, 1990; Liao et al., 2013; Pons
containing neurons that respond to stimulation et al., 1985). Subcortical connections include
of proprioceptors. In primates, these interdigita- those to VP of the thalamus, the superior col-
tions of proprioceptive cortex have been pushed liculus, and trigeminal and dorsal column nuclei
out as S1 (3b) compressed and elongated. Thus, (Liao et al., 2021).
cortex adjoining Area 3b (Area 3a) has become
more strip like, much like Area 3b. Although not
widely recognized as such, the proprioceptive cor-
tex along the anterior border of, and interdigitated
with Area 3b in non-primate mammals is the hom- AREA 1
ologue of Area 3a in primates (Ebner and Kaas,
2015; Krubitzer et al., 2011; Slutsky et al., 2000). Area 1 of primates contains a somatotopic repre-
As a further point on the somatotopy of area 3b of sentation of the contralateral body surface, that
primates, the most lateral part of the representa- closely mirrors that in Area 3b (Kaas, 1983).
tion includes, in sequence, the contralateral face, Thus, representations of the palm adjoin at the
teeth, and tongue, followed by a representation of 3b/1border, and the representation of the digits
the ipsilateral teeth and tongue (Kaas et al., 2006). point in opposite directions. The less distinct rep-
Thus, the sensory inputs from the teeth and tongue resentation along the posterior border of S1 in
are so important that both the ipsilateral and con- some rodents and other mammals contains what
tralateral teeth and tongue are represented in both may be a homolog of Area 1 (or Area 1/2) of pri-
hemispheres. This depends on both the contralat- mates (Ebner and Kaas, 2015; Krubitzer et al.,
eral and ipsilateral teeth and tongue being repre- 2011; Slutsky et al., 2000). Area 1 receives its
sented in the ventroposterior nucleus. driving input from Area 3b, while also receiving
Neurons in Area 3b reflect the relay of SA1, direct inputs from VP, which are thought to have
RA1 and RA2 classes of tactile afferents (Carter modulatory functions (Garraghty et al., 1990).
et al., 2014; Lieber and Bensmaia, 2019). While Other connections of Area 1 include soma-
cortical neurons have responses to tactile stimu- totopic interconnections with areas 3a and 2,
lation that reflect both SA1 and RA1 afferents, regions of the posterior parietal cortex, S2 and PV
layer 4 neurons are grouped in modules that are (Cerkevich and Kaas, 2019; Disbrow et al., 2003;
more dominated by either SA1 and RA1 classes of Krubitzer and Kaas, 1990), and a less widely
afferents (Sur et al., 1984). However, more super- recognized area of the lateral sulcus, the ventral
ficial neurons in S1 lose the slowly adapting com- somatosensory area (VS), and the rostroventral
ponent, perhaps as a result of local inputs from parietal area (PR) (Cerkevich and Kaas, 2019;
inhibitory neurons. The receptive fields of neurons Disbrow et al., 2003; Krubitzer and Kaas, 1990;
activated by touch on the hand, for example, have Padberg et al., 2019). Other connections are with
an excitatory center for specific locations on the motor and premotor cortex. The response prop-
glabrous hand, with an inhibitory surround, so that erties of neurons in Area 1 largely reflect those
the response to touch in the center is reduced by of the activating neurons in Area 3b, with some
touch elsewhere on the hand, and the magnitude convergence resulting in larger receptive fields
of the inhibition decreases with distance from (Rossi-Pool et al., 2021).
the excitatory center (Qi et al., 2016; Reed et al.,
2008). The suppressive surround does not extend
beyond the hand to parts of the arm. Surprisingly,
a weaker suppressive surround includes the gla-
brous skin of the opposite hand (Reed et al., 2011). AREAS 3A AND 2
Thus, these neurons in the hand representation in
primate S1 have excitatory receptive fields with Just rostral to Area 3b, area 3a is largely activated
bilateral inhibitory surrounds. The widespread by proprioceptive inputs (Krubitzer et al., 2004;
intrinsic connections within Area 3b likely con- Krubitzer and Kaas, 1990). Projections from VP
tribute to these widespread inhibitory surrounds and VPS activate neurons in area 3a (Padberg
(Liao et al., 2013). The pathways mediating the et al., 2009), and some from the same neurons
bilateral effects are not yet known. activate neurons in Area 2 (Cusick et al., 1985).
The cortical connections of S1 include includes Area 3a can be regarded as a primary area for
cortex along the anterior and posterior borders proprioceptive inputs, mainly from muscle spindle
of S1, which in primates is Area 3a and area 1 afferents. Area 2 is more responsive to tactile
(or Area 1/2 in some New World monkeys and inputs, although neurons also respond to stimula-
prosimians). Other connections are to areas S2 tion of proprioceptors (Pons et al., 1985).
374 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

While Area 3a is widely recognized in the primate Qi et al., 2002). In addition to connections with
taxa, homologs appear to exist in non-primate each other, and areas 3a, 3b, and 1, PV and S2
mammals (Krubitzer et al., 2004). Area 2 is not have connections with sensorimotor modules of
distinguished from Area 1 in all primates, and the posterior parietal cortex (Padberg et al., 2019;
region caudal to Area 3b is designated as Area 1/2 Stepniewska et al., 2009; Wang et al., 2021).
in some primates (Padberg et al., 2005); however, Neurons in these areas have large receptive fields
see Cerkevich and Kaas (2019). Area 2 receives and appear to have both tactile and propriocep-
much of its input from VPS of the thalamus, and tive components (Coq et al., 2004). S2 and PV
moderate inputs from other thalamic nuclei such are somatosensory areas that have been described
as VP and PLa (Padberg et al., 2009; Pons and in humans (Disbrow et al., 2000) as well as in a
Kaas, 1986). Cortical connections of Area 2 range of non-primate mammals (Beck et al.,
include those with Areas 3a, 3b, 1, S2, and regions 1996; Catania and Kaas, 2001; Ebner and Kaas,
of posterior parietal cortex (Padberg et al., 2009; 2015; Krubitzer and Calford, 1992; Krubitzer
Pons and Kaas, 1986). Neurons in Areas 3a and 2 et al., 2011). In humans, bilateral stimulation of
respond to manipulating joints and muscles, and the hands activates S2 and PV regions more than
movements of hands and digits (Krubitzer et al., stimulation of the contralateral hand alone
2004; Krubitzer and Kaas, 1990; Pons et al., (Disbrow et al., 2001).
1985). Especially in Area 2, the proprioceptive
signals interact with tactile signals (Kim et al.,
2015). Some neurons in Area 2 have matching
excitatory receptive fields on both hands (Iwamura
et al., 2001). Area 3a contains a somatotopic rep- SUMMARY AND CONCLUSIONS
resentation of the contralateral body that is paral-
lel to that in area 3b (Huffman & Krubitzer, 2001; All primates appear to have expanded somatosen-
Krubitzer et al., 2004), while Area 2 has a repre- sory networks at the cortical level. These include
sentation in parallel with Area 1 (Padberg et al., the traditional areas numbered after Brodmann
2007; Pons et al., 1985). (Brodmann, 1909), Areas 3a, 3b, 1, and 2; more
recently defined areas S2, PV, VSr, and VSc; an
array of multisensory domains in posterior parietal
cortex; and tactile and proprioceptive components
of domains in premotor and motor cortex. The
AREAS S2, PV, AND VS main peripheral afferents that provide information
to the vast network include the four members of
Areas S2, PV, and VS constitute a region of rapidly and slowly adaptive classes 1 and 2.
higher-order somatosensory processing located Afferents related to hair movements, muscle spin-
on the upper bank of the lateral sulcus dles, tendon stretch, nociception, and temperature
(Figure 23.3). Areas S2 and PV appear to directly add to this mixture. Tactile afferents terminate in
border S1 (area 3b) in some primates such as brainstem and spinal cord nuclei, where neurons
marmosets (Krubitzer and Kaas, 1990). In other have small receptive fields for locations on the
primates, such as titi monkeys, PV directly abuts body with antagonistic receptive field surrounds.
area 3b, and S2 abuts area 3b and area 1 (Coq Neurons in the ventroposterior nucleus of the thala-
et al., 2004); and in macaques and squirrel mon- mus and primary somatosensory cortex have simi-
keys, S2 adjoins area 1, and PV adjoins areas 3b lar receptive field sizes, while neurons in
and 1 along the representation of the face and higher-order areas have larger receptive fields,
mouth (Cerkevich and Kaas, 2019; Krubitzer more extensive antagonistic surrounds, and more
et al., 1995). Representations of the face and sources of influences on response properties,
mouth in PV and S2 are followed by that of the including those from roughly matching inputs
arm and forearm, and the leg somewhat deeper in stemming from both sides of the body. In posterior
the lateral sulcus. The representations of PV and parietal cortex and frontal motor areas, multisen-
S2 are mirror images of each other. Deeper in the sory neurons are often sensitive to visual and audi-
sulcus, PV and S2 are joined by additional repre- tory signals. Non-primate mammals often have less
sentations, the rostral (VSr) and caudal (VSc) complex somatosensory systems, but like primates,
divisions of the ventral somatosensory area VS they are specialized in various ways for specific
(Coq et al., 2004; Cusick et al., 1989; Krubitzer adaptations. However, at the cortical level, the
et al., 1995). These areas get thalamic inputs presence of S1 is universal and S1 is commonly
mainly from the ventroposterior inferior nucleus bordered by an anterior zone for proprioception
(VPI), but also from the anterior pulvinar much like Area 3a, a posterior zone suggestive of
(Disbrow et al., 2002; Krubitzer and Kaas, 1992; Area 1/2 in primates, and Areas S2 and PV.
Somatosensory Processing 375

REFERENCES with radiolabeled wheat-germ agglutinin and


wheat-germ agglutinin conjugated to horseradish-
peroxidase. Somatosensory Research, 3(1), 1–31.
Audette, N. J., Urban-Ciecko, J., Matsushita, M., &
Cusick, C. G., Wall, J. T., Felleman, D. J., & Kaas, J. H.
Barth, A. L. (2018). POm thalamocortical input
(1989). Somatotopic organization of the lateral
drives layer-specific microcircuits in somatosensory
sulcus of owl monkeys: Area-3b, S-II, and a ventral
cortex. Cerebral Cortex, 28(4), 1312–1328.
somatosensory area. Journal of Comparative
Beck, P. D., Pospichal, M. W., & Kaas, J. H. (1996).
Topography, architecture, and connections of Neurology, 282(2), 169–190.
somatosensory cortex in opossums: Evidence for Diamond, M. E., & Arabzadeh, E. (2013). Whisker
five somatosensory areas. Journal of Comparative sensory system: From receptor to decision.
Neurology, 366(1), 109–133. Progress in Neurobiology, 103, 28–40.
Bolanowski, S. J., Gescheider, G. A., & Verrillo, R. T. Diamond, M. E., von Heimendahl, M., Knutsen,
(1994). Hairy skin: Psychophysical channels and P. M., Kleinfeld, D., & Ahissar, E. (2008). ‘Where’
their physiological substrates. Somatosensory and and ‘what’ in the whisker sensorimotor system.
Motor Research, 11(3), 279–290. Nature Reviews: Neuroscience, 9(8), 601–612.
Brodmann, K. (1909). Vergleichende lokalisation- Disbrow, E., Litinas, E., Recanzone, G. H., Padberg, J.,
slehre der grosshirnrinde in ihren prinzipien darg- & Krubitzer, L. (2003). Cortical connections of the
estellt auf grund des zellenbaues. Barth, Leipzig. second somatosensory area and the parietal
Burton, H., Fabri, M., & Alloway, K. (1995). Cortical ventral area in macaque monkeys. Journal of
areas within the lateral sulcus connected to cuta- Comparative Neurology, 462(4), 382–399.
neous representations in Area-3b and Area-1: A Disbrow, E., Litinas, E., Recanzone, G. H., Slutsky, D. A.,
revised interpretation of the 2nd somatosensory & Krubitzer, L. A. (2002). Thalamocortical cnnec-
area in macaque monkeys. Journal of Comparative tions of the parietal ventral area (PV) and second
Neurology, 355(4), 539–562. somatosensory area (S2) in macaque monkeys.
Carter, A. W., Chen, S. C., Lovell, N. H., Vickery, R. M., Thalamus and Related System, 1(4), 289–302.
& Morley, J. W. (2014). Convergence across tactile Disbrow, E., Roberts, T., & Krubitzer, L. (2000). Soma-
afferent types in primary and secondary soma- totopic organization of cortical fields in the lateral
tosensory cortices. Plos One, 9(9). doi: 10.1371/ sulcus of Homo sapiens: Evidence for SII and PV.
journal.pone.0107617 Journal of Comparative Neurology, 418(1), 1–21.
Catania, K. C., & Kaas, J. H. (2001). Areal and callosal Disbrow, E., Roberts, T., Poeppel, D., & Krubitzer, L.
connections in the somatosensory cortex of the (2001). Evidence for interhemispheric processing
star-nosed mole. Somatosensory and Motor of inputs from the hands in human S2 and PV.
Research, 18(4), 303–311. Journal of Neurophysiology, 85(5), 2236–2244.
Cerkevich, C. M., & Kaas, J. H. (2019). Corticocortical Ebner, F. F., & Kaas, J. H. (2015). Somatosensory
projections to Area 1 in squirrel monkeys (Saimiri system. In G. Paxinos (Ed.), The rat nervous system
sciureus). European Journal of Neuroscience, 49(8), (4th ed., pp. 673–679). Oxford, UK: Elsevier.
1024–1040. Ergenzinger, E. R., Glasier, M. M., Hahm, J. O., &
Conner, J. M., Bohannon, A., Igarashi, M., Taniguchi, Pons, T. P. (1998). Cortically induced thalamic plas-
J., Baltar, N., & Azim, E. (2021). Modulation of ticity in the primate somatosensory system. Nature
tactile feedback for the execution of dexterous Neuroscience, 1(3), 226–229.
movement. Science, 374, 316–323. Florence, S.L., Wall, J.T., Kaas, J.H. (1989). Somato-
Coq, J. O., Qi, H., Collins, C. E., & Kaas, J. H. (2004). topic organization of inputs from the hand to the
Anatomical and functional organization of soma- spinal gray and cuneate nucleus of monkeys with
tosensory areas of the lateral fissure of the new observations on the cuneate nucleus of humans.
world titi monkey (Callicebus-moloch). Journal of Journal of Comparative Neurology, 286(1), 48-70.
Comparative Neurology, 476(4), 363–387. Garey, L. J. (1994). Brodmann’s localization in the
Craig, A. D. (2006). Retrograde analyses of spinotha- cerebral cortex. London, UK: Imperial College Press.
lamic projections in the macaque monkey: Input to Garraghty, P. E., Pons, T. P., & Kaas, J. H. (1990).
ventral posterior nuclei. Journal of Comparative Ablations of areas 3b (SI Proper) and 3a of soma-
Neurology, 499(6), 965–978. tosensory cortex in marmosets deactivate the 2nd
Craig, A. D. (2015). How do you feel? An interocep- and parietal ventral somatosensory areas. Soma-
tive moment with your neurobiological self. tosensory and Motor Research, 7(2), 125–135.
Princeton, NJ: Princeton University Press. Gharbawie, O. A., Stepniewska, I., Burish, M. J., &
Cusick, C. G., Steindler, D. A., & Kaas, J. H. (1985). Kaas, J. H. (2010). Thalamocortical connections of
Corticocortical and collateral thalamocortical con- functional zones in posterior parietal cortex and
nections of postcentral somatosensory cortical frontal cortex motor regions in New World
areas in squirrel-monkeys: A double-labeling study monkeys. Cerebral Cortex, 20(10), 2391–2410.
376 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Huffman, K. J., & Krubitzer, L. (2001). Area 3a: old-world monkeys. Journal of Comparative
Topographic organization and cortical connections in Neurology, 219(4), 384–419.
marmoset monkeys. Cerebral Cortex, 11(9), Kim, S. S., Gomez-Ramirez, M., Thakur, P. H., &
849–867. Hsiao, S. S. (2015). Multimodal interactions
Illing, R. B. (1992). Association of efferent neurons to between proprioceptive and cutaneous signals in
the compartmental architecture of the superior primary somatosensory cortex. Neuron, 86(2),
colliculus. Proceedings of the National Academy of 555–566.
Sciences of the United States of America, 89(22), Krubitzer, L. A., & Calford, M. B. (1992). 5 Topo-
10900–10904. graphically organized fields in the somatosensory
Iwamura, Y., Taoka, M., & Iriki, A. (2001). Bilateral cortex of the flying fox: Microelectrode maps,
activity and callosal connections in the somatosen- myeloarchitecture, and cortical modules. Journal
sory cortex. Neuroscientist, 7(5), 419–429. of Comparative Neurology, 317(1), 1–30.
Iyengar, S., Qi, H. X., Jain, N., & Kaas, J. H. (2007). Krubitzer, L. A., Campi, K. L., & Cooke, D. F. (2011).
Cortical and thalamic connections of the represen- All rodents are not the same: A modern synthesis
tations of the teeth and tongue in somatosensory of cortical organization. Brain Behavior and Evolu-
cortex of new world monkeys. Journal of tion, 78(1), 51–93.
Comparative Neurology, 501(1), 95–120. Krubitzer, L. A., Clarey, J., Tweedale, R., Elston, G., &
Johansson, R. S., & Flannagan, J. R. (2008). Tactile Calford, M. (1995). A redefinition of somatosen-
sensory control of object manipulation in humans. sory areas in the lateral sulcus of macaque mon-
In J. H. Kaas & E. Gardner (Eds.), The senses: A keys. Journal of Neuroscience, 15(5), 3821–3839.
comprehensive reference: Somatosensation (pp. Krubitzer, L. A., Huffman, K. J., Disbrow, E., & Recan-
67–86). Amsterdam: Elsevier. zone, G. (2004). Organization of area 3a in
Johnson, K. O. (2001). The roles and functions of macaque monkeys: Contributions to the cortical
cutaneous mechanoreceptors. Current Opinion in phenotype. Journal of Comparative Neurology,
Neurobiology, 11(4), 455–461. 471(1), 97–111.
Jones, E. G. (2007). The thalamus. Cambridge, UK: Krubitzer, L. A., & Kaas, J. H. (1987). Thalamic con-
Cambridge University Press. nections of 3 representations of the body-surface
Kaas, J. H. (1983). What, if anything, is SI? Organiza- in somatosensory cortex of gray squirrels. Journal
tion of 1st somatosensory area of cortex. Physio- of Comparative Neurology, 265(4), 549–580.
logical Reviews, 63(1), 206–231. Krubitzer, L. A., & Kaas, J. H. (1990). The organiza-
Kaas, J. H. (2008). The somatosensory thalamus and tion and connections of somatosensory cortex in
associated pathways. In J. H. Kaas & E. Gardner marmosets. Journal of Neuroscience, 10(3),
(Eds.), The senses: A comprehensive reference: 952–974.
Somatosensation (Vol. 6, pp. 117–141). Amsterdam: Krubitzer, L. A., & Kaas, J. H. (1992). The somatosen-
Elsevier. sory thalamus of monkeys: Cortical connections
Kaas, J. H. (2021). Somatosensory specializations in and a redefinition of nuclei in marmosets. Journal
mammals. Oxford encyclopedia of neuroscience of Comparative Neurology, 319(1), 123–140.
(pp. 1–12). Oxford, UK: Oxford University Press. Krubitzer, L. A., Sesma, M. A., & Kaas, J. H. (1986).
Kaas, J. H., & Huerta, M. F. (1988). The subcortical Microelectrode maps, myeloarchitecture, and cor-
visual system of primates. In H. D. Steklis & J. tical connections of three somatotopically organ-
Erwin (Eds.), Comparative primate biology: Neuro- ized representations of the body-surface in the
sciences (Vol. 4, pp. 327–391). New York: parietal cortex of squirrels. Journal of Comparative
Wiley-Liss. Neurology, 250(4), 403–430.
Kaas, J. H., Qi, H. X., & Iyengar, S. (2006). Cortical Krubitzer, L. A., & Stolzenberg, D. S. (2014). The
network for representing the teeth and tongue in evolutionary masquerade: Genetic and epigenetic
primates. Anatomical Record Part A Discoveries in contributions to the neocortex. Current Opinion in
Molecular Cellular and Evolutionary Biology, Neurobiology, 24, 157–165.
288(2), 182–190. Lenschow, C., & Brecht, M. (2018). Physiological and
Kaas, J. H., Qi, H. X., & Stepniewska, I. (2018). The anatomical outputs of rat genital cortex. Cerebral
evolution of parietal cortex in primates. Handbook Cortex, 28(4), 1472–1486.
of Clinical Neurology, 151, 31–52. Liao, C. C., DiCarlo, G. E., Gharbawie, O. A., Qi, H.
Keysers, C., Kaas, J. H., & Gazzola, V. (2010). Soma- X., & Kaas, J. H. (2015). Spinal cord neuron inputs
tosensation in social perception. Nature Reviews: to the cuneate nucleus that partially survive dorsal
Neuroscience, 11(10), 726–726. column lesions: A pathway that could contribute
Killackey, H. P., Gould, H. J., Cusick, C. G., Pons, T. P., to recovery after spinal cord injury. Journal of
& Kaas, J. H. (1983). The relation of corpus-callo- Comparative Neurology, 523(14), 2138–2160.
sum connections to architectonic fields and body- Liao, C. C., Gharbawie, O. A., Qi, H. X., & Kaas, J. H.
surface maps in sensorimotor cortex of new and (2013). Cortical connections to single digit
Somatosensory Processing 377

representations in Area 3b of somatosensory electrophysiological study. Journal of Comparative


cortex in squirrel monkeys and prosimian galagos. Neurology, 248(3), 313–335.
Journal of Comparative Neurology, 521(16), Pubols, B. H., & Pubols, L. M. (1969). Forelimb, hindlimb,
3768–3790. and tail dermatomes in spider monkey (Ateles).
Liao, C. C., Qi, H. X., Reed, J. L., Jeoung, H. S., & Brain, Behavior and Evolution, 2(2), 132–159.
Kaas, J. H. (2021). Corticocuneate projections are Qi, H. X., & Kaas, J. H. (2004). Myelin stains reveal an
altered after spinal cord dorsal column lesions in anatomical framework for the representation of
new world monkeys. Journal of Comparative Neu- the digits in somatosensory Area 3b of macaque
rology, 529(7), 1669–1702. monkeys. Journal of Comparative Neurology,
Liao, C. C., Reed, J. L., Qi, H. X., Sawyer, E. K., & 477(2), 172–187.
Kaas, J. H. (2018). Second-order spinal cord path- Qi, H. X., & Kaas, J. H. (2006). Organization of pri-
way contributes to cortical responses after long mary afferent projections to the gracile nucleus of
recoveries from dorsal column injury in squirrel the dorsal column system of primates. Journal of
monkeys. Proceedings of the National Academy of Comparative Neurology, 499(2), 183–217.
Sciences of the United States of America, 115(16), Qi, H. X., Lyon, D. C., & Kaas, J. H. (2002). Cortical
4258–4263. and thalamic connections of the parietal ventral
Lieber, J. D., & Bensmaia, S. J. (2019). High-dimen- somatosensory area in marmoset monkeys (Cal-
sional representation of texture in somatosensory lithrix jacchus). Journal of Comparative Neurology,
cortex of primates. Proceedings of the National 443(2), 168–182.
Academy of Sciences of the United States of Qi, H. X., Preuss, T. M., & Kaas, J. H. (2008). Soma-
America, 116(8), 3268–3277. tosensory areas of the cerebral cortex: Architec-
Mountcastle, V. B. (2005). The sensory hand: Neural tonic characteristics and modular organization. In
mechanisims of somatic sensation. Cambridge, J. H. Kaas & E. Gardner (Eds.), The senses: A
MA: Harvard University Press. comprehensive reference: Somatosensation (Vol. 6,
Olausson, H., Lamarre, Y., Backlund, H., Morin, C., pp. 193–169). Amsterdam: Elsevier.
Wallin, B. G., Starck, G., … Bushnell, M. C. (2002). Qi, H. X., Reed, J. L., Franca, J. G., Jain, N., Kajikawa,
Unmyelinated tactile afferents signal touch and Y., & Kaas, J. H. (2016). Chronic recordings reveal
project to insular cortex. Nature Neuroscience, tactile stimuli can suppress spontaneous activity of
5(9), 900–904. neurons in somatosensory cortex of awake and
Padberg, J., Cerkevich, C., Engle, J., Rajan, A. T., anesthetized primates. Journal of Neurophysiol-
Recanzone, G., Kaas, J., & Krubitzer, L. (2009). ogy, 115(4), 2105–2123.
Thalamocortical connections of parietal soma- Qi, H. X., Reed, J. L., Wang, F., Gross, C. L., Liu, X.,
tosensory cortical fields in macaque monkeys are Chen, L. M., & Kaas, J. H. (2021). Longitudinal
highly divergent and convergent. Cerebral Cortex, fMRI measures of cortical reactivation and hand
19(9), 2038–2064. use with and without training after sensory loss in
Padberg, J., Cooke, D. F., Cerkevich, C. M., Kaas, J. H., primates. NeuroImage, 236, doi: 10.1016/j.
& Krubitzer, L. (2019). Cortical connections of neuroimage.2021.118026
Area 2 and posterior parietal Area 5 in macaque Rausell, E., & Jones, E. G. (1991). Histochemical and
monkeys. Journal of Comparative Neurology, immunocytochemical compartments of the tha-
527(3), 718–737. lamic VPM nucleus in monkeys and their relation-
Padberg, J., Disbrow, E., & Krubitzer, L. (2005). The ship to the representational map. Journal of
organization and connections of anterior and pos- Neuroscience, 11(1), 210–225.
terior parietal cortex in titi monkeys: Do new Reed, J. L., Pouget, P., Qi, H. X., Zhou, Z. Y., Bernard,
world monkeys have an Area 2? Cerebral Cortex, M. R., Burish, M. J., … Kaas, J. H. (2008). Wide-
15(12), 1938–1963. spread spatial integration in primary somatosen-
Padberg, J., Franca, J. G., Cooke, D. F., Soares, J. G. M., sory cortex. Proceedings of the National Academy
Rosa, M. G. P., Fiorani, M., … Krubitzer, L. (2007). of Sciences of the United States of America,
Parallel evolution of cortical areas involved in 105(29), 10233–10237.
skilled hand use. Journal of Neuroscience, 27(38), Reed, J. L., Qi, H. X., & Kaas, J. H. (2011). Spatiotem-
10106–10115. poral properties of neuron response suppression in
Pons, T. P., Garraghty, P. E., Cusick, C. G., & Kaas, J. H. owl monkey primary somatosensory cortex when
(1985). The Somatotopic organization of area-2 in stimuli are presented to both hands. Journal of
macaque monkeys. Journal of Comparative Neuroscience, 31(10), 3589–3601.
Neurology, 241(4), 445–466. Reed, J. L., Qi, H. X., Pouget, P., Burish, M. J., Bonds,
Pons, T. P., & Kaas, J. H. (1986). Corticocortical con- A. B., & Kaas, J. H. (2010). Modular processing in
nections of Area-2 of somatosensory cortex in the hand representation of primate primary soma-
macaque monkeys: A correlative anatomical and tosensory cortex coexists with widespread
378 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

activation. Journal of Neurophysiology, 104(6), connections of physiologically identified zones


3136–3145. within anterior sensorimotor region. Journal of
Rossi-Pool, R., Zainos, A., Alvarez, M., Parra, S., Comparative Neurology, 517(6), 783–807.
Zizumbo, J., & Romo, R. (2021). Invariant time- Sur, M., Wall, J. T., & Kaas, J. H. (1984). Modular
scale hierarchy across the cortical somatosensory distribution of neurons with slowly adapting and
network. Proceedings of the National Academy of rapidly adapting responses in Area 3b of soma-
Sciences of the United States of America, 118(3). tosensory cortex in monkeys. Journal of Neuro-
doi: 10.1073/pnas.2021843118 physiology, 51(4), 724–744.
Sherman, S. M., & Guillery, R. W. (2006). Exploring Suresh, A. K., Greenspon, C. M., He, Q. P., Rosenow,
the thalamus and its role in cortical function (2nd J. M., Miller, L. E., & Bensmaia, S. J. (2021). Sen-
ed.). Cambridge, MA: MIT Press. sory computations in the cuneate nucleus of
Slutsky, D. A., Manger, P. R., & Krubitzer, L. (2000). macaques. Proceedings of the National Academy
Multiple somatosensory areas in the anterior pari- of Sciences of the United States of America,
etal cortex of the California ground squirrel (Sper- 118(49). doi: org/10.1073/pnas.2115772118
mophilus beecheyii). Journal of Comparative Wang, Q. M., Liao, C. C., Stepniewska, I., Gabi, M.,
Neurology, 416(4), 521–539. & Kaas, J. H. (2021). Cortical connections of the
Stepniewska, I., Cerkevich, C. M., Fang, P. C. Y., & functional domain for climbing or running in pos-
Kaas, J. H. (2009). Organization of the posterior terior parietal cortex of galagos. Journal of Com-
parietal cortex in galagos: II. Ipsilateral cortical parative Neurology, 529(10), 2789–2812.
24
Motor Control: Response
Preparation, Initiation,
and Inhibition
Vishal Rawji, John C. Rothwell,
and Marjan Jahanshahi

INTRODUCTION: WHAT IS THE ROLE OF It provides the means to navigate and interact
THE MOTOR SYSTEM? successfully in the world; in fact, there is a view
that the principal function of the human brain is to
produce movement.
Take the example of writing one’s name on a
whiteboard versus writing on a piece of paper. From the motor chauvinist’s point of view the
Writing on a whiteboard is predominantly executed entire purpose of the human brain is to produce
by proximal musculature, whereas writing one’s movement. Movement is the only way we have of
name on paper involves small muscles of the hand interacting with the world. All communication,
with stability of the shoulder. Hence despite the including speech, sign language, gestures and
same motor goal (i.e., writing one’s name), the writing, is mediated via the motor system. All
muscles involved, and movements required differ sensory and cognitive processes may be viewed as
greatly. This simple example emphasizes some of inputs that determine future motor outputs.
the intricacies that the motor system must
implement to make an appropriate movement: the (Wolpert et al., 2001)
correct dynamic and stabilizing forces must be
applied at specific muscles and around joints, with While the motor chauvinist’s perspective is rather
muscle contractions which must be performed polarizing, it nevertheless highlights that human
smoothly and be of appropriate size and duration. control of voluntary movement is a fundamental
Simultaneously, the motor system must keep track and impressive feature of brain function, gen-
of and account for the quality of the unfolding erating, and controlling a range of motor behav-
movement, such that subsequent actions can be iors including speech, gestures, writing, walking,
adjusted accordingly. Thus, the idea of a motor reaching, grasping, and an extensive motor reper-
system reiterates that it takes a series of intercon- toire (e.g., dancing, playing musical instruments,
nected brain regions to produce efficient and playing tennis, skiing, etc.) some of which set us
appropriate movements given a particular context. apart from other animals. Additionally, movement
More formally, the motor system describes the also has relevance to social cognition: commu-
central and peripheral components of the nerv- nicating with others and inferring the intentions
ous system that generate and support movement. of others from their verbal (tone of speech) and
380 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

subtle non-verbal motor behavior (facial expres- (basal ganglia and cerebellum) systems; the
sion, gaze, and eye focusing, gestures). extrapyramidal system supports higher-order fea-
As with most things, we come to appreciate tures of motor control such as appropriate effector
our motor systems when we recognize what it selection and error-mediated adjustment of
means not to have a fully functioning one (e.g., movements. In essence, the pyramidal system acts
after leg amputation or hemiplegia secondary to to generate movements, whereas the extrapyrami-
stroke), or to have one which does not function dal system acts to optimize movement and
properly (as in motor neuron disease, cerebellar behavior. The motor system also receives exten-
ataxias, or parkinsonism). In a more light-hearted sive afferent input from other cortical areas that
manner, our intact motor system is sorely missed further facilitate the selection and execution of
after the temporary state of alcohol-induced cer- appropriate movement, as will be discussed in
ebellar dysfunction that results in uncoordinated further sections. Dividing the motor system in this
walking and slurred speech. Furthermore, in the manner has functional value to clinicians, given
field of artificial intelligence, while building com- that distinct clinical syndromes are observed in
puters with higher “cognitive” capacity, faster patients with lesions of central vs. peripheral and
processing speed and ability for deep learning is pyramidal vs. extrapyramidal systems.
now a reality, building robots with the fine motor A major output of the motor system is the
skills of a human musician or the subtle motor pyramidal/corticospinal tract, formed from
coordination of a ballerina has proven to be a layer V of several cortical regions, including the
greater challenge. The excerpt above still runs true primary motor cortex, supplementary motor area
20 years after it was published, with smartphones and premotor cortex (Figure 24.1a). The larg-
and computers becoming a norm of communica- est contribution comes from the motor cortex,
tion and a means of interacting with the world; defined based on the cytoarchitectural subdivi-
after all, these devices and methods of commu- sion proposed by Brodmann (1909; cf. Elston
nication still require an intact motor system, that and Garey, 2013). Betz cells originating in layer
may actually require a more sophisticated motor V of the motor cortex terminate on interneurons
system and dextrous control of hand muscles than and anterior horn cells located in the spinal cord.
has previously been required. From there, spinal motor neurons fire to generate
In the sections that follow, we aim to outline the forces via their corresponding muscles. It is inter-
structure and function of the human motor system esting to note that there is a sense of decreasing
that allows such a wide repertoire of movement. dimensionality as one progresses from the motor
We discuss models of movement that highlight that cortex to muscle; cortical motor neurons vastly
rather than being driven in a top-down manner by outnumber lower motor neurons, which outnum-
a “movement centre,” effective movement unfolds ber the number of muscle fibres and muscles.
due to a constant interplay between several brain This arrangement likely affords the motor system
regions and with the extensive use of feedforward with a great deal of flexibility and resistance to
and feedback-driven control. We then consider neuronal loss (e.g., during ageing or after brain
how movements are prepared and initiated and injury) such that a number of different combina-
how they may be inhibited when necessary. tions of cortical neural activity can result in the
same movement. The imbalance in dimensionality
between components of the corticospinal tract has
placed attention on how neurons work together to
produce movement.
BUILDING BLOCKS OF Much of the early work on the motor cortex
THE MOTOR SYSTEM focused on how individual cortical motor neu-
rons generated movements, showing that the fir-
Several interconnected structures make up the ing of individual motor neurons preceded EMG
human motor system in a hierarchical manner. and closely correlated with force generated by
Broadly speaking, the motor system is divided corresponding muscles (Evarts and Tanji, 1976;
into its central (brain and spinal cord) and periph- Tanji and Evarts, 1976). However, not all neurons
eral (ventral nerve roots, peripheral nerves, neuro- in a patch of motor cortex will respond similarly
muscular junction, and muscle) parts. The preceding movement: more specifically, whilst
corticospinal (or pyramidal) tract represents the some motor neurons increase their activity, oth-
main pathway used to generate voluntary move- ers decrease theirs and some show no change
ment, starting in the brain and flowing via the at all. This discrepancy has been reconciled by
spinal cord to muscles. The central component of considering that neurons work together to per-
the motor system is further subdivided into the form computations necessary for generating and
pyramidal (corticospinal) and extrapyramidal supporting movement (rather than all neurons in
Motor Control: Response Preparation, Initiation, and Inhibition 381

Figure 24.1 Building blocks of the motor system.


Panel a: the corticospinal tract originates from a somatotopically organized motor “Homunculus.”
At the spinal cord level, descending corticospinal efferents terminate on anterior horn cells and
interneurons. Spinal reflexes are mediated by sensory afferents from muscle spindles making a
monosynaptic connection with alpha motor neurons.
Panel b: direct and indirect pathways via the basal ganglia allow appropriate response selection.
Panel c: proprioceptive inputs from spinal circuits (via the spinocerebellar tract), head posi-
tion sense inputs from vestibular nuclei and descending motor commands constitute affer-
ents to the cerebellar cortex. Efferents from the cerebellar cortex travel to the motor cortex
via the thalamus. (Created with BioRender.com, adapted from templates: “Direct and Indirect
Pathways of the Basal Ganglia” and “Brainstem and Diencephalon Structures Related to the
Cerebellum”)

an area responding in the same way). It is this that these actions can be called upon appropri-
fundamental shift in thinking that has resulted ately when the context arises again. Conversely,
in improved decoding of brain activity that may withholding dopamine transients after an action
pioneer advances in brain–machine interfaces is made does not reinforce those actions, which
(Willett et al., 2021). result in them not being selected in future itera-
As illustrated above, the human motor sys- tions of the scenario (Redgrave et al., 2016). In
tem can execute movements with huge degrees doing so, the basal ganglia act as a hub for learning
of freedom. But how does one decide which what action is best suited for a given situation and
movement to make from all the possibilities that offers a powerful way to optimize ongoing action
its motor system affords? A set of subcortical selection. A dichotomy has been classically drawn
nuclei comprises the basal ganglia, which help between two streams that act via the basal ganglia:
select actions based on the internal state, context/ the direct and indirect pathways, which have been
task rules and prior experiences (Figure 24.1b). purported to be pro- and anti-kinetic, respectively.
Dopamine transients are used to reinforce cer- While compelling evidence for this dichotomy
tain actions that are deemed advantageous, such has come from optogenetic stimulation of these
382 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

pathways in mice (Kravitz et al., 2010), this model INTERNAL FORWARD AND INVERSE
of separable pathways has been refined over time MODELS OF MOVEMENT
given that activity in both pathways is increased
during movement and decreased during periods
of immobility (Jin et al., 2014). Instead, the latter Equipped with the architecture required for move-
study supports a model whereby activities in the ment, a human must decide how to select a move-
direct and indirect pathways carefully sculpt basal ment, given all the possibilities that its motor
ganglia outflow to select optimal actions. system afford it; just think of the vast degrees of
Once an action has been selected, it is imple- freedom conferred by the brain’s many motor
mented via the pyramidal tracts, predominantly neurons and motor units. One option might be to
via the motor cortex. However, there are ancil- have a sort of look-up table, containing all possi-
lary structures that help facilitate the smooth ble movements that the body can make. Such a
execution of movement. One such complex of system, however, is intractable and would struggle
structures are the supplementary motor areas to keep up with the flexibility of movements
(SMA) that are located anterior to the motor cor- required by an ever-changing environment. An
tex and serve to facilitate complex components alternative solution would be to have a motor
of motor behavior such as sequence generation, system that calculates the muscle forces required
task switching, and response preparation and for successful movement, that is based on prior
inhibition. Support for the role of the SMA in the experience. Importantly, the calculation requires
aforementioned behaviors comes predominantly specification of a movement goal – that is, what
from imaging studies. Stimulation studies of the the motor system is trying to achieve.
SMA, on the other hand, have shown that this can Key to the above idea is that the brain holds a
result in complex movements (such as stepping), model of the environment in which the movement
the urge to move and even arrest of ongoing is performed, and of the how the motor system
action. It is improbable that the SMA is called itself is organized. Take the example of picking up
upon or its functions are restricted to those speci- a cup of tea. There is a model of the cup in its envi-
fied above; indeed, some authors have noted that ronment (if it’s on a table or the amount of tea in
the plurality of functions of the SMA is unified the cup) and a model of the human in that environ-
by an association between conditions and action ment (where the arms are relative to the cup or any
(Nachev et al., 2008). preferences such as handedness). One example of
The cerebellum is another structure support- internal models used by the brain are inverse mod-
ing smooth movement control that contains more els (Figure 24.2), which incorporate the current
neurons than the rest of the brain (Figure 24.1c). state of the body (joint positions, velocities, accel-
It integrates sensory information from proprio- erations) and future desired state (motor goal) to
ceptors, vestibular inputs from the otoliths and generate motor commands (activation of motor
descending motor commands from the motor neurons that lead to muscle activation and motor
cortex. In doing so, the cerebellum can compare torque) that are used by the motor system to bring
information about descending motor commands the body closer to the desired state (Kawato et al.,
and their sensory consequences to help modulate 1987). The final state can be specified as a number
ongoing movement (discussed below). The signif- of smaller states and as such, many inverse mod-
icance of the cerebellum’s role in motor control is els accumulate to result in the movement; in this
probably most notable during lesions, which result instantiation, forward models are continuously
in poor timing of muscular contractions eventually used to update the state of the body and error sig-
producing loss of co-ordination (i.e., ataxia) and nals are used to modify the ongoing movement.
dysdiadochokinesia. Less obvious from lesion Generation of effective motor commands by
studies is that the cerebellum also plays a role in the inverse model arises during motor learning.
the learning of motor skills and motor adaptation. In particular, the feedback-error learning model
While we have emphasized the role of the cor- posits that the discrepancy between the desired
ticospinal tract in dextrous hand and arm move- state and estimated state for a given action is used
ments, we must not forget how the remainder of as an error signal to generate a feedback motor
human motor control is mediated. For example, command, which goes on to modify the feedfor-
the reticulospinal and vestibulospinal tracts origi- ward motor command generated by the inverse
nate in the reticular formations and vestibular model (Kawato et al., 1987). Consequently, the
nuclei, respectively. They then descend via the signal generated by the feedback controller is
spinal cord to bilaterally innervate neck, proximal, used to train the inverse model and if there is no
and axial muscles. In all, these tracts are used to discrepancy between the desired and estimated
maintain posture and balance. states, there will be no error signal as the inverse
Motor Control: Response Preparation, Initiation, and Inhibition 383

Figure 24.2 Internal forward and inverse models during movement.


The desired movement is first specified, and an inverse model is used to generate the rele-
vant motor commands. These motor commands are passed to a motor system to be executed
and a copy is sent to be used by a forward model, which estimates the corresponding future
state and sensory feedback. The sensory system (peripheral sensory receptors such as muscle
spindles) conveys the sensory consequences of the actual movement, which is then com-
pared with the estimated state and sensory feedback generated by the forward model. Any
discrepancy between the actual and estimated state and sensory feedback is then used as an
error signal to modify subsequent motor commands. (Created with BioRender.com)

model is performing optimally. Explicit error models (Figure 24.2) state that the brain predicts
signals are also used to train the inverse model the future state (forward dynamics model) and
(Albert and Shadmehr, 2016) and it is likely that sensory consequences of the future state (forward
a number of learning mechanisms, such as basal sensory model – joint positions, velocities, accel-
ganglia-mediated reinforcement learning, are used erations) for a given action using an efference
to refine the inverse model. copy of the action and uses these predictions to
Human movement is commonly made in an optimize movement. In essence, the motor system
ever-changing environment, requiring a motor sends one copy of the motor command through
system that is adaptable and that can make con- its output structures and another (efference copy)
text-dependent adjustments depending on chang- to a dynamics predictor. The dynamics predictor
ing movement goals. The changes in environment uses the previous state of the human body and
are conveyed via sensory information (visual, the efference copy to predict what the next state
proprioceptive, task-based signals), which is inter- of the body and its sensory consequences will be,
preted by sensory cortices and incorporated into given the motor command (efference copy). The
the evolving movement, thereby creating a senso- predicted sensory consequences and real sensory
rimotor loop. However, there is a significant delay feedback are then compared, with any discrepancy
between the generation of sensory information between the two acting as an error signal. The
and incorporation into the movement that makes error signal is used by the motor system to enact
a sense-plan-act approach to motor control unvi- online adjustments to movements so that they are
able; by the time sensory information is received made accurately. Importantly, online adjustments
by the brain, it represents an outdated version of to movement in response to forward model error
the environment that may no longer be of value signals are specific to goals of the movement –
(and may even be deleterious). adjustments to movement are not made if the per-
Control theory has been used to inspire mod- turbation/error signal does not interfere with the
els which reconcile the human ability to make success of the movement (Nashed et al., 2012).
efficient movements despite sensory conduction A distinction must be made by the term error
delays (McNamee and Wolpert, 2019; Wolpert signal: in the context of forward models, error
and Ghahramani, 2000). Internal feedforward signals, and their consequences for voluntary
384 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

movement are generated continuously, throughout of the prior to result in a more accurate estima-
the movement, as opposed to error signals gener- tion of the state (van de Schoot et al., 2021).
ated after movement (such as those used in rein- Take the example of picking up an empty bottle
forcement learning literature). Therefore, feedback from the kitchen counter – visual information may
control in this instance is a continuous process set the prior belief that the bottle is more likely
throughout movement. The automaticity of online to be full. Motor commands will be produced for
adjustments to movement is made possible by each alternative (empty, full, and other intermedi-
the spinal motor system. Short and long-latency ate states), along with efference copies and their
stretch reflexes are tuned to movement goals such corresponding sensory consequences. Note that
that deviations from movement path may be cor- each motor command will require different forces
rected quickly and specifically (Doemges and to be produced by the effectors. As the grasping
Rack, 1992; Pruszynski et al., 2011). There are, movement begins and the bottle is lifted, affer-
however, some movements (such as golf swings ent sensory information is received and compared
or tennis serves) that unfold too quickly to benefit with the internal forward model sensory predic-
from corrections via spinal circuits. Instead, these tions for the full bottle and empty bottle scenarios
movements are likely controlled predominantly by (likelihoods). The forward model that minimizes
internal models. the prediction-afferent error is chosen as the most
Sensory inputs can be divided into afferents that probable state of the bottle; the corresponding
occur as a consequence of the environment and motor commands for this option are chosen and
reafferent (or efference copy) inputs, which arise combined with the prior to change the forces
as a consequence of the body’s own actions. These applied during ongoing movement to appropri-
two sources of sensory input cannot be distin- ately pick up an empty bottle. The idea is that mul-
guished from each other using the body’s sensory tiple inverse and forward models run in parallel
receptors. The forward model helps to differenti- and operate in a modular fashion to continuously
ate the two sources of sensory input by subtracting optimize movement (Wolpert and Kawato, 1998).
the predicted sensory consequences (efference) The use of afferent sensory information to
from the incoming sensory input, thereby leaving update forward and inverse models is an important
the signals generated by the environment. This is facet of control theory. This is emphasized by an
perhaps best appreciated as visual stability despite elegant study whereby participants were asked to
quick saccadic eye movements – an efference either drink from a cup of water using their hands
copy of the saccade is sent from the thalamus to or with a straw. Importantly, self-generated weight
the frontal eye fields to shift the neuron’s visual changes were experienced when the cup was lifted
receptive field spatially (Haarmeier et al., 2001). and not when participants drank using a straw;
This shift in receptive field accounts for the dis- they did have visual feedback that there was less
placement due to the upcoming saccade (Sommer water in the cup. Interestingly, the authors found
and Wurtz, 2006). Without forward models, the that subsequent forces generated to lift the cup
environment (as opposed to the eyes) may be were suboptimal when people first drank from a
perceived to move during saccades. straw vs. when they lifted the cup (Nowak and
Forward models are also hypothesized to allow Hermsdorfer, 2003). The idea was that cutaneous
for the next movement in a sequence to be planned and proprioceptive information were required to
without waiting for sensory feedback from the effectively update the internal model of the cup
first movement, which is especially useful in and its state. The significance of afferent pro-
speeded motor sequences such as piano playing prioceptive information is further highlighted by
and for anticipatory changes requiring pre-emp- considering that deafferentation can result in dif-
tive movement. For example, consider a pianist ficulty with complex voluntary movements, espe-
that is sight-playing, that plans the next sequence cially apparent when constant forces over longer
of keys as they are executing the current note. A (than one or two seconds) periods of time are
third use of feedforward models is in action selec- required (Rothwell et al., 1982).
tion: movements can be “trialed” internally with The cerebellum and parietal cortex have been
the predicted sensory consequences being used as proposed as candidate neural substrates to repre-
a dummy outcome. The action which is hypoth- sent internal models as described above. Much
esized to minimize this sensory prediction error evidence for the cerebellum’s role in holding inter-
can then be chosen and acted out, which is cru- nal forward models comes from studies of motor
cial for context-dependent modulation of move- learning and adaptation in patients with cerebel-
ment. Central to this idea is Bayesian modeling, lar lesions (Morton, 2006; Tseng et al., 2007).
which combines prior information about the state More specifically, whilst patients with cerebellar
of a system with the likelihood that sensory data lesions are able to make on-line corrections to
is produced by a given state to update estimations ongoing reaches (Smith et al., 2000), their motor
Motor Control: Response Preparation, Initiation, and Inhibition 385

adaptations following error trials are random and The modern prediction error theories overcome
uninformed by the error (Smith and Shadmehr, this problem by having multiple feedback loops at
2005). Non-invasive stimulation of the ipsilateral many levels of the CNS. Active inference models
cerebellum with transcranial magnetic stimulation notably get rid of the inverse model and are based
causes directional errors when suddenly reach- purely on forward modeling. Furthermore, they
ing toward a visual target (Miall et al., 2007). The pose an alternative account of the role of the motor
cellular architecture of the cerebellum also lends cortex – that integrates inputs from other brain
itself to acting as a hub for internal models, with regions (such as premotor and parietal cortices) to
extensive feedforward circuitry from mossy fibres output predictions about the sensory consequences
to dentate nucleus cells representing an internal of an intended movement. Active inference mod-
forward model, and cerebellar output via Purkinje eling with prior knowledge of neuroanatomy and
cells representing a time-delayed version of the functional connectivity has been used to gener-
current state (Tanaka et al., 2020). The parietal ate synthetic motor behavior that resembles that
cortex has similarly been suggested to hold inter- of humans, and even recapitulates the types of
nal models. In non-human primates performing clinical syndromes seen during lesioning (e.g.,
a joystick reaching task, neurons in the posterior hyperreflexia after upper motor neuron lesions –
parietal cortex collectively encode the dynamic Parr et al., 2021).
movement angle of the cursor, reflecting a forward
estimate of the state of the cursor (Mulliken et al.,
2008). As with studies in the cerebellum, lesion-
ing studies using cooling (Takei et al., 2021) or RESPONSE PREPARATION AND
non-invasive brain stimulation (Desmurget et al.,
1999) result in an impairment of error-based cor- INITIATION
rections and state estimation during reaching
movements. Thus, there is converging evidence Consider a tennis player hitting an approaching
that the cerebellum and parietal cortex hold inter- tennis ball. The brain must attend to and predict
nal copies of the motor command that have a role where the ball will end up at each time-step, there-
in adapting subsequent motor actions. fore establishing the future trajectory of the ball as
An alternative perspective of motor control it approaches. Given that the aim of the game is to
is active inference. This proposes that instead of win the point, relevant environmental features
being generated by descending motor commands must be considered, such as the location of the
from the motor cortex (as with inverse mod- player and the opponent on the court. With this
els), muscle forces are generated by spinal alpha information, the motor goal may be specified as
motor neurons, the activity and gain of which is returning the ball over the net, within the bound-
calibrated by the discrepancies between incoming ary lines at a location far from the opposite player,
proprioceptive information and the predictions of in order to maximize the likelihood of winning the
sensory consequences of movements sent down by point. Herein forms the motor goal or the “what”
the motor cortex. Understood in this way, move- of movement planning. Once specified, the player
ment serves to minimize proprioceptive predic- must decide upon which shot (from all the shots
tion errors. A simple example would equate to that can conceivably be played) is most appropri-
the “follow-up servo” model of Merton (1953). In ate (or most likely to win the point), given the
this model, the CNS activates intrafusal fibres of trajectory of the ball–action selection. With speci-
muscle spindles, which stretch the sensory end- fication of the desired action, the motor system
ings and cause an afferent discharge in muscle must plan how corresponding muscular contrac-
spindle afferents. This results in reflex contrac- tions and forces will be generated by the motor
tion of the extrafusal muscle in order to return cortex and other downstream effectors. Finally,
the spindle discharge to zero. The “prediction” the motor system uses this template to perform the
in this case is expressed as the desired length of desired action. As we have appreciated from for-
the muscle spindle; reflexes serve to minimize ward and inverse models, whilst the whole motor
the difference between the desired muscle length plan for the shot may be specified beforehand, it’s
and the current length. However, in this case, fur- subsequent planning and execution unfolds with
ther studies showed that the gain of the feedback feedback from the sensorium so that movement
was too small for reflexes to produce the desired can be adapted online.
change in length (Marsden et al., 1983) and the The example above highlights that move-
theory was abandoned. Furthermore, muscle ment generation has classically been parsed
spindle discharge starts after the onset of muscle into preparation (the what and how of move-
activity, contrary to a model whereby spindle dis- ment (Wong et al., 2015)) and initiation and
charge results in motor activity (Vallbo, 1970). execution (generation of the appropriate forces
386 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 24.3 The use of various types of reaction time (RT) tasks for behavioral assessment
of response preparation, initiation and execution.
Several behavioral tasks can be used to study movement preparation, initiation, and
execution.
Panel a: Simple reaction time task (SRT) – participants respond to a single go stimulus with
the same response.
Panel b: Choice reaction time task (CRT) with multiple response alternatives.
Panel c: Pre-cued choice reaction time task: Movement preparation and execution can be
parsed by introduction of delays: A pre-cue (left or right arrow) indicates the response to
be prepared. This is followed by a second, go cue, which signals the participant to make the
prepared response. Activity before and after the go cue represent activity during movement
preparation and execution, respectively. By changing the interval between the go cue and
the pre-cue arrow, the time allowed for movement preparation can be altered – the shorter
the interval, the less time allowed for movement preparation. Darker hands underneath each
task represent the correct/chosen responses. (Created with BioRender.com)

via muscular contractions). The neuroscience of movement preparation, initiation, and execu-
of movement is studied using behavioral para- tion (Jahanshahi, 2003).
digms that separate planning from execution In the choice reaction time task (CRT), partici-
phases of movement (Figures 24.3 and 24.4). pants are presented with one target stimulus of sev-
Employing mechanical or electronic devices, eral possible alternatives, each requiring a specific
these RT paradigms allow measurement of RT: response. Importantly, in each trial of the CRT, the
the interval between the presentation of a stimu- specific target stimulus is unknown until its pres-
lus and initiation of a response, and movement entation. Additionally, the CRT requires selection
time: the time taken for execution of a move- of the appropriate response once the stimulus has
ment from its initiation. In its simplest form, been presented. This is in contrast to the SRT,
the simple reaction time task (SRT), partici- where participants are aware of the response that
pants are presented with a single go stimulus, is to be performed prior to presentation of the
instructing them to initiate and execute the stimulus and can have it preprogramed and pre-
same prepared response, with the correspond- pared, only waiting for the go stimulus before it is
ing RT serving to represent a behavioral index initiated and executed. This difference in response
Motor Control: Response Preparation, Initiation, and Inhibition 387

Figure 24.4 The putative stages of processing in simple, un-cued and pre-cued choice
reaction time tasks.
Source: Adapted from Jahanshahi, 2003.
*S-R mapping is required only when the compatibility between stimuli and responses is low.

selection and extent of motor preparation between go cues. By changing the time between the pre-
the SRT and CRT manifests as longer RTs in cue and go-cue, the experimenter can change the
the latter. This observation points toward move- time allowed for movement preparation (shorter
ment preparation as conferring a RT advantage. pre-cue – go-cue interval results in less time for
The CRT can be modified further to distinguish preparation) and hence investigate the interaction
movement preparation and execution – namely between movement preparation and execution.
by inserting a delay between presentation of the Haith et al. (2016) used this method and com-
target stimulus and the imperative or go stimulus. pared a Free RT condition (participants instructed
In this pre-cued CRT, the delay between the pre- to respond as quickly as possible to the target
cue and the go stimulus presentation allows for presentation after a predictable sequence of audi-
movement preparation. Indeed, with sufficiently tory tones spaced 500 ms apart) and a Forced RT
long pre-cue intervals, reaction times are similar (participants were trained to initiate their reach-
in pre-cued CRT and SRT, owing to the affordance ing movement synchronously with the onset of the
conferred by movement preparation. fourth tone). They showed that in the Forced RT
A notable benefit of these task manipulations is task participants could make accurate responses
that different phases of movement (and their cor- approximately 80 ms faster than their fastest
responding neural activity) can be clearly deline- responses in the Free RT condition. The occur-
ated by varying and time-locking to pre-cue and rence of errors was also greater when the amount
388 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

of time allowed for preparation was reduced. The trials, where participants prepared a go response
authors reasoned that movement preparation and but did not end up making a response (Jong et al.,
initiation are independent from one another (Haith 1990). This finding provides further evidence that
et al., 2016). In fact, population-level recordings the LRP is a distinct cortical response related to
from macaque M1 and dorsal premotor cortex the preparation of movement.
show that sequences of movements are performed Preparation of a movement necessitates the
according to a model of independent movement specification of what movement must be made
preparation and execution and occur simultane- (given the state of the body, environment and
ously; movement preparation for the subsequent movement goals) and how the said movement
movement in a sequence occurs at the same time must be executed. Recordings from monkeys per-
as movement execution for the prior movement forming a pre-cued CRT show increases in neural
(Zimnik and Churchland, 2021). activity in the delay period (reflecting a movement
While the assumption of serial and independ- preparation period) distributed across the pre-
ent stages of processing in SRT and CRT tasks as motor, parietal and motor cortices. Interestingly,
illustrated in Figure 24.4, has been widely criti- activity during movement execution is also meas-
cized (see Jahanshahi, 2003), reaction time para- ured from these same regions, implying that,
digms have nevertheless been used extensively to despite being conceptually distinct, movement
study speed of movement initiation, preparation, preparation and execution are not clearly neuro-
and execution. Reaction time tasks have also lent anatomically separable. Nevertheless, some brain
themselves to be paired with electrophysiological regions hold greater weighting during specific
markers of motor preparation such as the contin- aspects of movement preparation, such as the role
gent negative variation (CNV), the negative EEG of the parietal cortex in holding internal models
potential which is recorded in the interval between and in state estimation described above (Mulliken
a warning stimulus and an imperative go stimulus. et al., 2008). Premotor cortices are involved in
The CNV represents the neural activity related to movement planning processes such as motor ini-
preparation and execution of externally-triggered, tiation, suppression and selection. Lesions of the
voluntary movements. Another neural marker of SMA lead to a variety of higher-order movement
movement preparation is the bereitschaftspotential deficits such as unconscious, disinhibited limb
(BP), a negative cortical potential measured using movements to grasp objects (as in alien-limb phe-
EEG over central regions, which is associated nomenon), impairments in motor selection and the
with the preparation of a self-generated movement loss of reaction time affordances provided by pre-
(Kornhuber and Deecke, 2016). The BP begins to cues in a pre-cued CRT. Furthermore, electrical
rise in both hemispheres approximately 1–1.5 sec- stimulation of the SMA in humans evokes an urge
onds preceding EMG onset and is characterized to move without resulting in overt motor activity
by an early, late, and peak component. Whereas whereas stimulation in monkeys (with different
the early part of the BP is measured in electrodes pulse parameters to those in humans) results in the
overlying the supplementary motor area (SMA), adoption of postures and complex movements.
late and peak components become maximal in An important distinction is between self-gener-
amplitude when measured over the hemisphere ated and externally triggered movements. There is
contralateral to the effector that moves, suggesting now a body of evidence from imaging and lesion-
that the locus of these components is the motor cor- ing studies that self-generated and externally-
tex (Jahanshahi and Hallett, 2003). In fact, when triggered movements are mediated by different
a pre-cue is used to signal an upcoming move- brain networks (Deiber et al., 1991; Playford
ment (such as a right-hand movement), a lateral- et al., 1992; Jahanshahi et al., 1995). Neurons
izing potential develops in the contralateral (left) in the SMA fire earlier during self-initiated than
hemisphere. This lateralized readiness potential externally generated movements and non-human
(LRP), calculated as the difference in EEG voltage primates with lesions of the SMA make fewer
between contralateral electrodes (e.g., C3 and C4 self-initiated movements, but retain the ability to
for hand movements), is proposed to represent the respond to external cues (Thaler and Passingham,
preparation for the forthcoming movement (Kutas 1995). In a study by Jahanshahi et al. (1995), par-
and Donchin, 1980) given that it appears well ticipants were asked to make finger movements
before the EMG response. Furthermore, LRPs are approximately every three seconds or the same
present even in the absence of movement but when movement was externally triggered by an auditory
a movement is prepared: LRPs have been recorded tone, while the BP was measured from scalp EEG
in the context of a stop-signal task whereby par- recordings. The amplitude of the late and peak
ticipants make responses to a go stimulus but must components of the BP were found to be greater
abort their developing response on a minority of when movements were self-initiated than when
stop trials. LRPs are still seen in successful stop they were predictably externally triggered, but the
Motor Control: Response Preparation, Initiation, and Inhibition 389

amplitude of the early BP did not differ between neural activity during movement preparation and
the two conditions, suggestive of similar extents of execution reflects the transition from one state to
motor preparation (Jahanshahi et al., 1995). More the next under some dynamical rule, and hence not
specifically, the BP seems related to the predict- all M1 activity need represent movement-related
ability of the forthcoming movement, given that activity. Notably, the dynamical system arises as
the BP was either absent or reduced in amplitude an interplay between populations of neurons dur-
during unpredictably cued movements relative ing motor preparation and execution and is not
to self-initiated movements (Jahanshahi et. al., appreciated from the single-neuron perspective,
1995). Measurements of regional cerebral blood which has traditionally driven theories of motor
flow clarified that this difference in BP amplitude control. The same methods have been applied to
may be related to activation of the rostral SMA, the understanding of how single neurons work
anterior cingulate cortex and dorsolateral prefron- together during movement preparation. A key
tal cortex during self-initiated movements that is feature of the dynamical systems perspective is
not present during movements triggered by unpre- that, during motor preparation, population activity
dictable cues (Jenkins et al., 2000). evolves along a “null space” where the activity of
Once preparation specifies what the appropri- individual neurons on corticospinal outputs sums
ate movement is and how it is to be achieved, it to zero through varying degrees of excitation and
needs to be executed via the pyramidal tract – the inhibition; this results in a new state primed for
bulk of which is derived from the motor cortex motor execution but in which there is no change
(although other cortical areas also contribute). in corticospinal output (Kaufman et al., 2014;
Using its extensive efference connections with Stavisky et al., 2017).
spinal interneurons and motor neurons, the corti-
cospinal tract generates the muscle activations and
forces required for successful movement initiation
and execution. Indeed, the firing patterns of corti-
cal motor neurons are closely related to the cor- RESPONSE INHIBITION
responding EMG output (Cheney and Fetz, 1980)
and display maximal firing to a preferred direction Effective motor behavior relies on the ability to
of limb movement. flexibly modulate movement depending on ever-
There have been recent developments in the changing motor goals. One example of this flexi-
understanding of how motor cortical neurons gen- ble modulation of movement includes behavioral
erate the patterns required to generate complex inhibition, a key component of normal human
movements. As mentioned earlier, the responses of functioning, serving to suppress inappropriate or
motor cortical neurons preceding movement vary unwanted actions (Hannah and Aron, 2021).
greatly, with some increasing and others decreas- Accordingly, motor inhibition pertains to how
ing firing rates before EMG onset. This oddity has motor actions are restricted or aborted. Motor
been reconciled by considering that the output of inhibition can be selective (a musician that stops
the motor cortex is a combination of the activity of singing but continues to play the piano) or global
the neurons that make it, and that the various dif- (freezing when faced with a bear in a forest). Most
ferent responses of neurons result in a pattern of typically, motor inhibition is dichotomized into
activity being generated by the motor cortex (Vyas reactive and proactive sub-types, each of which is
et al., 2020). Effectively, the motor cortex utilizes used depending on the behavioral demands/task
its many motor cortical neurons to flexibly gener- goals. Reactive inhibition is used when sudden
ate patterns of neural activity that drive EMG. The stopping of a response is required and is cued by
ability to record multiple neurons simultaneously, external events that require rapid cancellation of
along with dimensionality reduction methods has ongoing motor activity. For example, reactive
revealed that movement execution is marked by inhibition is called upon when a cyclist suddenly
motor neurons conforming to a particular “rota- stops in response to a wayward pedestrian coming
tional” pattern of dynamics (Churchland et al., into their path. Indeed, the cyclist might expect
2012). These findings are based on the dynamical that a pedestrian could walk out onto the road
systems perspective of motor control, which pro- without first looking and use this prior knowledge
poses that, instead of representing explicit features to appropriately modulate the cycle speed when
of the movement (such as direction or velocity), approaching busy areas. In anticipation of carefree
activity during motor preparation sets the initial pedestrians, the cyclist could cycle more slowly in
state of a dynamical system, that evolves into the central London as compared with in a quiet
desired movement (Churchland et al., 2010) upon ­country lane. This use of prior information about
the receipt of some trigger to move (Kaufman the environment or task to modulate speed and
et al., 2016; Nashef et al., 2021). Consequently, inhibit movement if deemed necessary is termed
390 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

proactive inhibition, a prospective and goal orien- inhibition more or less difficult, respectively. In
tated type of behavioral inhibition, concerned addition to the adjustment in the stop signal delay
with responding under restraint. Thus, rather than (interval between go and stop signals), as noted
operating independently, proactive and reactive above, the ratio of go and stop trials can be adjusted
inhibition act synergistically to enhance behavio- to change the difficulty of reactive inhibition.
ral inhibition, wherein engagement of proactive An important feature of the SST is that the time
inhibition enhances the efficacy of reactive inhibi- delay between the go and stop cues is dynamically
tion (Schall et al., 2017). adjusted based on the participant’s performance,
In everyday life, various sensory cues from the which normalizes the proportion of successfully
environment have the potential to trigger relevant stopped trials between participants. The efficacy
motor actions. This activation of a movement of reactive inhibition must be inferred in the SST,
triggered by simple external cues is an automatic given that successful reactive inhibition is achieved
mechanism, which prompts motor preparation by the absence of a response. The inferred meas-
in order to facilitate motor execution (Sumner ure is termed the stop signal reaction time (SSRT),
and Husain, 2008). Whilst these prompts afford which gives an indication of how well a partici-
certain motor advantages, they require a mecha- pant can stop reactively (Figure 24.5). Whilst both
nism to prevent us from responding to every cue the SST and go/no-go tasks are both used to meas-
in our environment. This introduces automatic ure reactive inhibition, they require inhibition of
inhibition which is used to avoid automatic cou- movements at different phases of movement: in
pling of sensory cues to motor actions. It was the go/no-go task reactive inhibition involves inhi-
initially identified when a patient with a visual bition of movement initiation, whereas in the SST,
cortex lesion could still differentiate between two reactive inhibition requires cancelling a movement
visual stimuli, despite not consciously perceiving that has already been initiated (see Verbruggen
them (Weiskrantz et al., 1974). Following this, it et al., 2019, for consensus view on the measure-
has been found that subliminal, sensory cues can ment of response inhibition).
modulate motor actions (Eimer and Schlaghecken, Proactive inhibition can be assessed by com-
2003; Obhi, 2007). Whilst reactive inhibition can paring reaction times on go trials between tasks
become automatic and habitual through learning with and without stop trials. The assumption is
and repetition, automatic inhibition differs in that that on tasks with no stop trials, no or little proac-
its effects are derived subliminally. Simply put, tive control is exerted whereas reaction time tasks
the sensory cues which evoke automatic inhibi- with stop trials involve proactive inhibition, since
tion are not perceived, whereas they are in reactive individuals slow their responses to the go signal
inhibition. in anticipation of a stop signal, despite instruc-
As with response initiation, reaction time para- tions not to do so. Hence the difference in reaction
digms form the mainstay of laboratory investiga- time on go trials between tasks with and without
tion of behavioral motor inhibition. Two popular stop trials gives an index of proactive control.
tasks to probe action restraint and reactive inhibi- More specifically, participants have faster reaction
tion are the go/no-go task and the stop-signal task times on go trials when they are performed in a
(SST), both of which require participants to with- task with no stop signals than in a task with stop
hold or suddenly abort prepotent motor responses. signals due to the anticipation of potentially stop-
In the go/no-go task, participants are asked to ping in the latter. One caveat to measuring pro-
respond as quickly as possible to either a go cue active inhibition in this manner is the difference
(e.g., a green circle) but to withhold responding to in attentional demands between tasks, which may
a stop cue (e.g., red cross, Figure 24.5). On an SST, confound measures of proactive control (Dimoska
participants are instructed to respond quickly to a and Johnstone, 2008; Elchlepp et al., 2016; Jahfari
go stimulus but to stop themselves from making et al., 2015; Liebrand et al., 2017; Verbruggen
a response when a stop signal (e.g., red cross or et al., 2014). A variant of the SST, the conditional
tone) is presented shortly after the go stimulus on stop-signal task (CSST), addresses this caveat by
a percentage of trials. simultaneously probing reactive and proactive
The ability to enact reactive inhibition can be inhibition in the same task (Obeso et al., 2014;
made more or less difficult by changing the ratio Rawji et al., 2020). Participants make responses
of go and stop trials; the greater the proportion to one of two go stimuli (e.g., right or left pointing
of go trials, the greater the chance of participants arrows) and stop signals are presented after the go
responding on a stop trial (making a stop failure stimuli, as in the SST. At the beginning of the task,
commission error), thereby making the response participants are instructed that they must follow
more prepotent and reactive inhibition more dif- the stopping rule if the stop signal appears after
ficult. The time delay between the go and stop one of the go stimuli only (right OR left point-
cues is made shorter or longer to make reactive ing arrow). In doing so, participants slow their
Motor Control: Response Preparation, Initiation, and Inhibition 391

Figure 24.5 The behavioral assessment of response inhibition.


Reactive inhibition is typically studied using the go/no-go or stop-signal task.
Panel a: In the go/no-go task, one of two signals can appear after the fixation cross – a go
(green circle) or no-go (red cross) signal.
Panel b: The stop-signal task asks participants to respond to a go cue on all trials (right
arrow). On a minority of trials, a stop signal (red cross) is shown after the go cue, instructing
participants to abort the ongoing response. Whilst both tasks measure reactive inhibition, an
important difference is that successful reactive inhibition in the go/no-go task is achieved
by inhibition of response initiation, whereas it is achieved by inhibition on an already initi-
ated response in the stop-signal task. Proactive inhibition can be measured by measuring the
reaction time difference between go reaction times in tasks with and without no-go/stop
signals, or by use of the conditional stop-signal task (see text).
Panel c: Automatic inhibition is measured using masked priming tasks. Primes are only shown
for short times (approximately 17 ms) and are then obscured by masks. Following the mask,
the target cue appears, which is shown for longer. Importantly, the prime is not consciously
perceived, yet its identity affects reaction times to targets such that if a prime and target
are the same, there is a reaction time shortening relative to a condition without priming or
if the prime and target are opposite. A reversal of this normal priming effect is seen when
the delay between prime and target reaches approximately 150 ms, believed to reflect auto-
matic motor inhibition. Darker hands underneath each task represent the correct/chosen
responses. Created with BioRender.com.

responses on go trials to the direction that they Automatic motor inhibition is assessed using the
might have to stop, thereby exerting proactive masked priming task (Eimer and Schlaghecken,
control; the reaction time difference on go trials 2003; Rawji et al., 2020). Participants are asked to
between the two conditions is the objective meas- make responses to one of two directions (left cue or
ure of proactive inhibition. right cue) following a go cue (baseline condition).
392 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Preceding this go cue, a prime (left cue or right 2015). Indeed, pathologies of the basal ganglia
cue) is presented for a short time (17 ms) followed such as Parkinson’s disease (Obeso et al., 2011)
by a non-sensical mask (randomly orientated and Huntington’s disease (Rao et al., 2014) have
lines), which occludes the prime. Importantly, the shown deficits in response inhibition.
prime is presented for such a short time that it is Functional neuroimaging studies have shown
not consciously perceived. Despite this, the iden- that reactive inhibition is implemented using
tity of the prime confers differential effects on the a circuit incorporating the right inferior fron-
reaction time to the go cue such that if the prime tal gyrus (rIFG), pre-SMA (Simmonds et al.,
and go cue are the same, responses are made faster 2008), and subthalamic nucleus (STN) via a
than the baseline reaction time. Conversely, if the cortico-subthalamic hyperdirect pathway (Frank
prime and go cue are different from each other, the et al., 2007; Nambu et al., 2002). In particular, the
reaction time is longer than baseline. STN is an important node in the inhibitory control
Given that the prime is not consciously per- network, given that its activity correlates with that
ceived, its effects are deemed as being automatic. of the rIFG and is negatively correlated with the
Indeed, the lack of generalization provides evi- SSRT (Aron et al., 2007). In addition, the strength
dence of its automaticity; primes only modulate of activity in these nodes is greater during suc-
performance if they are of the same kind as go cessful than unsuccessful stop trials (Aron, 2006).
cues. For example, prime-cue effects are seen if Patients with Parkinson’s disease who undergo
the combinations are both left/rightward facing deep brain stimulation have electrodes implanted
arrows, but not if the prime is an arrow and the in their STN, which can be recorded from during
target is a letter (L or R) denoting the effector to tasks of response inhibition. Patients with deep
be used (left or right, respectively). Interestingly, brain stimulation of the STN show impaired per-
reversal of the priming effect is seen as the time formance on motor tasks involving inhibitory con-
interval between prime and go cue is increased trol relative to patients without DBS and healthy,
above 150 ms, such that incompatible prime-cue age-matched controls (Hershey et al., 2004; Obeso
pairs now result in faster reaction times than com- et al., 2013; Georgiev et al, 2016). Notably, the
patible prime-cue pairs. The magnitude of the impairments in both reactive and proactive inhibi-
reaction time reversal above 150 ms is deemed to tion are observed in patients who have undergone
be the objective readout of automatic motor inhibi- a right-sided subthalamotomy only (Obeso et al.,
tion (Aron et al., 2003; Eimer and Schlaghecken, 2014) a finding that is consistent with the right lat-
1998, 2003; Lleras and Enns, 2004; Sohrabi and eralization of the stopping network (Aron, 2006).
West, 2009; Wilson et al., 2011). Electrophysiological recordings of the STN in
The proposed mechanism for automatic inhibi- patients with Parkinson’s disease who have had
tion is as follows: the subliminal prime causes an DBS surgery show an increase in beta-band power
automatic, initial motor facilitation correspond- during successful action restraint in a go/no-go task
ing to the prime choice alternative. The prime is (Kühn et al., 2004). Transcranial magnetic stimu-
then replaced by the mask, which decreases motor lation (TMS) of the motor cortex has been applied
activity for the prime and causes motor inhi- during tasks of reactive inhibition to provide an
bition. If the go cue is presented early after the insight into the cortical dynamics of reactive
prime, when motor facilitation is still active, then inhibition, finding that motor cortex excitability
responses are faster during compatible trials. If the is suppressed in response to stop signal presenta-
go cue is presented late after the prime (interval tion (MacDonald et al., 2014; Sohn et al., 2002;
above 150 ms), when motor inhibition has now Van Den Wildenberg et al., 2010). This decrement
occurred, then responses on compatible trials are in excitability is not only limited to the effector
now slower (Eimer and Schlaghecken, 1998). This that is to stop but also produces global influences
model of how the prime and go cue interact is on motor system output. For example, suddenly
supported by LRPs measured during the masked aborting speech decreases motor cortex activity in
priming task, which show a tri-phasic waveform. hand muscles, too (Badry et al., 2009; Greenhouse
The first two peaks are thought to form the exci- et al., 2012; Majid et al., 2012; Pouget et al., 2017;
tation-then-inhibition induced by the prime-mask Wessel et al., 2013). Of note, successful stopping
combination, with the final peak corresponding to results in motor cortical excitability reducing to a
motor execution (Seiss et al., 2014). level lower than that at baseline, suggesting that
Neuroimaging studies in humans have shown successful stopping is due to an active inhibitory
that the neuroanatomical substrates mediating process (Coxon, 2006; Sohn et al., 2002).
different forms of behavioral inhibition are ana- Proactive inhibition is implemented through
tomically distinct, although they all operate via the the cortico-striatal-thalamo-cortical indirect path-
basal ganglia to exert their influences on behavior way of the basal ganglia (Aron, 2006; Jahanshahi
(Jahanshahi and Rothwell, 2017; Jahanshahi et al., et al., 2015). As with tasks of reactive inhibition,
Motor Control: Response Preparation, Initiation, and Inhibition 393

functional neuroimaging has shown greater activa- corticospinal tract, which has varied inputs from
tion of the striatum (Vink et al., 2005; Zandbelt across the cortex. The basal ganglia help select
and Vink, 2010) and dorsolateral prefrontal cor- an optimal motor programme given a particular
tex (DLPFC) (Chikazoe et al., 2009; Jahfari et al., situation, and the execution of the ensuing move-
2010; Smittenaar et al., 2013) when a stop signal is ment is made smooth by the cerebellum. The
anticipated. Proactive inhibition measured during sensory consequences of ongoing movements are
the CSST is impaired in patients with Parkinson’s used to update subsequent motor plans, such that
disease who have undergone a right-sided sub- movement goals are met in an ever-changing
thalamotomy and as with reactive inhibition, this environment. The stages of voluntary movement
effect seems to lateralize to the right hemisphere have been parsed into preparation, initiation and
given that there was no differential effect on pro- execution, and reaction time paradigms have
active inhibition in patients with left-sided sub- been developed in an attempt to understand how
thalamotomies (Obeso et al., 2014). The STN may these processes are enacted by the brain.
be perfectly suited to implementing proactive inhi- Neuroimaging and neurophysiology have shown
bition given that it acts as a “hold-your-horses” a particular role of the SMA during movement
signal, which pauses movement until conflict can preparation, with its engagement depending on
be resolved (Frank et al., 2007). Proactive inhibi- whether movement is internally generated or
tion usually necessitates the inhibition of a spe- externally triggered, and the predictability of
cific response, as in the CSST. In these tasks of movement initiation. Finally, movements are
selective stopping that employ proactive inhibi- executed via the corticospinal tract, following a
tion, motor cortex excitability is held lower than “motor command” issued by the motor cortex.
the baseline value, again suggesting an active sup- Changes in the environment may necessitate ces-
pression as the movement is being prepared (Cai sation of ongoing motor output, requiring motor
et al., 2011; Claffey et al., 2010). Contrary to the inhibition. Conceptually, behavioral inhibition is
global suppression during reactive stopping, pro- subdivided depending on the type of inhibition
active inhibition is focal and specific to the effec- required and the circuits mediating each type of
tor that might need to stop (Cai et al., 2011). response inhibition have been shown to be neuro-
Patients with lesions have provided a rich anatomically distinct. The motor system has the
source to study the neuroanatomical basis of auto- degrees of freedom and flexibility to generate the
matic inhibition. Two such patients with small, multitude and range of gross (e.g., gait) and fine
focal lesions of the SMA and supplementary eye movements (e.g., playing the violin) that are
field (SEF) display impaired automatic inhibition necessary for us to move, act and interact with
specific to the effector supplied by the SMA/SEF. our environment and other people.
That is, the patient with the SMA lesion showed
impaired automatic inhibition if the task involved
responding with the contralateral limb but normal
automatic inhibition when the task was performed
with eye movements. The reverse was observed REFERENCES
in the patient with the SEF lesion (Sumner et al.,
2007). Functional neuroimaging during the Albert, S. T., & Shadmehr, R. (2016). The neural feed-
masked priming task has highlighted the basal back response to error as a teaching signal for the
ganglia (striatum) (D’Ostilio et al., 2012) and motor learning system. Journal of Neuroscience,
thalamus (Aron et al., 2003) although both studies 36, 4832–4845.
were performed in patients with neurological dis- Aron, A. R. (2006). Cortical and subcortical contribu-
ease. Furthermore, patients with primary tic disor- tions to stop signal response inhibition: Role of the
ders (believed to be due to pathologies of the basal subthalamic nucleus. Journal of Neuroscience, 26,
ganglia striatum) and SMA show impairments in 2424–2433.
automatic inhibition (Rawji et al., 2020). Aron, A. R., Behrens, T. E., Smith, S., Frank, M. J., &
Poldrack, R. A. (2007). Triangulating a cognitive
control network using diffusion-weighted mag-
netic resonance imaging (MRI) and functional MRI.
Journal of Neuroscience, 27, 3743–3752.
SUMMARY AND CONCLUSIONS Aron, A. R., Schlaghecken, F., Fletcher, P. C., Bullmore,
E. T., Eimer, M., Barker, R., … Robbins, T. W.
Human voluntary movement is co-ordinated by a (2003). Inhibition of subliminally primed responses
network of interconnected brain regions, each of is mediated by the caudate and thalamus: Evidence
which plays a particular role. The main outflow from functional MRI and Huntington’s disease.
of the motor system operates through the Brain, 126, 713–723.
394 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Badry, R., Mima, T., Aso, T., Nakatsuka, M., Abe, M., Eimer, M., & Schlaghecken, F. (1998). Effects of
Fathi, D., … Fukuyama, H. (2009). Suppression of masked stimuli on motor activation: behavioral
human cortico-motoneuronal excitability during and electrophysiological evidence. Journal of
the stop-signal task. Clinical Neurophysiology, Experimental Psychology: Human Perception and
120, 1717–1723. Performance, 24, 1737–1747.
Brodmann K (1909) Vergleichende lokalisationslehre Eimer, M., & Schlaghecken, F. (2003). Response facili-
der grosshirnrinde in ihren prinzipiendargestellt tation and inhibition in subliminal priming.
auf grund des zellenbaues. Leipzig, Germany: J.A. Biological Psychology, 64, 7–26.
Barth. Elchlepp, H., Lavric, A., Chambers, C. D., &
Cai, W., Oldenkamp, C. L., & Aron, A. R. (2011). A Verbruggen, F. (2016). Proactive inhibitory control:
proactive mechanism for selective suppression of A general biasing account. Cognitive Psychology,
response tendencies. Journal of Neuroscience, 31, 86, 27–61.
5965–5969. Elston, G. N., & Garey, L. J. (2013). The cytoarchitec-
Cheney, P. D., & Fetz, E. E. (1980). Functional classes tonic map of Korbinian Brodmann: Arealisation
of primate corticomotoneuronal cells and their and circuit specialisation. In S. Geyer & R. Turner
relation to active force. Journal of Neurophysiology, (Eds.), Microstructural parcellation of the human
44, 773–791. cerebral cortex. Heidelberg: Springer.
Chikazoe, J., Jimura, K., Hirose, S., Yamashita, K. -i., Evarts, E. V., & Tanji, J. (1976). Reflex and intended
Miyashita, Y., & Konishi, S. (2009). Preparation to responses in motor cortex pyramidal tract neurons
inhibit a response complements response inhibi- of monkey. Journal of Neurophysiology, 39,
tion during performance of a stop-signal task. 1069–1080.
Journal of Neuroscience, 29, 15870–15877. Frank, M. J., Samanta, J., Moustafa, A. A., & Sher-
Churchland, M. M., Cunningham, J. P., Kaufman, M. T., man, S. J. (2007). Hold your horses: Impulsivity,
Foster, J. D., Nuyujukian, P., Ryu, S. I., & Shenoy, K. V. deep brain stimulation, and medication in Parkin-
(2012). Neural population dynamics during reach- sonism. Science, 318, 1309–1312.
ing. Nature, 487, 51–56. Greenhouse, I., Oldenkamp, C. L., & Aron, A. R.
Churchland, M. M., Cunningham, J. P., Kaufman, M. T., (2012). Stopping a response has global or non-
Ryu, S. I., & Shenoy, K. V. (2010). Cortical prepara- global effects on the motor system depending on
tory activity: Representation of movement or first preparation. Journal of Neurophysiology, 107,
cog in a dynamical machine? Neuron, 68, 387–400. 384–392.
Claffey, M. P., Sheldon, S., Stinear, C. M., Haarmeier, T., Bunjes, F., Lindner, A., Berret, E., &
Verbruggen, F., & Aron, A. R. (2010). Having a Thier, P. (2001). Optimizing visual motion percep-
goal to stop action is associated with advance tion during eye movements. Neuron, 32,
control of specific motor representations. 527–535.
Neuropsychologia, 48, 541–548. Haith, A. M., Pakpoor, J., & Krakauer, J. W. (2016).
Coxon, J. P. (2006). Intracortical inhibition during Independence of movement preparation and
volitional inhibition of prepared action. Journal of movement initiation. Journal of Neuroscience, 36,
Neurophysiology, 95, 3371–3383. 3007–3015.
Desmurget, M., Epstein, C. M., Turner, R. S., Prablanc, Hannah, R., & Aron, A. R. (2021). Towards real-
C., Alexander, G. E., & Grafton, S. T. (1999). Role world generalizability of a circuit for action-
of the posterior parietal cortex in updating stopping. Nature Reviews: Neuroscience, 22(9),
reaching movements to a visual target. Nature 538–552.
Neuroscience, 2, 563–567. Hershey, T., Revilla, F. J., Wernle, A., Gibson, P. S.,
Dimoska, A., & Johnstone, S. J. (2008). Effects of Dowling, J. L., & Perlmutter, J. S. (2004). Stimula-
varying stop-signal probability on ERPs in the stop- tion of STN impairs aspects of cognitive control in
signal task: Do they reflect variations in inhibitory PD. Neurology, 62, 1110–1114.
processing or simply novelty effects? Biological Jahanshahi, M. (2003). Reaction time as an index of
Psychology, 77, 324–336. motor preparation/programming and speed of
Doemges, F., & Rack, P. M. (1992). Task-dependent response initiation. In M. Hallett (Ed.), Handbook
changes in the response of human wrist joints to of clinical neurophysiology (Vol. 1, pp. 203–229).
mechanical disturbance. Journal of Physiology, Amsterdam: Elsevier.
447, 575–585. Jahanshahi, M., & Frith, C. D. (1998). Willed action
D’Ostilio, K., Collette, F., Phillips, C., & Garraux, G. and its impairments. Cognitive Neuropsychology,
(2012). Evidence for a role of a cortico-subcortical 15, 483–533.
network for automatic and unconscious motor Jahanshahi, M., Hallett, M. (2003). (Eds.), The
inhibition of manual responses. PLoS One, 7. doi: Bereitschaftspotential-movement-related cortical
10.1371/journal.pone.0048007 potentials. New York: Kluwer-Plenum.
Motor Control: Response Preparation, Initiation, and Inhibition 395

Jahanshahi, M., Jenkins, I. H., Brown, R. G., Marsden, potentials. Pflügers Archiv: European Journal of
C. D., Passingham, R. E., & Brooks, D. J. (1995). Physiology, 468, 1115–1124.
Self-initiated versus externally triggered move- Kravitz, A. V., Freeze, B. S., Parker, P. R. L., Kay, K.,
ments: I. An investigation using measurement of Thwin, M. T., Deisseroth, K., & Kreitzer, A. C.
regional cerebral blood flow with PET and move- (2010). Regulation of parkinsonian motor behav-
ment-related potentials in normal and Parkinson’s iours by optogenetic control of basal ganglia cir-
disease subjects. Brain, 118, 913–933. cuitry. Nature, 466, 622–626.
Jahanshahi, M., Obeso, I., Rothwell, J. C., & Obeso, Kühn, A. A., Williams, D., Kupsch, A., Limousin, P.,
J. A. (2015). A fronto-striato-subthalamic-pallidal Hariz, M., Schneider, G. H., … Brown, P. (2004).
network for goal-directed and habitual inhibition. Event-related beta desynchronization in human
Nature Reviews: Neuroscience, 16, 719–732. subthalamic nucleus correlates with motor perfor-
Jahanshahi, M., & Rothwell, J. C. (2017). Inhibitory mance. Brain, 127, 735–746.
dysfunction contributes to some of the motor and Kutas, M., & Donchin, E. (1980). Preparation to
non-motor symptoms of movement disorders and respond as manifested by movement-related brain
psychiatric disorders. Philosophical Transactions of potentials. Brain Research, 21, 95–115.
the Royal Society B: Biological Sciences. 372. doi: Liebrand, M., Pein, I., Tzvi, E., & Krämer, U. M.
10.1098/rstb.2016.0198 (2017). Temporal dynamics of proactive and reac-
Jahfari, S., Stinear, C. M., Claffey, M., Verbruggen, F., tive motor inhibition. Frontiers in Human Neurosci-
& Aron, A. R. (2010). Responding with restraint: ence, 11. doi: 10.3389/fnhum.2017.00204
What are the neurocognitive mechanisms? Journal Lleras, A., & Enns, J. T. (2004). Negative compatibility
of Cognitive Neuroscience, 22, 1479–1492. or object updating? A cautionary tale of mask-
Jahfari, S., Waldorp, L., Ridderinkhof, K. R., & dependent priming. Journal of Experimental Psy-
Scholte, H. S. (2015). Visual information shapes chology: General, 133, 475–493.
the dynamics of corticobasal ganglia pathways MacDonald, H. J., Coxon, J. P., Stinear, C. M., &
during response selection and inhibition. Journal Byblow, W. D. (2014). The fall and rise of cortico-
of Cognitive Neuroscience, 27, 1344–1359. motor excitability with cancellation and reinitiation
Jenkins, I. H., Jahanshahi, M., Jueptner, M., Passing- of prepared action. Journal of Neurophysiology,
ham, R. E., & Brooks, D. J. (2000). Self-initiated 112, 2707–2717.
versus externally triggered movements: II. The Majid, D. S. A., Cai, W., George, J. S., Verbruggen, F., &
effect of movement predictability on regional cer- Aron, A. R. (2012). Transcranial magnetic stimula-
ebral blood flow. Brain: A Journal of Neurology, tion reveals dissociable mechanisms for global versus
123, 1216–1228. selective corticomotor suppression underlying the
Jin, X., Tecuapetla, F., & Costa, R. M. (2014). Basal stopping of action. Cerebral Cortex, 22, 363–371.
ganglia subcircuits distinctively encode the parsing Marsden, C. D., Rothwell, J. C., & Day, B. L. (1983).
and concatenation of action sequences. Nature Long-latency automatic responses to muscle
Neuroscience, 17, 423–430. stretch in man: Origin and function. Advances in
Jong, R. de, Coles, M., Logan, G., & Gratton, G. Neurology, 39, 509–539.
(1990). In search of the point of no return: The McNamee, D., & Wolpert, D. M. (2019). Internal
control of response processes. Journal of Experi- models in biological control. Annual Review of
mental Psychology: Human Perception and Perfor- Control, Robotics, and Autonomous Systems, 2,
mance, 16, 164–182. 339–364.
Kaufman, M. T., Churchland, M. M., Ryu, S. I., & Merton, P. A. (1953). Speculations on the servo-control
Shenoy, K. V. (2014). Cortical activity in the null of movement. In G. E. W. Wolstenholme (Ed.),
space: Permitting preparation without movement. Novartis Foundation symposia (pp. 247–260).
Nature Neuroscience, 17, 440–448. Chichester, UK: Wiley.
Kaufman, M. T., Seely, J. S., Sussillo, D., Ryu, S. I., Miall, R. C., Christensen, L. O. D., Cain, O., & Stanley, J.
Shenoy, K. V., & Churchland, M. M. (2016). The (2007). Disruption of state estimation in the
largest response component in the motor cortex human lateral cerebellum. PloS Biology, 5. doi:
reflects movement timing but not movement type. 10.1371/journal.pbio.0050316
eNeuro, 3. doi: 10.1523/ENEURO.0085-16.2016 Morton, S. M. (2006). Cerebellar contributions to
Kawato, M., Furukawa, K., & Suzuki, R. (1987). A locomotor adaptations during splitbelt treadmill
hierarchical neural-network model for control and walking. Journal of Neuroscience, 26, 9107–9116.
learning of voluntary movement. Biological Cyber- Mulliken, G. H., Musallam, S., & Andersen, R. A.
netics, 57, 169–185. (2008). Forward estimation of movement state in
Kornhuber, H. H., & Deecke, L. (2016). Brain poten- posterior parietal cortex. Proceedings of the
tial changes in voluntary and passive movements National Academy of Sciences of the United States
in humans: Readiness potential and reafferent of America, 105, 8170–8177.
396 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Nachev, P., Kennard, C., & Husain, M. (2008). Rawji, V., Rocchi, L., Foltynie, T., Rothwell, J. C., &
Functional role of the supplementary and pre- Jahanshahi, M. (2020). Ropinirole, a dopamine
supplementary motor areas. Nature Reviews: agonist with high D3 affinity, reduces proactive
Neuroscience, 9, 856–869. inhibition: A double-blind, placebo-controlled
Nambu, A., Tokuno, H., & Takada, M. (2002). study in healthy adults. Neuropharmacology, 179.
Functional significance of the corticosubthalamo- doi: 10.1016/j.neuropharm.2020.108278
pallidal “hyperdirect” pathway. Neuroscience Redgrave, P., Vautrelle, N., Overton, P. G., & Reynolds, J.
Research, 2, 111–117. (2016). Phasic dopamine signaling in action selec-
Nashed, J. Y., Crevecoeur, F., & Scott, S. H. (2012). tion and reinforcement learning. Handbook of
Influence of the behavioral goal and environmen- Behavioral Neuroscience, 24, 707–723.
tal obstacles on rapid feedback responses. Journal Rothwell, J. C., Traub, M. M., Day, B. L., Obeso, J. A.,
of Neurophysiology, 108, 999–1009. Thomas, P. K., & Marsden, C. D. (1982). Manual
Nashef, A., Mitelman, R., Harel, R., Joshua, M., & motor performance in a deafferented man. Brain,
Prut, Y. (2021). Area-specific thalamocortical syn- 105, 515–542.
chronization underlies the transition from motor Schall, J. D., Palmeri, T. J., & Logan, G. D. (2017).
planning to execution. Proceedings of the National Models of inhibitory control. Philosophical Trans-
Academy of Sciences of the United States of actions of the Royal Society B: Biological Sciences.
America, 118. doi: 10.1073/pnas.2012658118 doi: 10.1098/rstb.2016.0193
Nowak, D. A., & Hermsdorfer, J. (2003). Sensorimo- Seiss, E., Klippel, M., Hope, C., Boy, F., & Sumner, P.
tor memory and grip force control: Does grip force (2014). The relationship between reversed masked
anticipate a self-produced weight change when priming and the tri-phasic pattern of the lateral-
drinking with a straw from a cup? European ised readiness potential. PLoS One, 9. doi:
Journal of Neuroscience, 18, 2883–2892. 10.1371/journal.pone.0093876
Obeso, I., Wilkinson, L., Casabona, E., Bringas, M. L., Simmonds, D. J., Pekar, J. J., & Mostofsky, S. H.
Álvarez, M., Álvarez, L., & Jahanshahi, M. (2011). (2008). Meta-analysis of Go/No-go tasks demon-
Deficits in inhibitory control and conflict resolution strating that fMRI activation associated with
on cognitive and motor tasks in Parkinson’s response inhibition is task-dependent. Neuropsy-
disease. Experimental Brain Research, 212, chologia, 46, 224–232.
371–384. Smith, M. A., Brandt, J., & Shadmehr, R. (2000).
Obeso, I., Wilkinson, L., Casabona, E., Speekenbrink, Motor disorder in Huntington’s disease begins as a
M., Bringas, M. L., Álvarez, M., … Jahanshahi, M. dysfunction in error feedback control. Nature,
(2014). The subthalamic nucleus and inhibitory 403, 544–549.
control: Impact of subthalamotomy in Parkinson’s Smith, M. A., & Shadmehr, R. (2005). Intact ability to
disease. Brain, 137, 1470–1480. learn internal models of arm dynamics in Hunting-
Obhi, S. S. (2007). Evidence for feedback dependent ton’s disease but not cerebellar degeneration.
conscious awareness of action. Brain Research, Journal of Neurophysiology, 93, 2809–2821.
1161, 88–94. Smittenaar, P., Guitart-Masip, M., Lutti, A., & Dolan,
Parr, T., Limanowski, J., Rawji, V., & Friston, K. (2021). R. J. (2013). Preparing for selective inhibition
The computational neurology of movement under within frontostriatal loops. Journal of Neurosci-
active inference. Brain, 144, 1799–1818. ence, 33, 18087–18097.
Pouget, P., Murthy, A., & Stuphorn, V. (2017). Sohn, Y. H., Wiltz, K., & Hallett, M. (2002). Effect of
Cortical control and performance monitoring of volitional inhibition on cortical inhibitory mecha-
interrupting and redirecting movements. nisms. Journal of Neurophysiology, 88, 333–338.
Philosophical Transactions of the Royal Society B: Sohrabi, A., & West, R. L. (2009). Positive and negative
Biological Sciences. doi: 10.1098/rstb.2016.0201 congruency effects in masked priming: A neuro-
Pruszynski, J. A., Kurtzer, I., & Scott, S. H. (2011). The computational model based on representation, atten-
long-latency reflex is composed of at least two tion, and conflict. Brain Research, 1289, 124–132.
functionally independent processes. Journal of Sommer, M. A., & Wurtz, R. H. (2006). Influence of
Neurophysiology, 106, 449–459. the thalamus on spatial visual processing in frontal
Rao, J. A., Harrington, D. L., Durgerian, S., Reece, C., cortex. Nature, 444, 374–377.
Mourany, L., Koenig, K., … Rao, S. M. (2014). Stavisky, S. D., Kao, J. C., Ryu, S. I., & Shenoy, K. V.
Disruption of response inhibition circuits in prodro- (2017). Motor cortical visuomotor feedback activ-
mal Huntington disease. Cortex, 58, 72–85. ity is initially isolated from downstream targets in
Rawji, V., Modi, S., Latorre, A., Rocchi, L., Hockey, L., output-null neural state space dimensions.
Bhatia, K., … Jahanshahi, M. (2020). Impaired Neuron, 95, 195-208.
automatic but intact volitional inhibition in primary Sumner, P., & Husain, M. (2008). At the edge of
tic disorders. Brain: A Journal of Neurology, 143, consciousness: Automatic motor activation and
906–919. voluntary control. Neuroscientist, 14, 474–486.
Motor Control: Response Preparation, Initiation, and Inhibition 397

Sumner, P., Nachev, P., Morris, P., Peters, A. M., Jackson, (2019). A consensus guide to capturing the abil-
S. R., Kennard, C., & Husain, M. (2007). Human ity to inhibit actions and impulsive behaviors in
medial frontal cortex mediates unconscious inhibi- the stop-signal task. ELife, 8. doi: 10.7554/eLife.
tion of voluntary action. Neuron, 54, 697–711. 46323
Takei, T., Lomber, S. G., Cook, D. J., & Scott, S. H. Vink, M., Kahn, R. S., Raemaekers, M., Van Den
(2021). Transient deactivation of dorsal premotor Heuvel, M., Boersma, M., & Ramsey, N. F. (2005).
cortex or parietal Area 5 impairs feedback control Function of striatum beyond inhibition and execu-
of the limb in macaques. Current Biology, 31, tion of motor responses. Human Brain Mapping,
1476–1487. 25, 336–344.
Tanaka, H., Ishikawa, T., Lee, J., & Kakei, S. (2020). Vyas, S., Golub, M. D., Sussillo, D., & Shenoy, K. V.
The cerebro-cerebellum as a locus of forward (2020). Computation through neural population
model: A review. Frontiers in Systems Neurosci- dynamics. Annual Review of Neuroscience, 43,
ence, 14. doi: 10.3389/fnsys.2020.00019. 249–275.
Tanji, J., & Evarts, E. V. (1976). Anticipatory activity of Weiskrantz, L., Warrington, E. K., Sanders, M. D., &
motor cortex neurons in relation to direction of an Marshall, J. (1974). Visual capacity in the hemian-
intended movement. Journal of Neurophysiology, opic field following a restricted occipital ablation.
39, 1062–1068. Brain, 97, 709–728.
Thaler, D., & Passingham, R. E. (1995). The functions Wessel, J. R., Reynoso, H. S., & Aron, A. R. (2013).
of the medial premotor cortex. Experimental Brain Saccade suppression exerts global effects on the
Research, 102, 445–460. motor system. Journal of Neurophysiology, 110,
Tseng, Y., Diedrichsen, J., Krakauer, J. W., Shadmehr, 883–890.
R., & Bastian, A. J. (2007). Sensory prediction Willett, F. R., Avansino, D. T., Hochberg, L. R., Hen-
errors drive cerebellum-dependent adaptation of derson, J. M., & Shenoy, K. V. (2021). High-perfor-
reaching. Journal of Neurophysiology, 98, 54–62. mance brain-to-text communication via
Vallbo, Å. B. (1970). Discharge patterns in human handwriting. Nature, 593, 249–254.
muscle spindle afferents during isometric volun- Wilson, A. D., Tresilian, J., & Schlaghecken, F. (2011).
tary contractions. Acta Physiologica Scandinavica, The masked priming toolbox: An open-source
80, 552–566. MATLAB toolbox for masked priming researchers.
van de Schoot, R., Depaoli, S., King, R., Kramer, B., Behavior Research Methods, 43, 210–214.
Märtens, K., Tadesse, M. G., … Yau, C. (2021). Wolpert, Daniel M., & Ghahramani, Z. (2000). Com-
Bayesian statistics and modelling. Nature Reviews: putational principles of movement neuroscience.
Methods Primers, 1. doi: 10.1038/ Nature Neuroscience, 3, 1212–1217.
s43586-021-00017-2. Wolpert, Daniel M, Ghahramani, Z., & Flanagan, J. R.
Van Den Wildenberg, W. P. M., Burle, B., Vidal, F., (2001). Perspectives and problems in motor learn-
Van Der Molen, M. W., Ridderinkhof, K. R., & ing. Trends in Cognitive Sciences, 5, 487–494.
Hasbroucq, T. (2010). Mechanisms and dynamics Wolpert, D.M., & Kawato, M. (1998). Multiple paired
of cortical motor inhibition in the stop-signal para- forward and inverse models for motor control.
digm: A TMS study. Journal of Cognitive Neurosci- Neural Networks, 11, 1317–1329.
ence, 22, 225–239. Wong, A. L., Haith, A. M., & Krakauer, J. W. (2015).
Verbruggen, F., Stevens, T., & Chambers D., C. D. Motor planning. Neuroscientist, 21, 385–398.
(2014). Proactive and reactive stopping when dis- Zandbelt, B. B., & Vink, M. (2010). On the role of the
tracted: An attentional account. Journal of Experi- striatum in response inhibition. PLoS One, 5. doi:
mental Psychology: Human Perception and 10.1371/journal.pone.0013848.
Performance, 40, 1295–1300. Zimnik, A. J., & Churchland, M. M. (2021). Independ-
Verbruggen, F., Aron, A. R., Band, G. P., Beste, C., ent generation of sequence elements by motor
Bissett, P. G., Brockett, A. T., … Boehler, C. N. cortex. Nature Neuroscience, 24, 412–424.
25
Olfactory and Gustatory
Sensation-Perception
Anna Kristina Hernandez, Laiquan Zou,
and Thomas Hummel

INTRODUCTION older people as it is also an expression of over-


all health (Mackay-Sim et al., 2006). People who
Unlike vision and audition, the value of olfaction have “aged well” and do not take any medication
and gustation is often neglected, and only recog- seem to have essentially normal smell thresholds
nized when these senses are lost. Then it becomes (Almkvist et al., 1992). The slow, age-related loss
clear that olfaction and gustation play an impor- of smell is however not always noticed or com-
tant role in our daily life, including detection of plained about; and the loss of “fine taste” is appar-
environmental dangers (i.e., fire, toxic fumes, ently partly offset by gustatory and trigeminal
spoiled food, etc.), enjoyment of food, social com- sensations (Shu et al., 2009).
munication, and the overall quality of life (Schäfer In the general population, complete or partial
et al., 2021). loss of olfactory function have been reported in
Among many other factors, olfactory sensi- 3.6 to 5.8%, and 13 to 18% respectively (Hummel
tivity depends on age and gender. Women are et al., 2017), while gustatory dysfuction is less
superior to men in virtually all aspects of olfac- frequent and occurs with an estimated preva-
tory function (Sorokowski et al. 2019), although lence of 5% (Welge-Luessen et al., 2011) to 20%
the effect sizes are not very large. The exact rea- (Vennemann et al., 2008), depending on defini-
son for this is unclear. Hormonal effect has been tions or assessment tools used for smell or taste
discussed but remains a matter of debate (Doty loss. Previous studies showed that, apart from
and Cameron, 2009); possibly the higher social aging, olfactory dysfunction can have various
awareness of women also plays a role, in so far causes, including head trauma, infections of the
that women, more than men, show interest in upper respiratory tract, sinonasal diseases, drugs
odours as social signals (e.g., body odors, food or toxins, or neurological disease (Hummel et al.,
odors). Accordingly, women on average suffer 2017; Whitcroft and Hummel, 2019). In addi-
more than men from loss of smell (Frasnelli and tion, it is estimated that approximately 1 of 8000
Hummel, 2005). The reduced ability to smell with individuals have congenital anosmia (Abolmaali
increasing age is long known (Doty et al., 1984) et al., 2002). As for gustatory dysfunction, the
and is also partly due to the decrease in receptor main causes, apart from idiopathic gustatory dys-
expression patterns (Verbeurgt et al., 2014). The function, are as follows: direct nerve injuries (i.e.,
degree of presbyosmia can, however, vary among after dental/oral/ear surgery, neurologic diseases,
Olfactory and Gustatory Sensation-Perception 399

stroke, head trauma, etc.), vitamin and mineral etc.) may affect the flow of air to the olfactory
deficiencies, systemic diseases (i.e., cancer, dia- neuroepithelium. In particular, certain areas in the
betes, sarcoidosis, etc.), side effects from drugs nasal cavity, namely: the area of the olfactory cleft
(i.e., chemotherapy), post-infectious (i.e., Bell’s and the nasal valve, were found to be important
palsy), and idiopathic or psychologic causes (i.e., for individual variation of odor thresholds among
Burning mouth syndrome) (Hsieh et al., 2022; healthy individuals (Damm et al., 2002).
Bromley and Doty, 2015; Zhu and Hummel, In the human genome, there are approximately
2021; Fark et al., 2013). In recent years, olfactory 1000 olfactory receptors encoded, of which
and gustatory dysfunction have been considered approximately 380 are functionally expressed in
as dominant symptoms of the coronavirus dis- the human olfactory epithelium (Buck and Axel,
ease 2019 (COVID-19) infection (Parma et al., 1991; Firestein, 2001). Each olfactory receptor
2020; Saniasiaya et al., 2022; Trecca et al., 2022; neuron (ORN) expresses a specific receptor type.
Whitcroft and Hummel, 2020). A single odorant typically binds to different recep-
Over the past decades, many different tor types, which ultimately leads to different acti-
approaches have been available to evaluate olfac- vation patterns in the olfactory bulb. The axonal
tory and gustatory function, including subjective, projections are then conveyed with the relay in the
self-reported questionnaires or scales, psycho- olfactory bulb via the olfactory tract to the primary
physical tests, electrophysiological tests and neu- and secondary olfactory cortices, including the
roimaging (Hummel et al., 2017; Han et al., 2020). piriform cortex, the amygdala, the rostral entorhi-
To provide a better understanding of the olfactory nal cortex, orbitofrontal cortex (OFC), hippocam-
and gustatory sensation, this chapter contains a pus, parahippocampal gyrus, cingulate cortex,
summary of various testing options used to evalu- insula, striatum hypothalamus, and the mediodor-
ate olfactory and gustatory function in humans sal thalamus (Zhou et al., 2019; Gottfried, 2010).
and an overview on the description, epidemiol-
ogy, assessment, and treatment of olfactory and
gustatory disorders.
Orthonasal and Retronasal Olfaction
There are two different pathways for odor mole-
cules to reach the olfactory epithelium in the
OLFACTORY SENSATION human olfactory system (i.e., orthonasal and ret-
ronasal olfaction (Rozin, 1982)). In orthonasal
olfaction, odors are perceived during breathing
Anatomy and Physiology and sniffing through the nostrils; whereas in retro-
The perception of odor requires the activation of nasal olfaction, odors reach the olfactory mucosa
the olfactory nerve (Cranial Nerve I), and also the via the nasopharynx during eating or drinking.
trigeminal nerve (Cranial Nerve V) especially in
higher concentrations (Frasnelli and Manescu,
2017), by volatile chemicals through the
nasal cavity. Somatosensory and chemesthetic OLFACTORY ASSESSMENT
sensations of the nose (i.e., burning, warmth, cold-
ness, and pain) may accompany odor perception Different approaches can be used to evaluate
(Frasnelli and Manescu, 2017; Whitcroft and olfactory function: (1) Subjective, self-reported
Hummel, 2019). The presence of adequate nasal olfactory questionnaires or scales (Han et al.,
airflow and an intact peripheral and central olfac- 2020); (2) Psychophysical olfactory tests;
tory system are, likewise, important in this process. (3) Olfactory measures using electrophysiology
studies or magnetic resonance imaging. In the fol-
lowing sections, various methods of olfactory
assessment are described in detail.
Olfactory Neuroepithelium
The olfactory neuroepithelium, composed of
pseudostratified columnar epithelium (Young and Psychometric Questionnaires
Heath, 2000), is a small (∼2 to 5cm2) area of nasal
mucosa located inferior to the cribriform plate, on Frasnelli and Hummel (2005) developed the most
the upper nasal septum, the dorsal superior turbi- widely used self-report olfactory dysfunction
nate, and the middle turbinate, with approximately questionnaire (i.e., Questionnaire of Olfactory
5 to 15% of incoming airstream passing near it. Disorders; QOD). The QOD is divided into four
(Zhao and Frye, 2015). Alterations in the nasal subscales: four items on parosmia, 19 items on
anatomy (i.e., septal deviation, fractures, tumors, quality of life, four items on socially desired
400 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

responses, and five items on visual analogue scale 2019). It consists of 12 items divided into three
ratings on problems arising from olfactory dys- factors: food, imagination, and nature, which rep-
function related to work, to the family, and to resent the ability to experience pleasure while eat-
one’s social life. Due to the questionnaire’s good ing, anticipating food, and smelling natural scents,
reliability and validity, it has been increasingly respectively. The CPS has good validity and high
used by clinicians and researchers, and multi-lan- test-retest reliability in various versions for adults
guage versions have been validated, such as and adolescents/children (Qiu et al., 2021, 2022;
Chinese, English, Persian, and Korean (Choi Zhao et al., 2019).
et al., 2018; Jalessi et al., 2017; Langstaff et al.,
2019; Shu et al., 2011; Yang et al., 2016).
In order to have a broader clinical and research
utility, Mattos et al. (2019) developed a brief ver-
sion of the QOD-NS, which only includes seven PSYCHOPHYSICAL TESTS
items and maintains consistency with the 17-item
QOD-NS. Zou et al. (2022) further evaluated Orthonasal Olfactory Function
the reliability and validity of the brief QOD,
including seven items concerning quality of life A variety of olfactory tests have been developed to
(QOD-QOL), four items concerning parosmia assess orthonasal olfactory function for clinical
(QOD-P), and three visual analogue scales con- and research purposes (Hummel and Podlesek
cerning disease burden, awareness of the disorder, 2021). In this part, we focus on the two most
and issues related to professional life (QOD-VAS). widely used tests (i.e., the “Sniffin’ Sticks” test
The brief QOD showed suitable reliability and (Hummel et al., 1997) and the University of
validity for assessing the subjective severity of Pennsylvania Smell Identification Test [SIT-40]
olfactory dysfunction. (Doty et al., 1984)).
The QOD is widely used in both clinical and The “Sniffin’ Sticks” test battery is com-
research settings. It provides insight regard- prised of reusable felt-tip pens that are used to
ing both qualitative olfactory dysfunction (i.e., test for odor threshold (T), discrimination (D),
parosmia) and olfactory-related quality of life. and identification (I) (Hummel et al., 1997;
However, there is a need for shorter screening ver- see Figure 25.1). For odor threshold and dis-
sions of these established psychometric measures crimination tests, a three-alternative forced-
for easier use in a clinical context. Zou et al. (2020) choice paradigm is used. The tests include
developed the Self-Reported Mini Olfactory 16 triplets of pens that are presented at a distance
Questionnaire (Self-MOQ) to screen for quan- of about 2 cm from the nostrils. Scores for odor
titative olfactory dysfunction (i.e., anosmia and threshold and discrimination ranges from 1 or
hyposmia). It includes only five items, but these 0, respectively, to 16. For odor identification, a
items describe situations that are very common four-alternative forced-choice method is used.
and usually experienced in daily life, for example: Sixteen (16) pens containing suprathreshold
“I do not perceive the smell of coffee and fresh concentrations of common odors are presented
bread.” The Self-MOQ has good internal reliability and participants are tasked to select among four
and validity. The receiver operating characteristic (4) alternative descriptors (including both words
(ROC) analyses indicated that the Self-MOQ is an and pictures), the one that best corresponds to
effective measure for discriminating normosmic their perceived odor. Scores for odor identifica-
from hyposmic and anosmic patients. In China, tion ranges from 0 to 16. A composite TDI score,
Liu et al. (2021) developed the Self-reported composed of odor threshold, discrimination, and
Olfactory Dysfunction Questionnaire (SODQ) for identification scores, ranges from 1 to 48, with
screening quantitative olfactory dysfunction. The higher scores indicating better olfactory ability.
SODQ consists of 10 items with excellent reli- The “Sniffin’ Sticks” Test has good reliability
ability and high validity. Using the Self-MOQ or and validity, and normative data from a wide
SODQ, the optimal cut-off scores are 3.5 or 22, age range (from children to elderly population)
respectively, for distinguishing normosmic indi- are available (Hummel et al., 2007; Kobal et al.,
viduals from olfactory dysfunction patients (Liu 2000; Oleszkiewicz et al., 2019). It has also been
et al., 2021; Zou et al., 2020). adapted and validated in a number of different
Apart from the above-mentioned questionnaires countries (Catana et al., 2012; Dalton et al., 2013;
for qualitative or quantitative olfactory dysfunc- Delgado-Losada et al., 2020; Fjaeldstad et al.,
tion, a questionnaire to measure hedonic capac- 2015; Konstantinidis et al., 2008; Langstaff et al.,
ity for experiencing olfactory pleasure, namely, 2021; Neumann et al., 2012; Oleszkiewicz et al.,
the Chemosensory Pleasure Scale (CPS), has also 2016; Ribeiro et al., 2016; Sai-Guan et al., 2020;
been developed (CPS; Qiu et al., 2021; Zhao et al., Tekeli et al., 2013).
Olfactory and Gustatory Sensation-Perception 401

Figure 25.1 Various psychophysical olfactory and gustatory tests.


A. The “Sniffin’ Sticks” test battery (Hummel et al., 1997): Odor identification test in
foreground. Olfactory threshold test in the middle. Olfactory discrimination test in the back.
B & C. The retronasal test battery. B. Odor delivery container for retronasal olfactory threshold.
C. Tasteless powders for retronasal olfactory identification.
D. The Taste Strips test (Landis et al., 2009): Filter paper strips are infiltrated with four con-
centrations each of sucrose, citric acid, sodium chloride, and quinine hydrochloride. It can
test the taste function of the whole or only the left or right side of the tongue.
Sources: Yoshino et al., 2021; Zhu & Hummel, 2022.

The SIT-40 includes 40 “scratch and sniff” (i.e., flavor perception). Several established tools
microencapsulated odors and is a reliable, stand- used to assess retronasal flavor identification
ardized, forced-choice identification test (Doty function include the taste powders test (Heilmann
et al., 1984). The total SIT-40 score ranges from et al., 2002), Candy Smell Test (Renner et al.,
0 to 40, with higher scores indicating better olfac- 2009), and tasteless powders test (Pieniak et al.,
tory identification ability and extremely low 2021; Yoshino et al., 2021).
scores indicating possible malingering. Similar to The taste powders test contains 20 grocery-
other olfactory tests, it allows the categorization of available food powders (Heilmann et al., 2002).
olfactory function into normosmia, “microsmia”, It is a four-alternative forced-choice identification
and anosmia. Normative data from children to the test where powders are placed on the center of
elderly population are also available. It has been the tongue inside the oral cavity, and subjects are
adapted and validated for use in a number of dif- asked to identify the taste based on a list of four
ferent countries, such as China, Iran, Brazil, and verbal descriptors. Similarly, the Candy Smell
Portugal (Fornazieri et al., 2013, 2015; Jiang et al., Test (CST) consists of 23 different aromatized
2014; Kamrava et al., 2014). smell candies using four-alternative forced-choice
procedure (Renner et al., 2009). The CST is suit-
able for both children and adults. Furthermore, a
shortened self-administered version of the CST
Retronasal olfactory function consisting of seven items (7-CST) was developed
by Besser et al. (2020).
In contrast to orthonasal olfaction, there are lim- However, both the taste powders and CST
ited methods for assessing retronasal olfaction are limited when assessing retronasal olfactory
402 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

identification function, in that they have both may be suitable for children (Renner et al., 2009).
taste and/or texture components, which is espe- However, the reliability and validity of the other
cially true for the taste powders, and are not pure above-mentioned retronasal tests remain to be
tests of retronasal olfaction alone (Yoshino et al., investigated.
2021),. Therefore, to overcome the issue, a new
test for assessing retronasal identification with
tasteless powders was developed (Yoshino et al.,
2021). The tasteless powder test is comprised of ELECTROPHYSIOLOGY AND FUNCTIONAL
20 aroma powders which are selected to minimize
stimulation of taste. Based on the tasteless powder BRAIN IMAGING
test, Pieniak et al. (2021) developed the Q-Powers,
a 3-item version of the tasteless powders test used Subjective questionnaires/scales and psychophys-
for quick retronasal screening purposes. ical measures are widely used in clinical practice
The above-mentioned retronasal olfactory and research. However, these tests may be
tests can only assess odor identification function. influenced by patients’ response bias.
To assess retronasal function more completely, Electrophysiological (including electroencepha-
Yoshino et al. (2021) developed a retronasal test lography and electro-olfactogram) and imaging
battery comprised of the retronasal olfactory techniques provide non-invasive tools to assess
threshold and identification function (Figure 25.1) olfaction in a more objective way (Hummel et al.,
which has been examined for validity and reli- 2017). Electroencephalography (EEG) and EEG-
ability. The retronasal olfactory threshold was derived event-related potentials (ERP) are directly
assessed using a modified odor delivery container related to neuronal activation, have high temporal
procedure, whilst identification function was eval- resolution but low spatial resolution. Several stud-
uated using 16 tasteless powders. ies showed that this technique produces reliable
and valid results (Grosser et al., 2000; Hummel
and Heilmann, 2008; Hummel et al., 2007; Lötsch
and Hummel, 2006; Pause and Krauel, 2000;
Rombaux et al., 2010). Localization analysis iden-
OLFACTORY TESTING IN CHILDREN tified sources of olfactory ERP in mesial tempo-
ral, lateral temporal, and frontal structures
Although the “Sniffin’ Sticks” test and SIT-40 are (Lascano et al., 2010). Electro-olfactograms
widely used in both clinical and research contexts, (EOG) allow the recording of the olfactory
these tests may not accurately measure children’s mucosal potential though an electrode in contact
olfactory ability due, in part, to children’s limited with the olfactory neuroepithelium (Hummel
attention span and unfamiliarity with the odors et al., 1996; Lapid and Hummel 2013). A non-
(Zou et al., 2020). Several odor-identification tests invasive method of signal recording from the
have been developed for children, such as, the olfactory bulb through probes over the nasal
Sydney Children’s Hospital Odor Identification dorsum have been previously reported also
Test (SCHOT; Jiang et al., 2014), the Lyon (Iravani et al., 2020). However, the utility of EEG
Clinical Olfactory Test (LCOT; Monnery-Patris and EOG is limited to specific centers due to their
et al., 2009), the NIH Toolbox Odor Identification dependence on high-precision olfactometry to
Test (Dalton et al., 2013), the Smell Wheel deliver precisely known concentrations of odor
(Cameron and Doty, 2013), and the “Sniffin’ (Lundström et al., 2010).
Kids” Test (Schriever et al., 2014). However, these Functional imaging based on positron emis-
tests for children have not been validated in a sion tomography (PET) and magnetic resonance
cross-cultural context. Hence, Schriever and col- imaging (fMRI) map the brain activity via the
leagues developed the U-Sniff test as an interna- cerebral blood flow changes (Savic, 2002). These
tional 12-item odor identification test for children techniques have a high spatial resolution and have
(Schriever et al., 2018). Similar to the “Sniffin’ significantly contributed to investigate the brain
Sticks” Test, it is based on reusable felt-tip pen- activity in the processing of olfactory stimuli (Han
like devices. The U-Sniff Test for children shows et al., 2019). Similar to electrophysiological tech-
a high test-retest reliability on a global scale, niques, an olfactometer is needed to deliver the
normative data of the U-Sniff test has been estab- relevant odorants when investigating brain activ-
lished for 27 countries (Gellrich et al., 2019; Zou ity. Previous studies showed that odor-induced brain
et al., 2020). While the above olfactory tests for activation was decreased in patients with olfactory
children are used to assess orthonasal olfaction, dysfunction compared to healthy controls in olfac-
no methods have been developed to specifically tory related regions, including the piriform cor-
assess retronasal olfaction in children. The CST tex, amygdala, OFC, insula, and anterior cingulate
Olfactory and Gustatory Sensation-Perception 403

cortex (Han et al. 2018; Moon et al. 2018; Pellegrino matter integrity (Gullmar et al. 2017; Segura et al.
et al. 2016). In addition, functional connectivity of 2013; Woodward et al. 2017). Chen et al. (2020)
the olfactory related regions including the ante- further found that patients with olfactory dysfunc-
rior prefrontal cortex, anterior cingulate cortex, the tion demonstrated relevant white matter network
entorhinal cortex and the cerebellum was reduced dysfunction, although their structural integrity
in patients with olfactory dysfunction (Kollndorfer remained intact.
et al., 2015a,b). Still, as of today, olfactory FMRI
recordings cannot be used on an individual basis,
for example— to support diagnostical decisions
(Zang et al., 2021).
OLFACTORY DYSFUNCTION

Description and Epidemiology


STRUCTURAL BRAIN IMAGING Olfactory dysfunction can be distinguished as
quantitative and qualitative impairment.
The MRI technique can be used to measure the Quantitative olfactory dysfunction may be classi-
olfactory bulb volume and olfactory sulcus depth. fied into two: hyposmia and anosmia. Hyposmia
As the most important relay station in olfactory is defined as a reduced perception of odors, while
processing, a large number of studies have dem- anosmia is the inability to perceive odors (Hummel
onstrated that the OB volume is related to olfac- et al., 2017, Hernandez et al., 2023). On the other
tory function and the prognosis of olfactory loss hand, qualitative olfactory dysfunction can be
(Buschhüter et al., 2008; Lu et al., 2021; Mazal further classified as either parosmia or phantos-
et al., 2016; Rombaux et al., 2012; Seubert et al., mia. Parosmia is defined as distorted perception of
2013; Yousem et al., 1998). Furthermore, a recent an odor, while phantosmia refers to a perception
systematic review showed that strong associations of an odor in the absence of an odor stimulus
between OB volume and olfactory function were (Hummel et al., 2017; Leopold, 2002; Hernandez
consistently found across various etiologies of et al., 2023).
olfactory disorders in previous studies Prevalence of olfactory dysfunction can greatly
(Manan et al., 2022). vary depending on the demographic characteris-
The olfactory sulcus receives indirect projec- tics of the study population. In a review by Desiato
tions from the olfactory bulb and the depth of the et al. (2022), the prevalence of olfactory dysfunc-
sulcus can be used in evaluating olfactory function tion in the general population is as high as 22%.
(Abolmaali et al., 2002; Huart et al., 2011). It has In addition, some studies report the prevalence of
been shown that there is a relationship between anosmia and hyposmia in the general population
olfactory sulcus depth and olfactory function in to be 3.6 to 5.8% anosmia and 13 to 18%, respec-
healthy subjects (Hummel et al., 2003) and in tively (Hummel et al., 2017). Quantitative olfac-
olfactory dysfunction patients (Abolmaali et al., tory disorders can also be congenital, and may
2002; Huart et al., 2011; Hummel et al., 2015; occur as part of a genetic condition, prenatally
Karstensen et al., 2018; Miao et al., 2015); there- acquired diseases or syndromes, or as an isolated,
fore, it appears helpful to measure these routinely idiopathic, non-syndromic olfactory loss with-
in clinical settings. out a known genetic cause (Schäfer et al., 2021;
Apart from its relation to the olfactory bulb Whitcroft and Hummel, 2021). As for qualitative
and sulcus, olfactory dysfunction also results in impairment (e.g., parosmia), the prevalence is
structural changes in higher-order brain regions. estimated at around 3.9% in the general popula-
Manan et al. (2022) conducted a systematic tion (Nordin et al., 2007), although these estimates
review on the morphological variations in gray greatly depend on how “parosmia” is defined.
matter and white matter volume in both congenital
and acquired anosmia. Individuals with acquired
anosmia were found to have reduced gray matter
in the gyrus rectus, medial orbitofrontal cortex,
anterior cingulate cortex, and cerebellum, whereas
OLFACTORY CLINICAL EXAMINATIONS
those with congenital anosmia had larger volume AND TREATMENTS
and greater thickness in parts of the olfactory net-
work, including the piriform cortex, orbitofrontal Accurate assessment and diagnosis of patients
cortex, and insula. In addition, previous diffusion with olfactory disorders is very important.
tensor imaging (DTI) studies showed that olfac- Through measurement of olfactory function, clini-
tory function is positively correlated with white cians are better able to counsel patients on the
404 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

prognosis of their condition and provide guidance olfactory disorders secondary to traumatic brain
on treatment and other management approaches. injury, post-infectious olfactory loss, neurodegen-
A thorough and systematic clinical history taking erative diseases, and following exposure to toxins
and a full ENT examination should be done (Han et al., 2019). Moreover, a number of stud-
(Hummel et al., 2014, 2017). Questions related to ies have shown abnormal brain responses to odors
the nature of the impairment, onset, duration, fre- in patients with olfactory disorders (Han et al.,
quency, severity may already provide insight as to 2019). Currently, structural and functional MRI
a probable cause. Other symptoms (both sinonasal may help in investigating the underlying cause of
and non-sinonasal), past medical and family med- olfactory disorders. However, it is not common
ical history, medications or previous treatments, practice to use structural and functional MRI of
may help narrow down the list further (Hummel higher-order olfactory-related regions to diagnose
et al., 2017). As mentioned above, different olfactory disorders (Zang et al., 2021).
approaches can be used to comprehensively evalu- Patients with smell dysfunction should be
ate olfactory function. Psychometric question- counselled on general safety and may benefit from
naires and scales, such as Self-MOQ and SODQ installation of smoke/gas alarms and adherence
can be performed to screen for the presence of to use-by dates on food products (Hummel et al.,
olfactory disorders. These questionnaires have 2017). Depending on the underlying etiology,
been shown to be simple, reliable, and valid there are several interventions in the treatment of
screening tools for olfactory dysfunction in clini- olfactory disorders. Olfactory training remains to
cal practice. However, one should keep in mind have the strongest evidence for the treatment of
that it is best for questionnaires and scales to be olfactory loss, with 30% of individuals reporting
validated for the target population. Although these improvement in olfactory function after 12 weeks
measures may help to screen for olfactory dys- (Hummel et al., 2009; Patel et al. 2022). Varying
function, olfactory testing is considered an essen- degrees of effect have been reported depending
tial aspect of olfactory assessment. on the duration of training and etiology of olfac-
Standardized psychophysical orthonasal and tory dysfunction (Oleszkiewicz et al., 2018).
retronasal tests can be easily implemented in clini- Olfactory training has also been widely studied
cal practice as less-biased diagnostic tools – in in relation to various etiologies of olfactory loss,
comparison to ratings – to detect olfactory disor- including post-infectious (including COVID-19),
ders. The delineation for anosmia, hyposmia, and post-traumatic, and even in neurodegenerative
normosmia depends on which psychophysical test diseases (Pieniak et al., 2022). Other treatment
is used, and is often based on the performance of interventions include: corticosteroids (i.e., sys-
a sample of normal individuals in a particular age temic and topical), intranasal vitamin A, sodium
group. For example, in the “Sniffin’ Sticks” test, a citrate, phosphodiesterase inhibitors, monoclonal
composite TDI score was used to define anosmia antibodies, omega-3, platelet rich plasma, palmi-
(TDI ≤16), hyposmia (16 < TDI < 30.75), or nor- toyl ethanolamide/luteolin, or surgery (Hernandez
mosmia (TDI ≥30.75) (Oleszkiewicz et al., 2019). et al., 2022; Hummel et al., 2017; Pieniak et al.,
The composite score below 17 in the retronasal 2022; Whitcroft and Hummel 2019; Yan et al.,
test battery (Yoshino et al., 2021b) was the cut-off 2023). Patients who may experience anxiety or
value for olfactory disorders. depression, as well as those who have other sys-
In contrast to olfactory questionnaires and temic or neurologic symptoms should be referred
psychophysical olfactory tests, electrophysiologi- to specialists as appropriate. (Hummel et al., 2017)
cal tools, like olfactory ERP, are less dependent
on subjective response bias and can detect subtle
changes of olfactory function (Peters et al., 2003).
It has clinical value and is feasible for clinical Gustatory sensation
practice (Güdücü et al., 2019), although the setup
requires expertise in terms of both handling of Anatomy and Physiology
dynamic continuous air-flow oflactometry and the The sense of taste is based on the detection of chemi-
analysis and interpretation of the results. cals by specialized taste cells in the mouth. Taste
Structural MRI is the gold standard for the receptor cells are located within taste buds, which
diagnosis of congenital olfactory dysfunction again are contained in the visible papillae. Fungiform,
which is typically, but not always, associated with circumvallate, and foliate papillae contain taste buds,
olfactory bulb aplasia or hypoplasia. Previous while filiform papillae contain only keratinocytes.
studies showed that the depth of the olfactory Fungiform papillae are scattered across the tip and
sulcus was significantly shorter in patients with middle of the tongue, and usually each papilla con-
congenital anosmia (Abolmaali et al., 2002; Huart tains only one taste bud. The human tongue has
et al., 2011). Structural alterations in higher-order between 2,000 and 8,000 taste pores, each packed
olfactory related regions are also affected in the with approximately 50 to 100 taste cells (Chaudhari
Olfactory and Gustatory Sensation-Perception 405

and Roper, 2010; Witt et al., 2003). Taste cells have ingesting rotten or poisonous foods, devastating the
regenerative capabilities with an approximate life enjoyment of eating, resulting in changes in eating
span of 10 to 20 days. habits (Clark, 1998), and mental health (e.g.,
Taste cells are classified based on their taste depression and anxiety; Han et al., 2018; Hur et al.,
receptor expression profiles. Type I cells are most 2018). Malnutrition (Malaty and Malaty, 2013),
abundant, comprising about 50% of cells in taste metabolic, and cardiovascular disease (Sergi et al.,
buds. These cells provide support and protection 2017; Xue et al., 2020) and obesity (Nasser, 2001)
for other taste cells, and are often referred to as have been reported to follow taste dysfunction.
“glia-like cells.” Type I cells may also be respon- Taste dysfunction (dysgeusia) can be clinically
sible for the transduction of salty taste via ionic categorized as quantitative and qualitative taste
currents. Type II cells contain receptors for sweet, disorders (Landis et al., 2004). Quantitative taste
bitter, umami and possibly salty tastes. Type III disorders include ageusia (a complete loss of taste),
cells may respond directly to sour taste stimuli. hypogeusia (diminished sense of taste), and hyper-
Each taste bud also contains 7 to 13 Type IV cells, geusia (enhanced taste sensitivity). Qualitative
which are located at the basement membrane or on taste disorders include parageusia and phantogeu-
the outside the taste buds. These Type IV cells are sia. Parageusia is described as a taste distortion
thought to be pluripotent stem cells for regenera- where gustatory stimuli are reported to be differ-
tion of taste cells every 10 to 14 days (Chaudhari ent from what they used to be. They are perceived
and Roper, 2010; Witt et al., 2003). as bitter, sour, or metallic (e.g., metallic taste when
Type II cells in the taste bud express G-protein eating). Phantogeusia relates to a persistent taste
coupled receptor proteins (GCRPs) for sweet, bitter sensation in the absence of stimulation (Boesveldt
and umami (Chandrashekar et al., 2014). Type III et al., 2011). Taste phantoms (phantogeusia) have
cells express ionic taste receptors for salty or sour been reported in both epilepsy and schizophrenia
tastants but the exact receptors have not been fully (Hausser-Hauw and Bancaud, 1987). In clinical
elucidated (Kobayakawa and Ogawa, 2014; Niki practice, many patients are found to suffer from
et al., 2010). Afferent nerves make contact with the both quantitative and qualitative taste disorders
receptor cells at the base of the taste bud. A single (Fark et al., 2013).
taste bud may be innervated by several afferents, The three most frequent causes of taste dysfunc-
while a single fiber may innervate several taste tion depend on the investigated samples. A study
buds. There is not always a correlation between (Ikeda et al., 2008) based on 408 patients with taste
chemical stimulant and perceived taste quality; loss showed that the most frequent causative fac-
in turn, different types of chemicals can evoke tor was the administration of drugs (32%), followed
similar taste sensations (Smith, 1997; Smith and by idiopathic factors (28%) and systemic disease
St John, 1999). (13%). Another study reviewing 491 patients
Information is mainly carried by specific with quantitative and/or qualitative taste disorders
branches of three cranial nerves (CN). The facial showed that the three most frequent causes were
nerve (CN VII) innervates the anterior two-thirds idiopathic (34%), head trauma (24%), and surgery
of the tongue, the glossopharyngeal nerve (CN (e.g., tonsillectomy, 15%), with a much lower per-
IX) innervates the posterior one-third of the centage of patients with dysgeusia following admin-
tongue, and the vagal nerve (CN X) carries taste istration of drugs or contact with toxic substances
information from the back part of the mouth (4%) (Fark et al., 2013).
including the upper third of the esophagus. The
tongue is also innervated by the trigeminal nerve
(CN V), which is involved in tasting through the
perception of touch, pressure, temperature, and GUSTATORY CLINICAL EXAMINATIONS
pain (e.g., spicy foods). The first nuclear region
in the brainstem is the nucleus of the solitary AND TREATMENTS
tract of the medulla from which the information
runs to the posteromedial ventral nucleus of the Many individuals do not spontaneously report
thalamus, and then to the cortex, where taste their taste disorder, particularly if other symptoms
information is integrated with other sensory are present (Soter et al., 2008; Welge-Lüssen
information to form a “flavor” (Rolls, 2019). et al., 2011). Thus, it appears extremely important
to ask “effective” questions. For example, the
question “Do you have difficulties in recognizing
food or beverages as sweet, sour, bitter, or salty?”
GUSTATORY DYSFUNCTION is evidenced to be more effective than the simple
question “Do you have a taste problem?” in
Taste dysfunction can greatly affect an individual’s detecting real taste problems and rule out non-
safety, quality of life (e.g., increasing risk of taste problems (Gent et al., 1987).
406 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

However, asking about taste function is unreli- warning us of potential danger, allowing social com-
able and becomes meaningful only with concurrent munication, and facilitating the appreciation of food
testing (Landis et al., 2004). Clinically, psychophys- and drinks. These functions are also reflected in some
ical tests are most commonly used, which involve complaints of patients with smell disorders: occur-
different subtypes. For example, based on different rence of accidents related to foods, inability to smell
aims, there are taste threshold tests, taste identifi- other people like family members, and the loss of
cation tests (e.g., Taste strips; Landis et al., 2009, flavour perception. Still, approximately 5% of the
Zhu & Hummel, 2022; see Figure 25.1) and taste general population live without the sense of smell
intensity tests. Based on the use of either chemical (with “old age” being a major factor in smell loss).
stimuli or electric stimuli, there are chemogustom- Other causes of olfactory loss include: acute infec-
etry and electrogustometry. Based on different test- tions of the upper respiratory tract (like COVID-19),
ing regions, there could be whole-mouth testing and sinonasal diseases (including chronic rhinosinusitis
regional testing (Zhu and Hummel, 2021). or allergies), or head trauma. Of course, people can
Taking clinical history from patients is important live without their sense of smell, but their quality of
(Gent et al., 1987). The history should focus on prob- life diminishes, with a proportion of them showing
able causes and accompanying diseases (e.g., diabe- signs of depression.
tes mellitus, hypothyroidism, or cancer; Malaty and The discrimination of odor qualities originates
Malaty, 2013). For example, if abnormalities in the from the olfactory mucosa with olfactory recep-
nervous pathways or lesions of brain are suspected, tor neurons (which can regenerate) expressing
electrophysiological tests (Jääskeläinen et al., 1997) one specific olfactory receptor from a variety of
or structural imaging (e.g., MRI) can be employed approximately 400 of them. Odorants typically
(Heckmann et al., 2003). In addition, analysis of activate not just one, but several types of recep-
saliva (Bradley and Beidler, 2003), and assessment tors, which then leads to a specific pattern of acti-
of morphological changes of the tongue tissues are vation in the olfactory bulb ultimately resulting
also available if necessary (Hummel et al., 2004). in an olfactory percept. Apart from the olfactory
Blood tests to screen for systemic diseases or defi- bulb, numerous other brain structures are involved
ciency states may be warranted in selected patients in processing of odor-related information (i.e., the
(Hsieh et al., 2022). primary olfactory cortex including the entorhinal
Counseling on food safety should be routine, and the piriform cortex, the amygdala, and the sec-
also possibly referring to other specialists (i.e., ondary olfactory cortex including the hippocam-
psychologists, psychiatrist, neurologist, dentist, pus and the orbitofrontal cortex. In the gustatory
etc.) as appropriate (Hummel et al., 2017). Patients system, with taste cells on the tongue, few recep-
should be warned against overseasoning with salt tors are involved in the mediation of sensations
or sugar, and instead may be taught to accentu- like sweet, umami, sour, and salty, while approxi-
ate other sensory experiences of meals (texture, mately 25 receptors detect bitterness. Information
temperature, color, aroma, spicyness; Bromley from the mouth is then mediated to the brainstem
& Doty, 2015). Supplementations with zinc glu- through the chorda tympani, the glossopharyngeal
conate and alpha lipoic acid for several months nerve, and the vagus nerve, and is finally received
have been reported to help restore taste in a subset at the insula, also the primary gustatory cortex.
of patients (Femiano and Scully, 2002; Takaoka Being highly interactive, taste, smell, and the
et al., 2010). In addition, both corticosteroids and trigeminal system (burning, stinging, or tickling)
vitamin A have been used to treat taste disorders, are typically integrated within the central nervous
despite the lack of convincing clinical studies. system (CNS) in order to form holistic percepts.
Thus, in gustatory disorders, the focus is on the In recent years, there have been major improve-
search for, and therapy of, possible underlying ments in the assessment of olfactory and gus-
diseases. This approach also includes the thorough tatory abilities in chemosensory dysfunction,
review of drugs taken by the patient (Naik et al., which is important in the development of treat-
2010). If there is no specific treatment option, par- ment options. We discussed the various tests used
ticularly in the case of idiopathic dysgeusia, zinc to evaluate olfactory and gustatory function in
may be promising according to preliminary data humans, including questionnaires, psychophysical
(Heckmann et al., 2005; Stoll and Oepen, 1994). tests, as well as electrophysiological and imaging
techniques. Most questionnaires and psychophysi-
cal tests are used for evaluation of adults, while a
few standardized (especially gustatory) tests have
SUMMARY AND CONCLUSIONS been developed for children. Numerous child-
hood disorders are associated with olfactory and
The chemical olfactory and gustatory senses are gustatory dysfunction, so further development and
important in one’s daily life. Generally, three func- implementation of simple, useful tools for chil-
tions are assigned to the sense of smell, namely: dren is necessary.
Olfactory and Gustatory Sensation-Perception 407

In contrast to questionnaires and psychophysi- Cameron, E. L., & Doty, R. L. (2013). Odor identifica-
cal tests, electrophysiological and imaging tech- tion testing in children and young adults using the
niques are objective measuring techniques that can smell wheel. International Journal of Pediatric
detect subtle neurological changes of olfactory Otorhinolaryngology, 77(3), 346–350.
and gustatory function. Structural and functional Catana, I., Negoiaș, S., Maniu, A., Porojan, M., &
MRI help us understand olfactory and gustatory Cosgarea, M. (2012). A modified version of
disorders. However, it is far from clinical practice “Sniffin’Sticks” odor identification test: The Roma-
to investigate higher-order brain regions using nian cultural adaptation. Medicine and Pharmacy
functional MRI as individual markers for diagno- Reports, 85(2), 218–223.
sis of olfactory and gustatory disorders. Chandrashekar, J., Hoon, M., Ryba, N., & Zuker, C.
Overall, the chemical senses contribute greatly (2006). The receptors and cells for mammalian
to one’s social life, enjoyment of eating and drink- taste. Nature, 444, 288–294.
ing, and quality of life. They are different from Chaudhari, N., & Roper, S. D. (2010). The cell biology
other senses in many aspects, one of them being of taste. Journal of Cell Biology, 190(2),
that they exhibit strong regenerative capacities. 285–296.
Based on the evolution of knowledge regarding the Chen, B., Akshita, J., Han, P., Thaploo, D., Kitzler, H. H.,
biology of chemical senses over the last 20 years, & Hummel, T. (2020). Aberrancies of brain net-
there remains more to discover and develop in this work structures in patients with anosmia. Brain
constantly evolving field. Topography, 33(3), 403–411.
Choi, W. R., Jeong, H. Y., & Kim, J. H. (2018). Reliabil-
ity and validity of the Korean version of the Ques-
tionnaire of Olfactory Disorders. International
REFERENCES Forum of Allergy and Rhinology, 8(12),
1481–1485.
Abolmaali, N. D., Hietschold, V., Vogl, T. J., Hütten- Clark, J. E. (1998). Taste and flavour: their impor-
brink, K.-B., & Hummel, T. (2002). MR evaluation tance in food choice and acceptance. Proceedings
in patients with isolated anosmia since birth or of the Nutrition Society, 57(4), 639–643.
early childhood. American Journal of Neuroradiol- Croy, I., Negoias, S., Novakova, L., Landis, B. N., &
ogy, 23(1), 157–164. Hummel, T. (2012). Learning about the functions
Almkvist, O., Berglund, B., & Nordin, S. (1992). Odor of the olfactory system from people without a
detectability in successfully aged elderly and young sense of smell. PloS One, 7(3). doi: 10.1371/jour-
adults. Reports from the Department of Psychol- nal.pone.0033365
ogy, Stockholm University, 744, 1–12. Dalton, P., Doty, R. L., Murphy, C., Frank, R.,
Besser, G., Liu, D. T., Prem, B., Iabloncsik, D., Pablik, Hoffman, H. J., Maute, C., … Slotkin, J. (2013).
E., Mueller, C. A., & Renner, B. (2020). Retronasal Olfactory assessment using the NIH Toolbox.
olfactory testing using candies sent by post and for Neurology, 80(11, S3), S32–S36.
screening purposes: A feasibility study. Rhinology, Damm, M., Vent, J., Schmidt, M., Theissen, P., Eckel,
58(3), 218–225. H. E., Lötsch, J., & Hummel, T. (2002). Intranasal
Boesveldt, S., Lindau, S. T., Mcclintock, M. K., volume and olfactory function. Chemical Senses,
Hummel, T., & Lindstrom, J. N. (2011). Gustatory 27(9), 831–839.
and olfactory dysfunction in older adults: A Delgado-Losada, M. L., Delgado-Lima, A. H., &
national probability study. Rhinology, 49(3), Bouhaben, J. (2020). Spanish validation for olfac-
324–330. tory function testing using the Sniffin’Sticks
Bradley, R. M., & Beidler, L. M. (2003). Saliva: Its role olfactory test: Threshold, discrimination, and identi-
in taste function. In R. L. Doty, (Ed.). Handbook of fication. Brain Sciences, 10(12). doi: 10.3390/
olfaction and gustation (pp. 639–650). New York: brainsci10120943
Marcel Dekker. Doty, R. L., Shaman, P., & Dann, M. (1984).
Buck, L., & Axel, R. (1991). A novel multigene family Development of the University of Pennsylvania
may encode odorant receptors: A molecular basis Smell Identification Test: A standardized microen-
for odor recognition. Cell, 65(1), 175–187. capsulated test of olfactory function. Physiology
Bromley, S. M., & Doty, R. L. (2015). Clinical disorders and Behavior, 32(3), 489–502.
affecting taste: An update. In R. L. Doty (Ed.), Hand- Doty, R. L., & Cameron, E. L. (2009). Sex differences and
book of olfaction and gustation (pp. 887–910). reproductive hormone influences on human odor
Hoboken, NJ: Wiley. perception. Physiology & Behavior, 97(2), 213–228.
Buschhüter, D., Smitka, M., Puschmann, S., Gerber, J. C., Fark, T., Hummel, C., Hhner, A., Nin, T., & Hummel,
Witt, M., Abolmaali, N. D., & Hummel, T. (2008). T. (2013). Characteristics of taste disorders. Euro-
Correlation between olfactory bulb volume and pean Archives of Oto-Rhino-Laryngology, 270(6),
olfactory function. NeuroImage, 42(2), 498–502. 1855–1860.
408 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Femiano, F., & Scully, C. (2002). Burning mouth syn- Han, P., Su, T., Qin, M., Chen, H., & Hummel, T.
drome (BMS): Double blind controlled study of (2021). A systematic review of olfactory related
alpha-lipoic acid (thioctic acid) therapy. Journal of questionnaires and scales. Rhinology, 59(2),
Oral Pathology & Medicine, 31(5), 267–269. 133–143.
Firestein, S. (2001). How the olfactory system makes Han, P., Georgi, M., Cuevas, M., Haehner, A.,
sense of scents. Nature, 413, 211–218. Gudziol, V., & Hummel, T. (2018). Decreased elec-
Fjaeldstad, A., Kjaergaard, T., Van Hartevelt, T. J., trogustometric taste sensitivity in patients with
Moeller, A., Kringelbach, M. L., & Ovesen, T. acquired olfactory dysfunction. Rhinology, 56(2),
(2015). Olfactory screening: Validation of 158–165.
Sniffin’Sticks in Denmark. Clinical Otolaryngology, Han, P., Winkler, N., Hummel, C., Hähner, A., Gerber,
40(6), 545–550. J., & Hummel, T. (2018). Impaired brain response
Fornazieri, M. A., Doty, R. L., Santos, C. A. dos, to odors in patients with varied severity of olfac-
Pinna, F. de R., Bezerra, T. F. P., & Voegels, R. L. tory loss after traumatic brain injury. Journal of
(2013). A new cultural adaptation of the Univer- Neurology, 265(10), 2322–2332.
sity of Pennsylvania Smell Identification Test. Clin- Han, P., Zang, Y., Akshita, J., & Hummel, T. (2019).
ics, 68, 65–68. Magnetic resonance imaging of human olfactory
Fornazieri, M. A., Santos, C. A. dos, Bezerra, T. F. P., dysfunction. Brain Topography, 32(6), 987–997.
Pinna, F. de R., Voegels, R. L., & Doty, R. L. (2015). Hausser-Hauw, C., & Bancaud, J. (1987). Gustatory
Development of normative data for the Brazilian hallucinations in epileptic seizures: Electrophysio-
adaptation of the University of Pennsylvania Smell logical, clinical and anatomical correlates. Brain,
Identification Test. Chemical Senses, 40(2), 110(2), 339–359.
141–149. Heckmann, J. G., Heckmann, S. M., Lang, C., &
Frasnelli, J., & Hummel, T. (2005). Olfactory dysfunc- Hummel, T. (2003). Neurological aspects of taste
tion and daily life. European Archives of Oto- disorders. Archives of Neurology, 60(5), 667–671.
Rhino-Laryngology, 262(3), 231–235. Heckmann, S. M., Hujoel, P., Habiger, S., Friess, W.,
Frasnelli, J., & Manescu, S. (2017). The intranasal Wichmann, M., Heckmann, J. G., & Hummel, T.
trigeminal system. In A. Buettner (Ed.), Springer (2005). Zinc gluconate in the treatment of dysgeu-
handbook of odor (pp. 881–895), Cham, Switzer- sia: A randomized clinical trial. Journal of Dental
land: Springer. Research, 84(1), 35–38.
Gellrich, J., Sparing-Paschke, L. M., Thieme, T., Heilmann, S., Strehle, G., Rosenheim, K., Damm,
Schwabe, K., Dworschak, A., Hummel, T., & M., & Hummel, T. (2002). Clinical assessment of
Schriever, V. A. (2019). Normative data for olfac- retronasal olfactory function. Archives of Otolar-
tory threshold and odor identification in children yngology: Head and Neck Surgery, 128(4),
and adolescents. International Journal of Pediatric 414–418.
Otorhinolaryngology, 123, 5–9. Hernandez, A. K., Landis, B., Altundag, A., Fjaeldstad, A.
Gent, J. F., Goodspeed, R. B., Zagraniski, R. T., & Cata- W., Gane, S., Holbrook, E. H., … Hummel, T.
lanotto, F. A. (1987). Taste and smell problems: Vali- (2023). Olfactory nomenclature: An orchestrated
dation of questions for the clinical history. Yale effort to clarify terms and definitions of dysosmia,
Journal of Biology and Medicine, 60(1), 27–35. anosmia, hyposmia, normosmia, hyperosmia, olfac-
Gottfried, J. A. (2010). Central mechanisms of odour tory intolerance, parosmia, and phantosmia/olfactory
object perception. Nature Reviews: Neuroscience, hallucination. Journal for Oto-Rhino-Laryngology:
11(9), 628–641. Head and Neck Surgery. doi: 10.1159/000530211
Grosser, K., Oelkers, R., Hummel, T., Geisslinger, G., Hernandez, A. K., Woosch, D., Haehner, A., Hummel,
Brune, K., Kobal, G., & Lötsch, J. (2000). Olfactory T. (2022). Omega-3 supplementation in postviral
and trigeminal event-related potentials in migraine. olfactory dysfunction: A pilot study. Rhinology,
Cephalalgia, 20(7), 621–631. 60(2), 139–144.
Güdücü, C., Olcay, B. O., Schäfer, L., Aziz, M., Hsieh, J. W., Daskalou, D., Macario, S., Voruz, F., &
Schriever, V. A., Özgören, M., & Hummel, T. Landis, B. N. (2022). How to manage taste disor-
(2019). Separating normosmic and anosmic ders. Current Otorhinolaryngology Reports. doi:
patients based on entropy evaluation of olfactory 10.1007/s40136-022-00428-z
event-related potentials. Brain Research, 1708, Huart, C., Meusel, T., Gerber, J., Duprez, T., Rom-
78–83. baux, P., & Hummel, T. (2011). The depth of the
Güllmar, D., Seeliger, T., Gudziol, H., Teichgräber, U. olfactory sulcus is an indicator of congenital anos-
K., Reichenbach, J. R., Guntinas-Lichius, O., & mia. American Journal of Neuroradiology, 32(10),
Bitter, T. (2017). Improvement of olfactory func- 1911–1914.
tion after sinus surgery correlates with white Hummel, T, Hummel, C., & Welge-Luessen, A.
matter properties measured by diffusion tensor (2004). Assessment of olfaction and gustation. In
imaging. Neuroscience, 360, 190–196. T. Hummel & A. Welge-Luessen (Eds.),
Olfactory and Gustatory Sensation-Perception 409

Management of smell and taste disorders Jääskeläinen, S. K., Forssell, H., & Tenovuo, O.
(pp. 58–75). Stuttgart: Thieme. (1997). Abnormalities of the blink reflex in burn-
Hummel, T, Knecht, M., & Kobal, G. (1996). Peripher- ing mouth syndrome. Pain, 73(3), 455–460.
ally obtained electrophysiological responses to Jalessi, M., Kamrava, S. K., Amini, E., Rafiei, F.,
olfactory stimulation in man: Electro-olfactograms Nasouti, M. A., Moosavi, N., & Farhadi, M. (2017).
exhibit a smaller degree of desensitization com- Is the Persian version of the “Olfactory Disorder
pared with subjective intensity estimates. Brain Questionnaire” reliable and valid? Iranian Journal
Research, 717(1–2), 160–164. of Otorhinolaryngology, 29(4), 209–213.
Hummel, T., Kobal, G., Gudziol, H., & Mackay-Sim, A. Jiang, R.-S., Kuo, L.-T., Wu, S.-H., Su, M.-C., & Liang,
(2007). Normative data for the “Sniffin’ Sticks” K.-L. (2014). Validation of the applicability of the
including tests of odor identification, odor dis- traditional Chinese version of the University of
crimination, and olfactory thresholds: An upgrade Pennsylvania Smell Identification Test in patients
based on a group of more than 3,000 subjects. with chronic rhinosinusitis. Allergy & Rhinology,
European Archives of Oto-Rhino-Laryngology, 5(1), 28–35.
264(3), 237–243. Kamrava, S. K., Farhadi, M., Jalessi, M., Khosravian,
Hummel, T., Reden, K. R. J., Hähner, A., Weidenbe- B., Pousti, B., & Hemami, M. R. (2014). University
cher, M., & Hüttenbrink, K. B. (2009). Effects of of Pennsylvania smell identification on Iranian
olfactory training in patients with olfactory loss. population. Iranian Red Crescent Medical Journal,
Laryngoscope, 119(3), 496–499. 16(1). doi: 10.5812/ircmj.7926
Hummel, T., & Podlesek, D. (2021). Clinical assess- Kobal, G., Klimek, L., Wolfensberger, M., Gudziol, H.,
ment of olfactory function. Chemical Senses, 46. Temmel, A., Owen, C. M., … Hummel, T. (2000).
doi: 10.1093/chemse/bjab053 Multicenter investigation of 1,036 subjects using a
Hummel, T., Whitcroft, K. L., Andrews, P., Altundag, standardized method for the assessment of olfac-
A., Cinghi, C., Costanzo, R. M., … Welge-Luessen, tory function combining tests of odor identifica-
A. (2017). Position paper on olfactory dysfunction. tion, odor discrimination, and olfactory thresholds.
Rhinology, 54, 1–30. European Archives of Oto-Rhino-Laryngology,
Hummel, T., & Heilmann, S. (2008). Olfactory event- 257(4), 205–211.
related potentials in response to ortho-and retro- Kobayakawa, T., & Ogawa, H. (2014). Functional
nasal stimulation with odors related or unrelated anatomy of the gustatory system: From the taste
to foods. International Dairy Journal, 18(8), papilla to the gustatory cortex. In T. Hummel & A.
874–878. Welge-Lüssen (Eds.), Management of smell and
Hummel, T., Bensafi, M., Nikolaus, J., Knecht, M., taste disorders (pp. 150–167). Stuttgart, Germany:
Laing, D. G., & Schaal, B. (2007). Olfactory func- Thieme.
tion in children assessed with psychophysical and Kollndorfer, K., Fischmeister, F. P.,, Kowalczyk, K.,
electrophysiological techniques. Behavioural Brain Hoche, E., Mueller, C. A., Trattnig, S. & Schopf, V.
Research, 180(2), 133–138. (2015a). Olfactory training induces changes in
Hummel, T., Damm, M., Vent, J., Schmidt, M., Theis- regional functional connectivity in patients with
sen, P., Larsson, M., & Klussmann, J.-P. (2003). long-term smell loss. NeuroImage, 9, 401–410.
Depth of olfactory sulcus and olfactory function. Kollndorfer, K., Jakab, A., Mueller, C. A., Trattnig, S.,
Brain Research, 975(1–2), 85–89. & Schopf, V. (2015b). Effects of chronic peripheral
Hummel, T., Sekinger, B., Wolf, S. R., Pauli, E., & olfactory loss on functional brain networks. Neu-
Kobal, G. (1997). “Sniffin’ sticks”: Olfactory per- roscience, 310, 589–599.
formance assessed by the combined testing of Konstantinidis, I., Printza, A., Genetzaki, S., Mamali,
odor identification, odor discrimination and olfac- K., Kekes, G., & Constantinidis, J. (2008). Cultural
tory threshold. Chemical Senses, 22(1), 39–52. adaptation of an olfactory identification test: The
Hur, K., Choi, J. S., Zheng, M., Shen, J., & Wrobel, B. Greek version of Sniffin’Sticks. Rhinology, 46(4),
(2018). Association of alterations in smell and 292–296.
taste with depression in older adults. Laryngo- Landis, B. N., Konnerth, C. G., & Hummel, T. (2004).
scope Investigative Otolaryngology, 3(2), 94–99. A study on the frequency of olfactory dysfunction.
Ikeda, M., Ikui, A., Komiyama, A., Kobayashi, D., & Laryngoscope, 114(10), 1764–1769.
Tanaka, M. (2008). Causative factors of taste dis- Landis, B. N., Welge-Luessen, A., Brämerson, A.,
orders in the elderly, and therapeutic effects of Bende, M., Mueller, C. A., Nordin, S., & Hummel, T.
zinc. Journal of Laryngology & Otology, 122(2), (2009). “Taste Strips”: A rapid, lateralized, gusta-
155–160. tory bedside identification test based on impreg-
Iravani, B., Arshamian, A., Ohla, K., Wilson, D. A., & nated filter papers. Journal of Neurology, 256(2),
Lundström, J. N. (2020). Non-invasive recording from 242–248.
the human olfactory bulb. Nature Communications, Langstaff, L., Clark, A., Salam, M., & Philpott, C. M.
11(1). doi: 10.1038/s41467-020-14520-9. (2021). Cultural adaptation and validity of the
410 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Sniffin’Sticks psychophysical test for the UK set- European Archives of Oto-Rhino-Laryngology,


ting. Chemosensory Perception, 14, 102–108. 273(1), 1–7.
Langstaff, L., Pradhan, N., Clark, A., Boak, D., Salam, Monnery-Patris, S., Rouby, C., Nicklaus, S., & Issan-
M., Hummel, T., & Philpott, C. M. (2019). Valida- chou, S. (2009). Development of olfactory ability
tion of the olfactory disorders questionnaire for in children: Sensitivity and identification. Develop-
English-speaking patients with olfactory disorders. mental Psychobiology, 51(3), 268–276.
Clinical Otolaryngology, 44(5), 715–728. Moon, W. J., Park, M., Hwang, M., & Kim, J. K.
Lapid H, Hummel T. (2013). Recording odor-evoked (2018). Functional MRI as an objective measure of
response potentials at the human olfactory epithe- olfaction deficit in patients with traumatic anos-
lium. Chemical Senses 38, 3–17. mia. American Journal of Neuroradiology, 39(12),
Lascano, A. M., Hummel, T., Lacroix, J.-S., Landis, B. 2320–2325.
N., & Michel, C. M. (2010). Spatio–temporal Naik, B. S., Shetty, N., & Maben, E. (2010). Drug-
dynamics of olfactory processing in the human induced taste disorders. Drug Safety, 21(3),
brain: An event-related source imaging study. 240–243.
Neuroscience, 167(3), 700–708. Nasser, J. (2001). Taste, food intake and obesity.
Leopold, D. (2002). Distortion of olfactory percep- Obesity Reviews, 2(4), 213–218.
tion: Diagnosis and treatment. Chemical Senses, Neumann, C., Tsioulos, K., Merkonidis, C., Salam,
27(7), 611–615. M., Clark, A., & Philpott, C. (2012). Validation
Liu, X., Huang, J., Tian, P., Hu, J., & Zou, L. (2021). study of the “Sniffin’ Sticks” olfactory test in a
Development of a Self-reported Olfactory Dysfunc- British population: A preliminary communication.
tion Questionnaire (SODQ) to screen olfactory Clinical Otolaryngology, 37(1), 23–27.
disorders in China. Rhinology, 59(4), 393–397. Niki, M., Yoshida, R., Takai, S., & Ninomiya, Y. (2010).
Lötsch, J., & Hummel, T. (2006). The clinical signifi- Gustatory signaling in the periphery: Detection,
cance of electrophysiological measures of olfac- transmission, and modulation of taste informa-
tory function. Behavioural Brain Research, 170(1), tion. Biological & Pharmaceutical Bulletin, 33(11),
78–83. 1772–1777.
Lu, R., Aziz, N. A., Reuter, M., Stöcker, T., & Breteler, Nordin, S., Brmerson, A., Millqvist, E., & Bende, M.
M. M. (2021). Evaluation of the neuroanatomical (2007). Prevalence of parosmia: The Skövde popu-
basis of olfactory dysfunction in the general popu- lation-based studies. Rhinology, 45(1), 50–53.
lation. JAMA Otolaryngology: Head & Neck Sur- Oleszkiewicz, A., Hanf, S., Whitcroft, K. L., Haehner, A.,
gery, 147(10), 855–863. & Hummel, T. (2018). Examination of olfactory
Lundström, J. N., Gordon, A. R., Alden, E. C., Boes- training effectiveness in relation to its complexity
veldt, S., & Albrecht, J. (2010). Methods for build- and the cause of olfactory loss. Laryngoscope,
ing an inexpensive computer-controlled 128(7), 1518–1522.
olfactometer for temporally-precise experiments. Oleszkiewicz, A., Schriever, V. A., Croy, I., Hähner, A.,
International Journal of Psychophysiology, 78(2), & Hummel, T. (2019). Updated Sniffin’ Sticks nor-
179–189. mative data based on an extended sample of 9139
Mackay-Sim, A., Johnston, A. N., Owen, C., & Burne, subjects. European Archives of Oto-Rhino-Laryn-
T. H. (2006). Olfactory ability in the healthy popu- gology, 276(3), 719–728.
lation: Reassessing presbyosmia. Chemical Senses, Oleszkiewicz, A., Taut, M., Sorokowska, A., Radwan,
31, 763–771. A., Kamel, R., & Hummel, T. (2016). Development
Malaty, J., & Malaty, I. A. (2013). Smell and taste of the Arabic version of the “Sniffin’ Sticks” odor
disorders in primary care. American Family Physi- identification test. European Archives of Oto-
cian, 88(12), 852–859. Rhino-Laryngology, 273(5), 1179–1184.
Manan, H. A., Yahya, N., Han, P., & Hummel, T. Parma, V., Ohla, K., Veldhuizen, M. G., Niv, M. Y.,
(2022). A systematic review of olfactory-related Kelly, C. E., Bakke, A. J., …Hayes, J. E. (2020).
brain structural changes in patients with congeni- More than smell: COVID-19 is associated with
tal or acquired anosmia. Brain Structure and Func- severe impairment of smell, taste, and chemesthe-
tion, 227(1), 177–202. sis. Chemical Senses, 45(7), 609–622.
Mattos, J. L., Edwards, C., Schlosser, R. J., Hyer, M., Patel, Z. M., Holbrook, E. H., Turner, J. H., Adappa, N.
Mace, J. C., Smith, T. L., & Soler, Z. M. (2019). A D., Albers, M. W., Altundag, A., … Yan, C. H.
brief version of the questionnaire of olfactory dis- (2022). International consensus statement on
orders in patients with chronic rhinosinusitis. allergy and rhinology: Olfaction. International
International Forum of Allergy and Rhinology, Forum of Allergy & Rhinology 12, 327–680.
9(10), 1144–1150. Pause, B. M., & Krauel, K. (2000). Chemosensory
Mazal, P. P., Haehner, A., & Hummel, T. (2016). event-related potentials (CSERP) as a key to the
Relation of the volume of the olfactory bulb to psychology of odors. International Journal of Psy-
psychophysical measures of olfactory function. chophysiology, 36(2), 105–122.
Olfactory and Gustatory Sensation-Perception 411

Pellegrino, R., Haehner, A., Bojanowski, V., Hummel, Rozin, P. (1982). Taste-smell confusions and the dual-
C., Gerber, J., & Hummel, T. (2016). Olfactory ity of the olfactory sense. Perception & Psycho-
function in patients with hyposmia compared to physics, 31, 397–401.
healthy subjects: An fMRI study. Rhinology, 54(4), Sai-Guan, L., Husain, S., Zahedi, F. D., Ahmad, N., &
374–381. Gendeh, B. S. (2020). Cultural adaptation of Snif-
Peters, J. M., Hummel, T., Kratzsch, T., Lötsch, J., fin’ Sticks smell identification test: The Malaysian
Skarke, C., & Frölich, L. (2003). Olfactory function version. Iranian Journal of Otorhinolaryngology,
in mild cognitive impairment and Alzheimer’s dis- 32(111), 213–222.
ease: An investigation using psychophysical and Saniasiaya, J., Islam, M. A., & Abdullah, B. (2021).
electrophysiological techniques. American Journal Prevalence of olfactory dysfunction in coronavirus
of Psychiatry, 160(11), 1995–2002. disease 2019 (COVID-19): A meta-analysis of
Pieniak, M., Oleszkiewicz, A., Avaro, V., Calegari, F., 27,492 patients. Laryngoscope, 131(4), 865–878.
& Hummel, T. (2022). Olfactory training: Thirteen Savic, I. (2002). Imaging of brain activation by odor-
years of research reviewed. Neuroscience & Biobe- ants in humans. Current Opinion in Neurobiology,
havioral Reviews, 141. doi: 10.1016/j. 12(4), 455–461.
neubiorev.2022.104853 Schäfer, L., Schriever, V. A., & Croy, I. (2021). Human
Pieniak, M., Oleszkiewicz, A., Klockow, M., Yoshino, olfactory dysfunction: Causes and consequences.
A., & Hummel, T. (2021). q-Powders: A quick test Cell and Tissue Research, 383(1), 569–579.
for screening retronasal olfactory disorders with Schriever, V. A., Agosin, E., Altundag, A., Avni, H.,
tasteless powders. European Archives of Oto-Rhino- Cao Van, H., Cornejo, C., … Hummel, T. (2018).
Laryngology. doi: 10.1007/s00405-021-06849-8 Development of an international odor identifica-
Qiu, Y. Q., Ma, Q. W., Huang, Y. Y., Lin, J. W., Zhao, tion test for children: The Universal Sniff Test.
J. B., & Zou, L. Q. (2022). Validation of the chem- Journal of Pediatrics, 198, 265–272.
osensory pleasure scale for kids (CPS-K) and its Schriever, V. A., Mori, E., Petters, W., Boerner, C.,
mediating role in the relationship between autistic Smitka, M., & Hummel, T. (2014). The Sniffin’ Kids
traits and eating behaviors. Food Quality and Pref- Test: A 14-item odor identification test for chil-
erence, 95. doi: 10.1016/j.foodqual.2021.104341 dren. Plos One, 9(6). doi: 10.1371/journal.
Qiu, Y.-Q., Huang, G.-J., Zhao, J.-B., Ma, Q.-W., & pone.0101086
Zou, L.-Q. (2021). The chemosensory pleasure Segura, B., Baggio, H. C., Solana, E., Palacios, E. M.,
scale for children (CPS-C): Factor structure, reliabil- Vendrell, P., Bargalló, N., & Junqué, C. (2013).
ity, and validity. Food Quality and Preference, 92. Neuroanatomical correlates of olfactory loss in
doi: 10.1016/j.foodqual.2021.104214 normal aged subjects. Behavioural Brain Research,
Renner, B., Mueller, C. A., Dreier, J., Faulhaber, S., 246, 148–153.
Rascher, W., & Kobal, G. (2009). The Candy smell Sergi, G., Bano, G., Pizzato, S., Veronese, N., &
test: A new test for retronasal olfactory perfor- Manzato, E. (2017). Taste loss in the elderly: Pos-
mance. Laryngoscope, 119(3), 487–495. sible implications for dietary habits. Critical
Ribeiro, J. C., Simões, J., Silva, F., Silva, E. D., Reviews in Food Science and Nutrition, 57(17),
Hummel, C., Hummel, T., & Paiva, A. (2016). Cul- 3684–3689.
tural adaptation of the Portuguese version of the Seubert, J., Freiherr, J., Djordjevic, J., & Lundström, J. N.
“Sniffin’Sticks” smell test: Reliability, validity, and (2013). Statistical localization of human olfactory
normative data. PloS One, 11(2). doi: 10.1371/ cortex. NeuroImage, 66, 333–342.
journal.pone.0148937 Shu, C. H., Hummel, T., Lee, P. L, Chiu, C. H., Lin, S. H.,
Rolls, E. T. (2019). Taste and smell processing in the & Yuan, B. C. (2009). The proportion of self-rated
brain. In R. L. Doty (Ed.), Handbook of clinical olfactory dysfunction does not change across the
neurology (Vol. 164, pp. 97–118). Amsterdem: life span. American Journal of Rhinology & Allergy,
Elsevier. 23, 413–416.
Rombaux, P., Huart, C., Collet, S., Eloy, P., Negoias, Shu, C. H., Lee, P. L., Lan, M. Y., & Lee, Y. L. (2011).
S., & Hummel, T. (2010). Presence of olfactory Factors affecting the impact of olfactory loss on
event-related potentials predicts recovery in the quality of life and emotional coping ability.
patients with olfactory loss following upper res- Rhinology, 49(3), 337–341.
piratory tract infection. Laryngoscope, 120(10), Smith, D. V, & St John, S. J. (1999). Neural coding of
2115–2118. gustatory information. Current Opinion in Neuro-
Rombaux, P., Huart, C., Degouj, N., Duprez, T., & biology, 9(4), 427–435.
Hummel, T. (2012). Prognostic value of olfactory Smith, D. V. (1997). Basic anatomy and physiology of
bulb volume measurement for recovery in postin- taste. In A. Seiden (Ed.), Taste and smell disorders
fectious and posttraumatic olfactory loss. Otolar- (pp. 128-145). New York: Thieme
yngology-Head and Neck Surgery, 147(6), Sorokowski, P., Karwowski, M., Misiak, M., Marczak,
1136–1141. M. K., Dziekan, M., Hummel, T., & Sorokowska, A.
412 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

(2019). Sex differences in human olfaction: A Woodward, M. R., Dwyer, M. G., Bergsland, N.,
meta-analysis. Frontiers in Psychology, 10. doi: Hagemeier, J., Zivadinov, R., Benedict, R. H., &
10.3389/fpsyg.2019.00242 Szigeti, K. (2017). Olfactory identification deficit
Soter, A., Kim, J., Jackman, A., Tourbier, I., Kaul, A., predicts white matter tract impairment in Alzhei-
Doty, R. L. (2008). Accuracy of self-report in mer’s disease. Psychiatry Research: Neuroimaging,
detecting taste dysfunction. Laryngoscope, 118, 266, 90–95.
611–617. Xue, Y., Wen, Q., Xu, C., Zhang, X., Zeng, J., Sha, A.
Stoll, A. L., & Oepen, G. (1994). Zinc salts for the M., …Zeng, C. (2020). Elevated salt taste thresh-
treatment of olfactory and gustatory symptoms in old is associated with increased risk of coronary
psychiatric patients: A case series. Journal of Clini- heart disease. Journal of Cardiovascular Transla-
cal Psychiatry, 55(7), 309–311. tional Research, 13(6), 1016–1023.
Takaoka, T., Sarukura, N., Ueda, C., Kitamura, Y., Yan C. H., Jang S. S., Lin, H. C., Ma, Y., Khanwalkar,
Kalubi, B., Toda, N., … Takeda, N. (2010). Effects A. R., Thai, A. & Patel, Z. M. (2023). Use of
of zinc supplementation on serum zinc concentra- platelet-rich plasma for COVID-19-related olfac-
tion and ratio of apo/holo-activities of angiotensin tory loss: A randomized controlled trial. Interna-
converting enzyme in patients with taste impair- tional Forum of Allergy & Rhinology, 13(6),
ment. Auris Nasus Larynx, 37(2), 190–194. 989–997.
Tekeli, H., Altundağ, A, Salihoğlu, M., Cayönü, M., & Yang, D. H., Wang, J., Ni, D. F., Liu, J. F., & Wang, X.
Kendirli, M. T. (2013). The applicability of the (2016). Reliability and validity of the Chinese ver-
“Sniffin’ Sticks” olfactory test in a Turkish popula- sion of the Questionnaire of Olfactory Disorders
tion. Medical Science Monitor, 19, 1221–1226. (QOD) when used with patients having olfactory
Trecca, E. M., Cassano, M., Longo, F., Petrone, P., dysfunction. European Archives of Oto-Rhino-Lar-
Miani, C., Hummel, T., & Gelardi, M. (2022). yngology, 273(10), 3255–3261.
Results from psychophysical tests of smell and Yoshino, A., Goektas, G., Mahmut, M. K., Zhu, Y.,
taste during the course of SARS-CoV-2 infection: Goektas, O., Komachi, T., … Hummel, T. (2021). A
A review. Acta Otorhinolaryngologica Italica, new method for assessment of retronasal olfactory
42(S1), S20–S35. function. Laryngoscope, 131(2), E324–E330.
Vennemann, M. M., Hummel, T., & Berger, K. (2008). Yoshino, A., Pellegrino, R., Luckett, C. R., & Hummel, T.
The association between smoking and smell and (2021). Validation study of a novel approach for
taste impairment in the general population. Jour- assessment of retronasal olfactory function with
nal of Neurology, 255(8), 1121–1126. combination of odor thresholds and identification.
Verbeurgt, C., Wilkin, F., Tarabichi, M., Gregoire, F., European Archives of Oto-Rhino-Laryngology,
Dumont, J. E., & Chatelain, P. (2014). Profiling of 278, 3847–3856.
olfactory receptor gene expression in whole Young, B., & Heath, J. W. (2000). Special sense
human olfactory mucosa. PLoS One, 9(5). doi: organs. In B. Young & J. W. Heath (Eds.), Wheat-
10.1371/journal.pone.0096333 er’s functional histology (4th ed, p. 380). Edin-
Welge-Lüssen, A., Dörig, P., Wolfensberger, M., burgh: Churchill Livingstone.
Krone, F., & Hummel, T. (2011). A study about the Yousem, D. M., Geckle, R. J., Bilker, W. B., & Doty, R. L.
frequency of taste disorders. Journal of Neurology, (1998). Olfactory bulb and tract and temporal lobe
258(3), 386–392. volumes: Normative data across decades. Annals
Whitcroft, K. L., & Hummel, T. (2019). Clinical diag- of the New York Academy of Sciences, 855(1),
nosis and current management strategies for 546–555.
olfactory dysfunction: A review. JAMA Otolaryn- Zang, Y., Han, P., Joshi, A., & Hummel, T. (2021). Indi-
gology: Head and Neck Surgery, 145(9), vidual variability of olfactory fMRI in normosmia
846–853. and olfactory dysfunction. European Archives of
Whitcroft, K. L., & Hummel, T. (2021). Olfactory func- Oto-Rhino-Laryngology, 278, 379–387.
tion and dysfunction. In P. Flint, H. Francis, B. Zhao, K., & Frye, R. E. (2015). Nasal patency and the
Haughey, M. Lesperance, V. Lund, K. Robbins & R. aerodynamics of nasal airflow in relation to olfac-
Thomas (Eds.), Cummings otolaryngology (7th tory function. In R. L. Doty (Ed.), Handbook of
ed.). Amsterdam: Elsevier. olfaction gustation (3rd ed., pp. 353-374). Hobo-
Whitcroft, K. L., &, Hummel, T. (2020). Olfactory ken, NJ: Wiley.
dysfunction in COVID-19: Diagnosis and manage- Zhao, J. B., Wang, Y. L., Ma, Q. W., Zhao, J. B.,
ment. Journal of the American Medical Associa- Zhang, X. Y., & Zou, L. Q. (2019). The Chemosen-
tion, 323(24), 2512–2514. sory Pleasure Scale: A new assessment for measur-
Witt, M., Reutter, K., & Miller Jr, I. J. (2003). Morphol- ing hedonic smell and taste capacities. Chemical
ogy of the peripheral taste system. In R. L. Doty Senses, 44(7), 457–464.
(Ed.). Handbook of olfaction and gustation (pp. Zhou, G., Lane, G., Cooper, S. L., Kahnt, T., & Zelano, C.
651-678). New York: Taylor & Francis. (2019). Characterizing functional pathways of the
Olfactory and Gustatory Sensation-Perception 413

human olfactory system. Elife, 8. doi:10.7554/ Zou, L. Q., Dworschak, A., Alizadeh, R., Kamrava, S.
eLife.47177 K., Alwashahi, M., Bock, M. A., … Schriever, V. A.
Zhu, Y., & Hummel, T. (2022). Assessment of taste (2020). U-sniff-the international odor identifica-
function. In K. Palmer & G. Servant (Eds.), The tion test for children: An extension of its normative
pharmacology of taste: Handbook of experimental database and study of global reliability. Rhinology,
pharmacology (Vol. 275, pp. 295–315). Cham, 58(5), 471–476.
Switzerland: Springer. Zou, L. Q., Haehner, A., Menzel, S., Gunder, N., &
Zou, L. Q., Linden, L., Cuevas, M., Metasch, M. L., Hummel, T. (2022). Reliability and validity of a
Welge-Lüssen, A., Hähner, A., & Hummel, T. brief version of the Questionnaire of Olfactory
(2020). Self-reported mini olfactory questionnaire Disorders (brief QOD) in patients with olfactory
(Self-MOQ): A simple and useful measurement for dysfunction. Rhinology, 60(1), 56–62.
the screening of olfactory dysfunction. Laryngo-
scope, 130(12), E786–E790.
26
Interoception and
Thermoreception
H u g o D . C r i t c h l e y , Yo k o N a g a i , a n d L i s a Q u a d t

INTRODUCTION the position of the body within the external world.


However, boundaries between interoception and
these other senses are blurred: For example, sens-
Functions ing the environment through tactile, thermal, and
chemoreceptive channels “internalizes” informa-
Interoception is the sense that concerns the inter-
tion salient to physiological health (Craig, 2014).
nal physiological state of the body, and encapsu-
Moreover, acting on the environment changes
lates the afferent signaling, sensing, transmission,
one’s internal state, since proprioceptive and ves-
and perceptual representation of internal bodily
tibular feedback of skeletomotor activity is tightly
signals, transmitted via neural and humoral path-
coupled to efferent autonomic responses, ena-
ways (Cameron, 2002; Craig, 2014; Khalsa et al.,
bling the fine dynamic cardiovascular support of
2018). Interoception drives simple homeostatic
movement (Yates et al., 2014).
(autonomic and neuroendocrine) reflexes, and
more complex allostatic responses, including
adaptive behavior. In part through the experience
of motivational and emotional feelings, interocep-
tion coordinates physiological changes across PERCEPTION
organ systems, and higher-level motivational and
emotional behaviors. By extension, interoception Although interoception includes the conscious
shapes longer-term behavioral repertoires, to perception and appraisal of inner bodily sensa-
ensure health and welfare by guiding the selection tions, for the most part, we are unaware of the
and timing of action policies that help maintain majority of interoceptive signals, when perceived
stable access to resources (in, for example, the consciously (e.g., when feeling the need to void
planning of routine meals including what to eat, bladder or bowel), visceromotor sensations are
when to eat, and with whom). mostly rather diffuse, and lack the perceptual
Interoception is distinct from the exteroceptive clarity of exteroceptive senses. When we do
senses that sample and describe the external envi- become consciously aware of our internal bodily
ronment. Interoception is also conceptually dis- signals, this often means that we need to select a
tinguishable from proprioception, which encodes behavior to help maintain homeostatic state
Interoception and Thermoreception 415

(i.e., the internal changes of a magnitude that determined by context (Cantrill and Hunt, 1932;
exceeds the potential for autonomic reflexes to Schachter and Singer, 1962). Happiness is
sustain homeostasis on their own) (Critchley and reported when arousal is experienced in a happy
Garfinkel, 2017). Perhaps one exception is the emotional context, and anger is reported when
good conscious access we have to respiratory arousal is experienced in an angry emotional
sensations. Speculatively, this ability to perceptu- context.
ally track and volitionally regulate breathing may Thus, an emotion may be triggered and inten-
be tied to the evolutionary development of human sified by interoceptive signaling of physiological
speech. The perception of interoceptive respira- arousal, yet the explanatory (psychosocial) context
tory sensations is also closely coupled to more in which the arousal occurs allows the emotion to
accessible somatosensory signals of pharyngeal be specifically defined and labelled. A related con-
airflow (Sozansky and Houser, 2014) and chest structionist account of human emotion from Lisa
wall motion. When internal changes are intense Feldman Barrett (Barrett, 2017), still grounded
enough to generate viscerosensory pain, soma- on the primacy of interoception, has emerged as a
tosensory pathways are also often recruited. This powerful riposte to the competing idea that there
can amplify the perceptual precision of the are individual “basic” emotions (such as fear, dis-
strength and attributed location of that pain gust, sadness, anger, and happiness) that are deter-
(Gebhart and Bielefeldt, 2016). mined by evolution for specific functions. Such
emotions are typically viewed as fundamental
“natural kinds” that have their own specific neu-
ral-circuity and labelled-line responses (Darwin,
1872; Ekman et al., 1983; Panksepp, 2011). The
EMOTION clinical, experimental and theoretical work of
Antonio Damasio (Damasio, 1999, 2010) revived
In modern Western psychological thought, interest an interest in body–brain interaction, placing it
in interoception has flourished within the context once more toward the foreground of human neu-
of emotion. Physiological sensations, notably of ropsychological thinking. Their studies, compar-
strong fast heartbeats, have been associated with ing people with and without specific brain lesions
passions since at least the time of Aristotle. James as they performed gambling tasks, showed that
(1884) and Lange (1907) proposed a “peripheral implicit physiological arousal responses (somatic
theory” of emotion that encouraged people to markers) can predict and guide adaptive motiva-
consider the perception of physiological sensa- tional (“microeconomic”) decisions even before
tions as the fundamental basis to emotional feel- an individual can consciously describe why they
ings, while James highlighted the link between made a particular decision (Bechara et al., 1997,
somatomotor feedback and fear in his suggestion 2005). This work highlighted the influence of
that “fear is running from the bear.” Lange physiological state and bodily feelings on both
emphasized more the relationship between vascu- cognition and emotion, and thus reinvigorated the
lar responses and diffuse emotional feeling states. field of interoception.
Exploration of, and challenges to, this reductionist
model ensued: Evidence against it included
Sherrington’s (1900) observation that emotional/
affective reactivity remained in dogs after the
brain had been (partially) isolated from the physi- BROADER ASPECTS OF INTEROCEPTION
ological state of the body by spinal transection.
Cannon (1927) and Bard (1934) also argued Psychology has weighted interoceptive science
against James and Lange’s theory by focusing on toward the study of how body-to-brain signals
the lack of strong emotion-specific patterning to are perceived consciously; with perhaps undue
peripheral bodily responses. However, evidence in emphasis on individual differences in people’s
support of the James–Lange therapy included ability to perceive their heart beating at rest. This
masking studies that showed physiological reac- continues as a topic of rich debate. In parallel,
tions to emotive stimuli can be evoked automati- other areas of interoceptive science have devel-
cally with minimal conscious appraisal (Dimberg oped independently: Advances in the understand-
et al., 2000). The issue of lack of specificity of ing of the gut–brain axis (including microbiome;
emotion-related physiological responses was Dinan and Cryan, 2017; Meyer 2011), humoral
partly resolved by “two-factor” models. These signaling from tissues (e.g., in energy storage
were backed by studies showing that the emo- and metabolism; Sun et al., 2014), and immune-
tional reaction to heightened states of physiologi- brain signaling (Dantzer et al., 2008) are influen-
cal arousal in the body (adrenalin-induced) were tial fields that carry their own momentum.
416 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

These channels of interoceptive information flow pathways at the same spinal level/dorsal
typically operate on slower timescales than the horn (somatotome) as the strong viscerosensory
neural signaling of autonomically controlled input. Intraspinal sensitization also contributes to
visceromotor responses. They nonetheless illus- the cross-processing of such sensations.
trate the powerful expression of interoceptive Characteristically, internal organs do not carry the
influences on brain and behavior (Critchley and variety and density of receptor nerve terminals
Harrison 2013). when compared to the skin surface or specialist
sensory organs. Within the smooth muscle of the
gastrointestinal tract, there are broadly two types
of receptor terminal; intramuscular laminar arrays
(IMAs) and intraganglionic laminar endings
INTEROCEPTORS (IGLEs). Both terminal types respond to tissue
stretch and also have chemoreceptive properties
Charles Sherrington (1906) first proposed the (Bethoud and Neuhuber, 2000). Other visceral
notion of interoceptors, referring to sensors detect- organs possess similar afferent terminals, the car-
ing changes in internal states of the body. diac sensory endings showing greater complexity
He implied that this concept largely referred to (Marron et al., 1995).
chemoreception (e.g., from gut) and encompassed Overall, visceral afferent sensors (visceros-
taste responses (now not generally considered ensory terminals) appear less specialized and
interoceptive). Present conceptualizations of inter- more “primitive” when compared to soma-
oception recognize multiple sensors (tuned to tosensory and kinaesthetic receptors in skin,
mechanical, chemical, osmotic, and thermal muscle, tendons, and joints (e.g., Ruffini end-
changes) and a range of transduction mechanisms ings, Merkel cells, and Paccinian corpuscles)
(that include free nerve endings, general and spe- and generally cannot convey perceptions of
cialized chemoreception, PEIZO mechanosensi- nuanced touch, pinch, cutting, tickle, or vibra-
tive channels, thermal sensing). tion. Viscerosensory terminals are innervated
Visceromotor signals pertaining to mechani- by thin myelinated (A delta) and unmyelinated
cal function (stretch and compression) of visceral (C) nerve fibres that relay information to brain-
organs (including vessels) are conveyed along stem via prevertebral plexuses and paravertebral
neural pathways (Bernston and Khalsa, 2021), ganglia projections into ascending spinal cord
which also convey peripheral chemosensory infor- tracts (Craig, 2004), or more directly via cranial
mation about luminal contents and tissue integrity. nerves, most importantly though the vagus nerve
The carotid bodies represent one specialist site (Berthoud and Neuhuber, 2000; Jaenig, 2008).
of chemosensation. Chemoreceptive signals are
also sampled directly from circulating blood by
the brain itself, via specialized circumventricular
organs and by specific populations of neuroglia
and neurons (Critchley and Harrison, 2013). CHEMORECEPTION IN THE PERIPHERY

Specialist organs such as the carotid bodies


(innervated by a branch of the glossopharyngeal
cranial nerve) show chemoreceptive sensitivity to
MECHANORECEPTION CO2, O2 and pH levels (e.g., through TWIK-
related acid-sensitive K+ channels; Fagerlund
Many interoceptive sensations have unique quali- et al., 2010) and provide strong interoceptive
ties, distinct from non-visceral somatosensory drive for cardiorespiratory reflex control.
sensations: Feelings of bloating, organ distension, Chemoreceptors are also widely distributed across
breathlessness, and nausea are “visceral” (typi- bodily organs, expressed often on specialized
cally dull or spasmodic and poorly localized). cells (e.g., neuropods projecting into the gut
Even when such sensations intensify to the point lumen) that sample the local chemical environ-
of pain, their origin can remain difficult to localize, ment. Such sensory cells are innervated by vis-
with the exception of referred pain, i.e., where pain ceral afferents ultimately entering the vagus nerve
is perceived at a different location than its origin or spinal cord. Viscerosensory nerves themselves,
and radiating into adjacent regions. Thus, cardiac through “free nerve endings,” can also directly
angina is felt in the left anterior shoulder, medial respond to specific (e.g., glucose, immune, and
upper arm, and throat; pancreatic or renal pain is hormone receptors) and non-specific (pH, osmo-
felt in the back. These referred locations reflect the larity, cytokine) changes in peripheral chemical
recruitment of more localizable somatosensory environment.
Interoception and Thermoreception 417

CENTRAL CHEMORECEPTION or prostaglandin E2 activate microglia through


Toll-like receptors and tumour necrosis factor
The brain directly samples internal states via the release to initiate a cascade of immune reactions in
circulation: changes in the constitution of the brain (Nadeau and Rivest, 2000; cf. Critchley and
blood, including the presence of nutrients, hor- Harrison, 2013). This central response to bodily
mones, immune modulators, and inflammatory infection and inflammation is associated with the
proteins are detected directly by the brain at the triggering of sickness behaviors, a constellation
level of the sensory circumventricular organs: of adaptive allostatic responses affecting moti-
The area postrema (AP), organum vasculosum of vational and emotional state, attenuating social
the lamina terminalae (OVLT), and subfornical engagement and reallocating the energy utilization
organ (SFO) are located in the walls of the third across the body in a sustained reaction to internal
and fourth cerebral ventricles and consist of per- interoceptive threat.
meable fenestrated capillary tissue within perivas-
cular spaces. The tissue surfaces express multiple
chemoreceptor types, sensitive to systemic
osmotic, metabolic, and inflammatory status. DETECTING TEMPERATURE
There is a degree of specialization: The AP is
linked to feeding behavior (including vomiting), The regulation of bodily temperature is crucial to
satiety and hunger (Hindmarch et al., 2011) health and wellbeing, ensuring efficient skeletomo-
though reciprocal connections with the Nucleus of tor, neural, and organ function. Core body tempera-
the Solitary Tract (NTS) and hypothalamus. The ture is tightly controlled within a narrow range
OVLT has intrinsic osmosensory properties (Ciura through homeostatic reflexes and behaviors.
et al., 2011) and corresponding coupling to hypo- Elevated temperature does occur in allostatic sick-
thalamic supraoptic nucleus (SON) vasopressin ness states but arguably as a compromise that car-
release. The SFO appears to have a broader role in ries significant risk. Temperature sensing is thus a
viscerosensing (reviewed by Critchley and particular important form of interoception
Harrison, 2013). (Crucanelli et al., 2022). Both peripheral and
Many circulating molecules also cross the blood central temperature sensing occur and are linked to
brain barrier (either actively or passively) to stimu- local and systemic physiological reflex mecha-
late chemoreceptors on neurons and glia. NTS neu- nisms for temperature regulation (increasing or
rons express receptors for Angiotensin II and the decreasing skin blood flow, sweat production, and
satiety factor GLP-1, while neurons in hypothalamic thermogenic shivering or metabolism). In addition,
nuclei respond to hormones, vascular and immune temperature sensing drives adaptive behavioral
mediators, and to glucose levels. Glucose-sensing responses. Temperature in the periphery fluctuates
in hypothalamus is linked to feeding (lateral arcu- more directly with the environment, and changes
ate nucleus) and satiety (ventromedial hypothala- are sensed by hot and cold sensitive neurons
mus) (Oomura et al., 1969). Glucose sensing also located mainly in the skin with cell bodies in the
occurs in other brain regions, e.g., medial amyg- spinal dorsal root ganglia (body) or trigeminal gan-
dala nucleus and in monoaminergic (dopamine and glion (head, face). Cold neurons respond to cooling
noradrenaline) neurons expressing ATP-sensitive through activation of the Transient Receptor
potassium channels (Dunn-Meynell et al., 1998). In Potential Melastantin 8 channel (TRPM8; also
fact, glucose sensing may be an intrinsic feature of sensitive to menthol). Warmth sensitivity is medi-
the brain reward system (Koekkoek et al., 2017), ated by neurons expressing Transient Receptor
wherein motivational learning is grounded on Potential Vanilloid 1 (TRPV1), TRPM2 and per-
metabolic homeostatic foundations. Interoceptive haps other related TRPV and TRPM receptors.
signaling regarding the availability and allocation While the bare skin of face, hands, and feet enables
of energy resources is fundamental to shaping moti- tactile discrimination of the temperature of external
vational behavior, in part through feeling of need, objects, hairy skin elsewhere is important for
want, and pleasure. This is expressed at multiple interoceptive thermoregulatory control. There is
levels in the relationship between interoceptive also some peripheral temperature sensing that
signaling pathways and the brain circuitry encod- occurs in visceral organs and within the spinal cord
ing reward and punishment, motivation drive and itself (Ran et al. 2016). Thermosensory input from
satiety and the selection of behaviors through rein- the periphery terminates in the brain, at the level of
forcement learning. the preoptic area (POA) of the hypothalamus.
Microglia are also important to immune aspects Importantly, this region also contains neurons that
of interoception, continuously sampling the are directly sensitive to brain (hence systemic
brain’s extracellular space. Circulating cytokines bodily) temperature (Tan and Knight, 2018).
418 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

INTEROCEPTIVE ANATOMY with correlated “quasi-interoceptive” somatosen-


sory information e.g., regarding chest movement,
pharyngeal airflow, cardiac percussion of chest, or
Pathways to Brain skin pulsation. Such sensations may be appraised
consciously with greater spatial and temporal pre-
The unmyelinated (C fibre) or thin (Að fibre)
cision. Moreover, when spinal interoceptive sign-
sensory neurons that innervate the vasculature and
aling intensifies (e.g., when organs are damaged,
viscera travel to the brain via cranial nerves
visceral pain may be referred, or perceived as an
(mostly via vagus and glossopharyngeal nerves)
exteroceptive somatosensory sensation at a loca-
and via afferents that ascend the spinal cord
tion different to its origin): Thus, pain around the
(Jaenig, 2008) The latter track back in
left neck, anterior shoulder, and medial upper arm
post-ganglionic sympathetic nerves to preverte-
may signal cardiac angina; pain in the back may
bral plexuses and paravertebral ganglia projec-
signal pancreatic and renal damage (Gebhart and
tions into the spinal cord. Autonomic spinal
Bielefeld, 2016).
reflexes provide some proximate homeostatic
control; yet, the consequences of such reflexes
may also be systemic, for example in dysreflexic
responses to tissue injury or bladder filling in
patients with spinal cord transection (Rabchevsky,
BRAIN ANATOMY OF INTEROCEPTION
2006). Voiding and sexual functions also depend
on visceral afferent feedback into spinal reflexes, AND THERMORECEPTION
processes that are under descending control.
Within the spinal cord, cell bodies lie in dorsal Within the medulla, the Nucleus of the Solitary
root ganglia and axons ascend both ipsilaterally Tract (NTS) receives ascending visceral afferent
and contralaterally spinothalamic tracts. Spinal inputs form spinal neurons with cell bodies in the
Lamina 1, inputting into the dorsal spinothalamic dorsal root ganglion and from cranial nerves,
tract, conveys temperature and pain sensations. notably the vagus and glossopharyngeal nerves.
This is argued to be a specialized channel dedicated The NTS is a primary site of anatomical conver-
to carrying motivationally salient viscerosensory gence of gustatory, gastrointestinal, cardiovascu-
information that ultimately give rise to affectively lar, hepatic, renal, respiratory, and immune system
laden sensations (Craig, 2004, 2014). Slow, light, and is critically important to homeostasis; its
repeated mechanical tactile stimulation and warm destruction is lethal (Doba and Reis, 1973). The
skin sensations, ultimately giving rise to positive NTS supports specific functions dependent upon
feelings (e.g., “sensual touch”) also are carried this primary viscerosensory role. These include
by unmyelinated fibres originating within cuta- the coordination of simple autonomic reflexes
neous and mucosal epithelia. This information generating respiratory and cardiovascular rhythms
is purportedly also relayed via spinal Laminar 1 and peristalsis of the upper gastrointestinal tract.
(Craig, 2004, 2014), though ablation of this tract In addition, it relays signals of taste, satiety, and
does not necessarily dispute affective aspects of visceral pain (Boscan et al., 2001; Cutsforth-
touch (Marshall et al., 2019). Information about Gregory and Bennarroch 2017). The NTS also
bodily temperature, from both skin and internal contributes to the coordination of stress responses,
organs, also reaches Laminar 1. This tract projects via inputs that modulate the homeostatic auto-
strongly into “homeostatic” brainstem centres in nomic rhythms (e.g., generating a state of cardio-
the medulla, notably the Nucleus of the Solitary vascular arousal through baroreflex suppression
Tract (NTS) and onto parabrachial nucleus and allowing blood pressure and heart rate to rise
thalamus. The vagus (in which over 80% of fibres together to facilitate action-related behaviors), and
are afferent) and glossopharyngeal (innervating through efferent modulation of hypothalamic-
the carotid sinus and body) nerves also project into pituitary-adrenal (HPA) axis activity (correspond-
the NTS, the “classical visceral receiving area” ingly NTS neurons express glucocorticoid
of the brainstem (Bethoud and Neuhuber, 2000; receptors) (Gosal et al., 2014).
Jaenig, 2008). There is broad rostrocaudal viscerotopic
While much of interoceptive signaling is covert, organization, yet NTS neurons with distinct vis-
a number of interoceptive sensations (e.g., those ceroceptor input (e.g., arterial baroreceptors or
relating to voiding and breathing) are commonly gastrointestinal chemoreceptors) often lie in close
perceived. Feelings of bloating, organ distension, proximity (Paton et al., 1999) whereby crossmodal
breathlessness, and nausea are also uniquely vis- integration may occur. Interoceptive information
ceral, hence are poorly localized. Such sensations from the NTS projects through ventroposterior
are likely to be conveyed via the spinal route along medial thalamus to cortex, branching to synapse
Interoception and Thermoreception 419

within pons and midbrain (e.g., parabrachial Critchley et al., 2004). Orbitofrontal and anterior
nucleus, locus coeruleus, hypothalamus) to influ- cingulate cortices, amygdala and hippocampus
ence ascending and descending neuromodulatory receive viscerosensory information both directly
pathways and subcortical substrates of motiva- (mostly from insular projections) and indirectly
tional behavior. At this level also, humoral infor- (e.g., via subcortical and alternative dorsome-
mation, sampled from the systemic circulation by dial thalamic branch and via the subcortical and
the brain, is integrated with other viscerosensory neuromodulatory pathways).
signals to influence the neurosecretory regulation
of fluid balance and hormonal status, and shape
descending autonomic control of the cardiovas-
cular system, digestive tract, and genitourinary
HUMORAL AND THERMOSENSORY
organs.
The NTS proximally supports autonomic INFORMATION
reflexes through output to brainstem centres con-
trolling autonomic function. In the baroreflex, for Wide-reaching connections from the circumven-
example, the phasic input from arterial (aortic tricular organs enable information concerning
arch, carotid body) baroreceptors enhances para- humoral physiology (i.e., systemic metabolic
sympathetic output to the heart via stimulation of inflammatory or osmotic status) to integrate with
the dorsal motor nucleus of the vagus to reduce other interoceptive representations). Much of this
heartrate (Benarroch, 2008). These baroreceptor is achieved at the level of the hypothalamus,
signals also inhibit efferent sympathetic outflow which, as noted, also contains the brain’s “ther-
at the level of the rostral ventrolateral medulla mometer” (POA). The contribution of the AP to
(RVLM) reducing vasoconstriction within mus- cardiovascular regulation and immune reactivity
cle vascular beds. This mechanism permits blood is also achieved through close reciprocal connec-
pressure regulation at a beat-by-beat level, yet is tions with the NTS, parabrachial nucleus, sympa-
further modulated by multiple other viscerosen- thetic medullary nuclei, and the parasympathetic
sory inputs (nociceptors, pH, CO2, O2, lactate, dorsal motor nucleus and nucleus ambiguus of the
hormones, prostaglandins, cytokines, and temper- vagus (van der Kooy and Koda 1983). The OVLT
ature) via GABAergic action on the second order projects to the supraoptic nucleus of the hypo-
NTS neurons that project to the rostral ventrolat- thalamus to support osmoregulation, yet it also
eral medulla. projects to NTS, parabrachial nucleus and other
The NTS has close bidirectional connections regions of hypothalamus. Moreover, its projection
with the parabrachial nucleus, periaqueductal gray to midline thalamus, and onto insula and cingulate
matter, hypothalamus, and bed nucleus of the stria cortices, is proposed to underpin the motivational
terminalis. These centers, in turn, link function- feeling of thirst.
ally to forebrain regions, notably amygdala, adja- Thermal information about body tempera-
cent basal ganglia, and insula cortex, cingulate, ture from peripheral tissues (notably the skin,
hippocampus, and prefrontal cortices (Benarroch, Crucianelli et al., 2022) projects from both lami-
1993; Blessing, 1997). The flow of interoceptive nar 1 and classical somatosensory spinothalamic
information to these regions is paralleled by neu- tract into the NTS, lateral parabrachial and thal-
romodulator projections (notably cholinergic and amus. Parabrachial lesions abolish autonomic
monoaminergic) originating in adjacent brainstem responses to skin warming and cooling. Thus, the
and midbrain nuclei (Bernston et al., 2003) the parabrachial nucleus is important for thermoregu-
likely means by which arousal, attention, and moti- latory responses to environmental temperature
vation are coupled to internal state. The Lamina 1 (Nakamura and Morrison, 2008). At the cortical
spinothalamic tract, along with the cranial nerve level, peripheral temperature is represented in
visceral afferent input into NTS, projects via the posterior insular (Craig et al., 2000) and soma-
ventroposterior thalamus into posterior and dorsal tosensory cortices. Interestingly, lesions to this cor-
mid insular cortices. The dorsal insula is viewed ticothalamic pathway have only a limited impact
as the primary viscerosensory cortex; represent- on thermoregulatory responses. The projection of
ing internal bodily sensations (Craig, 2002, 2014). temperature information from lateral parabrachial
The re-representation of interoceptive information nucleus to the POA in midline hypothalamus is
in more anterior insular regions, and its integration more salient: The intrinsically thermosensitive
with contextual information from exteroceptive neurons in POA, when heated, initiate reflexes
perceptions, mnemonic and contextual informa- and behaviors to promote heat loss. Conversely,
tion is linked to conscious awareness of internal cooling the POA triggers systemic physiological
physiological state, and associated interoceptive and behavioral responses to preserve and gener-
motivational and emotional feelings (Craig, 2002; ate body heat. Thus, POA appears to be critical to
420 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

integrating central and peripheral thermosensory PSYCHOLOGICAL REPRESENTATIONS OF


information for temperature regulation (Tan and INTEROCEPTIVE INFORMATION
Knight, 2016).
Human neuroimaging studies, together with
lesion studies and stimulation studies, have Interoceptive information is fundamental for life,
mapped neural correlates of peripheral arousal driving organ- and system-level homeostatic
and interoceptive processing, often coupled to reflexes and organism-level coordination of
autonomic control, to brain regions throughout the allostatic responses. The latter encompasses moti-
neuraxis (Beissner et al., 2013). These findings are vational behaviors through to relatively simple
consistent with information from animal physiol- affective feelings and primordial emotional states
ogy, identifying brainstem, midbrain, and thalamic such as hunger, thirst, warmth- or cold-seeking to
relays of detailed viscerosensory/interoceptive more elaborate levels of emotional affiliative,
information to insular cortex and related regions, social, emotional, and cognitive experiences that
including forebrain components of the reward sys- are coupled to action decisions and adaptive behav-
tem including amygdala and anterior hippocam- ioral policies. Thus, at multiple levels, mental pro-
pus, orbitofrontal cortices, and both dorsal and cesses are influenced by interoceptive signals
subgenual anterior cingulate cortex. Co-activation representing physiological integrity and need.
of, and interaction between, visceromotor dorsal Perhaps interoception’s biggest influence on
anterior cingulate cortex and viscerosensory insula psychological processes occur implicitly, without
have been framed in terms of the detection of the need for conscious appraisal: Fluctuating inter-
“interoceptive surprise.” Here, insula is proposed oceptive states (e.g., changing blood glucose level,
to compare the afferent (interoceptive) informa- osmolality, bodily temperature) re-orientate moti-
tion about current bodily states with the efference vations and entrain behavioral routines (including
copies of descending visceromotor signals from food-seeking and voiding) before even sensations
anterior cingulate cortex (predictions). The latter of early hunger, thirst, satiety, and arousal are per-
typically drive sympathetically dominant allo- ceived. These preconscious signals also dictate
static states of cardiorespiratory arousal, linked to dominant sensory and behavioral interests (e.g.,
the facilitation of motor action (Critchley, 2005, in food, action, seeking shelter, clothing, or hugs)
2009). Neuroimaging and lesions studies further and set the state of self-efficacy, motivational drive
support the role of insular cortex as a basis for and capacity to learn. There are multiple examples
conscious viscerosensory representations that are of this, but rarely confined to discrete organ spe-
elaborated as motivational and emotional feelings cific level. Blood pressure fluctuations can gate
(Craig, 2004, 2014; Critchley, 2005; Critchley pain processing (Gray et al., 2009), the signaling
et al., 2004). of cardiovascular arousal enhances threat percep-
Brain-wise associations between interregional tion (Garfinkel and Critchley, 2016; Garfinkel
functional connectivity, inferred from enhanced et al., 2001). Psychological “repertoires,” includ-
correlations in haemodynamic signatures of neu- ing the drop in mood, decreased social interest,
ral activity (and also electrocortical or magnetoen- and anorexia can be elicited by low-grade inter-
cephalographic neurophysiological measures) oceptive immune challenges, (Harrison et al.,
indicate distribution of interoceptive informa- 2009) influence and bias foreground sensorimotor
tion across brain networks (Jarrahi and Kollias, functions. Physiological arousal states influence
2020). Some, for example related to electrogas- cognitive alertness and performance. Such pre-
tric activity (Rebollo et al., 2018), may represent perceptual interoceptive influences are revealed
independent functional brain networks that can experimentally in the interaction between pha-
be distinct from the acknowledged association sic physiology signals (e.g., heartbeats, respira-
between salience/executive attentional networks tion, gastric motility) or more tonic physiological
or the default mode network and states of high states (glycaemia, cytokine response) and/or their
and low peripheral arousal. Electrocortical and neural representation (e.g., as heartbeat evoked
magnetocortical correlates of heartbeats (heart- potentials) and with perceptual sensitivity to
beat evoked potential, HEP, and heartbeat evoked exteroceptive stimuli or changing cognitive or
response; HER ) present specific challenges with emotional responses (Garfinkel and Critchley,
respect to artefact removal and interpretation. 2016; Garfinkel et al., 2014; Saloman et al., 2018).
Nevertheless, these indices similarly suggest that Such theoretical notions were earlier formalized
interoceptive information concerning heartbeats in the somatic marker hypothesis (Bechara et al.,
(potentially driven by the activation of fast arterial 2005). More broadly, the continuous background
baroreceptors) has a broad reach over frontotem- hum of interoceptive activity interacts with senso-
poral and insular cortices (Luft and Battacharya, rimotor functions across all domains. Whether or
2015; Park et al., 2018). not this is definitively or explicitly perceived as
Interoception and Thermoreception 421

an internal bodily sensation, the implicit impact people’s insight (“metacognitive” awareness) into
of interoceptive signaling and the relentlessness their own interoceptive ability (Garfinkel and
of dynamic interoceptive control is proposed to Critchley, 2013, Garfinkel et al., 2015; Quadt
be fundamental to the sense of “self” as a unitary et al., 2018). These approaches were formulated
actor and observer navigating and interacting with into a three-level categorization of psychological
the external world (Craig, 2014; Critchley, 2005; aspects of interoception that attempted to over-
Damasio, 1999, 2010; Seth et al., 2012; Tallon- come previous discrepancies in this research field
Baudry et al., 2018). that clouded inference about interoception’s
Clearly, however, many interoceptive sensa- impact on emotion and cognition (although note
tions are consciously perceived, but usually with- novel iterations of this approach, e.g., Suksasilp
out fine spatiotemporal precision. The role of these and Garfinkel, 2022).
sensations as the basis for motivational behaviors As an easily identifiable interoceptive stimulus,
often conflates afferent representation with affec- the heartbeat has been the mainstay of “objec-
tive drive. Patterns of interoceptive response are tive” tasks of interoceptive sensitivity. Heartbeat
important; the sensation of hunger goes beyond detection tasks seek to quantify an individual’s
feelings of stomach emptiness. In their influential ability to perceive of their own heartbeats at rest,
theory of emotion, James (1884) and (especially) by counting, tapping, or by judging heartbeat tim-
Lange (1907) proposed the perception of bod- ing relative to an external stimulus. Many of these
ily changes, often triggered automatically by the tasks have long-recognized psychometric limita-
“emotive stimulus,” is the basis of emotional feel- tions, particularly where prior knowledge influ-
ings that “color” the otherwise cold perception ences the performance of the “heartbeat counting
of the stimulus with a value-laden sensation. The task” (Ring and Brener, 1996). Concerns about
conscious experience of interoceptive sensations the “two-interval heartbeat discrimination task”
as emotional feelings is commonly enhanced by are more about task difficulty (most people expe-
correlated somatosensory signals (e.g., the feel rience heartbeats at around the same time within
of the skin’s piloerection, shivering or sweating the cardiac cycle, Betka et al., 2021; Wiens and
accompanies interoceptive signaling of tempera- Palmer, 2001) (see Figure 26.1). Although they
ture regulation and fear) including more obvious may often conflate interoceptive beliefs with
feedback from emotion-triggered behaviors (like interoceptive perception, heartbeat detection tasks
running from a bear). remain widely used for reasons of practicality,
and inferences are necessarily constrained and
contextualized. Newer related heartbeat tasks are
emerging that attempt to improve on the objec-
tive validity of the measure of interoceptive per-
LEVELS OF INTEROCEPTIVE PERCEPTION formance accuracy (Betka et al., 2021, Larsson
et al., 2021), and beliefs (Legrand et al., 2021).
The proposal that interoceptive signaling may be There are similar tests for gastrointestinal and res-
an important component of emotional processing piratory interoceptive sensitivity (Garfinkel et al.,
found some qualified support in the observed 2016), but there is a dearth of methods to measure
effects of adrenaline injections (Cantril and Hunt, objectively other aspects of interoceptive percep-
1932; Marañon, 1924). The proposal also moti- tion, including thermal sensitivity.
vated research to quantify individual differences The partitioning of interoception along three
in sensitivity to internal bodily changes. If people psychological dimensions, as described above,
vary in the degree to which they are more or less represented a leap forward for interoception
sensitive to internal bodily changes, they may research (Garfinkel and Critchley, 2013; Garfinkel
consequently feel emotion with lesser or greater et al., 2015; Quadt et al., 2018; see also Table 26.1).
intensity. By extension, this may have clinical This framework distinguished between: (1) objec-
relevance in that they may be more prone to anxi- tive measures of interoception (i.e. interoceptive
ety and panic, may somatize emotional experi- performance (e.g., behavioral score on a heartbeat
ences more, or conversely may be better at detection task); (2) subjective measures of intero-
self-regulating their state of emotional arousal. ception (how well a person thinks they perceive
Approaches to quantify individual differences in interoceptive signals (e.g., self-rated confidence
interoceptive sensitivity have centered on: on heartbeat detection, or score on self-reported
(1) performance measures in tasks that ask people questionnaires); and (3) metacognitive intero-
to judge “objectively” measured changes in their ceptive awareness insight (e.g., correspondence
body; (2) self-ratings (e.g., questionnaires) of between actual objective performance and subjec-
judged sensitivity or confidence in their experi- tive sense of interoceptive ability). These psycho-
ence of bodily sensations; (3) examination of logical dimensions of interoception are typically
422 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 26.1 When and where people perceive heartbeats


A multi-interval heartbeat task is a proposed alternative means of measuring interoceptive
performance accuracy (sensitivity to feeling one’s heart beating at rest) that overcomes
psychometric limitations of heartbeat counting and 2-interval heartbeat discrimination task
(Ring and Brener 1996; Wiens and Palmer 2001). Here, Betka et al. (2021) asked healthy par-
ticipants to match the timing of when they felt their heartbeat to external tones presented
at different time intervals relative to the electrocardiogram (ECG) R-wave (measuring the
myoelectric trigger of ventricular contraction). Participants then indicated where on their
body they felt their heart beating.
A. There was consistency in the timing of heartbeat perception; most heartbeats were per-
ceived between 150 and 350 ms (peak approximately 250ms after the ECG R-wave) by the
majority of participants.
B. There was less consistency in the reported site of heartbeat perception; nevertheless,
the most common location endorsed by participants was the left side front of the chest.
Interestingly there was no systematic correspondence between when and where people
reported heartbeat sensations.
Source: From data published in Betka et al., 2021.

dissociable from each other. Moreover, the degree 2016; Garfinkel et al., 2014, 2020). The model
to which subjective and objective dimensions align also can be extended beyond metacognition, to
can predict emotion states and affective psycho- encompass executive processes, including flexible
pathology (Garfinkel et al., 2015). An extended switching of interoceptive attention (Table 26.1).
dimensional model of interoception incorporates The impact of phasic interoceptive signals on
lower-level measures of visceral afferent neural emotional processes is perhaps best described
traffic (to quantify afferent signal strength objec- for the cardiovascular channel, though, as noted
tively (e.g., through heartbeat evoked potentials above, respiratory and gastric signals modulate
(Gray et al., 2007), and the preconscious impact brain responses and associated cognition and
of interoceptive signals on sensory processing perception. Cardiovascular arousal is signaled by
(e.g., modification of startle response (Schulz arterial baroreceptors in aorta and carotids that
et al., 2017) or augmentation of threat processing fire with each heartbeat as ventricular contraction
(Critchley et al., 2002; Garfinkel and Critchley, pumps blood out of the heart. Thus, baroreceptors
Interoception and Thermoreception 423

Table 26.1 Dimensions of interoception


Dimension Level Measurement Example

Metacognition Execution/Attribution Metacognitive Attention modulation/ Attributing aversive


execution/ attribution of outcomes to heart
attribution bodily signals pounding
Interoceptive Metacognitive Insight into Confidence-accuracy
awareness accuracy performance correspondence
aptitude
Behaviour Interoceptive Subjective belief Self-report Questionnaires
sensibility
Interoceptive accuracy Behavioral Performance on Heartbeat detection
performance behavioural tasks tasks
Brain/Body Preconscious impact Brain-body interaction Degree of impact of Cardiac modulation
of afferent signal signal of fear
Afferent signal Interoceptive signal Strength of signal Heartbeat evoked
potentials

signal the timing and strength of each heartbeat INTEROCEPTIVE DIMENSIONS AND
to brainstem via the vagus and glossopharyngeal CLINICAL SYMPTOMS
nerves and inform the reflexive control of blood
pressure through the baroreflex. In states of car-
diovascular arousal (including emotional stress), Different aspects of interoception interact in how
the baroreflex is suppressed, allowing heart rate they shape emotional states and behaviors. The
and blood pressure to rise together. The impact impact of phasic cardiac signals may be tempered
of this channel of interoceptive information on by interoceptive accuracy (i.e., the degree to
brain processes can be assessed by exploiting the which an individual can perform a heartbeat
phasic nature of baroreceptor firing. Responses to detection task), mitigating the deleterious impact
brief stimuli presented around systole, when the of bodily arousal signals on memory (Pfeifer
baroreceptors are active, can be contrasted with et al., 2017) and enhancing the processing of self-
responses to stimuli presented at diastole, when related signals (Seth and Tsakiris, 2018). Good
the baroreceptors are quiescent. Differences are interoceptive accuracy is also generally associated
thus attributable to the presence, versus absence, with better affective regulation. Positive shifts in
of cardiac interoceptive signals. Historically, interoceptive accuracy induced by contemplative
such experiments highlight the inhibitory nature training are associated with improved capacity to
of cardiac afferent signals. Interoceptive inhi- verbalize emotional states (decreased alexithy-
bition is also apparent in suppression of startle mia). Disruption of interoceptive signaling, accu-
responses and attenuation of memory for words racy, and subjective sensibility are observed in
encoded at systole (Garfinkel et al., 2013; Schulz clinical disorders that affect self-focused emo-
et al., 2009). However, these baroreceptor signals tional control (Bonaz et al., 2018; Khalsa et al.,
amplify threat processing, enhancing the detec- 2018). Moreover, the alignment of subjective and
tion and perception of fear and threat (Garfinkel subjective components of emotion appears to pre-
and Critchley, 2016; Garfinkel et al., 2014, dict emergence of affective symptoms. For exam-
2020). Importantly also, when it comes to the ple, in autistic people, the degree of “mismatch”
learning and recollection of bodily threat, signals between perceived sensitivity to interoceptive
pertaining to cardiovascular arousal can act both state and observed accuracy in performing an
as an amplifier of the threat (that may general- interoceptive task (heartbeat detection) predicts
ize to related stimuli), and as a context, such that anxiety symptoms and deficits in interpersonal
threats learned in a state of high or low cardio- emotional interaction (Garfinkel et al., 2016).
vascular arousal will be preferentially evoked Beyond anxiety, interoceptive dysfunction
in the same physiological context (Garfinkel expressed differentially across different interocep-
et al., 2020). This dual interoceptive influence tive levels (and organ systems) is implicated in the
may have relevance in the expression of clinical expression of a wider range of clinical disorders,
symptoms and the shaping of perceptions that from dysregulation of metabolism, immune
underpin “better safe than sorry” behavioral pat- responses or respiration, dysautonomia, to func-
terns (Van den Bergh et al., 2021). tional neurological symptoms (see below).
424 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

COMPUTATIONAL MODELING IN Recent computational models of interoception


INTEROCEPTIVE SCIENCE extend more general principles of computational
neuroscience applied to sensation and perception,
motivational learning and sensorimotor control,
The control of the internal physiological state of albeit within the unique and varied properties
the body is achieved through the regulation of of interoceptive signaling. An early assumption
interoception. Organ-level homeostatic reflexes is the hierarchical nature of interoception and
are enacted through sensory control loops, and its control: Afferent (sensory) signals from vis-
coordinated across organs at a system level. This ceral organs support local and spinal autonomic
higher-order coordination is shaped and allo- reflexes and hypothalamus/brainstem-level
statically adjusted by purposeful action-selection autonomic and neuroendocrine reactions within
and the pursuit of longer-term behavioral strate- “hard-wired” negative neural and humoral feed-
gies. Fundamental homeostatic processes, such back loops. The set-points of these are neverthe-
as the baroreflex, cardiorespiratory control, and less subject to top-down modulation to engage
the regulation of bodily temperature, are inher- these dynamic adjustments in support of allo-
ently complex, even before one considers how static policies that ensure future stability through
physiology might anticipate prospective internal change. These polices (sequences of responses or
and external challenges. Computational mode- actions) take place at a higher, more integrated,
ling seeks to provide deeper insight into the level of representation, for example representing
observable data relating to interoceptive signal- the organism as a whole, rather than a single organ
ing and its influence upon motivational, behav- system. Here, both the reflex set-point and the
ioral, psychological, and physical processes and higher-order policy reveal an internal (generative
their dysfunction (Allen et al., 2019; Petzschner or inverse) model, i.e., a representation of what
et al., 2021; Smith et al., 2017). Complementing the internal state ought to be. More generally, it is
experimental and clinical observations, compu- proposed that the brain makes sense of (typically
tational modeling is useful in systematically noisy) sensory information by comparison with an
organizing and testing the feasibility of differ- internal model that can be viewed as an expecta-
ent, and potentially competing, mechanistic tion or prediction of the source of the sensation.
hypotheses and making predictions about As applied to interoception, this is a prediction
responses at multiple levels. There is the poten- of the desired internal physiological state. Many
tial for model parameters (which may otherwise current computational neuroscience approaches
not be measurable) to reveal hidden or emergent to perception and sensorimotor control imple-
mechanisms and “computational biomarkers” to ment Bayesian statistical principles of inference
support diagnosis or treatment represents one as predictive coding: Here, afferent information
aim of computational models of interoception. is evaluated against a priori sensory predictions
The mathematical modeling of physiological (expectation or beliefs) to generate prediction
processes has a rich history. Interoceptive relevant errors (sensory surprise). The system as a whole
examples include detailed simulation of cardiopul- is conceptualized as a hierarchically, with predic-
monary interaction bringing together biophysical tion errors (initially from low-level reflex loops)
aspects of respiration with those of blood circu- ascending against descending higher-level pre-
lation, the modeling of the baroreflex as a func- dictions) primed to diminish prediction errors
tional arc or broken down into the afferent control by either adjusting the prediction itself (through
system interacting with the pulsating heart allows learning) or changing the sensory signal (through
for modeling of complex processes at different action) (Petzschner et al., 2017; Pezzulo et al.,
levels of description while always providing a 2015; Seth and Critchley, 2013).
simplification to retain and highlight fundamental Physiological control through interoception
mechanistic insight. Assumptions, conditions and is fundamental to survival, hence the updat-
constraints need to be broadly correct at a work- ing of interoceptive predictions often equates to
able level of complexity for the question in hand. reinforcement learning of reward/punishment.
These can range from simplified models of neural Alongside internal efferent autonomic responses,
firing such as “leaky integrate and fire” (Mahdi actions that change interoceptive predictions
et al., 2019), vascular viscoelasticity (Raghu include appetitive and aversive motivational
et al., 2011), or the passive thermal properties behaviors (e.g., feeding, warmth-seeking, social
of cylindrical body segments in heat convection affiliation) behaviors. Both can be viewed as
and conduction (Katic et al., 2016). The accu- interoceptive active inference; a process that sup-
rate simulation of physiological control process ports allostatic adaptation: Allostatic (autonomic,
may valuably be used to understand higher-order behavioral) policies enable short-term adjust-
mind–body interaction (Van Roon et al., 2004). ment of homeostatic reflex set-points in order to
Interoception and Thermoreception 425

Figure 26.2 Interoceptive inference


A. This panel shows a model of the dynamics of active interoceptive inference in health.
Predictions (“P”) about interoceptive states are passed down via backward connections and
compared against actual interoceptive input. Mismatches between predicted and actual
states lead to the generation of prediction errors (“PE”). In order to inhibit autonomic
control, ascending error signals are attenuated so not to change predictions. When, in turn,
prediction errors are quashed by autonomic control, changed interoceptive input if fed
forward to inform future predictions.
B. A model depicting the dynamics of faulty interoceptive inference in the case of depression
or chronic fatigue. The over-prediction of metabolic energy demands leads to a downregu-
lation of noisy error signals, thus inhibiting the possibility of correcting these faulty
predictions, and resulting in a “locked-in” state that perpetuates this cycle.
Source: Adapted from Quadt et al., 2019.

permit longer-term maintenance of integrity of the computational neuroscience methods to intero-


whole organism. This survival-related primacy of ception can help advance clinical understanding
interoception means that interoceptive prediction and design of interventions relevant to biopsycho-
likely underpins cognition, emotion and even the social determinants of physical and mental health
conscious continuity and unitary sense of self- outcomes (Petzschner et al., 2017).
hood. A higher-level interoceptive representation
can also support an integrated prediction about the
effectiveness of coordinated interoceptive control,
and, correspondingly, self-efficacy (i.e., the capac-
ity to engage in physiological and behavioral acts
DISORDERS OF INTEROCEPTION AND
of self-regulation). This metacognitive index of THERMOREGULATION
allostatic self-efficacy is a proposed basis to trig-
gered states of fatigue and depression (Stephan While basic interoceptive functioning is essential
et al., 2016), illustrating how the application of for homeostasis (and is hence a prerequisite for
426 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

life), the abnormal integration and control of account for non-cardiac chest pains and symptom
interoceptive processes can manifest clinically maintenance in other chronic medical conditions.
across psychological and physical conditions. A structural framework to consider how differ-
Disordered interoception is increasingly impli- ent levels of interoceptive processing contrib-
cated in the pathophysiology and expression of ute to symptom-expression across mental health
symptoms of psychiatric, neurodegenerative, and related disorders is needed. Computational
developmental, and neurological conditions approaches may be usefully leveraged to param-
(Bonaz et al., 2021). Problems may arise at dis- eterize, and hence quantify, such contributions in
tinct levels of interoceptive processing, for exam- order to plan and tailor specific interventions, and
ple in peripheral signaling (e.g., neuropathies; also to gain a richer insight into how psychoso-
following vagotomy; spinal cord transection), in cial factors influence physical and mental health
the central discriminative representation, sensory outcomes.
gating and impact on other sensory processes While interoceptive influences are brain-wide,
(e.g., pain), and in the prediction, interpretation, central neurological brain disorders may specifi-
and general awareness of sensations from the cally impact interoceptive pathways or hubs. For
internal body. Fundamental problems with intero- example, epilepsies involving insular cortex may
ception may give rise to metabolic disturbances give rise to interoceptive sensations and feelings
and dysautonomia control of the body, often (including ecstatic experiences (Picard and Craig,
co-occuring and interacting. 2009)) and compromise allostatic control of car-
Reflecting core contributions to motivation diac function to jeopardize health through arrhyth-
and emotion, interoceptive dysfunction is increas- mia and/or sudden death (Cheung and Hachinski,
ingly considered in relation to psychological and 2000). Neurodegenerative disorders can also
psychiatric disorders, particularly where there is impact the central processing and representation
disruption in motivated behavior, often accom- of interoceptive information. In so doing, these
panied by affective symptoms (Khalsa et al., may shed a light more generally on interoceptive
2018, Critchley et al., 2023). Eating disorders mechanism in pathology (Garcia-Cordero et al.,
(Jenkinson et al., 2018) and drug addiction (Paulus 2016). Amotivational and fatigue states, flattened
and Stewart, 2014) are representative expressions affective reactivity, and reduction in drug-crav-
of aberrant processing of embodied motivational ing are reported alongside reduced interoceptive
signals (e.g., hunger, satiety, craving, withdrawal). sensitivity to bodily signals following lesions to
Interoceptive signaling of exaggerated states of insula, ventral prefrontal, and cingulate cortices
physiological arousal is a prominent feature of (Darby et al., 2018; Fuchs et al., 2019). Such
anxiety disorders, including Panic Disorder, and symptoms occur also in dementia, where loss of
Post Traumatic Stress Disorder (PTSD), and often self-awareness may be prominent in later stages
there is a more general and over-focusing on such (Mograbi et al., 2021).
signals (Adams et al., 2022). Depression is often Thermoregulatory disorders represent an inter-
accompanied by a flattening of interoceptive esting expression of interoceptive dysfunction.
processing (e.g., Eggart et al., 2019) (potentially Mostly, thermoregulatory problems arise from the
leading to feelings of emptiness and hollowness) inability to lose or retain heat as an expression of
while dissociative states (e.g., depersonalization autonomic problems and/or extreme behavioral
and derealization, often arising clinically as a con- and environmental challenges, rather than from
sequence of extreme anxiety, suggest perturbation primary deficits in the interoceptive sensing of
in interoceptive coherence, such that the under- temperature (Cheshire, 2016). Nevertheless, small
lying “biological” self-representation diverges fibre neuropathies (e.g., with diabetes) may not
from metacognitive appraisal (Khalsa et al., 2018; only impact (efferent) sweat gland function but
Table 26.2). Interoceptive perceptual deficits are lead to abnormal temperature signaling (including
reported in the context of schizophrenia (Ardizzi sensations of burning) through compromised vis-
et al., 2016) yet metacognitive interoceptive defi- cerosensory and cutaneous sensory fibres (Flores
cits appear better able to explain perturbations et al., 2015). Central neurological disorders may
of self-representation intrinsic to the dissociative also impair thermosensation: Examples include
symptoms of psychosis (Garfinkel et al., 2018). demyelination in multiple sclerosis (fatigue in
The dysregulation of interoceptive control multiple sclerosis is associated with raised central/
features in stress-sensitive medical conditions, brain temperature) and dementias. Both cold and
including irritable bowel syndrome, fibromy- heat insensitivity are reported. Certain drugs, not
algia, chronic pelvic pain, and ME/CFS that are least monoaminergic-acting psychomotor stimu-
often accompanied by dysautonomia symptoms lants such as cocaine and amphetamines, perturb
(Csecs et al., 2022). Misattribution and/or gener- central thermosensation, while increasing activ-
alization of interoceptive sensations similarly may ity levels. The pyrexia associated with immune
Interoception and Thermoreception 427

Table 26.2 Illustrative disorders of interoception


Disorder Interoceptive signature Bodily/affective expression

Eating disorders Aberrant processing of motivational signals Insensitivity to hunger signals, food anxiety
and compulsions
Panic and anxiety disorders Over-focus on / misinterpretation of Exaggerated physiological states, fear
internal signals
Depression Flattening of interoceptive signals Feelings of hollowness/emptiness,
exhaustion, psychomotor slowing
Dissociative disorders Perturbation in interoceptive coherence Feelings of detachment, tingling sensations
Substance abuse disorders Aberrant processing of motivational signals Exaggerated physiological states,
withdrawal, craving
Multiple sclerosis Impaired thermosensation Increased heat/cold sensitivity, fatigue
Diabetes Abnormal temperature signaling Sensations of burning, tingling sensations
Epilepsy Compromised central allostatic control / Seizure propagation
afferent gating of cortical arousal Pro-ictal states; evocation of bodily
sensations and emotional states

responses to inflammatory interoceptive chal- symptoms, opportunities arise to improve physical


lenges, mediated by prostaglandin E2 acting on and psychological wellbeing and function through
POA neurons, also reflects a down-regulation of interventions that act upon the representation and
thermostatic reflex function that is typically adap- control of internal bodily states. There are some
tive within the constellation of sickness behaviors noteworthy established examples: Implanted vagus
yet may be as life threatening as externally induced nerve stimulation is presumed to reduce seizure
causes of hyperthermia leaning to heat exhaustion, frequency in (treatment resistant) epilepsy through
organ/tissue damage, and death (Cheshire, 2016; action on ascending interoceptive influences on
Skok et al., 2020). thalamocortical excitability (Henry, 2002). The
As noted, certain areas of interoceptive science impact of this interoceptive therapy on treatment
(not least interactions between the immune system resistant depression indicates an effect on mood
and brain, and the gut and brain) are developing stability, arousal, and positive affect attributable to
rapidly, often ahead of general understanding and ascending monoaminegic neuromodulatory
appreciation of viscerosensory aspects of intero- systems (Sackheim et al., 2001). Exercise interven-
ception. Topic areas including immunopsychiatry tions, breath training, biofeedback approaches, and
and “the microbiota and behavior” have emerged. even evoking the diving reflex by immersing one’s
These are consequently finding independent appli- head in cold water, a recommended strategy in
cation to understanding and treatment of clinical dialectical behavioral therapy (Lineman et al.,
disorders. Overall, interoceptive mechanisms are 2007), have a long history of use to enhance mood
increasingly considered within mechanistic refor- and emotional regulation. Each of these techniques
mulations of clinical conditions, particularly those are associated with enhanced engagements of
disorders at the interface between mental and vagus afferents and vagally mediated parasympa-
physical health, and where chronicity and com- thetic cardiac outflow.
plexity limit quick-fix interventions that target spe- Treatments directed at interoceptive processing
cific organ systems in isolation. Emergent models may target distinct levels of this sensing hierarchy,
of interoception are providing fresh insights into targeting in the periphery the signaling of pain,
established therapeutic approaches and leading to inflammatory status, visceromotor activity, ther-
new treatments. mal change, and humoral contents or modifying
the function of pathways communicating intero-
ceptive information to the brain. Interventions can
target neural brain systems, while both behav-
ioral and psychological interventions can focus
THERAPEUTIC TARGETING OF on changing the perception and interpretation of
INTEROCEPTION interoceptive signals. The purpose of the interven-
tion is also important to consider: An intervention
As we learn more about interoceptive processes may seek to decrease or increase the symptoms
and their role in the expression of (often interact- arising as direct consequences of the interoceptive
ing) physical, motivational, and emotional sensation or to modify the impact of interoceptive
428 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

sensation on other functions or symptoms (includ- unhelpful feedback contributions of arousal on


ing somatic, neural, psychological, or behavioral behavior (Azevedo et al., 2017; Morera Maiquez
processes). Many interventions modify autonomic et al., 2020; Ventureya, 2000). Thermal biofeed-
(re)activity in order to evoke the therapeutic back is typically used for clinical conditions such
benefits of effects of an altered (attenuated or as migraine, hypertension, and Raynaud’s syn-
amplified) interoceptive state. drome (Blanchard et al., 1984; Karavidas et al.,
Pharmacological treatments can target periph- 2006; Nakao et al., 2003; Sharff et al., 2002).
eral aspects of interoception via autonomic Hand warming biofeedback demonstrated clinical
responses (e.g., smooth muscle relaxants, beta- improvement of migraine in paediatric popula-
blockers), or afferent signaling (e.g., pain killers, tion in a controlled trial. Biofeedback to increase
and antinflammatory drugs, local anaesthetics, temperature is also effective for hypertension and
antacids, intravesical capsaicin, and Botulinus Raynaud’s syndrome, increasing the circulation of
toxin). Surgical approaches extend beyond neu- peripheral blood flow.
rodestructive vagotomy (for gastric and cardiac The identified association of symptoms, such
symptoms) to neuromodulatory approaches as anxiety, with discrepancies in objective and
including sacral nerve stimulation (Dasgupta subjective axes (Garfinkel et al., 2015, 2016)
et al., 2005) and vagus nerve stimulation that also (and interoceptive metacognitive insight) has ena-
interrupt visceral afferent signaling (Sackheim bled development of cognitive and biobehavioral
et al., 2001). However, the impact may extend remediating strategies that specifically target the
beyond proximate interoceptive symptoms to mismatch across interoceptive dimensions (opera-
affect neurological function and mood state. tionalized for example in heartbeat detection per-
Neurostimulatory approaches to brain, neverthe- formances accuracy versus subjective confidence
less, become more removed from clear interocep- or questionnaire self-rating of interoceptive sensi-
tive targets. However, such mechanisms are likely tivity) (Quadt et al., 2021). This focused approach
to play a part in, for example, the improvement in likely targets mechanisms that are also implicit in
neurovegetative symptoms of psychomotor slow- the beneficial consequences of exercise, “breath-
ing in depression following deep brain stimulation work” and interoceptive exposure therapies.
applied to subgenual cingulate cortex and associ- There, the conceptual framework is consistent
ated regions (Sankar et al., 2020). with computational notions of interoceptive pre-
Biobehavioral approaches including biofeed- dictive control in which subjective report may be
back and cognitive behavioral strategies have viewed as expressing top-down predictions tested
found particular use: Biofeedback, by “closing against ascending objective interoceptive signals
the interoceptive loop” on typically covert auto- (see earlier). A large clinical trial for anxiety, in
nomic control signals and associated interocep- an adult autistic population, attests to the value
tive responses, can be used to train the control and acceptability of interoceptive training of this
of internal state and its integration with psycho- nature (Quadt et al., 2021).
logical and behavioral processes. In this con-
text, electrodermal biofeedback has been used to
diminish sympathetic activity in stress states, e.g.,
anxiety, panic, conditions such as irritable bowel
syndrome and may be useful in managing tic dis- SUMMARY AND CONCLUSIONS
order (Gilbert, 1986; Leahy et al., 1988; Nagai
et al., 2014). Training the capacity to volition- The representation of the internal physiological
ally increase sympathetic arousal can empower state of the body is fundamental to the processes
patients with epilepsy to employ effective coun- through which an individual senses, interprets,
termeasures to mitigate seizures in pro-ictal states, integrates, and regulates informative signals from
leveraging apparent inverse coupling of interocep- within itself and applies this representation to navi-
tive coupling of bodily arousal states to thalamo- gating and adapting to an uncertain external world.
cotical excitability (Nagai et al., 2018). Heart rate A maturing framework for understanding intero-
variability biofeedback, aiming to amplify para- ception includes the signaling and processing
sympathetic “neurovisceral integration,” has been through the neuraxis including expression at dis-
applied to enhance the management of affective tinct levels of perception and cognition.
symptoms (Goessl et al., 2013; Schumann et al., Interoception is an important component of
2019) and sport performance (Jiminez-Morgan somatic, neurological, psychiatric, and behavioral
and Molina-Mora, 2017). There is much prom- disorders and the expression of symptoms in
ise in the application of “proxy” somatosensory chronic and complex conditions that span physical
stimulation that can compete with or entrain and mental health. Better understanding of the
central interoceptive representation to attenuate neural basis of interoception may refine existing
Interoception and Thermoreception 429

treatments offer new focused therapeutic targets for Berntson, G. G., & Khalsa S. S. (2021). Neural circuits
somatic, psychological and neurological disorders. of interoception. Trends in Neurosciences, 44(1),
17–28.
Berthoud, H. R., & Neuhuber, W. L. (2000). Func-
tional and chemical anatomy of the afferent vagal
system. Autonomic Neuroscience: Basic & Clinical,
REFERENCES 85(1–3), 1–17.
Betka, S., Lukowska, M., Silva, M., King, J., & Garfin-
Adams, K. L., Edwards, A., Peart, C., Ellett, L., kel, S. N., Critchley, H. D. (2021). Evidence toward
Mendes, I., Bird, G., & Murphy, J. (2022). The the potential absence of relationship between tem-
association between anxiety and cardiac intero- poral and spatial heartbeats perception. Scientific
ceptive accuracy: A systematic review and meta- Reports, 11(1). doi: 10.1038/s41598-021-90334-z
analysis. Neuroscience & Biobehavioral Reviews, Blanchard, E. B., McCoy, G. C., Andrasik, F., Acerra,
140. doi: 10.1016/j.neubiorev.2022.104754 M., Pallmeyer, T. P., Gerardi, R., Halpern, M., &
Allen, M. Levy, A. Parr, T., & Friston, K. (2019). In the Musso, A. (1984). Preliminary results from a con-
body’s eye: The computational anatomy of intero- trolled evaluation of thermal biofeedback as a
ceptive inference. BioRxiv, doi: 10. 1101/603928 treatment for essential hypertension. Biofeedback
Ardizzi, M., Ambrosecchia, M., Buratta, L., Ferri, F., and Self-Regulation, 9(4), 471–495.
Peciccia, M., Donnari, S., Mazzeschi, C., & Gallese, V. Blessing, W. W. (1997). The lower brainstem and
(2016). Interoception and positive symptoms in bodily homeostasis. Oxford, UK: Oxford University
schizophrenia Frontiers in Human Neuroscience, Press.
10. doi: 10.3389/fnhum.2016.00379. Bonaz, B., Lane, R. D., Oshinsky, M. L., Kenny, P. J.,
Azevedo, R. Bennett, N. Bilicki, A. Hooper, J. Marko- Sinha, R., Mayer, E. A., & Critchley, H. D. (2021).
poulou, F. & Tsakiris, M. (2017). The calming effect Diseases, disorders, and comorbidities of intero-
of a new wearable device during the anticipation ception. Trends in Neurosciences, 44(1), 39–51.
of public speech. Scientific Reports, 7(1). doi: Boscan, P., Pickering, A. E., & Paton, J. F. (2002). The
10.1038/s41598-017-02274-2 nucleus of the solitary tract: An integrating station
Bard, P. (1934). On emotional expression after decor- for nociceptive and cardiorespiratory afferents.
tication with some remarks on certain theoretical Experimental Physiology. 87(2), 259–266.
views, Part I. Psychological Review, 41, 309–329. Cameron, O. G. (2002). Visceral sensory neurosci-
Barrett, L. F. (2017). The theory of constructed emo- ence: Interoception. Oxford, UK: Oxford University
tion: An active inference account of interoception Press.
and categorization. Social Cognitive and Affective Cannon, W. B. (1927). The James-Lange theory of
Neuroscience, 12(1), 1–23. emotions: A critical examination and an alterna-
Bechara, A. Damasio, H., Tranel, D., & Damasio, A. tive theory. American Journal of Psychology, 39(1),
(1997). Deciding advantageously before knowing 106–124.
the advantageous strategy. Science, 275, Cantril, H., & Hunt, W. A. (1932). Emotional effects
1293–1295. produced by injection of adrenalin. American Jour-
Bechara, A. Damasio, H., Tranel, D., & Damasio, A. nal of Psychology, 44(2), 300–307.
(2005). The Iowa Gambling Task and the somatic Cheshire, W. P. Jr. (2016). Thermoregulatory disorders
marker hypothesis: Some questions and answers. and illness related to heat and cold stress.
Trends in Cognitive Sciences, 9(4), 159–162. Autonomic Neuroscience, 196, 91–104.
Beissner, F., Meissner, K., Bär K. J., & Napadow, V. Cheung, R. T., & Hachinski, V. (2000). The insula and
(2013). The autonomic brain: An activation likeli- cerebrogenic sudden death. Archives of Neurology,
hood estimation meta-analysis for central process- 57(12), 1685–1688.
ing of autonomic function. Journal of Neuroscience, Ciura, S., Liedtke, W., & Bourque, C. W. (2011).
33(25), 10503–10511. Hypertonicity sensing in organum vasculosum
Benarroch, E. E. (1993). The central autonomic net- lamina terminalis neurons: A mechanical process
work, functional organization, dysfunction, and involving TRPV1 but not TRPV4. Journal of Neuro-
perspective. Mayo Clinic Proceedings, 68(10), science, 31(41), 14669–14676.
988–1001. Craig, A. D. (2014). How do you feel? An interocep-
Benarroch, E. E. (2008). The arterial baroreflex: Func- tive moment with your neurobiological self.
tional organization and involvement in neurologic Princeton, NJ: Princeton University Press.
disease. Neurology, 71(21), 1733–1738. Craig, A. D., Chen, K., Bandy, D., & Reiman, E. M.
Berntson, G. G., Sarter, M., & Cacioppo, J. T. (2003). (2000). Thermosensory activation of insular cortex.
Ascending visceral regulation of cortical affective Nature Neuroscience, 3(2), 184–190.
information processing. European Journal of Neu- Craig, A. D. (2002). How do you feel? Interoception:
roscience, 8(8), 2103–2109. The sense of the physiological condition of the
430 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

body. Nature Reviews: Neuroscience, 3(8), Darby, R. R., Joutsa, J., Burke, M. J., & Fox, M. D. (2018).
655–666. Lesion network localization of free will. Proceedings
Craig, A. D. (2004). Distribution of trigeminothalamic of the National Academy of Science of the United
and spinothalamic lamina I terminations in the States of America. 115(42), 10792–10797.
macaque monkey. Journal of Comparative Neurol- Darwin, C. R. (1872). The expression of the emotions
ogy, 477(2), 119–148. in man and animals. London: John Murray.
Critchley, H. D., & Garfinkel, S. N. (2017). Interocep- Dasgupta, R., Critchley, H. D., Dolan, R. J., & Fowler,
tion and emotion. Current Opinion in Psychology, C. J. (2005). Changes in brain activity following
17, 7–14. sacral neuromodulation for urinary retention. Jour-
Critchley, H. D., & Harrison, N. A. (2013). Visceral nal of Urology, 174(6), 2268–2272.
influences on brain and behavior. Neuron, 77(4), Dimberg, U., Thunberg, M., & Elmehed, K. (2000).
624–638. Unconscious facial reactions to emotional facial
Critchley, H. D., Mathias, C. J., & Dolan, R. J. (2002). expressions. Psychological Science, 11(1), 86–89.
Fear conditioning in humans, the influence of Dinan, T. G., & Cryan, J. F. (2017). Brain-gut micro-
awareness and autonomic arousal on functional biota axis and mental health. Psychosomatic
neuroanatomy. Neuron, 33(4), 653–656. Medicine, 79(8), 920–926.
Critchley, H. D. (2005). Neural mechanisms of auto- Doba, N., & Reis, D. J. (1973). Acute fulminating neu-
nomic, affective, and cognitive integration. Jour- rogenic hypertension produced by brainstem lesions
nal of Comparative Neurology, 493, 154–166. in the rat. Circulation Research, 32(5), 584–593.
Critchley, H. D. (2009). Psychophysiology of neural Dunn-Meynell, A. A., Rawson, N. E., & Levin, B. E.
cognitive and affective integration, fMRI and auto- (1998). Distribution and phenotype of neurons
nomic indicants International Journal of Psycho- containing the ATP-sensitive K+ channel in rat
physiology, 73, 88–94. brain. Brain Research, 814(1–2), 41–54.
Critchley, H. D., Wiens, S., Rotshtein, P., Ohman, A., Eggart, M., Lange, A., Binser, M., Queri, S., & Müller-
& Dolan, R. J. (2004). Neural systems supporting Oerlinghausen, B. (2019). Major depressive disorder
interoceptive awareness. Nature Neuroscience, 7, Is associated with impaired Interoceptive accuracy:
189–195. A systematic review. Brain Sciences, 9(6), 131. doi:
Critchley, H. D., Sherrill, S. P., Ewing, D. L., van Praag, 10.3390/brainsci9060131
C. G., Habash-Bailey, H., Quadt, L., … Garfinkel, Ekman, P., Levenson, R. W., & Friesen, W. V. (1983).
S. N. (2023). Cardiac interoception in patients Autonomic nervous system activity distinguishes
accessing secondary mental health services: A among emotions. Science, 2211, 1208–1210.
transdiagnostic study. Autonomic Neuroscience, Fagerlund, M. J., Kåhlin, J., Ebberyd, A., Schulte, G.,
245. doi: /10.1016/j.autneu.2023.103072 Mkrtchian, S., & Eriksson, L. I. (2010). The human
Crucianelli, L., Enmalm, A., & Ehrsson, H. H. (2022) carotid body: Expression of oxygen sensing and
Interoception as independent cardiac, thermosen- signaling genes of relevance for anesthesia. Anes-
sory, nociceptive, and affective touch perceptual thesiology, 113(6), 1270–1279.
submodalities. Biological Psychology. 172. doi: Flores, S., Davis, M. D., Pittelkow, M. R., Sandroni, P.,
10.1016/j.biopsycho.2022.108355. Weaver, A. L., & Fealey, R. D. (2015). Abnormal
Csecs, J. L. L., Iodice, V., Rae, C. L., Brooke, A., Sim- sweating patterns associated with itching, burning
mons, R., Quadt, L., … Eccles, J. A. (2022). Joint and tingling of the skin indicate possible underly-
hypermobility links neurodivergence to dysautono- ing small-fibre neuropathy. British Journal of Der-
mia and pain. Frontiers in Psychiatry, 12. doi: matology, 172(2), 412–418.
10.3389/fpsyt.2021.786916 Fuchs, T. A., Vaughn, C. B., Benedict, R. H. B.,
Cutsforth-Gregory, J. K., & Benarroch, E. E. (2017). Weinstock-Guttman, B., Choudhery, S., Carolus,
Nucleus of the solitary tract, medullary reflexes, K., … Dwyer, M. G. (2019). Lower self-report
and clinical implications Neurology. 88(12), fatigue in multiple sclerosis is associated with
1187–1196. localized white matter tract disruption between
Damasio, A. R. (1999). The feeling of what happens: amygdala, temporal pole, insula, and other con-
Body and emotion in the making of conscious- nected structures. Multiple Sclerosis and Related
ness. New York: Harcourt. Disorders, 27, 298–304.
Damasio, A. R. (2010). Self comes to mind: Construct- García-Cordero, I., Sedeño, L., de la Fuente, L.,
ing the conscious brain. New York: Pantheon. Slachevsky, A., Forno, G., Klein, F., … Ibañez, A.
Dantzer, R., O’Connor, J. C., Freund, G. G., Johnson, (2016). Feeling, learning from and being aware of
R. W., & Kelley, K. W. (2008). From inflammation inner states: Interoceptive dimensions in neurode-
to sickness and depression: When the immune generation and stroke. Philosophical Transactions
system subjugates the brain. Nature Reviews: Neu- of the Royal Society of London: B Biological Sci-
roscience, 9(1), 46–56. ences, 371. doi: 10.1098/rstb.2016.0006
Interoception and Thermoreception 431

Garfinkel, S. N., Barrett, A. B., Minati, L., Dolan, R. J., training on stress and anxiety: A meta-analysis.
Seth, A. K., & Critchley, H. D. (2013). What the Psychological Medicine, 47(15), 2578–2586.
heart forgets: Cardiac timing influences memory Ghosal, S., Bundzikova-Osacka, J., Dolgas, C. M.,
for words and is modulated by metacognition and Myers, B., & Herman, J. P. (2014) Glucocorticoid
interoceptive sensitivity. Psychophysiology, 50(6), receptors in the nucleus of the solitary tract (NTS)
505–512. decrease endocrine and behavioral stress responses.
Garfinkel, S. N., & Critchley, H. D. (2013). Interocep- Psychoneuroendocrinology. 45, 142–153.
tion, emotion and brain, new insights link internal Gray, M. A., Taggart, P., Sutton, P. M., Groves, D.,
physiology to social behaviour: Commentary on Holdright, D. R., Bradbury, D., Brull, D., & Critch-
“Anterior insular cortex mediates bodily sensibility ley, H. D. (2007). A cortical potential reflecting
and social anxiety” by Terasawa et al. (2012). cardiac function. Proceedings of the National
Social Cognitive and Affective Neuroscience, 8(3), Academy of Sciences of the United States of
231–234. America, 104(16), 6818–6823.
Garfinkel, S. N., Manassei, M. F., & Hamilton-Fletcher, Gray, M. A., Rylander, K., Harrison, N. A., Wallin, B.
G., den Bosch, Y., Critchley, H. D., & Engels, M. G., & Critchley, H. D. (2009). Following one’s
(2016). Interoceptive dimensions across cardiac heart: Cardiac rhythms gate central initiation of
and respiratory axes. Philosophical Transactions of sympathetic reflexes. Journal of Neuroscience,
the Royal Society of London: B Biological Sciences. 29(6), 1817–1825.
371. doi: 10.1098/rstb.2016.0014 Harrison, N. A., Brydon, L., Walker, C., Gray, M. A.,
Garfinkel, S. N., Seth, A. K., Barrett, A. B., Suzuki, K., Steptoe, A., Dolan, R. J., & Critchley, H. D. (2009).
& Critchley, H. D. (2015). Knowing your own Neural origins of human sickness in interoceptive
heart: Distinguishing interoceptive accuracy from responses to inflammation. Biological Psychiatry.
interoceptive awareness. Biological Psychology, 66(5), 415–422.
104, 65–74. Henry, T. R. (2002). Therapeutic mechanisms of
Garfinkel, S. N., Tiley, C., O’Keeffe, S., Harrison, N. vagus nerve stimulation. Neurology, 59(6, S4),
A., Seth, A. K., & Critchley, H. D. (2016). Discrep- S3–S14.
ancies between dimensions of interoception in Hindmarch, C. C., Fry, M., Smith, P. M., Yao, S. T.,
autism: Implications for emotion and anxiety. Hazell, G. G., Lolait, S. J., … Murphy, D. (2011).
Biological Psychology, 114, 117–126. The transcriptome of the medullary area pos-
Garfinkel, S. N., & Critchley, H. D. (2016). Threat and trema: The thirsty rat, the hungry rat and the
the body: How the heart supports fear processing. hypertensive rat. Experimental Physiology, 96,
Trends in Cognitive Sciences, 20(1), 34–46. 495–504.
Garfinkel, S. N., Greenwood, K., Rae, C., Davies, G., James, W. (1884). What is an emotion? Mind, 9,
van Praag, C. G., Seth, A., Nick, M., & Critchley, 188–205.
H. (2018). T149. Metacognitive deficits in intero- Jaenig, W. (2008). The integrative action of the auto-
ception are associated with dissociative experi- nomic nervous system, Neurobiology of homeo-
ences in patients with first episode psychosis. stasis. Cambridge, UK: Cambridge University
Schizophrenia Bulletin, 44(S1), S173. Press.
Garfinkel, S. N., Gould van Praag, C. D., Engels, M., Jarrahi, B., & Kollias, S. (2020). Effects of visceral
Watson, D., Silva, M., Evans, S. L., Duka, T., & interoception on topological properties of the
Critchley, H. D. (2020). Interoceptive cardiac sig- brain: A graph theory analysis of resting state
nals selectively enhance fear memories. Journal of fMRI. 42nd Annual International Conference of
Experimental Psychology General, doi: 10. 1037/ the IEEE Engineering in Medicine & Biology Society
xge0000967. (EMBC), (pp. 1116-1119). doi: 10.1109/EMBC
Garfinkel, S. N., Minati, L., Gray, M. A., Seth, A. K., 44109.2020.9175465
Dolan, R. J., & Critchley, H. D. (2014). Fear from Jenkinson, P. M., Taylor, L., & Laws, K. R. (2018). Self-
the heart: Sensitivity to fear stimuli depends on reported interoceptive deficits in eating disorders:
individual heartbeats. Journal of Neuroscience, A meta-analysis of studies using the eating disor-
34(19), 6573–6582. der inventory. Journal of Psychosomatic Research,
Gebhart, G. F., & Bielefeldt, K. (2016). Physiology of 110, 38-45.
visceral pain. Comprehensive Physiology, 6(4), Jiménez Morgan, S., & Molina Mora, J. A. (2017).
1609–1633. Effect of heart rate variability biofeedback on
Gilbert, C. (1986). Skin conductance feedback and sport performance: A systematic review. Applied
panic attacks. Biofeedback and Self-Regulation, Psychophysiology and Biofeedback, 42,
11, 251–254. 235–245.
Goessl, V. C., Curtiss, J. E., & Hofmann, S. G. (2017). Karavidas, M. K., Tsai, P. S., Yucha, C., McGrady, A.,
The effect of heart rate variability biofeedback & Lehrer, P. M. (2006). Thermal biofeedback for
432 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

primary Raynaud’s phenomenon: A review of the Mograbi, D., Huntley, J., & Critchley, H. D. (2021). Self-
literature. Applied Psychophysiology and Biofeed- awareness in dementia, a taxonomy of processes:
back, 31(3), 203–216. Overview of findings and integrative framework.
Khalsa, S. S., Adolphs, R., Cameron, O. G., Critchley, Current Neurology and Neuroscience Reports,
H. D., Davenport, P. W., Feinstein, J. S., … Zucker, N. 21(12). doi: 10.1007/s11910-021-01155-6.
(2018). Interoception and mental health: A road- Morera Maiquez, B., Sigurdsson, H. P., Dyke, K.,
map. Biological Psychiatry: Cognitive Neuroscience Clarke, E., McGrath, P., Pasche, M., Rajendran, A.,
and Neuroimaging, 3(6), 501–513. Jackson, G. M., & Jackson, S. R. (2020). Entraining
Koekkoek, L. L., Mul, J. D., & La Fleur, S. E. (2017). movement-related brain oscillations to suppress
Glucose-sensing in the reward system. Frontiers in tics in Tourette syndrome. Current Biology, 30(12),
Neuroscience, 11. doi: 10.3389/fnins.2017.00716 2334–2342.
Lange, K. G. (1907). Les émotions: Étude psycho- Nadeau, S., & Rivest, S. (2000). Role of microglial-
physiologique. Paris: Alcan. derived tumor necrosis factor in mediating CD14
Larsson, D. E. O., Esposito, G., Critchley, H., Dienes, transcription and nuclear factor kappa B activity in
Z., & Garfinkel, S. (2021). Sensitivity to changes in the brain during endotoxemia. Journal of
rate of heartbeats as a measure of interoceptive Neuroscience, 20(9), 3456–3468.
ability. Journal of Neurophysiology. doi: 10.31234/ Nagai, Y., Cavanna, A. E., Critchley, H. D., Stern, J. J.,
osf.io/v34a7 Robertson, M. M., & Joyce E. M. (2014). Biofeed-
Leahy, A., Clayman, C., Mason, I., Lloyd, G., & back treatment for Tourette syndrome: A prelimi-
Epstein, O. J. (1988). Computerised biofeedback nary randomized controlled trial. Cognitive and
games: A new method for teaching stress man- Behavioral Neurology, 27(1), 17–24.
agement and its use in irritable bowel syndrome. Nagai, Y., Aram, J., Koepp, M., Lemieux, L., Mula,
Journal of the Royal College of Physicians London, M., Critchley, H., Sisodiya, S., & Cercignani, M.
32(6), 552–556. (2018). Epileptic seizures are reduced by auto-
Legrand, N., Nikolova, N., Correa, C., Brændholt, M., nomic biofeedback therapy through enhancement
Stuckert, A., Kildahl, A., Vejlø, M., Fardo, F., & of fronto-limbic connectivity: A controlled trial and
Allen, M. (2021). The heart rate discrimination neuroimaging study. EBioMedicine, 27, 112–122.
task: A psychophysical method to estimate the Nakamura, K., & Morrison, S. F. (2008). A thermosen-
accuracy and precision of interoceptive beliefs. sory pathway that controls body temperature.
BioRxiv, doi: 2021.02.18.431871 Nature Neuroscience, 11(1), 62–71.
Lineman, M. M., Bohus, M., & Lynch, T. R. (2007). Nakao, M., Yano, E., Nomura, S., & Kuboki, T. (2003).
Dialectical behavior therapy for pervasive emotion Blood pressure-lowering effects of biofeedback
dysregulation: Theoretical and practical underpin- treatment in hypertension: A meta-analysis of
nings. In J. J. Gross (Ed.), Handbook of emotion randomized controlled trials. Hypertension
regulation (pp. 581–605). New York: Guilford. Research, 26(1), 37–46.
Luft, C. D., & Bhattacharya, J. (2015). Aroused with Oomura, Y., Ono, T., Ooyama, H., & Wayner, M. J.
heart: Modulation of heartbeat evoked potential (1969). Glucose and osmosensitive neurones of
by arousal induction and its oscillatory correlates. the rat hypothalamus. Nature, 222, 282–284.
Scientific Reports, 5. doi: 10.1038/srep15717 Panksepp, J. (2011). The basic emotional circuits of
Mahdi, A., Sturdy, J., Ottesen, J. T., & Olufsen, M. S. mammalian brains: Do animals have affective
(2013). Modelling the afferent dynamics of the lives? Neuroscience and Biobehavioral Reviews,
baroreflex control system. PLoS Computational 35(9), 1791–1804.
Biology, 9(12). doi: 10.1371/journal.pcbi.1003384 Park, H. D., Bernasconi, F., Salomon, R., Tallon-
Marañon, G. (1924). Contribution a l’étude de Baudry, C., Spinelli, L., Seeck, M., Schaller, K., &
l’action emotive de l’adrénaline. Revue Française Blanke, O. (2018). Neural sources and underlying
d’Endocinologie, 2, 301–325. mechanisms of neural responses to heartbeats,
Marron, K., Wharton, J., Sheppard, M. N., Fagan, D., and their role in bodily self-consciousness: An
Royston, D., Kuhn, D. M., … Polak, J. M. (1995). Dis- intracranial EEG study. Cerebral Cortex, 28(7),
tribution, morphology, and neurochemistry of endo- 2351–2364.
cardial and epicardial nerve terminal arborizations in Paton, J. F., Li, Y. W., & Kasparov, S. (1999). Reflex
the human heart. Circulation, 92(8), 2343–2351. response and convergence of pharyngoesophageal
Marshall, A. G., Sharma, M. L., Marley, K., Olausson, and peripheral chemoreceptors in the nucleus of
H., & McGlone, F. P. (2019). Spinal signalling of the solitary tract. Neuroscience, 93(1), 143–154.
C-fiber mediated pleasant touch in humans. Elife, Paulus, M. P., & Stewart, J. L. (2014). Interoception
8. doi: 10.7554/eLife.51642 and drug addiction. Neuropharmacology, 76, 342–
Mayer, E. (2011). Gut feelings: the emerging biology 350. doi: 10.1016/j.neuropharm.2013.07.002
of gut–brain communication. Nature Reviews: Petzschner, F., Garfinkel, S. N., Paulus, M. P., Koch,
Neuroscience, 12, 453–466 C., & Khalsa, S. S. (2021). Computational models
Interoception and Thermoreception 433

of interoception and body regulation. Trends in Sackeim, H. A., Rush, A. J., George, M. S., Marangell,
Neurosciences, 44(1), 63–76. L. B., Husain, M. M., Nahas, Z., … Goodman, RR.
Petzschner, F. H., Weber, L. A. E., Gard, T., & Stephan, (2001). Vagus nerve stimulation (VNS) for treat-
K. E. (2017). Computational psychosomatics and ment-resistant depression, efficacy, side effects,
computational psychiatry: Toward a joint frame- and predictors of outcome. Neuropsychopharma-
work for differential diagnosis. Biological Psychia- cology, 25(5), 713–728.
try, 82(6), 421–430. Salomon, R., Ronchi, R., Dönz, J., Bello-Ruiz, J., Her-
Pezzulo, G., Rigoli, F., & Friston, K. (2015). Active belin, B., Faivre, N., Schaller, K., & Blanke, O.
Inference, homeostatic regulation and adaptive (2018). Insula mediates heartbeat related effects
behavioural control. Progress in Neurobiology, on visual consciousness. Cortex, 101, 87–95.
134, 17–35. Sankar, T., Chakravarty, M. M., Jawa, N., Li, S. X.,
Pfeifer, G., Garfinkel, S. N., Gould van Praag, C. D., Giacobbe, P., Kennedy, S. H., Rizvi, S. J., Mayberg,
Sahota, K., Betka, S., & Critchley, H. D. (2017). H. S., Hamani, C., & Lozano, A. M. (2020). Neuro-
Feedback from the heart: Emotional learning and anatomical predictors of response to subcallosal
memory is controlled by cardiac cycle, interocep- cingulate deep brain stimulation for treatment-
tive accuracy and personality. Biological Psychol- resistant depression. Journal of Psychiatry and
ogy, 126, 19–29. Neuroscience. 45(1), 45–54.
Picard, F., & Craig, A. D. (2009). Ecstatic epileptic Schachter, S., & Singer, J. E. (1962). Cognitive, social,
seizures: A potential window on the neural basis and physiological determinants of emotional state.
for human self-awareness. Epilepsy & Behavior, Psychological Review, 69, 379–399.
16(3), 539–546. Scharff, L., Marcus, D. A., & Masek, B. J. (2002). A
Quadt, L., Critchley, H. D., & Garfinkel, S. N. (2018). controlled study of minimal-contact thermal bio-
The neurobiology of interoception in health and feedback treatment in children with migraine.
disease. Annals of the New York Academy of Sci- Journal of Pediatric Psychology, 27(2), 109–119.
ences, 1428(1), 112–128. Schulz, A., Reichert, C. F., Richter, S., Lass-Henne-
Quadt, L., Critchley, H. D., & Garfinkel, S. N. (2019). mann, J., Blumenthal, T. D., & Schächinger, H.
Interoception and emotion: Shared mechanisms (2009). Cardiac modulation of startle: Effects on
and clinical implications. In M. Tsakiris & H. De eye blink and higher cognitive processing. Brain
Preester (Eds.), The interoceptive mind: From and Cognition, 71(3), 265–271.
homeostasis to awareness. (pp.123–143). Oxford, Schulz, A., van Dyck, Z., Lutz, A. P. C., Rost, S., &
UK: Oxford University Press. Vögele, C. (2017). Gastric modulation of startle
Quadt, L., Garfinkel, S. N., Mulcahy, J. S., Larsson, D. eye blink. Biological Psychology, 127, 25–33.
E., Silva, M., Jones, A. M., Strauss, C., & Critchley, Schumann, A., Köhler, S., Brotte, L., & Bär, K. J.
H. D. (2021). Interoceptive training to target anxiety (2019). Effect of an eight-week smartphone-
in autistic adults (ADIE): A single-center, superiority guided HRV-biofeedback intervention on auto-
randomized controlled trial. EClinicalMedicine. 39. nomic function and impulsivity in healthy controls.
doi: 10.1016/j.eclinm.2021.101042 Physiological Measurement, 40(6). doi:
Rabchevsky, A. G. (2006). Segmental organization of 10.1088/1361-6579/ab2065
spinal reflexes mediating autonomic dysreflexia Seth, A. K., Suzuki, K., & Critchley, H. D. (2012). An
after spinal cord injury. Progress in Brain Research, interoceptive predictive coding model of conscious
152, 265–274. presence. Frontiers in Psychology, 2. doi: 10.3389/
Raghu, R., Vignon-Clementel, I. E., Figueroa, C. A., & fpsyg.2011.00395
Taylor, C. A. (2011). Comparative study of viscoe- Seth, A. K., & Critchley, H. D. (2013). Extending pre-
lastic arterial wall models in nonlinear one-dimen- dictive processing to the body: Emotion as intero-
sional finite element simulations of blood flow. ceptive inference. Behavioral Brain Science, 36(3),
Journal of Biomechanical Engineering, 133(8). doi: 227–228.
10.1115/1.4004532 Seth, A. K., & Tsakiris, M. (2018). Being a beast
Ran, C., Hoon, M. A., & Chen, X. (2016). The coding machine: The somatic basis of selfhood. Trends in
of cutaneous temperature in the spinal cord. Cognitive Sciences, 22(11), 969–981.
Nature Neuroscience, 19(9), 1201–1209. Sherrington, C. S. (1900). Experiments on the value
Rebollo, I., Devauchelle, A. D., Béranger, B., & Tallon- of vascular and visceral factors for the genesis of
Baudry, C. (2018). Stomach-brain synchrony reveals emotion Proceedings of the Royal Society London,
a novel, delayed-connectivity resting-state network 66, 424–433.
in humans. eLife. 21(7). doi: 10.7554/eLife.33321 Sherrington, C. S. (1906). The integrative action of
Ring, C., & Brener, J. (1996). Influence of beliefs the nervous system. New York: Scribner.
about heart rate and actual heart rate on heart- Skok, K., Duh, M., Stožer, A., Markota, A., & Gosak,
beat counting. Psychophysiology, 33(5), M. (2020). Thermoregulation: A journey from physi-
541–546. ology to computational models and the intensive
434 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

care unit. Wiley Interdisciplinary Reviews: Systems first-person perspective in conscious vision. Cortex,
Biology and Medicine. doi: 10.1002/wsbm.1513 102, 139–149.
Smith, R., Thayer, J. F., Khalsa, S. S., & Lane, R. D. Tan, C. L., & Knight, Z. A. (2018). Regulation of body
(2017). The hierarchical basis of neurovisceral inte- temperature by the nervous system. Neuron,
gration. Neuroscience and Biobehavioral Reviews, 98(1), 31–48.
75, 274–296. Van den Bergh, O., Brosschot, J., Critchley, H.,
Sozansky, J., & Houser, S. M. (2014). The physiologi- Thayer, J. F., & Ottaviani, C. (2021). Better safe
cal mechanism for sensing nasal airflow: A litera- than sorry: A common signature of general vulner-
ture review. International Forum of Allergy & ability for psychopathology. Perspectives on Psy-
Rhinology, 4(10), 834–838. chological Science, 16(2), 225–246.
Stephan, K. E., Manjaly, Z. M., Mathys, C. D., Weber, van der Kooy, D., & Koda, L. Y. (1983). Organization
L. A., Paliwal, S., Gard, T., … Petzschner, F. H. of the projections of a circumventricular organ,
(2016). Allostatic self-efficacy: A metacognitive the area postrema in the rat. Journal of Compara-
theory of dyshomeostasis-induced fatigue and tive Neurology, 219(3), 328–338.
depression. Frontiers in Human Neuroscience, 10. Van Roon, A. M., Mulder, L. J., Althaus, M., &
doi: 10.3389/fnhum.2016.00550 Mulder, G. (2004). Introducing a baroreflex model
Suksasilp, C., & Garfinkel, S. N. (2022) Towards a for studying cardiovascular effects of mental
comprehensive assessment of interoception in a workload. Psychophysiology, 41(6), 961–981.
multi-dimensional framework. Biological Psychol- Ventureyra, E. C. (2000). Transcutaneous vagus nerve
ogy, 168. doi:10.1016/j.biopsycho.2022.108262 stimulation for partial onset seizure therapy: A
Sun, X., Veldhuizen, M. G., Wray, A. E., de Araujo, I. new concept. Child’s Nervous System, 16(2),
E., Sherwin, R. S., Sinha, R., & Small, DM. (2014). 101–102.
The neural signature of satiation is associated with Wiens, S., & Palmer, S. N. (2001). Quadratic trend
ghrelin response and triglyceride metabolism. analysis and heartbeat detection. Biological
Physiology and Behavior. 136, 63–73. Psychology, 58(2), 159–175.
Tallon-Baudry, C., Campana, F., Park, H. D., & Yates, B. J., Bolton, P. S., & Macefield, V. G. (2014).
Babo-Rebelo, M. (2018). The neural monitoring of Vestibulo-sympathetic responses. Comprehensive
visceral inputs, rather than attention, accounts for Physiology, 4(2), 851–887.
27
Peripersonal Space
Representation:
Neural Bases, Properties, and
Functional Significance
Claudio Brozzoli, Nadia Bolognini,
Alessandro Zanini, and Alessandro Farnè

INTRODUCTION space that span from the seminal discovery of


multisensory neurons with specific spatial proper-
The brain creates multiple representations of ties in monkey models to neuroimaging evidence
space, which encode sensory information in a of the existence of a peripersonal space system in
diversity of frames of references centered on the the human brain. Starting from neurophysiology,
body or on the environment, depending on the new theoretical frameworks have been developed
source of sensory stimulation and the nature of to define the functional and dynamic features
interaction between the individual and the objects of the human peripersonal space in relation to
it contains. The body constitutes the origin of perception, action, and social cognition.
most of spatial representations, with different
neuronal populations representing two main sec-
tors of space in terms of distance from the body:
the peripersonal space, which is the space imme- NEURAL BASES OF PERIPERSONAL
diately surrounding our bodies and the extra-­
SPACE REPRESENTATION IN
personal space, which is far from our body. Such
dichotomy dates back to the last century, when MONKEY MODELS
Brain (1941) proposed to distinguish between a
“grasping” space from a “walking” space, an idea The definition of peripersonal space originates
that followed his investigation of selective impair- from single-unit electrophysiological studies in
ments of these two spatial representations in macaque monkeys that have provided an influential
brain-damaged patients. Since then, almost a cen- framework for the demonstration of the existence
tury of interdisciplinary neuroscientific research of a selective specific system to represent the space
has developed such intuition, yielding insights on closely surrounding the human body. Single-unit
the neural foundations and psychological mecha- electrophysiological studies in macaque monkeys
nisms unique to the representation of the space revealed the existence of a class of multisensory
close to the body. neurons suggested to represent the neural basis of
The goal of this chapter is to provide an over- peripersonal space representations (Graziano,
view of modern conceptions of the peripersonal 2001; Rizzolatti et al., 1981a). Their most relevant
436 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

feature is to respond both to visual and tactile


stimuli, showing visually evoked responses modu-
lated by the distance between the object and the
tactile receptive field (RF). Some of these neurons
also respond to auditory stimuli (Graziano, 2001).
The visual RFs are usually, though not always,
confined to the space near the body, and they are
better represented like bubbles of space anchored
to the body surface (Figure 27.1). This allows for
the coding of visual information that is dependent
(centered) on the body part that contains the tac-
tile RF. These body-centered RFs of visuo-tactile
neurons likely form the neural mechanism for the
psychological concept of peripersonal space.
Visuo-tactile neurons with body-centered Figure 27.2 Neural bases of peripersonal
RFs have been reported in several regions of the space in the macaque brain.
monkey brain (Figure 27.2), including the premo- Lateral view of a macaque’s brain showing
tor Area 6, parietal areas (Broadmann’s Area 7b
the parietal area VIP (the ventral sec-
and the ventral intraparietal area, VIP) and the
putamen (Fogassi et al., 1999; Graziano, 2001; tion of the intraparietal sulcus) and
Rizzolatti et al., 1997). 7b (in inferior posterior parietal lobe), the
Peripersonal visuo-tactile neurons in the F4 sub- polysensory zone in the posterior half of F4
region of ventral premotor cortex (Matelli et al., (a subregion of inferior Area 6 in the ventral
1985) present with large tactile RFs located primar- premotor cortex) and the putamen (dotted
ily on the monkey’s face, neck, arm, hand, or both line). Single-cell recordings highlighted the
hands and face (Gentilucci et al., 1989; Rizzolatti presence, in all these areas, of visuo-tactile
et al., 1981a). A large proportion (85%) of these neurons with body-part-centered RFs.
neurons also respond to visual stimuli according (from Graziano, 2001).
to their distance from the body because their vis-
ual RFs are limited in depth (from ∼5 to ∼50 cm).
About half of these visuo-tactile neurons are
named pericutaneous (54%) since they respond
to visual stimuli presented in the close proxim-
ity (a few centimeters, 10 cm or less) to the skin,
while the so-called distant peripersonal neurons
(46%) responds to visual stimuli within the mon-
key’ reaching distance (Rizzolatti et al., 1981b,c).
Another key property of the visual RFs of premo-
tor neurons is their independence from the posi-
tion of the eyes, being anchored to the tactile
RFs (Fogassi et al., 1992; Gentilucci et al., 1983;
Graziano et al., 1994, 1997). For instance, in a
neuron with a tactile RF on the arm, the corre-
sponding visual RF shifts along with the arm if it
is moved (Figure 27.3), while remaining relatively
fixed near the arm if only the eyes are moved.
Visuo-tactile neurons are also present in the
adjacent area F5, along the inferior branch of the
arcuate sulcus. They have smaller tactile RFs (than
that of F4 neurons) located on the face, the hand,
Figure 27.1 Schematic illustration of the
or both, and respond stronger to visual stimuli
peripersonal space (dark gray area; the light close to the body but the type of the stimuli is a
gray area depicts the extrapersonal-far - critical factor in driving the activity of F5 neurons.
space) surrounding the monkey’s body. Indeed, the visual response of F5 neurons mostly
Darkest circles depict body-part-centered relies on whether the viewed object is graspable
(hand- and face-centered) peripersonal or not, rather than on their distance from the body
space representations. (Bonini et al., 2014).
PERIPERSONAL SPACE REPRESENTATION 437

Figure 27.3 Schematic of receptive fields (RFs) of visuo-tactile neuron.


(A) The tactile RF is centered on the hand and the visual RF (red area) on the area surround-
ing the hand. (B) When the monkey’s limb is passively moved over the midline, the visual RF
shifts accordingly. (C) Graphic representation of the responses of a premotor bimodal neuron
to four trajectories of an approaching visual stimulus (arrows 1–4); The arm is visible to the
monkey. When the left hand is on the left (dark gray line), neuron’s response (spike/second)
is maximal for trajectory “3,” which is approaching the neuron’s tactile RF. When the left
hand is on the right (light gray line), the response is maximal for trajectory “2,” which is now
approaching the tactile RF (from Brozzoli et al., 2012a).

The posterior parietal lobe of the macaque brain Multisensory neurons have also been found in
contains two regions involved in peripersonal space the monkey area VIP, an audio-visuo-tactile con-
representations: Area 7b of the inferior posterior vergence site in the fundus of the intraparietal
parietal lobe and the ventral section of the intrapari- sulcus (Avillac et al., 2005; Colby et al., 1993;
etal sulcus (VIP). Area 7b contains a coarse soma- Duhamel et al., 1998). VIP neurons respond to
totopic organization, with separate face, arm, and tactile and visual stimulation presented within a
hand representations: the face map lies on the upper few centimeters of the tactile RF. Unlike area 7b
inferior parietal convexity, at the border with area neurons, tactile RFs in VIP are primarily located
7a; adjacent to the face area, along the inferior pari- on the face and head, and visual RFs are anchored
etal convexity, there are arm and hand representa- to a region of space around the face. Moreover,
tions, followed by the foot ones (Hyvarinen, 1981; VIP neurons encode visual information in a gradi-
Hyvarinen and Shelepin, 1979). In the face and ent of eye- to head-frame of reference, while tac-
arm region of Area 7b, about 33% of the cells have tile stimuli are encoded in a head-centered frame
visual RFs spatially aligned with their tactile RFs of reference. Consequently, while for some VIP
(Hyvarinen, 1981; Hyvarinen and Shelepin, 1979; neurons the visual and tactile RFs are spatially
Leinonen, 1980; Leinonen and Nyman, 1979). The aligned irrespectively of eye position, for others
visual responses of 7b neurons emerge prior to the their alignment depends on gaze direction.
neuron’s tactile RF is touched, suggesting a sort Notably, visuo-tactile neurons within area 7b
of “anticipatory” function of these bimodal cells and VIP not only respond to objects close to the
(Hyvarinen and Poranen, 1974). Most of these neu- body, but even when another individual (a human
rons respond to visual stimuli moving toward the experimenter) body part is approached by a vis-
tactile RF, if within a reachable distance (about 10 ual stimulus: Ishida and colleagues (Ishida et al.,
cm), while the 20% of them are active only during 2009) found that some visuo-tactile neurons with
the monkey’s own movements (in particular, grasp- RFs anchored on the monkey’s body showed
ing, and manipulation with fingers) (Leinonen and visual responses matched to corresponding body
Nyman, 1979). parts of the experimenter observed by the monkey,
438 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

regardless (at least for some of these neurons) of of the extinguishing stimulus to the relevant body
the position of the experimenter with respect to part in external space, suggesting that extinction
the monkey. arises in a spatial reference frame that is anchored
In the putamen, visuo-tactile neurons are soma- to the hand and it can even remap when the hand
totopically organized and present with tactile RFs moves. This neuropsychological evidence demon-
on the arm, hand, and face to which the visual ones strates that crossmodal extinction takes place
are anchored. A large portion (82%) of face neu- within a limited sector of space closely surrounding
rons responds best to visual stimuli presented in a the body surface, namely the peripersonal space.
region of space within 10–20 cm from the tactile Moreover, the investigation of crossmodal extinc-
RF, whereas neurons with tactile RFs on the arm tion demonstrated the plastic nature of the human
and hand present even more shallow (up to 5 cm) peripersonal space, which can extend after tool use
visual RFs (Graziano and Gross, 1993). similarly to what had been shown in monkeys
An important characteristic of some visuo- (Ladavas and Farne, 2004). Overall, the findings on
tactile cells is the plastic feature of their visual crossmodal extinction are entirely consistent with
RFs. For instance, the extent in depth of the visual the functional properties of the monkey multisen-
RFs of F4 visuo-tactile neurons increases with the sory neuronal cells.
velocity (20–80 cm/s) of an approaching visual The neuropsychological investigation of unilat-
stimulus (Fogassi et al., 1996). Iriki and col- eral spatial neglect syndrome further showed the
leagues revealed that, after training monkeys to modularity of spatial representations, document-
use a rake as a tool to reach food pellets placed ing double dissociations between peripersonal and
outside their reaching space, some neurons start extrapersonal, farther space. The basic defining
to respond to stimuli near the far end of the tool, feature of this syndrome is a deficit in the abil-
a sector of the space unreachable without it (Iriki ity to orient attention toward the contralesional
et al., 1996). These and other dynamic properties (usually the left) side of space, that manifest as
of the visual RFs seem to depend on the execution inability to report sensory events occurring in that
of specific motor actions (Rizzolatti et al., 1998). side of space and to perform actions in that portion
The above-mentioned regions form a densely of space (Vallar and Bolognini, 2014). Of inter-
interconnected cortical and subcortical network est, some individuals present with neglect that is
supporting various body-part-centered representa- restricted to near space, whilst others show the
tions of the near space, which relies on the neural reverse pattern (Berti and Frassinetti 2000; Berti
integration of the tactile stimuli on a given body et al. 2001; Bisiach et al., 1989; Cowey et al. 1994;
part with visual information related to external Halligan and Marshall, 1991; Vuilleumier et al.,
objects close to the body. 1998; cf. Halligan et al., 2003).
A large body of evidence in healthy humans
confirmed the neuropsychological findings
(Maravita et al., 2003). In healthy humans, the
Human Peripersonal Space and peripersonal space may be assessed with various
its Neural Bases multisensory behavioral paradigms (Figure 27.4),
as the crossmodal congruency task (Spence
The initial studies aimed to identify a peripersonal et al., 2004a,b). The crossmodal congruency task
space representation in humans, functionally requires to discriminate, as quickly as possible,
homologous to that described in non-human pri- the elevation (up or down) of a tactile stimulus
mates, were carried out on stroke patients suffering applied to the thumb or the index fingers while a
from “spatial extinction” after unilateral cortical or synchronous visual stimulus appears near either
subcortical brain injury, typically affecting the right finger. Participants discriminate faster the tactile
cerebral hemisphere (Ladavas and Farne, 2004). elevation when visuo-tactile stimuli are congruent,
Patients showing spatial extinction can typically rather than incongruent. This strength of this effect,
detect a single sensory stimulus regardless of which decays with the increased distance of visual
whether it is presented in the ipsilesional or contral- stimuli from the fingers, is considered a proxy of
esional hemispace. However, when presented with the extent of space where visuo-tactile integration
two stimuli concurrently, the contralesional stimu- occurs and this of the peripersonal space. Indeed,
lus is “extinguished” from awareness. This phe- if the hand is moved to a different location, as
nomenon can arise within each sensory modality whit hand crossed, the crossmodal congruency
and crossmodally, so that an ipsilesional visual or effects swaps accordingly. Crossmodal ­congruency
auditory event may extinguish awareness of a touch effects can also be elicited by far visual distractors
on the contralesional hand that would otherwise after tool-use (Maravita et al., 2003).
have been felt (Mattingley et al., 1997). The extent The behavioral findings on brain-damaged
of crossmodal extinction depends on the proximity and healthy subjects have led to hypothesize that
PERIPERSONAL SPACE REPRESENTATION 439

Figure 27.4 Example of tasks to measure peripersonal space in humans.


(A) Some paradigms assess crossmodal facilitation. In this case, a visual stimulus (yellow star)
is randomly presented at different distances from the body (e.g., participant’s hand). The
visual stimulus may be static or moving (approaching to or receding from the body). Tactile
targets (red circles) are applied to the participant’s hand. Participants have to respond to
the tactile target as quickly as possible. Auditory, instead of visual, stimuli may be also used.
Typically, participants respond faster to touches when the visual/auditory stimulus is near the
hand, as compared to when it is far away from it. (B) The crossmodal congruency task instead
assesses the crossmodal interference between visual and tactile stimuli delivered in the
proximity of the body. In the original design, participants hold a sponge cube in either hand,
in which two vibrotactile stimulators (red circles) and two visual distractors (red circles) are
embedded. Participants have to discriminate the elevation of vibrotactile targets presented
unpredictably to the thumb or index finger (lower versus upper elevation, respectively) of
either hand. A visual distractor is presented randomly from one of the four possible loca-
tions from which a vibrotactile stimulus can be delivered. Thus, the visual distractor can be
congruent or incongruent in elevation (e.g., a “congruent” upper light together with upper
touch, versus an “incongruent” lower light together with the same upper touch). Incongruent
visual distractors delay tactile elevation judgments and produce more errors. This interfer-
ence effect (crossmodal congruency effect) is larger for a visual distractor near the tactually
stimulated hand than for one near the opposite, unstimulated, hand.

the neural underpinnings of peripersonal space the lateral-anterior portion of IPS (Huang et al.,
could involve a similar network across species. 2012). Furthermore, the level of activation of the
In a seminal human fMRI study, Bremmer et al. peripersonal space network, and in particular of
(2001) investigated the pattern of neural responses the premotor cortex, can predict and explain inter-
to tactile stimulations applied to the face and vis- individual differences in the extent of the periper-
ual and auditory stimulations moving toward the sonal space boundary. In particular, an increased
face. They observed an overlap of the responses variability of BOLD responses in the premotor
elicited by the three sensory stimuli in the deeper cortex to far – rather than near – approaching
part of the intraparietal sulcus (IPS), in the ven- (auditory) stimuli predicts the individual extent of
tral premotor cortex (PMv), and in the lateral part peripersonal space (Ferri et al., 2015).
of the postcentral gyrus (Bremmer et al., 2001). Considering the primary role of hands in eve-
Subsequent studies have confirmed the existence ryday actions, a large amount of neuroimaging
of a somatotopic mapping of close visual stimuli research in humans focused on the study of the
within the inferior parietal cortex, with the face peripersonal space centered on the hands, the so-
represented at the level of the IPS, the lower part called “peri-hand” space. Such peri-hand space
of the body more medially and the upper limbs in is encoded in the human IPS and lateral occipital
440 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

complex (LOC), with a sort of transition from a the peripersonal space network as a function of its
strictly visual representation of the hand in the proximity to the hand.
posterior IPS (adjoining the transverse occipital Crucially, the brain regions identified to
sulcus) and in LOC regions to a representation respond to a visual object in the peri-hand space
based on both visual and proprioceptive informa- also integrate multisensory signals (Gentile et al.,
tion in the anterior IPS. Indeed, the anterior IPS is 2011), as shown by the presence of super- and sub-
featured by a proprioceptive representation of the additive responses to the combination of stimuli
hand, as well as by a tactile hand-specific activa- of different sensory modalities, as compared to
tion, suggesting a homology with monkey parietal their responses to unisensory stimuli (Stein and
hand-centered areas (Bolognini and Maravita, Standord, 2008), again in line with findings in ani-
2007; Makin et al., 2007). By taking advantage of mals (Avillac et al., 2005). Indeed, the concurrent
the neuronal adaptation phenomenon in fMRI,1 a presentation of visual and tactile stimuli increases
better characterization of the human brain network the activity of peripersonal space network, while
selectively activated by objects in the peri-hand introducing temporal and/or spatial mismatch
space has been reached, showing that the perip- between modalities disrupts its response (Gentile
ersonal space network includes not only IPS, but et al., 2013).
also the supramarginal gyrus (SMG), dorsal and Even within the human visual cortex preferen-
ventral premotor cortices, cerebellum and puta- tial responses to near vs. far stimuli were found
men (Brozzoli et al., 2011). (Previc, 1990). Indeed, already in low-level visual
The increased sensitivity of the fMRI adap- cortical areas such as V2 and V3 it is possible to
tion method also highlighted the hand-centered observe the presence of distinct neural populations
nature of the visual processing occurring in these representing the lower and upper visual field that
areas (Figure 27.5): maintaining the same fixa- respond selectively to stimuli presented in the near
tion point and the same absolute position of the or far space respectively (Nasr and Tootell, 2018,
external stimulus (a 3D object), the proximity of 2020).
the hand to the object induces adaptation within Thus, in humans, neuroimaging studies have
the peripersonal space network, while its displace- revealed a peripersonal space network including
ment in a position far away does not generate such bilateral fronto-parietal regions and the putamen,
a response. By moving the object closer to the which is thought to represent a putative homo-
new hand position, the peripersonal space network logue of the premotor-parietal network described
response reappears (Brozzoli et al., 2012b). These in monkeys, sharing similar functional proper-
results indicate that an object presented within the ties. The areas forming the human peripersonal
reachable space elicits – or not – adaptation within space network are densely interconnected and are

Figure 27.5. Neural bases of the human peripersonal space.


(A) Experimental conditions: The participant’s right hand was placed on the table, on either
the left or the right of a central fixation point (black cross). A 3D object was presented to
either the left or the right of the fixation point. (B) Blood oxygen level–dependent (BOLD)
adaptation to the 3D visual stimulus moving near the hand, using a gradient indexing the
degree of hand-centered encoding (0 = absence of hand-centered responses, 5 = stronger
hand-centered encoding). The posterior parietal and premotor cortices showed a high
degree of hand-centered encoding. IPS = intraparietal sulcus; SMG = supramarginal gyrus;
PMv = ventral premotor cortex; PMd = dorsal premotor cortex.
Source: Modified from Brozzoli and others (2012b).
PERIPERSONAL SPACE REPRESENTATION 441

usually activated during sensory-motor tasks, sug- space representation is also implicated in the guid-
gesting that the peripersonal space could act as a ance of voluntary, object-oriented actions, the
sensory-motor interface. By contrast, a distinct peripersonal space has been also called “action
ventral network is dedicated to far space process- space.” Objects in the vicinity of the body are
ing, which includes occipital, temporal, parietal, indeed more relevant by virtue of the possible
posterior cingulate as well as orbitofrontal regions interactions our body can establish with them, as
not activated by near space, likely subserving the compared to distant objects. Body-centered repre-
processing of the shape and identity of objects and sentations of the space around us allow to access
visually guided action in the far space (Aimola to accurate information regarding the spatial posi-
et al., 2012; Cléry et al. 2015, 2018; Lane et al. tion of objects with respect to our body-parts, in
2013; Weiss et al., 2000). turn facilitating and fastening our actions toward
objects within reaching distance. By acting as an
anticipatory sensory-motor interface, by which
our brain can successfully predict the arrival of
Functional Significance of the environmental stimuli, the peripersonal space also
Peripersonal Space allows an early and fast detection of objects
approaching the body, assisting automatic reac-
Why does our brain need to represent the periper- tions (Fogassi and Luppino, 2005; Graziano and
sonal space, distinct from the extrapersonal space? Cooke, 2006).
Most likely because many benefits derive from the The same anticipatory mechanism supports
availability of a brain system specifically dedi- also voluntary object-oriented actions (Rizzolatti
cated to peripersonal space coding and monitor- et al., 1997). In everyday life, the location of
ing. The space around us is indeed the arena of our external objects as inside or outside the periper-
interactions with the external world, whether we sonal space depends not only on objects dynam-
want to reach a glass of wine or avoid an unidenti- ics, but also on the movements of the body and
fied insect flying toward our hand, or pass the its parts. This requires an online remapping of
glass we grabbed over to our dear friend. Hence, the peripersonal space as a function of voluntary
the peripersonal space has an inherent “appeti- goal-directed actions. This has been shown behav-
tive” function, being involved in the execution of iorally in healthy humans by using a modified,
actions toward entities in the proximity of the dynamic, version of the crossmodal congruency
body, but also a “defensive” function, dealing with task (Spence et al., 2004a,b), which included a
avoidance or protective actions meant to preserve reaching-to-grasp movement toward an object.
the integrity of our body. Furthermore, the perip- The strength of visuo-tactile interaction increases
ersonal space representation entails also a poten- during, and even immediately before, the hand
tial “social” function and may be linked to the movement, meaning that a far object affects tactile
representation of the self. processing at the hand whenever a hand move-
ment is executed, or even just planned, toward
it (Brozzoli et al., 2009). It follows that the spa-
tial constraints of multisensory interactions are
Appetitive Function of the recoded in an anticipatory fashion. This is the case
Peripersonal Space not only for hand movements, but also for whole
body movements (Noel et al., 2015). Moreover,
Thanks to its functional properties and its ana- the visuo-tactile effect is influenced by the spe-
tomical position at the interface with motor cificities of the sensorimotor demands (Brozzoli
regions, the peripersonal space may serve many et al., 2010) and the goal of the action sequence
motor functions related to body–objects interac- (Senna et al., 2019), thus adjusting peripersonal
tions. The visuo-tactile neurons of the parietal space processing to include the target of goal-
Area 7b, the premotor Area 6 and putamen are directed actions.
activated not only when a visual or tactile stimula- The involvement of the peripersonal space in
tion is passively perceived, but also when a motor action planning and execution would also predict
response is implemented (Brozzoli et al., 2014). it plays a role in situations where, instead of mov-
Furthermore, both the sensory and motor activities ing the hand toward an object, the object moves
are expressed in a system of common spatial coor- toward the hand. An object approaching the hand
dinates, which places in a common spatial repre- might thus interfere with an ongoing manual motor
sentation the position of the body-parts, the target program. This was shown by assessing the “readi-
to be reached and the movement necessary to ness” of the motor cortical hand representation to
reach it (Caminiti et al., 1991). To underline that an approaching object by means of Transcranial
the cerebral network involved in peripersonal Magnetic Stimulation (TMS) and the recording
442 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

of Motor Evoked Potentials (MEPs): an object function of the peripersonal space is related to
falling near the hand suppresses the ongoing defensive (reactions, dealing with the protection
hand motor plan a few milliseconds after object of our body.
vision (Makin et al., 2009). This indicates that the Studies in non-human primates had shown that
peripersonal space not only serves goal-directed electrical stimulation of the VIP area in the intrapa-
actions, but also updates quickly our motor system rietal sulcus and of the “polysensory zone” within
about the relevant events arising in surrounding the premotor cortex causes movement patterns
space. Despite the peripersonal space may share that are compatible with defensive, or avoidance
some characteristics with the arm-reaching space, responses (Graziano and Cooke, 2006). By chemi-
which is the space reachable by extending the arm cally modulating the activity of these regions, it is
without moving the trunk, the two representations possible to provoke a consequent modulation of
are not equivalent. Indeed, the peripersonal space the defensive behavior, inhibiting or enhancing it
(as measured with a visuo-tactile detection task) is (Cooke and Graziano, 2004b). Defensive reactions
smaller and codes for the target of our actions in include movements of the eyes, face, shoulders,
a hand-centered reference frame, while the arm- head and arms aimed at protecting the body part
reaching space (as measured with a reachability where the sensory RFs of the neurons are located
judgment task) is larger and not hand-centered (Cooke and Graziano, 2003, 2004a). Similar
(Zanini et al., 2021a). response patterns have also been reproduced in the
The plasticity of the peripersonal space for presence of real external threatening stimuli, such
action has been extensively studied with respect as sudden air puffs on the surface of the body, with
to the use of mechanical tools, and more recently peripersonal space neurons showing a strong pref-
even with the use of technological devices, to erence for moving stimuli (Cooke and Graziano,
reach targets outside the physical limits of their 2003; Fogassi et al., 1996). These properties led
body (Serino, 2019). Different lines of behavioral some authors to describe the neurons coding the
investigations in humans showed an extension of surrounding space as “looming detectors” and the
the extent of multisensory interactions to include peripersonal space as a “bubble” around our body
the tool’s action space, hence demonstrating a that creates a margin of safety from approaching
dynamical shaping of the extension of the periper- dangers (Graziano and Cooke, 2006).
sonal space (Maravita and Iriki, 2004). Critically, Evidence in humans supporting the defensive
the extension of peripersonal space representation function of the peripersonal space comes from
is determined by where and how the tool is func- investigations of the hand blink reflex (HBR), a
tionally used, not by its physical length or structure contraction of the orbicularis oculi muscle elicited
(Farne et al., 2005). The plasticity of the periper- by a threatening touch to the hand (i.e., electrical
sonal space is bidirectional, since it can expand stimulation of the median nerve). The magnitude
or contract depending on the motor effort/ability of the HBR increases in a non-linear fashion with
and on the degree of limb use (Bassolino et al., the reduction of the distance between the hand and
2015; Lourenco and Longo, 2009). Peripersonal the face, as if, beyond a certain distance from the
space changes after tool-use emerge quickly, after face, the stimulus applied to the hand suddenly
few seconds of use (Holmes et al., 2007), but also becomes threatening (Sambo and Iannetti, 2013),
reverse quickly, disappearing after a few minutes unless there is a protection between the hand and
of no tool-use (Farne et al., 2007), a finding fur- the face (Sambo et al., 2012). Modeling the mag-
ther stressing the role of motor experience. On nitude of the HBR as a function of the position of
the other hand, extensive (daily use for long time the hand in the space around the head shows that
periods) and functionally relevant tool-use could the representation of the head-centered peripersonal
promote a much longer lasting expansion of the space is symmetrical on the horizontal axis, but not
peripersonal space, as found in the case of blind on the vertical one, resulting in a half-ellipsoidal
cane users (Serino et al., 2007). shape toward the top (Bufacchi et al., 2016). This
configuration is likely due to the fact that our inter-
actions with the world are shaped by physical laws,
including that of gravity. If we throw something, or
if something is thrown toward us, its trajectory will
DEFENSIVE FUNCTION OF PERIPERSONAL tend downward. It is therefore conceivable that even
SPACE a mechanism designed to protect our body takes
this property into consideration. In accordance with
The mechanism that quickly informs our motor this, by changing the posture of the participants, for
system about the presence of an object in the near example by making them lie down on their back,
space is also useful when a potential threat the peripersonal space reshapes, extending upward
approaches our body. Indeed, another main motor (Bufacchi and Iannetti, 2016).
PERIPERSONAL SPACE REPRESENTATION 443

Figure 27.6 (A) The hand blink reflex (HBR) with the hand far from (on the left) or near
(on the right) the face. (B) Half-ellipsoidal shape of the peripersonal space centered on
the head, as evidenced by the gradual reduction in magnitude of the HBR elicited by the
electrical stimulation of the hand as function of the proximity of the hand to the face. Note
the dramatic increase of the HBR when the hand is inside the peripersonal space surrounding
the face (closest dark areas).
Source: Modified from Bufacchi et al., 2016.

The extent of the peripersonal space is also the peripersonal space allows to learn new associa-
influenced by the identity of the approaching tions of stimuli in body part-centered coordinates.
stimulus, namely its valence and in particular its In a more general perspective, these findings show
threatening value, as well as by the characteristic that the peripersonal space mechanism might be
of the observer, such as the level of fear or anxi- exploited by a larger range of functions which
ety (de Haan et al., 2016; Poliakoff et al., 2007; needs to build on such a strategic information
Rossetti et al., 2015; Sambo and Iannetti, 2013). as the distance between body parts and external
For instance, we respond faster to tactile stimuli events (Zanini et al., 2021b).
on our hand if a spider is approaching that hand,
as compared to a butterfly, especially if we are
afraid of spiders; this occurs only for stimuli loom-
ing toward the hand, and not for stimuli moving
away. Similarly, cynophobic individuals react faster
SOCIAL SIDE OF THE PERIPERSONAL
to touches at their hand if they hear an approach- SPACE
ing growling dog, as compared to no dog-phobic
individuals. The examples indicate that the “safety” The peripersonal space representation is also
peripersonal space enlarges depending on what affected by social factors, such as the presence of
situation scares us, clearly supporting its defensive people near us and by the nature of interactions
functions whereby the peripersonal space acts by we have with them. Depending on the context,
preparing our motor reactions to approaching aver- peripersonal space boundary enlarges, shrinks, or
sive events. Conversely, approaching visual stimuli remaps in relation to others according to our and
with a positive valence reduce the extension of the their individual characteristics, such as familiarity,
peripersonal space facilitating our appetitive motor proximity, and personality traits (Bogdanova
responses (Spaccasassi et al., 2021). et al., 2021). The neural substrate of this social
To accurately serve defensive or appetitive side of the peripersonal space is possibly linked to
functions, the peripersonal space mechanism the above-mentioned mirror-like properties of
could also contribute to learning the positive and visuo-tactile neurons, by which they respond not
negative value of the stimuli. Such a feature would only to visual stimuli approaching the monkey’s
allow to learn associations in hand-centered coor- own body but even to visual stimuli approaching
dinates to update and exploit them when the hand the body of another person (Ishida et al., 2009), as
moves. Indeed, fear conditioning, a form of asso- well by the existence of neural subpopulations
ciative learning, has been shown to arise in hand- sensitive to the sector of space (within or outside
centered coordinates (Zanini et al., 2021b). Thus, the peripersonal space) where others’ actions
not only an external threat is encoded as a function occurs (Bonini et al., 2014). The activity of these
of its position relative to a specific body part, but neurons may explain some social effects linked to
444 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

the human peripersonal space, such as that touches PERIPERSONAL SPACE AND
on our hand are boosted by visual stimuli near SELF-REPRESENTATION
someone else’s hand (Brozzoli et al., 2013), or the
fact that social interactions (e.g. performing a task
with another person) and the way we perceive The sensation of owning a body and of its integrity
people near us modulate visual-tactile processing has as fundamental basis the continuous integration
in peripersonal space, in turn changing its extend of coherent multisensory signals coming not only
(Heed et al., 2010). For instance, if a nearby from the body, but also from the environment sur-
person is perceived as collaborative or moral, our rounding it. The multisensory integration mecha-
peripersonal space expands (Pellencin et al., nism underlying the peripersonal space indeed
2018). Even the amplitude of the HBR is enhanced contributes to the sense of body ownership and to
when the stimulated hand of the participant is other components of bodily self-consciousness
brought close to the face of another person and (Blanke et al., 2015). Much of the evidence sup-
this interpersonal modulation depends on the par- porting this view comes from the use of the experi-
ticipant’s ability to empathize with the other mental paradigm of the rubber hand illusion.
(Fossataro et al., 2016). Basically, the synchronous visuo-tactile stimulation
The social side of peripersonal space recalls of a fake hand, positioned next to the real (hidden)
the concept of personal space, also called inter- hand, induces in the participant the illusion of
personal space in social psychology, that is the owning the fake hand (Botvinick and Cohen,
space we keep between us and others to avoid 1998). The illusory feeling of owning the fake hand
discomfort. However, some caution must be used has been related to the activity of the neurons
in superimposing the peripersonal space to the coding for the peri-hand space and in particular to
interpersonal space, since they are dissociable and a re-tuning of peripersonal space neurons whose
show opposite dynamic features: the expansion of RFs are centered on the hand (Blanke et al., 2015;
the peripersonal space during positive social inter- Makin et al., 2008). During the rubber hand illu-
actions indeed is paralleled by a reduction of the sion, the activation of the tactile RF of these neu-
interpersonal distance (Pellencin et al., 2018). rons would be systematically associated, due to
Beyond other individuals, other social features synchronicity of the visuo-tactile stimulation, with
can modulate the peripersonal space representation. the activation of a visual RF not perfectly overlap-
When object ownership is at stake, for example, the ping the hand location. But thanks to the plastic
recruitment of peripersonal space may be modu- properties of the peripersonal space, the prolonged
lated when we grasp the object as a function of stroking of the hand coupled with visual stimula-
whether it belongs to us or to another person. Most tion from a farther location (namely, from the
notably, this ownership effect emerges also dur- rubber hand) is able to re-tune the visuo-tactile RFs
ing action observation, that is when we watch oth- of peripersonal space neurons toward the location
ers grasping their own object (Patane et al., 2020). of the visual inputs. Thus, changes in body owner-
Additionally, observing others’ actions induces a ship can be induced by manipulating the spatio-
sensory resonance (Bolognini et al., 2014b), sup- temporal coherence of multisensory bodily inputs
porting the view that one’s own peripersonal space processed by the peripersonal space system. This
also implements a mechanism to map others’ perip- account is strongly supported by neuropsychologi-
ersonal space; such mapping may allow the shar- cal evidence showing a remission from somatopara-
ing of a common reference frame to interact with phrenia, a somatic delusion usually following
the people around us and anticipate their actions right-hemisphere damage, which typically mani-
(Brozzoli et al., 2014). This evidence is in line with fests as a feeling of disownership of one’s contral-
electrophysiological work on nonhuman primates, esional body parts, by applying synchronous
showing agent selectivity of visuomotor neurons touches to the patient’s visible disowned hand and
in the monkey’s area F6. This neuronal population to the invisible unimpaired hand (Bolognini et al.,
responds selectively to the presentation of an object 2014a). Moreover, the induction and the strength of
as a function of whether it is grasped by the mon- the illusory feeling of owning a rubber hand corre-
key, by another agent, or by both. When the object late with the activity of neurons in the peripersonal
is presented outside of the monkey’s peripersonal space network, in particular at the level of the pre-
space, the same neurons do not discharge (Livi motor and parietal cortices (Ehrsson et al., 2004).
et al., 2019). The fact that ownership modulates the Convergent evidence that the peripersonal space
activity of peripersonal space network both dur- is involved in generating our feeling of body owner-
ing action execution and observation is compatible ship derives from another illusory effect, the enface-
with the notion that peripersonal space is sensitive ment illusion (Tsakiris, 2008). In this illusion, the
to both which object is grasped and who is the agent participant perceives a touch on the face while see-
of the action. ing a stranger’s face being touched, in a spatially
PERIPERSONAL SPACE REPRESENTATION 445

and temporally coherent way. After few seconds of advantage of fMRI-adaptation is to reveal the pres-
synchronous stimulation, the illusion of ownership ence of neuronal populations selective to a stimu-
extends to the stranger’s face. The onset of this illu- lus feature, such as its proximity to the hand.
sion is linked to the activation of the right intrapa-
rietal sulcus (Apps et al., 2015), the area putatively
corresponding to the monkey VIP region. This
area of the peripersonal space network is strongly
involved in the coding of tactile stimuli on the face REFERENCES
and of visual stimuli close to it, and its activation
during the enfacement illusion leads to recoding Aimola, L., I. Schindler, I., & Venneri A. (2012). Task-
the space near an unknown face as the peripersonal and response related dissociations between
space of one’s own face (Huang et al., 2012). neglect in near and far space: A morphometric
By applying a visuo-tactile stimulation similar case study. Behavioural Neurology, 27(3),
to that used for the rubber hand and the enface- 245–257.
ment illusions to the trunk it is also possible to Apps, M. A., Tajadura-Jimenez, A., Sereno, M.,
elicit the sensation of ownership toward a dif- Blanke, O., & Tsakiris, M. (2015). Plasticity in uni-
ferent whole body (body swap illusion; Petkova modal and multimodal brain areas reflects multi-
and Ehrsson, 2008), or a dislocation of the “self” sensory changes in self-face identification. Cerebral
toward a position external to one’s body (full- Cortex, 25(1), 46–55.
body illusion; Lenggenhager et al., 2007). These Avillac, M., Deneve, S., Olivier, E., Pouget, A., &
illusory experiences indicate that the peripersonal Duhamel, J. R. (2005). Reference frames for repre-
space does not necessarily reflect the position of senting visual and tactile locations in parietal
the physical body, rather it may reflect the position cortex. Nature Neuroscience, 8(7), 941–949.
of one’s self. Bassolino, M., Finisguerra, A., Canzoneri, E.,
Serino, A., & Pozzo, T. (2015). Dissociating effect
of upper limb non-use and overuse on space and
body representations. Neuropsychologia, 70,
SUMMARY AND CONCLUSIONS 385–392.
Berti, A., & Frassinetti, F. (2000). When far becomes
near: Remapping of space by tool use. Journal of
Our brain has inherited a highly flexible system to Cognitive Neuroscience, 12(3), 415–420.
represent the space closely surrounding the body, Berti, A., Smania, N., & Allport, A. (2001). Coding of
the peripersonal space. This space has a clearly far and near space in neglect patients. NeuroIm-
adapting meaning as it is where all physical inter- age, 14(1), S98–S102.
actions with objects in the environment occur: it Bisiach, E., Perani, D., Vallar, G., & Berti A. (1986).
allows to detect and anticipate potential interac- Unilateral neglect: personal and extra-personal.
tions between the body and an external event, Neuropsychologia, 24(6), 759–767.
triggering appropriate motor reactions thanks to Blanke, O., Slater, M., & Serino, A. (2015). Behavio-
its direct access to the motor system. The periper- ral, neural, and computational principles of bodily
sonal space features itself as an important antici- self-consciousness. Neuron, 88(1), 145–166.
patory sensory-motor interface mediating Bogdanova, O. V., Bogdanov, V. B., Dureux, A.,
body–object interactions for appetitive and defen- Farnè, A., & Hadj-Bouziane, F. (2021). The periper-
sive purposes. The plastic boundaries of the perip- sonal space in a social world. Cortex, 142,
ersonal space are efficiently and dynamically 28–46.
shaped by the ongoing sensorimotor experience, Bolognini, N., & Maravita, A. (2007). Proprioceptive
and taking into account environmental and social alignment of visual and somatosensory maps in
changes relevant for our body safety and the rep- the posterior parietal cortex. Current Biology,
resentation of ourselves as distinct from the envi- 17(21), 1890–1895.
ronment and the others. Bolognini, N., Ronchi, R., Casati, C., Fortis, P., &
Vallar, G. (2014a). Multisensory remission of
somatoparaphrenic delusion: My hand is back!
Note Neurology: Clinical Practice, 4(3), 216–225.
Bolognini, N., Rossetti, A., Fusaro, M., Vallar, G., &
1  Neuronal adaptation consists in a suppression of Miniussi, C. (2014b). Sharing social touch in the
the response of those neurons that are selective primary somatosensory cortex. Current Biology,
to one characteristic of the stimulation, when a 24(13), 1513–1517.
stimulus featuring such characteristic is presented Bonini, L., Maranesi, M., Livi, A., Fogassi, L., & Riz-
repeatedly (Grill-Spector et al., 2006). The main zolatti, G. (2014). Space-dependent representation
446 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

of objects and other’s action in monkey ventral Cléry, J., Guipponi, O., Wardak, C., & Hamed, S. B.
premotor grasping neurons. Journal of Neurosci- (2015). Neuronal bases of peripersonal and extrap-
ence, 34(11), 4108–4119. ersonal spaces, their plasticity and their dynamics:
Botvinick, M., & Cohen, J. (1998). Rubber hands Knowns and unknowns. Neuropsychologia, 70,
“feel” touch that eyes see. Nature, 391, 756. 313–326.
Brain, W. R. (1941). A form of visual disorientation Cléry, J., Guipponi, O., Odouard, S., Wardak, C., &
resulting from lesions of the right cerebral hemi- Ben Hamed, S. (2018). Cortical networks for
sphere. Proceedings of the Royal Society of Medi- encoding near and far space in the non-human
cine, 34(12), 771–776. primate. NeuroImage, 176, 164–178.
Bremmer, F., Schlack, A., Shah, N. J., Zafiris, O., Colby, C. L., Duhamel, J. R., & Goldberg, M. E.
Kubischik, M., Hoffmann, K., … Fink, G. R. (2001). (1993). Ventral intraparietal area of the macaque:
Polymodal motion processing in posterior parietal Anatomic location and visual response properties.
and premotor cortex: A human fMRI study strongly Journal of Neurophysiology, 69(3), 902–914.
implies equivalencies between humans and mon- Cooke, D. F., & Graziano, M. S. (2003). Defensive
keys. Neuron, 29(1), 287–296. movements evoked by air puff in monkeys. Journal
Brozzoli, C., Makin, T. R., Cardinali, L., Holmes, N. P., of Neurophysiology, 90(5), 3317–3329.
& Farnè, A. (2012a). Peripersonal space: A multi- Cooke, D. F., & Graziano, M. S. (2004a). Sensorimo-
sensory interface for body–object interactions. In tor integration in the precentral gyrus: Polysensory
M. M. Murray & M. T. Wallace (Eds.), The neural neurons and defensive movements. Journal of
bases of multisensory processes. Boca Raton, FL: Neurophysiology, 91(4), 1648–1660.
CRC Press/Taylor & Francis. Cooke, D. F., & Graziano, M. S. (2004b). Super-
Brozzoli, C., Cardinali, L., Pavani, F., & Farnè, A. flinchers and nerves of steel: Defensive move-
(2010). Action-specific remapping of peripersonal ments altered by chemical manipulation of a
space. Neuropsychologia, 48(3), 796–802. cortical motor area. Neuron, 43(4), 585–593.
Brozzoli, C., Ehrsson, H. H., & Farnè, A. (2014). Mul- Cowey, A., Small, M., & Ellis, S. (1994). Left visuo-
tisensory representation of the space near the spatial neglect can be worse in far than in near
hand: From perception to action and interindivid- space. Neuropsychologia, 32(9), 1059–1066.
ual interactions. Neuroscientist, 20(2), 122–135. de Haan, A. M., Smit, M., Van der Stigchel, S., &
Brozzoli, C., Gentile, G., Bergouignan, L., & Ehrsson, Dijkerman, H. C. (2016). Approaching threat
H. H. (2013). A shared representation of the space modulates visuotactile interactions in peripersonal
near oneself and others in the human premotor space. Experimental Brain Research, 234(7),
cortex. Current Biology, 23(18), 1764–1768. 1875–1884.
Brozzoli, C., Gentile, G., & Ehrsson, H. H. (2012b). di Pellegrino, G., Ladavas, E., & Farné, A. (1997).
That’s near my hand! Parietal and premotor Seeing where your hands are. Nature, 388,
coding of hand-centered space contributes to 730–730.
localization and self-attribution of the hand. Jour- Duhamel, J. R., Colby, C. L., & Goldberg, M. E.
nal of Neuroscience, 32(42), 14573–14582. (1998). Ventral intraparietal area of the macaque:
Brozzoli, C., Gentile, G., Petkova, V. I., & Ehrsson, H. Congruent visual and somatic response properties.
H. (2011). FMRI adaptation reveals a cortical Journal of Neurophysiology, 79(1), 126–136.
mechanism for the coding of space near the hand. Ehrsson, H. H., Spence, C., & Passingham, R. E.
Journal of Neuroscience, 31(24), 9023–9031. (2004). That’s my hand! Activity in premotor
Brozzoli, C., Pavani, F., Urquizar, C., Cardinali, L., & cortex reflects feeling of ownership of a limb. Sci-
Farnè, A. (2009). Grasping actions remap periper- ence, 305, 875–877.
sonal space. Neuroreport, 20(10), 913–917. Farnè, A., Demattè, M. L., & Làdavas, E. (2005). Neu-
Bufacchi, R. J., & Iannetti, G. D. (2016). Gravitational ropsychological evidence of modular organization
cues modulate the shape of defensive peripersonal of the near peripersonal space. Neurology, 65(11),
space. Current Biology, 26(21), R1133–R1134. 1754–1758.
Bufacchi, R. J., Liang, M., Griffin, L. D., & Iannetti, G. D. Farnè, A., Serino, A., & Làdavas, E. (2007). Dynamic
(2016). A geometric model of defensive periper- size-change of peri-hand space following tool-use:
sonal space. Journal of Neurophysiology, 115(1), Determinants and spatial characteristics revealed
218–225. through cross-modal extinction. Cortex, 43(3),
Caminiti, R., Johnson, P. B., Galli, C., Ferraina, S., & 436–443.
Burnod, Y. (1991). Making arm movements within Ferri, F., Costantini, M., Huang, Z., Perrucci, M. G.,
different parts of space: The premotor and motor Ferretti, A., Romani, G. L., & Northoff, G. (2015).
cortical representation of a coordinate system for Intertrial variability in the premotor cortex accounts
reaching to visual targets. Journal of Neurosci- for individual differences in peripersonal space.
ence, 11(5), 1182–1197. Journal of Neuroscience, 35(50), 16328–16339.
PERIPERSONAL SPACE REPRESENTATION 447

Fogassi, L., Gallese, V., di Pellegrino, G., Fadiga, L., stimulus-specific effects. Trends in Cognitive Sci-
Gentilucci, M., L Luppino, G., … Rizzolatti, G. ences, 10(1), 14–23.
(1992). Space coding by premotor cortex. Experi- Halligan, P. W., & Marshall, J. C. (1991). Left neglect
mental Brain Research, 89(3), 686–690. for near but not far space in man. Nature, 350,
Fogassi, L., Gallese, V., Fadiga, L., Luppino, G., 498–500.
Matelli, M., & Rizzolatti, G. (1996). Coding of Halligan, P. W., Fink, G. R., Marshall, J. C., & Vallar,
peripersonal space in inferior premotor cortex G. (2003). Spatial cognition: Evidence from visual
(Area F4). Journal of Neurophysiology, 76(1), neglect. Trends in Cognitive Sciences, 7(3),
141–157. 125–133.
Fogassi, L., & Luppino, G. (2005). Motor functions of Heed, T., Habets, B., Sebanz, N., & Knoblich, G.
the parietal lobe. Current Opinion in Neurobiol- (2010). Others’ actions reduce crossmodal integra-
ogy, 15(6), 626–631. tion in peripersonal space. Current Biology, 20(15),
Fogassi, L., Raos, V., Franchi, G., Gallese, V., Luppino, 1345–1349.
G., & Matelli, M. (1999). Visual responses in the Holmes, N. P., Calvert, G. A., & Spence, C. (2007).
dorsal premotor Area F2 of the macaque monkey. Tool use changes multisensory interactions in sec-
Experimental Brain Research, 128(1–2), 194–199. onds: Evidence from the crossmodal congruency
Fossataro, C., Sambo, C. F., Garbarini, F., & Iannetti, task. Experimental Brain Research, 183(4),
G. D. (2016). Interpersonal interactions and empa- 465–476.
thy modulate perception of threat and defensive Huang, R. S., Chen, C. F., Tran, A. T., Holstein, K. L.,
responses. Scientific Reports, 6, doi: 10.1038/ & Sereno, M. I. (2012). Mapping multisensory
srep19353. parietal face and body areas in humans. Proceed-
Gentile, G., Guterstam, A., Brozzoli, C., & Ehrsson, H. ings of the National Academy of Sciences
H. (2013). Disintegration of multisensory signals of the United States of America, 109(44),
from the real hand reduces default limb self-attri- 18114–18119.
bution: An fMRI study. Journal of Neuroscience, Hyvarinen, J. (1981). Regional distribution of func-
33(33), 13350–13366. tions in parietal association Area 7 of the monkey.
Gentile, G., Petkova, V. I., & Ehrsson, H. H. (2011). Brain Research, 206(2), 287–303.
Integration of visual and tactile signals from the Hyvarinen, J., & Poranen, A. (1974). Function of the
hand in the human brain: An fMRI study. Journal parietal associative Area 7 as revealed from cellular
of Neurophysiology, 105(2), 910–922. discharges in alert monkeys. Brain, 97(4),
Gentilucci, M., Fogassi, L., Luppino, G., Matelli, M., 673–692.
Camarda, R., & Rizzolatti, G. (1989). Somatotopic Hyvarinen, J., & Shelepin, Y. (1979). Distribution of
representation in inferior Area 6 of the macaque visual and somatic functions in the parietal asso-
monkey. Brain, Behavior and Evolution, 33(2–3), ciative Area 7 of the monkey. Brain Research,
118–121. 169(3), 561–564.
Gentilucci, M., Scandolara, C., Pigarev, I. N., & Riz- Iriki, A., Tanaka, M., & Iwamura, Y. (1996). Coding of
zolatti, G. (1983). Visual responses in the post- modified body schema during tool use by
arcuate cortex (Area 6) of the monkey that are macaque postcentral neurones. Neuroreport,
independent of eye position. Experimental Brain 7(14), 2325–2330.
Research, 50(2–3), 464–468. Ishida, H., Nakajima, K., Inase, M., & Murata, A.
Graziano, M. S. (2001). A system of multimodal areas (2009). Shared mapping of own and others’
in the primate brain. Neuron, 29(1), 4–6. bodies in visuotactile bimodal area of monkey
Graziano, M. S., & Cooke, D. F. (2006). Parieto-fron- parietal cortex. Journal of Cognitive Neuroscience,
tal interactions, personal space, and defensive 22(1), 83–96.
behavior. Neuropsychologia, 44(13), 2621–2635. Làdavas, E., Pellegrino, G. D., Farnè, A., & Zeloni, G.
Graziano, M. S., & Gross, C. G. (1993). A bimodal (1998). Neuropsychological evidence of an inte-
map of space: Somatosensory receptive fields in grated visuotactile representation of peripersonal
the macaque putamen with corresponding visual space in humans. Journal of Cognitive Neurosci-
receptive fields. Experimental Brain Research, ence, 10(5), 581–589.
97(1), 96–109. Làdavas, E., & Farnè, A. (2004). Visuo-tactile repre-
Graziano, M. S., Hu, X. T., & Gross, C. G. (1997). sentation of near-the-body space. Journal of Physi-
Visuospatial properties of ventral premotor cortex. ology, 98(1–3), 161–170.
Journal of Neurophysiology, 77(5), 2268–2292. Lane, A. R., Ball, K., Smith, D. T., Schenk, T., & Ellison,
Graziano, M. S., Yap, G. S., & Gross, C. G. (1994). A. (2013). Near and far space: Understanding the
Coding of visual space by premotor neurons. Sci- neural mechanisms of spatial attention. Human
ence, 266, 1054–1057. Brain Mapping, 34(2), 356–366.
Grill-Spector, K., Henson, R., & Martin, A. (2006). Leinonen, L. (1980). Functional properties of neu-
Repetition and the brain: Neural models of rones in the posterior part of Area 7 in awake
448 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

monkey. Acta Physiologica Scandinavica, 108(3), remapping of peripersonal space: The case of
301–308. walking. Neuropsychologia, 70, 375–384.
Leinonen, L., & Nyman, G. (1979). II. Functional prop- Patanè, I., Brozzoli, C., Koun, E., Frassinetti, F., &
erties of cells in anterolateral part of Area 7 asso- Farnè, A. (2020). Me, you, and our object: Perip-
ciative face area of awake monkeys. Experimental ersonal space recruitment during executed and
Brain Research, 34(2), 321–333. observed actions depends on object ownership.
Lenggenhager, B., Tadi, T., Metzinger, T., & Blanke, O. Journal of Experimental Psychology: General. doi:
(2007). Video ergo sum: Manipulating bodily self- 10.1037/xge0001001
consciousness. Science, 317, 1096–1099. Pellencin, E., Paladino, M. P., Herbelin, B., & Serino,
Livi, A., Lanzilotto, M., Maranesi, M., Fogassi, L., Riz- A. (2018). Social perception of others shapes one’s
zolatti, G., & Bonini, L. (2019). Agent-based repre- own multisensory peripersonal space. Cortex, 104,
sentations of objects and actions in the monkey 163–179.
pre-supplementary motor area. Proceedings of the Petkova, V. I., & Ehrsson, H. H. (2008). If I were you:
National Academy of Sciences of the United States Perceptual illusion of body swapping. PLoS One,
of America, 116(7), 2691–2700. 3(12). doi: 10.1371/journal.pone.0003832.
Lourenco, S. F., & Longo, M. R. (2009). The plasticity Poliakoff, E., Miles, E., Li, X., & Blanchette, I. (2007).
of near space: Evidence for contraction. Cognition, The effect of visual threat on spatial attention to
112(3), 451–456. touch. Cognition, 102(3), 405–414.
Makin, T. R., Holmes, N. P., Brozzoli, C., Rossetti, Y., Previc, F. H. (1990). Functional specialization in the
& Farnè, A. (2009). Coding of visual space during lower and upper visual fields in humans: Its eco-
motor preparation: Approaching objects rapidly logical origins and neurophysiological implications.
modulate corticospinal excitability in hand-cen- Behavioral and Brain Sciences, 13(3), 519–542.
tered coordinates. Journal of Neuroscience, Rizzolatti, G., Fadiga, L., Fogassi, L., & Gallese, V.
29(38), 11841–11851. (1997). The space around us. Science, 277,
Makin, T. R., Holmes, N. P., & Ehrsson, H. H. (2008). 190–191.
On the other hand: dummy hands and periper- Rizzolatti, G., Luppino, G., & Matelli, M. (1998). The
sonal space. Behavioural Brain Research, 191(1), organization of the cortical motor system: New
1–10. concepts. Electroencephalography and Clinical
Makin, T. R., Holmes, N. P., & Zohary, E. (2007). Is Neurophysiology, 106(4), 283–296.
that near my hand? Multisensory representation Rizzolatti, G., Scandolara, C., Gentilucci, M., &
of peripersonal space in human intraparietal Camarda, R. (1981a). Response properties and
sulcus. Journal of Neuroscience, 27(4), 731–740. behavioral modulation of “mouth” neurons of the
Maravita, A., & Iriki, A. (2004). Tools for the body postarcuate cortex (area 6) in macaque monkeys.
(schema). Trends in Cognitive Sciences, 8(2), Brain Research, 225(2), 421–424.
79–86. Rizzolatti, G., Scandolara, C., Matelli, M., & Genti-
Maravita, A., Spence, C., & Driver, J. (2003). Multi- lucci, M. (1981b). Afferent properties of periarcu-
sensory integration and the body schema: Close to ate neurons in macaque monkeys. I. Somatosensory
hand and within reach. Current Biology, 13(13), responses. Behavioural Brain Research, 2(2),
R531–539. 125–146.
Matelli, M., Luppino, G., & Rizzolatti, G. (1985). Pat- Rizzolatti, G., Scandolara, C., Matelli, M., & Genti-
terns of cytochrome oxidase activity in the frontal lucci, M. (1981c). Afferent properties of periarcu-
agranular cortex of the macaque monkey. Behav- ate neurons in macaque monkeys. II. Visual
ioural Brain Research, 18(2), 125–136. responses. Behavioural Brain Research, 2(2),
Mattingley, J. B., Driver, J., Beschin, N., & Robertson, 147–163.
I. H. (1997). Attentional competition between Rossetti, A., Romano, D., Bolognini, N., & Maravita,
modalities: Extinction between touch and vision A. (2015). Dynamic expansion of alert responses
after right hemisphere damage. Neuropsycholo- to incoming painful stimuli following tool use.
gia, 35(6), 867–880. Neuropsychologia, 70, 486–494.
Nasr, S., & Tootell, R. B. (2018). Visual field biases for Sambo, C. F., Forster, B., Williams, S. C., & Iannetti,
near and far stimuli in disparity selective columns G. D. (2012). To blink or not to blink: Fine cogni-
in human visual cortex. NeuroImage, 168, tive tuning of the defensive peripersonal space.
358–365. Journal of Neuroscience, 32(37), 12921–12927.
Nasr, S., & Tootell, R. B. (2020). Asymmetries in Sambo, C. F., & Iannetti, G. D. (2013). Better safe
global perception are represented in near-versus than sorry? The safety margin surrounding the
far-preferring clusters in human visual cortex. Jour- body is increased by anxiety. Journal of Neurosci-
nal of Neuroscience, 40(2), 355–368. ence, 33(35), 14225–14230.
Noel, J. P., Grivaz, P., Marmaroli, P., Lissek, H., Blanke, Senna, I., Cardinali, L., Farnè, A., & Brozzoli, C.
O., & Serino, A. (2015). Full body action (2019). Aim and plausibility of action chains remap
PERIPERSONAL SPACE REPRESENTATION 449

peripersonal space. Frontiers in Psychology, 10. Stein, B. E., & Stanford, T. R. (2008). Multisensory
doi: 10.3389/fpsyg.2019.01681 integration: Current issues from the perspective of
Serino, A. (2019). Peripersonal space (PPS) as a mul- the single neuron. Nature Reviews: Neuroscience,
tisensory interface between the individual and the 9(4), 255–266.
environment, defining the space of the self. Tsakiris, M. (2008). Looking for myself: Current mul-
Neuroscience and Biobehavioral Reviews, 99, tisensory input alters self-face recognition. PLoS
138–159. One, 3(12). doi: 10.1371/journal.pone.0004040.
Serino, A., Bassolino, M., Farnè, A., & Làdavas, E. Vallar, G., & Bolognini, N. (2014). Unilateral spatial
(2007). Extended multisensory space in blind cane neglect. In A. C. Nobre & S. Kastner (Eds.), The
users. Psychological Science, 18(7), 642–648. Oxford handbook of attention. Oxford, UK: Oxford
Serino, A., Noel, J. P., Galli, G., Canzoneri, E., Mar- University Press.
maroli, P., Lissek, H., & Blanke, O. (2015). Body Vuilleumier, P., Valenza, N., Mayer, E., Reverdin, A., &
part-centered and full body-centered peripersonal Landis, T. (1998). Near and far visual space in uni-
space representations. Scientific Reports, 5. lateral neglect. Annals of neurology, 43(3),
doi: 10.1038/srep18603 406–410.
Spaccasassi, C., Dijkerman, H. C., Maravita, A., Fer- Weiss, P. H., Marshall, J. C., Wunderlich, G., Tell-
rante, O., & de Jong, M. C. (2021). Body-space mann, L., Halligan, P. W., Freund, H. J., … Fink, G.
interactions: same spatial encoding but different R. (2000). Neural consequences of acting in near
influence of valence for reaching and defensive versus far space: A physiological basis for clinical
purposes. Journal of Cognitive Neuroscience, dissociations. Brain, 123(12), 2531–2541.
33(10), 2149–2166. Zanini, A., Patanè, I., Blini, E., Salemme, R., Koun, E.,
Spence, C., Pavani, F., & Driver, J. (2004a). Spatial Farnè, A., & Brozzoli, C. (2021a). Peripersonal and
constraints on visual-tactile cross-modal distractor reaching space differ: Evidence from their spatial
congruency effects. Cognitive, Affective, & Behav- extent and multisensory facilitation pattern. Psy-
ioral Neuroscience, 4(2), 148–169. chonomic Bulletin & Review. doi: 10.3758/
Spence, C., Pavani, F., Maravita, A., & Holmes, N. s13423-021-01942-9.
(2004b). Multisensory contributions to the 3-D Zanini, A., Salemme, R., Farnè, A., & Brozzoli, C.
representation of visuotactile peripersonal space in (2021b). Associative learning in peripersonal
humans: Evidence from the crossmodal congru- space: Fear responses are acquired in hand-­
ency task. Journal of Physiology, 98(1–3), centered coordinates. Journal of Neurophysiology,
171–189. 126(3), 864–874.
This page intentionally left blank
PART VI
Methodological Advances
This page intentionally left blank
28
Neural Recordings and
Time Series Analyses
R a n d o l p h F. H e l f r i c h a n d R o b e r t T. K n i g h t

INTRODUCTION et al., 2013) in terms of spatiotemporal resolution


and approaches the spatial resolution of functional
Modern day neuroimaging provides unprece- magnetic resonance imaging (fMRI). This wealth
dented spatiotemporal resolution to study cogni- of data poses a challenge for extraction of mean-
tive processing at the level of whole-brain ingful information about the functional architec-
large-scale network interactions. For decades ture underlying human perception, cognition and
whole-head electroencephalography (EEG) was action.
restricted to only 19 channels as defined by the In this chapter, we focus on analysis techniques
international 10–20 electrode layout (a practice of time series data. First, we provide a brief over-
that prevails in the clinical setting). In contrast, view of current methods that enable imaging the
modern day amplifiers enable simultaneous human brain with high spatial and temporal reso-
recordings of a few hundred channels (Biasiucci lution. Throughout the chapter we emphasize that
et al., 2019; Cohen, 2017; Lopes da Silva, 2013). time series analyses can be applied to different
For example, current high-density EEG systems types of electrophysiological data. Second, we
offer up to 256 channels, which can be sampled at review analyses strategies for high-dimensional
>1,000 data points per second. When combined time-series data. Methods are introduced accord-
with inverse solutions to project 2D sensor-level ing to their practical importance during data anal-
data into 3D source space, as informed by indi- ysis (i.e., univariate analysis approaches in the
vidual high-resolution structural MRI (magnetic time-domain are covered first, before advancing
resonance imaging) scans, one can obtain densely into spectral decomposition, bivariate connectivity
sampled time series data for >4,000 voxels inside analyses, and finally multivariate analysis strate-
the brain at 1 mm3 resolution. A thorough discus- gies). We then review methods that go beyond
sion of inverse solutions exceeds the scope of this established linear time- and/or frequency analyses
chapter, for a detailed discussion see (Lopes da and discuss non-linear approaches, including infor-
Silva, 2013; Pascual-Marqui et al., 1994; Van mation-theoretical approaches as well as recent
Veen et al., 1997). This development now places machine-learning inspired strategies. Finally, we
high-density EEG recordings on par with magne- take recent developments of the last five years into
toencephalography (MEG; Baillet, 2017; Gross account, as exemplified by strategies to analyze
454 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

background “noise,” which has recently been will not be reviewed in detail (Biasiucci et al.,
shown to contain important behaviorally relevant 2019), we provide a more in-depth account of
information. In addition, we highlight how analy- intracranial recordings in humans (Fried et al.,
sis strategies can be synergistically combined to 2014; Parvizi and Kastner, 2018).
maximize insight into neurophysiological pro- Intracranial electrophysiological signals can be
cesses underlying human cognition. Throughout obtained from electrodes placed within the human
the chapter, we highlight potential caveats, with brain for diagnostic and/or therapeutic purposes
the goal to provide a roadmap for state-of-the-art (Figure 28.1). The two most common entities that
electrophysiological data analysis. require a surgical placement of leads in the human
brain are either placement to deliver therapeutic
deep brain stimulation electrodes for Parkinson’s
disease (target: subthalamic nucleus or internal
globus pallidus; Bronstein et al., 2011), dystonia
NEURAL RECORDINGS (ventral intermediate nucleus of the thalamus) or
epilepsy (anterior nucleus of the thalamus; Fisher
Traditionally, neural recordings mainly referred to et al., 2010)). In addition, implanted electrodes
invasive single unit and local field potential record- are used to guide surgical decision-making for
ings in animal models (Buzsáki et al., 2012). In the intratractable epilepsy (Parvizi and Kastner, 2018).
human literature, there is often a distinction Here, leads are inserted into multiple nodes of the
between direct neurophysiological recordings, as suspected seizure network to identify the seizure
for instance EEG, and indirect measures, such as onset zone. Target areas are identified according
the fMRI BOLD (blood oxygen level dependent) to the non-invasive work-up, which includes scalp
response. The goal of this chapter is not to provide EEG, high-resolution imaging and neuropsychol-
an exhaustive list of imaging modalities, but rather ogy and can be complemented by various other
to survey the analytical possibilities in the context diagnostic tools, including positron-emission
of time-series analyses and highlight the similari- tomography (PET), voxel-based morphometry
ties between different methods. To accomplish (VBM), or MEG (Zijlmans et al., 2019). An impor-
this, we adopt a liberal definition of neural record- tant feature of some electrodes is that they feature
ings, which encompasses every method that ena- a hollow lumen, enabling insertion of additional
bles quantification of a brain process, irrespective wire bundles, which protrude by 2–4 mm at the
whether it refers to direct electrophysiological electrode tip for recording local field potentials and
recordings (voltage differences in scalp and intrac- unit activity (Fried et al., 2014). Several studies
ranial EEG), magnetic fields (MEG), indirect have demonstrated the feasibility and safety of this
oxygen-dependent responses (fMRI BOLD) or approach (Carlson et al., 2018; Chari et al., 2020;
functional near-infrared spectroscopy; fNIRS) or Despouy et al., 2020; Hefft et al., 2013). Over the
motor signals that are obtained at the output stage, last decade, intracranial recordings in humans have
such as time-resolved behavior (quantified by yielded important insights into the functional archi-
time-resolved hit rates or reaction times), electro- tecture of cognition, such that the method is now
myography (EMG; voltage trace) or pupillometry widely regarded as an ideal tool to bridge the gap
and pupil size (recorded in millimeters, degree between invasive recordings in animal models and
angles or pixels). These considerations can also be non-invasive recordings in humans.
extrapolated to other types of recordings, such as Ultimately, only a few factors determine the
two-photon calcium imaging data in rodents. One application of time-series analysis strategies.
exception that we discuss in detail is that certain Foremost are the sampling rate and the duration of
analysis approaches are limited by the sampling the recordings. These factors determine the result-
rate and recording location, such as extracting ing Nyquist frequency (the highest frequency that
single unit responses from continuous local field can be resolved from the data is half the sampling
potential recordings, which require recordings at a rate – at 1,000 Hz sampling rate, all frequencies up
fine-grained spatial scale at high temporal to 500 Hz can be resolved; practically one should
resolution above 30,000 Hz. rather aim for a 3rd or 4th of the sampling rate).
To illustrate the analytical approaches, we focus The duration further determines the Rayleigh fre-
on electrophysiological recordings in humans, quency resolution, which is defined by 1 divided
both by means of scalp EEG as well as intracra- by the temporal window. For example, a two
nial electrophysiology, which encompasses intrac- second segment at 1,000 Hz can be analyzed in
ranial EEG, local field potentials, and single unit steps of 0.5 Hz (1/2), while a 10 second segment
activity (Parvizi and Kastner, 2018). While most allows a more fine-grained resolution at 0.1 Hz
readers will be familiar with scalp EEG, which (1/10). The signal duration also determines the
Neural Recordings and Time Series Analyses 455

Figure 28.1 Intracranial human electrophysiology.


(A) Upper row: Subdural grid electrode placement from three representative subjects cover-
ing large portions of frontal, temporal, and motor areas. Lower row: Stereotactically placed
depth electrodes targeting medial temporal, orbitofrontal, or medial frontal structures. (B)
Single unit activity can be recorded from additional wire bundles that are inserted through a
hollow lumen of the clinical macro-electrode.
Source: Figure reproduced with permission (Helfrich & Knight, 2019).

digital filtering that can be applied to the data. The Keeping those theoretical principles in mind
lower cut-off needs to match at the least one whole now enables applying spectral analyses to dif-
cycle of the lower boundary (i.e., filtering a 1 s ferent recording modalities with theoretical
second segment at 1 Hz is theoretically possible, and practical implications (Prerau et al., 2017).
but pushes the limits of signal processing leading For example, intracranial EEG data that was
to unstable results). However, filtering at a lower recorded over one hour at a frequency resolution
cut-off of 10 Hz (10 cycles in 1 second) is feasible, of 5,000 Hz can easily be filtered and spectrally
while a cut-off of 0.5 Hz is impossible (only half decomposed in a broad-range of frequencies
a cycle can be fit into the segment). Lastly, one up to 2,500 Hz at a fine-grained resolution of
needs to be aware of the noise floor of both the ≪0.1 Hz (in practice an upper threshold of 250
environment as well the recording equipment. For Hz and a resolution of 0.5 Hz is often sufficient).
typical EEG amplifiers, the noise floor where the In contrast, whole-brain fMRI BOLD over one
amplifier yields meaningful results is > 100 Hz. hour typically provides one data point per voxel
However, the environmental noise floor is already every second, thus, resulting in a frequency
present at ∼30–40 Hz given muscle activity in this resolution of 0.33 Hz, which limits both the
recording range. This issue is largely mitigated ability to filter as well as to spectrally decom-
in intracranial EEG recordings, which are less pose the data (Fox et al., 2005). For fMRI using
impacted by muscle artifacts. For instance, analy- these parameters, he upper frequency cut-off is
ses in the high-frequency activity band (70–200 at 0.16 Hz, so frequencies between 0.01 and 0.1
Hz) have yielded important insights into cognitive Hz can best analyzed at a resolution in the 10-2
functioning (Leszczyński et al., 2020). to 10-3 Hz range.
456 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

ELECTROPHYSIOLOGICAL DATA events. In the context of EEG, averaging can be


ANALYSIS done on the raw trace, thus, yielding event-related
potentials (ERP; Handy, 2005). Likewise, averag-
ing can be performed after extraction of a distinct
The improved spatiotemporal resolution of human spectral component. For example, it had been
electrophysiological recordings has benefited the observed that the 70–150 Hz (termed high gamma
development of numerous algorithms and meth- or high frequency band activity, HFA/HFB) in
ods to extract behaviorally meaningful informa- intracranial EEG recordings reflects a proxy of
tion. However, these high-dimensional and multi-unit activity and contains more behaviorally
complex data sets that are governed by non-linear relevant information than the broadband signal
dynamics, posing unique challenges. For exam- (Edwards et al., 2005; Flinker et al., 2015; Kanth
ple, there are multiple correct answers for ques- and Ray, 2020; Leszczyński et al., 2020; Ray and
tions regarding (1) how to analyze the data, (2) Maunsell, 2011; Rich and Wallis, 2017). Thus,
which method to choose, and (3) how to statisti- filtering and extracting the signal envelope by
cally quantify the results. More commonly, the means of (e.g., a Hilbert transform; Figure 28.2),
right answer will be “it depends.” Here, we pro- are used to distil the relevant signal component
vide a practical road map with an initial focus on prior to subsequent averaging (Figure 28.2A).
EEG analyses that helps narrow the immense In continuous task designs, such as resting
search space and justify analytical choices and state or sleep recordings, there are no external
their interpretations. temporal structure for time locking analyses.
Therefore, researchers rely on identification of
intrinsically generated events for subsequent
analysis. Many events have first been described in
GENERAL ANALYSIS STRATEGIES the time domain (e.g., alpha waves by Berger as
characteristic 10 Hz bursts of activity in the raw
Data analysis does not begin once data collection trace). Likewise, slow waves (< 4 Hz) and sleep
ended, but rather starts with data recording and the spindles (∼12–16 Hz) have been identified based
experimental structure. During data recording, on the characteristic waveform shape in continu-
researchers make a number of explicit (e.g., ous recordings (Buzsáki, 1996; Diekelmann and
number of EEG sensors, sampling rate) as well as Born, 2010). Detection of these events in the
implicit choices (task design, duration of trials, time domain can be algorithmically formalized,
event structure) that limit the subsequent analyses. typically by introducing constrains with respect to
Important considerations include whether distinct (a) the frequency content (through band pass fil-
events are present (task-based time-locked events tering), (b) an amplitude criterion, (c) a duration
that can be contrasted) or whether a continuous criterion, and (d) exclusion of unrelated activity
design (e.g., resting state or experience sampling) (Helfrich et al., 2019; Staresina et al., 2015). For
were employed. Within the historical context of example, detection of sleep spindles only consid-
EEG, Hans Berger’s seminal experiments on ers bursts in the amplitude series of the 12–16 Hz
alpha oscillations (cf. Berger, 2004) first consti- filtered signal that exceed a z-score of (e.g., 3 SDs
tuted a continuous analysis (spontaneous fluctua- above the signal mean) for anywhere between 0.5
tions in the trace were quantified). Subsequently, to 3 seconds. Based on the individual detections,
he employed an event-locked approach (eyes averaging can be performed in the time domain to
open/close), which later gave rise to the discovery obtain the average waveform shape (Figure 28.3).
of event-related potentials by averaging across Furthermore, the detected event can be con-
several repetitions (Polich, 2007). To illustrate ceptualized as a point process, i.e. an event that
these methods, we will assume a task-based occurred precisely at one moment in time (e.g.,
design with distinct events that require a behavio- activity peak). To again employ the example of
ral response and thereby, enable time-locked sleep spindles, time-locking of the raw signal
analyses either relative to the event or the action. relative to spindle peaks reveals the presence of a
second spectral signatures in the raw signal. The
spindle does not occur in isolation, but is nested
with a slow wave. This approach exemplifies how
detection of endogenous temporal events can
EVENT-RELATED ANALYSIS IN THE
reveal temporal regularities that otherwise could
TIME-DOMAIN not be detected from the raw trace.
The same principle applies to the extraction
Averaging across several events is typically done of single unit spikes from broadband local field
to improve the signal-to-noise ratio and to quan- potentials (LFP). LFPs are sampled at >30,000
tify the average response across a number of Hz, spikes are again extracted based on amplitude
Neural Recordings and Time Series Analyses 457

signal has revealed that spikes preferentially occur


at distinct phases of the underlying population
oscillation, which are already present in the down-
sampled signals. Hence, subsequent analyses can
be carried out on signals sampled at 1000 Hz and
not 30,000 Hz, enhancing computational effi-
cacy and speed. Furthermore, from these point
events (“Spikes”), peri-stimulus time histograms
(PTSH) can be created, which are commonly
smoothed into a trace of overall spiking activity;
hence, yielding a signal that is comparable to an
event-related potential, only based on a number of
distinct neuronal spikes. Collectively, this demon-
strates how temporal events can be extracted from
continuous data and then again be analyzed in a
time-locked fashion, either relative to external or
internal events.

SPECTRAL ANALYSIS

Electrophysiological signals are rich and com-


plex. Ever since Berger’s seminal observation, the
community was well aware that different fre-
quency bands might contain distinct information.
Over recent decades, several methods were intro-
duced to either spectrally decompose the signal to
obtain activity in multiple frequency bands or to
isolate activity in a distinct frequency band.
The most common approach is based on the
Fourier transformation, which decomposes the
signal into (co-)sines and provides estimates of
Figure 28.2 High frequency band activity. the relative contribution of every frequency band
to the entire signal. Using this approach, the time
(A) Average high frequency band activity domain is typically lost (translation of time- into
recorded from a motor cortex electrode frequency-domain), but can be recovered by means
relative to the detection of the onset of a of repeating the analysis in different time windows
visual target reveals activity after approxi- by means of convolution. Electrophysiological
mately 500ms. (B) Stacked single trials that power spectra exhibit distinct characteristics,
are sorted relative to reaction times (black including a steep 1/fx drop-off, where x typically
scales in the range from –2 to –4 in the healthy
trace) reveals that HFA tracks behavior on a
brain (for extended discussion see section on
single trial basis. aperiodic activity; He et al. (2010); Miller et al.
Source: Unpublished (Helfrich & Knight). (2009)). Band-limited oscillations arise as distinct
“bumps” (Donoghue et al., 2020) above the back-
ground activity (Figure 28.4).
and time criteria (sharp transient in the range In addition to the Fourier transform, several
from 2–4 ms) before all waveform shapes are fur- other methods have been introduced, such as
ther characterized in a process called spike sort- Morlet wavelets (sines multiplied with Gaussians,
ing (Fried et al., 2014; Rutishauser et al., 2006). which are convoluted with the signal) or the
Here, different waveform shapes are disentangled Hilbert transform, which requires band-pass fil-
in order to isolate activity from distinct neurons. tering first and then estimates the instantaneous
Subsequently, every spike from every identified amplitude and phase series. One important caveat
neuron is regarded as one distinct time stamp; thus, is that all spectral decomposition techniques only
the dimensionality of the data has been reduced, provide “estimates,” which differ as a function
benefiting subsequent analyses. For example, of (e.g., signal length, sampling rate, noise and
spike-triggered averaging of the raw broadband pre-processing). A common approach to improve
458 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 28.3 Visualization of cross-frequency coupling and raw waveform shapes.


(A) Slow waves in a group of older adults (black) on top of the respective time-frequency
decomposition, indicating an activity peak in the ∼14 Hz spindle range just prior to the slow
wave peak. Inset: Visualization in circular space of the same data, indicating that spindle
activity peaked prior to the slow wave peak (approx. 45°, corresponding to 50–100ms).
(B) Same visualization for a cohort of younger adults, indicating that spindles are precisely
locked to the peak of the slow wave. (C) Visualization relative to detected spindles (colored)
with the slow wave superimposed in black. Note that spindle peak prematurely. Inset:
Illustration of the spindle peak relative to the slow wave (as exemplified by a cosine).
(D) Histogram-based visualization of coupling: Relative amplitude in the spindle-band rela-
tive to different binned slow-wave phases, reveals a non-uniform distribution that is skewed
in older adults. Note all panels depict the same data and illustrate different approaches to
cross-frequency coupling and waveform shape analyses.
Source: Figure reproduced with permission from (Helfrich, Mander, et al., 2018).

estimates is to “window” the data (also called are not sinusoidal in nature, but are characterized
taper; multiplication of a data segment with, e.g., by biased rise- and decay-times with skewed and
Boxcar window, where the edges are Gaussian often sharp waveform shapes (Cole and Voytek,
shaped, thus, attenuating edge artifacts). The 2017). Application of sinusoidal methods can
signal-to-noise ratio can be improved if multiple introduce severe artifacts, which are prone to
windows are combined and estimates are averaged misinterpretation (Aru et al., 2015; Gerber et al.,
(as for instance done when using Welch’s method 2016). One example is sensorimotor Mu-Rhythm
or the multi-taper approach based on discrete pro- at ∼8–12 Hz, which is named “Mu” given that is
late spheroidal sequences (Prerau et al., 2017; often shaped like a “M.” The Fourier transform
Figure 28.5). will extract the main component at ∼10 Hz, but
Another major drawback that all methods share also yield peaks at ∼20 Hz as well as all other
is that they are based on sinusoidal basis functions subsequent harmonics, given that the sharp peak
(i.e., if these methods are applied to brain data, is incompletely captured by a single sine wave
then the results will be systematically biased to (Voytek et al., 2010). In electrophysiological
reveal sinusoidal oscillations). Inspection of raw recordings, researchers are then faced with the
EEG traces reveals that most neuronal oscillations presence of true beta-band activity at ∼20 Hz
Neural Recordings and Time Series Analyses 459

Figure 28.4 Relationship of oscillatory and broadband activity.


(A) Illustration of Fourier transformation of electrophysiological data that exhibits both an
oscillation in 8-12 Hz alpha range (“bump” exceeding the 1/f background activity within the
gray shaded area) as well as broadband 1/f component. A true increase in oscillatory power
between two conditions (red to green) is illustrated. (B) A broadband shift (i.e., change of
overall offset along y-axis) can mimic an alpha power increase, which is not oscillatory in
nature. (C) Likewise, a change in peak frequency can mimic power changes when power is
averaged within a predefined range. (D) Similarly, a rotation of power spectrum (change in
spectral exponent) can introduce changes in oscillatory power if spectra are not properly
parameterized. Illustrations created using the FOOOF toolbox.
Source: Donoghue et al., 2020.

(a hallmark of the motor system), which is con- has gained more traction recently, since several
taminated by Mu-harmonics at ∼20 Hz (Stolk groups demonstrated that the background activity
et al., 2019). In recent years, several methods have – which had been previously considered to mainly
been introduced to disentangle true from spurious reflect neuronal noise – in fact contains behavio-
oscillatory brain activity (Donoghue et al., 2020; rally relevant information (Donoghue et al., 2020;
Kosciessa et al., 2020; Wen and Liu, 2016). For He et al., 2010; Lendner et al., 2020; Voytek et al.,
example, empirical mode decomposition (EMD) 2015). Importantly, the level of information con-
does not rely on sinusoidal basis functions and can tent about the behavioral state is on par with neu-
also model a change in the precise peak frequency ronal information encoded in band-limited
over time (Quinn et al., 2021). The same issues oscillatory activity. To date, the exact physiologi-
apply when the interaction of multiple frequency cal role of both components is not fully under-
bands is assessed. To date, there is no unique solu- stood, but they can be conceptualized as providing
tion for this issue, but several recent publications complementary insights into cognitive processes
suggest that a set of criteria could be applied to the (Wainio-Theberge et al., 2021, 2022). Over the
data to infer whether non-sinusoidality is present last decade, several approaches have been intro-
(Aru et al., 2015). duced to parameterize oscillations and to disen-
tangle oscillatory from broadband (also termed
aperiodic, fractal or scale-free) background activ-
ity. The simplest solution to isolate oscillations
entails a linear fit to the power spectrum in dou-
SEPARATING APERIODIC FROM ble-log space (thus the linear fit reflects the aperi-
OSCILLATORY ACTIVITY odic component) and subtraction from the original
spectrum (Lendner et al., 2020; Miller et al.,
The electrophysiological power spectrum encom- 2009). More sophisticated algorithms employ
passes oscillations (discrete “bumps”), exceeding robust fitting (Kosciessa et al., 2020), irregular
the general 1/fx drop-off. For decades, both phe- spectral resampling (Wen and Liu, 2016) or addi-
nomena have been studied together and were not tive fitting of Gaussian components to the oscilla-
explicitly disentangled (Donoghue et al., 2021). tory peaks (Donoghue et al., 2020). All methods
Hence, elevated “alpha power” could either be the have in common that they now provide two com-
results of an amplitude increase of the oscillatory ponents that are derived from the same underlying
component or a general increase in activity in all signal, which can be related to behavior and brain
frequency bands (Figure 28.4). This distinction state. Currently, these concepts are actively being
460 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 28.5 Effect of different spectral decomposition methods.


Upper row: Sleep hypnogram from single case across 8 hours of sleep. Below: Spectral
decomposition of entire night of sleep using either a multi-taper or single-taper approach,
the periodogram or Welch’s method. The resulting estimates become progressively more
coarse-grained. (B) Signal-to-noise ratio per frequency reveals a clear advantage of multi-
tapered spectral analysis.
Source: From Lendner et al., 2020.

explored, with several new notions, such as the dependent, such as oscillatory phase. Hence, it is
concept of population time constants of relative best practice to return from the spectral to the time
stability (also termed temporal integration win- domain to quantify and assess the oscillatory fea-
dows, autocorrelation window or intrinsic neural tures after the presence of an oscillation was
timescales; Gao et al., 2020; Golesorkhi et al., established using spectral methods. These analy-
2021; Ito et al., 2020; Raut et al., 2020; Wolff ses can either be applied on band-limited or
et al., 2022), which can be approximated by char- broadband data. It is important to consider that
acteristic bends in the shape of the power spec- both linear as well as non-linear variables can be
trum in the frequency domain or from the decay of obtained from time- and frequency-domain data,
the autocorrelation function in the time domain. with amplitude reflecting a linear variable, while
Population time constants are thought to provide phase constitutes a circular, and thereby, non-
the necessary means for temporal integration and linear variable. Both features can be extracted
exhibit a clear cortical gradient with short time- from the Hilbert transform and can be related to
scales in sensory cortex and longer timescales in behavior using either linear or circular-linear cor-
association areas (Gao et al., 2020; Raut et al.; relations (Berens, 2009; Fiebelkorn et al., 2018;
2020; Wolff et al., 2022). Helfrich, Fiebelkorn, et al., 2018). In this case,
one needs to assume a fixed frequency band to
obtain reliable phase estimates. Again, Fourier
methods are ill suited to assess if the peak fre-
quency changes over time (jitter in peak frequency
OSCILLATORY AND WAVEFORM SHAPE reflects a broadening of the spectral peak in the
FEATURES electrophysiological power spectrum). Several
recent developments, including the EMD, now
Definition of oscillations, including their band- aim at mitigating these effects (Quinn et al., 2021;
width and amplitude (i.e., power), is most com- Watrous and Buchanan, 2020).
monly done in the spectral domain. However, as Another recent development is the appre-
outlined above, spectral analysis also omit impor- ciation of waveform shapes (Cole et al., 2017).
tant wave form features as well as instantaneous Previously, waveform shapes have largely been
signal characteristics, which are inherently time ignored given that the Fourier basis functions were
Neural Recordings and Time Series Analyses 461

Figure 28.6 Effects of non-sinusoidal sharp transients on cross-frequency coupling esti-


mates, (A–H) eight examples of simulated EEG traces (upper left) that are composed of
broadband EEG activity (center left) and sharp transients (lower left) as well as the accom-
panying phase-amplitude comodulogram (right; black outlines indicate significant coupling)
revealing that sharp transients introduce spurious cross-frequency coupling at the primary
frequency (10 Hz) as well as at the subsequent harmonics.
Source: From Gerber et al., 2016.

sinusoidal. The role of physiologic non-sinusoidal interaction between multiple regions. This interac-
waveform shapes came into focus after it was real- tion is often also termed functional connectivity, to
ized that non-sinusoidality introduces artifacts contrast it to structural connectivity, which can be
in (e.g., cross-frequency coupling analyses; see obtained from tracing or fiber tracking studies
below; Aru et al., 2015; Gerber et al., 2016; Figure (Buzsáki and Draguhn, 2004; Engel et al., 2001;
28.6). Novel tools now enable assessing waveform Varela et al., 2001). Functional connectivity is
shapes with the goal to relate distinct features (such undirected (i.e., it is not quantified who drives the
as rise- or decay-times, asymmetries or amplitude interaction). Directed connectivity is referred to
bias) to distinct physiological processes. However, effective connectivity implying information flow
there is currently only limited evidence that sup- from node A to node B. Here, we first discuss undi-
ports a distinct role in cortical processing. rected connectivity between different nodes, and
then we discuss directed connectivity methods
across spatial scales. The concept of connectivity
can also be extended to interactions in the temporal
NETWORK CONNECTIVITY ACROSS
domain (i.e., the interaction between different
SPATIAL AND TEMPORAL SCALES oscillations or between the LFP and spikes. Lastly,
we discuss the use of information theoretical
We have focused on univariate analyses that can be approaches in this context.
carried out at the single electrode or single voxel Undirected connectivity is most commonly stud-
level. In contrast, network approaches quantify the ied by means of coherence or magnitude-squared
462 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

coherence as it is more correctly termed (Bastos to B. Again, this approach is not causal, but ena-
and Schoffelen, 2015). This distinction indicates bles a statistical comparison based on empirical
that the coherence formula encompasses both the signal distributions. One disadvantage of infor-
relationship of amplitude as well the relationship mation theoretical metrics is that they typically
of phase. Collectively, this relationship is then nor- require binning of the data and thus, reduce the
malized to yield a number between 0 and 1. If the signal complexity into a finite number of bins at
amplitude term in the formula is replaced by a 1, the expense of losing fine-grained details (Panzeri
then the formula become amplitude-­independent et al., 2015).
and only the contribution of phase synchroniza- Lastly, the concepts that apply to interactions
tion is estimated; this normalized variant is also between spatial nodes can be extended to the
known as the phase-locking value (Lachaux spectral domain to infer if two frequency bands
et al., 1999). Likewise, one can focus only on the interact (Figure 28.7). This coupling across tem-
amplitude contribution by (e.g., correlation of the poral scales is widely known as cross-frequency
amplitude time series; Hipp et al., 2012). The lit- coupling (CFC (Canolty and Knight, 2010)) and
erature often distinguished between phase-based has most prominently studied in the context of
and amplitude-based connectivity and several phase-amplitude coupling (PAC), where the phase
theoretical accounts postulated distinct roles for of slow frequency (e.g., theta or alpha) predicts
cortical communication (Engel et al., 2013). Both broadband high frequency activity (Canolty et al.,
metrics have in common that they are suscepti- 2006). However, amplitude-amplitude as well as
ble to volume spread in the cortical tissue, which phase-phase couplings have also been described
inflates connectivity metrics. Solutions by means for CFC (Aru et al., 2015). CFC analyses are sus-
of orthogonalized amplitude correlations (Hipp ceptible to signal artifacts (cf. Figure 28.6) and
et al., 2012) or imaginary coherence (Nolte et al., signal processing choices; hence, several papers
2004) as well as several variants have been pro- formulated concrete guidelines to circumvent
posed (Bastos and Schoffelen, 2015), which mini- these issues (Aru et al., 2015; Gerber et al., 2016).
mize this confound by attenuating the contribution Similar considerations apply also for LFP-spike
of zero phase-lag interactions. coupling.
Both approaches share that they are undirected
in nature, so it remains equivocal whether node A
is driving node B, or vice versa. Several metrics
that take advantage of statistical regularities in the
data have been introduced to infer directionality,
MULTIVARIATE REPRESENTATIONS AND
such as Granger causality (model-based assess- ANALYSES
ment (Seth et al., 2015)) or the phase slope index
(dependence across multiple frequency bands The analysis of electrophysiological data consti-
(Nolte et al., 2008)). However, these methods tutes an extensive multivariate problem. In order
operate on statistical dependencies, are suscep- to better understand the data and help the interpre-
tible to noise and do not provide a true “causal” tation, the most common analytical approaches
explanation. condense this complex analytical space into serial
The concept of directed and undirected con- univariate tests, which are oftentimes easier to
nectivity has also been formalized in an infor- interpret. From a conceptual standpoint, univari-
mation-theoretical framework (Ince et al., 2017; ate analyses seem reasonable when data is
Panzeri et al., 2015). Shannon Information approached with the “Neuron Doctrine” in mind,
Theory (Shannon and Weaver, 1998) is centered which states that the single neuron is the central
on entropy to quantify the observed distribution computational unit of the nervous system (cf.
of a given variable (here activity in one region). Yuste, 2015). Within this framework, it is reason-
Connectivity between two regions can be inferred able to employ univariate analyses approaches.
if knowledge about the activity in one region However, in recent years it became obvious that
reduces uncertainty about the state of the other the single neuron framework falls short in explain-
region. This interaction has also been termed ing several cognitive phenomena, such as flexible
Mutual Information and is undirected. An impor- context-dependent behavior, which inspired the
tant extension of this idea called sample entropy concept of a “Population Doctrine” (Eichenbaum,
basically takes a third signal into account to infer 2017; Ebitz and Hayden, 2021; Saxena and
directionality (Lobier et al., 2014): Does knowl- Cunningham, 2019; Yuste, 2015). This population
edge about the past of region A provide more framework suggests that transient coalitions of
information about the future of region B than the neurons form the central computational unit of the
past of region B alone? If the answer is yes, then nervous system (Eichenbaum, 2017; Siegel et al.,
this can be interpreted as information flow from A 2012). Therefore, gaining a deeper understanding
Neural Recordings and Time Series Analyses 463

Figure 28.7 Analysis strategy for cross-frequency coupling analyses.


(A) Raw EEG data (black) can be decomposed into different frequency bands through band-
pass filtering. Instantaneous phase and amplitude estimates are extracted from the Hilbert
transform and can be correlated using either linear or circular correlation analyses.
(B) Visualization of the proposed coupling mechanism, where overall power remains con-
stant, but power is modulated as a function of phase.
Source: From Helfrich et al., 2015.

into cognitive mechanisms requires large-scale distance to either the center point of coordinate
recordings, which are not amendable to classic system or to adjacent time points. Here the criti-
univariate analyses. This novel framework was cal advantage is that Euclidean distance is again
motivated by a series of new artificial-intelligence a single number, hence, the high dimensional data
inspired algorithms, which enable uncovering the can effectively be condensed into a single vector.
organizing principles of seemingly chaotic popu- Thus, the entire matrix can be regarded as a trajec-
lation activity. This is a rapidly emerging field tory traveling through N-dimensional space and
with many new technical innovations in recent given that Euclidean distances can be calculated,
years, and we are unable to provide a thorough one can also infer (e.g., velocity (distance/time)
review, but will focus on two representative exam- or acceleration (velocity/time) of the system –
ples to illustrate how to approach population- Figure 28.8).
based analyses. As humans, we cannot effectively visualize
The first important approach is that multivari- more than three dimensions, hence, it is com-
ate data can be represented in a so-called state- mon practice to visualize state space trajectories
space representation (Ebitz and Hayden, 2021; in 2D or 3D, and after dimensionality reduction
Gervasoni et al., 2004). Instead of analyzing indi- has been performed (Ebitz and Hayden, 2021).
vidual time series, the entire channel (or neuron) This conceptualization of population activity gave
x time matrix is conceptualized as a single time rise to several emerging concepts in neuroscience,
series of adjacent points which travels through including attractor states (points in the system
a N-dimensional coordinate system along a tra- where the activity patterns are naturally drawn to
jectory, which is spanned by the individual data and converge on) or neuronal manifolds (activity
points. Here N refers to the number of observa- patterns fall onto certain planes in the state space
tions (e.g., channels or neurons). If one considers and are not completely randomly distributed along
recordings from three neurons, then the first time all possible dimensions, hence, all possible activity
point is a point in a 3D coordinate system. Hence, patterns are constrained to a subspace that houses
using linear algebra, we can infer the Euclidean most observations). To date, these techniques have
464 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 28.8 2D State-space representations of cognitive dynamics.


(Left) Tangled trajectories of three different conditions (varying degree of predictability)
along two dimensions in human PFC indicate a complex representational pattern. (Right) In
contrast, activity in motor cortex is neatly organized along a ring structure, indicating low-
dimensional coding schemes.
Source: From Weber et al., 2021.

mainly been applied in the motor domain, where chance. In contrast to engineering, the goal is not
they provide additional explanatory power over to maximize classifier performance, but the goal is
univariate analyses, but they are gaining traction to test if the classifier performs significantly bet-
in the cognitive domain (Chaisangmongkon et al., ter than chance. If above-chance performance is
2017; Goudar et al., 2021; Murray et al., 2017; observed, this is taken as evidence that the brain
Weber et al., 2021). The second common technique contains information about the classified behavior
to address multivariate brain data relies on multi- (i.e., context or movement execution).
variate pattern analysis (MVPA (Grootswagers In practice, state-space and classification
et al., 2017; Hebart and Baker, 2018; Kriegeskorte analyses are often combined (Mante et al., 2013).
et al., 2006; Quian Quiroga and Panzeri, Jointly these methods enable extraction of cod-
2009)), which is also sometime called pattern ing dimensions (i.e., finding a latent or hidden
­classification or more commonly “brain decoding” dimension in the data that cannot be accessed
(Figure 28.9). using univariate analyses, that maximizes a
Here, researchers take advantage of classifi- given behavioral contrast). This approach ena-
cation algorithms, such as the linear discrimi- bles disentangling (i.e., cognitive from motor
nant analysis (LDA) or support vector machines contributions, which can appear superimposed
(SVM), which learn patterns that are associated when using univariate analysis tools; Vyas et al.,
with certain conditions or responses. Importantly, 2020). Disentangling the respective contributions
the classifiers require splitting the data into a train- provides the opportunity to study their contribu-
ing dataset, where the classifier has access to the tion to the overall population activity and pro-
ground truth, and a testing dataset, which consists vides new insights into the coding mechanisms.
of held-out samples. Based on the performance For example, it had repeatedly been shown that
on the held-out samples, one can infer the accu- motor activity is confined to a low-dimensional
racy of the classifier. Critically, these algorithms sub-space (i.e., can be described by only few
often work as a “black box,” where the algorithm activity patterns, which often cycle along a low-
learns a given association and then is used to pre- dimensional ring structure; Churchland et al.,
dict held-out data points, but one cannot visualize 2012; Shenoy et al., 2013). In contrast, more
the classifier per se. Therefore, it often remains complex higher-order cognitive operations also
ambiguous whether the classifier utilizes informa- require higher dimensional neuronal representa-
tion that is also accessible by the brain itself or tions, since they rely on distributed computing
whether it picks up epiphenomenal or noise influ- (Ebitz and Hayden, 2021). Collectively, novel
ences. In the context of neurophysiological experi- population-based analysis strategies extend the
ments, classifiers are used to infer if a population explanatory power of univariate tests and open a
contains behaviorally relevant information or not, new conceptual space to interpret the functional
i.e. whether the classifier performs better than architecture underlying human behavior.
Neural Recordings and Time Series Analyses 465

Figure 28.9 Information-theoretical and decoding approaches.


(A) Illustration of representational similarity. Across several repetitions, similarity in neu-
ronal representations is inferred by correlation, indicating that the same or related concepts
share similar spatiotemporal patterns in electrophysiological recordings. (B) Decoding or
pattern classification analysis relies on training classifiers on distinct features, which then
enable a validation on a held-out testing data set. Here a linear discrimination analysis is
depicted with single points representing observations that are characterized by features
along two dimensions. (C) Information-theoretical analyses between object and EEG features
can be conceptualized as a non-linear correlation analysis.

SCALE-FREE PHENOMENA After applying a Fourier transform, one can


easily appreciate the general 1/f drop-off (cf. ape-
Scale-free phenomena refer to the analysis of sta- riodic activity in EEG), which indicates a stable
tistical regularities in electrophysiological data relationship between frequency and amplitude,
that are not governed by a defining temporal scale irrespective of the precise frequency and there-
(He et al., 2010). For example, scalp EEG is gov- fore irrespective of the precise temporal scale. In
erned by alpha oscillations at ∼10 Hz as their the past two decades, several methods have been
defining temporal scale, while many intracranial introduced to quantify this type of behavior, either
EEG contact often do not exhibit a clear oscilla- in the temporal or spatial domain. For example,
tory peak. The absence of oscillatory peaks raises linear fitting of the 1/f background activity is a
the question if there is any underlying hidden common approach to extract the spectral exponent
temporal structure present and if yes, then how and thereby, quantify the underlying statistical
could this be quantified. An important concept regularities. A related approach is the detrended
that borrowed from physics is fractality, which fluctuation analysis (Hardstone et al., 2012),
describes the self-similarity of a signal irrespec- which assesses long-range temporal correlations,
tive of the chosen temporal window (Pritchard, i.e. signal correlations that obey the same scaling
1992). Critically, this self-similarity follows a behavior irrespective of the chosen temporal win-
power-law (i.e., a law of nature; Miller et al., dow. Similar 1/f phenomena have been observed
2009). A typical example from nature is the struc- in the spatial domain, where activity peaks across
ture of a Romanesco cauliflower, which exhibits many electrodes coincide in time and the distri-
characteristic florets, which always look similar, bution of the duration and amplitude again follow
irrespective of their size or magnification a power law with 1/f-like characteristics (Palva
(Hardstone et al., 2012; Linkenkaer-Hansen et al., et al., 2013). Going beyond the activity peaks,
2001). In the context of electrophysiological data, analysis of microstates (i.e., quasi-static scalp
this means that an EEG trace always exhibits simi- topographies in EEG) revealed that brain activity
lar fluctuations, irrespective of whether one hour, alternates between four major scalp topographies,
one minute or one second is displayed. While this where again a 1/f-like power law can describe the
phenomenon is difficult to describe in the tempo- duration of each topography (Ville et al., 2010). To
ral domain, it can easily be captured in the fre- date, it remains unclear how these different phe-
quency domain (Figure 28.10). nomena are related, but several lines of research
466 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 28.10 Power-law scaling in intracranial recordings.


(A) Experimental approach. (B) 1/fx power law fits of electrophysiological brain activity in
relationship to recording hardware noise floors.
Source: From Miller et al., 2009.

suggest that they might constitute emergent ana- all observations to distinct groups (i.e., shuffling
lytical tools to quantify temporal regularity, which the condition label), then to repeat the analysis
cannot be appreciated by the naked eye, unlike 1,000–10,000 times and then compare the true
neuronal oscillations (Cocchi et al., 2017). It has observed value to the surrogate distribution. If the
been argued that the characterization of temporal observed value falls outside of the mean ± 2 SDs,
regularities in the form of scale-free dynamics then significance can be assumed (at p < 0.05). This
constitutes a promising avenue to understanding approach is elegant, since it allows that any type of
the neural correlates of consciousness (Lendner primary test statistic can be employed to build the
et al., 2020; Tagliazucchi et al., 2013, 2016; Zhang surrogate distribution. Hence, this test is inherently
et al., 2018). non-parametric and can be used in a wide variety
of contexts. An alternative solution that emerged
in recent years, is the use of general linear mixed
effects models, which however, are often difficult
to interpret and difficult to visualize and therefore,
STATISTICAL CONSIDERATIONS have not been adopted widely.

Since brain activity is non-linear, non-stationary,


and non-normally distributed, classic analyses
such as t-tests, analysis-of-variance (ANOVA),
linear correlation or regression often fall short in SUMMARY AND CONCLUSIONS
capturing the data complexity (Maris and
Oostenveld, 2007). However, these methods are In the present chapter, we have reviewed the cur-
still widely employed, since they are easy to use rent state-of-the-art of neuronal time series analy-
and most researchers know how to interpret their sis through the lens of scalp and intracranial EEG
outcomes (Piai et al., 2015). recordings. Notably, as outlined in beginning, the
To mitigate the issue of unknown distributions same analyses can be applied to other imaging
of brain data, which violate the assumptions of modalities that produce time series data and most
many statistical tests, the currently most accepted analyses are only constrained by sampling rate,
approach includes cluster-based permutation tests signal duration and event types. From a practical
(Maris and Oostenveld, 2007). Here a new surrogate standpoint, we emphasized the need to go from
distribution is build based on randomly assigning simple to more complicated analyses, i.e. going
Neural Recordings and Time Series Analyses 467

from univariate analyses in the time domain to ACKNOWLEDGMENTS


univariate analyses in the frequency domain,
before branching off into bivariate connectivity or This work was supported by the German Research
multivariate classification analyses. In order to Foundation (Emmy Noether Program, DFG
interpret the result of a bivariate connectivity or HE8329/2-1; R.F.H.), the Hertie Foundation
multivariate classification analysis in a meaning- (Network for Excellence in Clinical Neuroscience;
ful way, one needs to ensure that the observed R.F.H.), the Baden Württemberg Foundation
differences are not solely driven by univariate dif- (PostDoc Program; R.F.H.), the Ernst Jung
ferences, in (e.g., the overall amplitude), but in Foundation for Science and Research (Career
fact can only be attributed to their joint activity. Development Award; R.F.H.), and NINDS grants
The example of aperiodic activity analyses R01 NS021135 and U19NS107609-01 (R.T.K.).
nicely illustrates how one can teach an old dog
(the Fourier transform) a few new tricks that pro-
vide additional explanatory power and enable
linking macroscopic signals to underlying circuit
properties (Gao et al., 2017). In the case of aperi-
odic activity, computationally modeling suggested REFERENCES
that the steepness of the exponential decay of the
power spectrum reflects the excitation-inhibition- Aru, J., Aru, J., Priesemann, V., Wibral, M., Lana, L.,
balance of the underlying neuronal population, Pipa, G., Singer, W., & Vicente, R. (2015). Untan-
which otherwise could not have been inferred gling cross-frequency coupling in neuroscience.
(Chini et al., 2021). Current Opinion in Neurobiology, 31, 51–61.
The wealth of available methods also enables Baillet, S. (2017). Magnetoencephalography for brain
reanalysis of available datasets, especially if the electrophysiology and imaging. Nature Neurosci-
primary analysis was mainly concerned with uni- ence, 20(3), 327–339.
variate analysis approaches. As an example, a Bastos, A. M., & Schoffelen, J.-M. (2015). A tutorial
classic working memory experiment in two mon- review of functional connectivity analysis methods
keys indicated that single neurons in prefrontal and their interpretational pitfalls. Frontiers in
cortex code distinct aspect of working memory Systems Neuroscience, 9. doi: 10.3389/fnsys.
(Warden and Miller, 2007, 2010). Through careful 2015.00175
reanalysis of the same dataset, a new concept of Berens, P. (2009). CircStat: A MATLAB toolbox for
working memory emerged. In the first follow-up circular statistics. Journal of Statistical Software,
publication, the authors demonstrated that spik- 31(10). doi: 10.18637/jss.v031.i10
ing is coupled to the underlying LFPs (Siegel Berger, H. (2004). Über das elektrenkephalogramm
et al., 2009). Subsequently, these LFP signatures des menschen: Die vierzehn originalarbeiten von
were described in more detail, which triggered 1929-1938. Berlin: Deutsche Gesellschaft für Kli-
follow-up investigations on neuronal bursts and nische Neurophysiologie.
how these differ from sustained oscillatory activ- Biasiucci, A., Franceschiello, B., & Murray, M. M.
ity (Lundqvist et al., 2016, 2018). Furthermore, (2019). Electroencephalography. Current Biology,
the same dataset was used to infer the dimen- 29(3), R80–R85.
sionality of spiking activity, which supported the Bronstein, J. M., Tagliati, M., Alterman, R. L., Lozano,
notion that high-dimensional representations are A. M., Volkmann, J., Stefani, A., … Krack, P.
desirable for cognitive flexibility (Rigotti et al., (2011). Deep brain stimulation for Parkinson dis-
2013). With the emergence of more analytical ease: An expert consensus and review of key
tools and the availability of more open datasets issues. Archives of Neurology, 68(2), 165–165.
novel concepts will be generated based on existing Buzsáki, G. (1996). The hippocampo-neocortical
data (Fusi et al., 2016). Overall, machine learn- dialogue. Cerebral Cortex, 6(2), 81–92.
ing is now established as an independent field, Buzsáki, G., Anastassiou, C. A., & Koch, C. (2012).
thus, we foresee that more multivariate tools will The origin of extracellular fields and currents: EEG,
become available to experimental neurophysiolo- ECoG, LFP and spikes. Nature Reviews: Neurosci-
gists to assess their data beyond classical univari- ence, 13(6), 407–420.
ate analyses (Grootswagers et al., 2017; Hebart Buzsáki, G., & Draguhn, A. (2004). Neuronal oscilla-
and Baker, 2018). tions in cortical networks. Science, 304,
Overall, experimentalists are nowadays faced 1926–1929.
with a wide-array of analysis tools, which requires Canolty, R. T., Edwards, E., Dalal, S. S., Soltani, M.,
careful justification of the employed methods. Nagarajan, S. S., Kirsch, H. E., … Knight, R. T.
However, the wealth of tools also provides an (2006). High gamma power is phase-locked to
exciting avenue to unravel the mechanisms behind theta oscillations in human neocortex. Science,
human cognition, perception, and action. 313, 1626–1628.
468 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Canolty, R. T., & Knight, R. T. (2010). The functional Ebitz, R. B., & Hayden, B. Y. (2021). The population
role of cross-frequency coupling. Trends in Cogni- doctrine in cognitive neuroscience. Neuron,
tive Sciences, 14(11), 506–515. 109(19), 3055–3068.
Carlson, A. A., Rutishauser, U., & Mamelak, A. N. Edwards, E., Soltani, M., Deouell, L. Y., Berger, M. S.,
(2018). Safety and utility of hybrid depth elec- & Knight, R. T. (2005). High gamma activity in
trodes for seizure localization and single-unit response to deviant auditory stimuli recorded
neuronal recording. Stereotactic and Functional directly from human cortex. Journal of Neuro-
Neurosurgery, 96(5), 311–319. physiology, 94(6), 4269–4280.
Chaisangmongkon, W., Swaminathan, S. K., Freed- Eichenbaum, H. (2017). Barlow versus Hebb: When is
man, D. J., & Wang, X.-J. (2017). Computing by it time to abandon the notion of feature detectors
robust transience: How the fronto-parietal net- and adopt the cell assembly as the unit of cogni-
work performs sequential, category-based deci- tion? Neuroscience Letters, 680, 88–93.
sions. Neuron, 93(6), 1504–1517. Engel, A. K., Fries, P., & Singer, W. (2001). Dynamic
Chari, A., Thornton, R. C., Tisdall, M. M., & Scott, R. C. predictions: Oscillations and synchrony in top-
(2020). Microelectrode recordings in human epi- down processing. Nature Reviews: Neuroscience,
lepsy: A case for clinical translation. Brain Communi- 2(10), 704–716.
cations, 2(2). doi: 10.1093/braincomms/fcaa082 Engel, A. K., Gerloff, C., Hilgetag, C. C., & Nolte, G.
Chini, M., Pfeffer, T., & Hanganu-Opatz, I. L. (2021). (2013). Intrinsic coupling modes: Multiscale inter-
Developmental increase of inhibition drives decor- actions in ongoing brain activity. Neuron, 80(4),
relation of neural activity. bioRxiv. doi: 867–886.
10.1101/2021.07.06.451299 Fiebelkorn, I. C., Pinsk, M. A., & Kastner, S. (2018). A
Churchland, M. M., Cunningham, J. P., Kaufman, M. dynamic interplay within the frontoparietal net-
T., Foster, J. D., Nuyujukian, P., Ryu, S. I., & Shenoy, work underlies rhythmic spatial attention. Neuron,
K. V. (2012). Neural population dynamics during 99(4), 842–853.
reaching. Nature, 487, 51–56. Fisher, R., Salanova, V., Witt, T., Worth, R., Henry, T.,
Cocchi, L., Gollo, L. L., Zalesky, A., & Breakspear, M. Gross, R., … SANTE Study Group. (2010). Electri-
(2017). Criticality in the brain: A synthesis of neu- cal stimulation of the anterior nucleus of thalamus
robiology, models and cognition. Progress in Neu- for treatment of refractory epilepsy. Epilepsia,
robiology, 158, 132–152. 51(5), 899–908.
Cohen, M. X. (2017). Where Does EEG Come from Flinker, A., Korzeniewska, A., Shestyuk, A. Y.,
and what does it mean? Trends in Neurosciences, Franaszczuk, P. J., Dronkers, N. F., Knight, R. T., &
40(4), 208–218. Crone, N. E. (2015). Redefining the role of Broca’s
Cole, S. R., van der Meij, R., Peterson, E. J., de Hemp- area in speech. Proceedings of the National Acad-
tinne, C., Starr, P. A., & Voytek, B. (2017). Nonsi- emy of Sciences of the United States of America,
nusoidal beta oscillations reflect cortical 112(9), 2871–2875.
pathophysiology in Parkinson’s disease. Journal of Fox, M. D., Snyder, A. Z., Vincent, J. L., Corbetta, M.,
Neuroscience, 37(18), 4830–4840. Essen, D. C. V., & Raichle, M. E. (2005). The
Cole, S. R., & Voytek, B. (2017). Brain oscillations and human brain is intrinsically organized into dynamic,
the importance of waveform shape. Trends in anticorrelated functional networks. Proceedings of
Cognitive Sciences, 21(2), 137–149. the National Academy of Sciences of the United
Despouy, E., Curot, J., Reddy, L., Nowak, L. G., States of America, 102(27), 9673–9678.
Deudon, M., Sol, J.-C., … Barbeau, E. J. (2020). Fried, I., Rutishauser, U., Cerf, M., & Kreiman, G.
Recording local field potential and neuronal activ- (2014). Single neuron studies of the human brain:
ity with tetrodes in epileptic patients. Journal of probing cognition. Cambridge, MA: MIT Press.
Neuroscience Methods, 341. doi: 10.1016/j. Fusi, S., Miller, E. K., & Rigotti, M. (2016). Why neu-
jneumeth.2020.108759 rons mix: High dimensionality for higher cognition.
Diekelmann, S., & Born, J. (2010). The memory func- Current Opinion in Neurobiology, 37, 66–74.
tion of sleep. Nature Reviews. Neuroscience, Gao, R., Peterson, E. J., & Voytek, B. (2017). Inferring
11(2), 114–126. synaptic excitation/inhibition balance from field
Donoghue, T., Haller, M., Peterson, E. J., Varma, P., potentials. NeuroImage, 158, 70–78.
Sebastian, P., Gao, R., … Voytek, B. (2020). Param- Gao, R., van den Brink, R. L., Pfeffer, T., & Voytek, B.
eterizing neural power spectra into periodic and (2020). Neuronal timescales are functionally
aperiodic components. Nature Neuroscience, dynamic and shaped by cortical microarchitecture.
23(12), 1655–1665. ELife, 9. doi: 10.7554/eLife.61277
Donoghue, T., Schaworonkow, N., & Voytek, B. Gerber, E. M., Sadeh, B., Ward, A., Knight, R. T., &
(2021). Methodological considerations for study- Deouell, L. Y. (2016). Non-sinusoidal activity can
ing neural oscillations. European Journal of produce cross-frequency coupling in cortical sig-
Neuroscience, 55(11-12):3502–3527. nals in the absence of functional interaction
Neural Recordings and Time Series Analyses 469

between neural sources. PloS One, 11(12). doi: Helfrich, R. F., Lendner, J. D., Mander, B. A., Guillen,
10.1371/journal.pone.0167351 H., Paff, M., Mnatsakanyan, L., … Knight, R. T.
Gervasoni, D., Lin, S.-C., Ribeiro, S., Soares, E. S., (2019). Bidirectional prefrontal-hippocampal
Pantoja, J., & Nicolelis, M. A. L. (2004). Global dynamics organize information transfer during
forebrain dynamics predict rat behavioral states sleep in humans. Nature Communications, 10(1).
and their transitions. Journal of Neuroscience, doi: 10.1038/s41467-019-11444-x
24(49), 11137–11147. Helfrich, R. F., Mander, B. A., Jagust, W. J., Knight, R.
Golesorkhi, M., Gomez-Pilar, J., Tumati, S., Fraser, T., & Walker, M. P. (2018). Old brains come uncou-
M., & Northoff, G. (2021). Temporal hierarchy of pled in sleep: Slow wave-spindle synchrony, brain
intrinsic neural timescales converges with spa- atrophy, and forgetting. Neuron, 97(1), 221–230.
tial core-periphery organization. Commu­ Hipp, J. F., Hawellek, D. J., Corbetta, M., Siegel, M.,
nications Biology, 4(1). doi: 10.1038/s42003-021- & Engel, A. K. (2012). Large-scale cortical correla-
01785-z tion structure of spontaneous oscillatory activity.
Goudar, V., Peysakhovich, B., Freedman, D. J., Buf- Nature Neuroscience, 15(6), 884–890.
falo, E. A., & Wang, X.-J. (2021). Elucidating the Ince, R. A. A., Giordano, B. L., Kayser, C., Rousselet, G.
neural mechanisms of learning-to-learn. bioRxiv, A., Gross, J., & Schyns, P. G. (2017). A statistical
doi: 10.1101/2021.09.02.455707 framework for neuroimaging data analysis based
Grootswagers, T., Wardle, S. G., & Carlson, T. A. on mutual information estimated via a gaussian
(2017). Decoding dynamic brain patterns from copula. Human Brain Mapping, 38(3), 1541–1573.
evoked responses: A tutorial on multivariate pat- Ito, T., Hearne, L. J., & Cole, M. W. (2020). A cortical
tern analysis applied to time series neuroimaging hierarchy of localized and distributed processes
data. Journal of Cognitive Neuroscience, 29(4), revealed via dissociation of task activations, con-
677–697. nectivity changes, and intrinsic timescales.
Gross, J., Baillet, S., Barnes, G. R., Henson, R. N., NeuroImage, 221. doi: 10.1016/j.neuroimage.
Hillebrand, A., Jensen, O., … Schoffelen, J.-M. 2020.117141
(2013). Good practice for conducting and report- Kanth, S. T., & Ray, S. (2020). Electrocorticogram
ing MEG research. NeuroImage, 65, 349–363. (ECoG) is highly informative in primate visual
Handy, T. C. (2005). Event-related potentials: A cortex. Journal of Neuroscience, 40(12),
methods handbook. Cambridge, MA: MIT Press. 2430–2444.
Hardstone, R., Poil, S.-S., Schiavone, G., Jansen, R., Kosciessa, J. Q., Grandy, T. H., Garrett, D. D., &
Nikulin, V. V., Mansvelder, H. D., & Linkenkaer- Werkle-Bergner, M. (2020). Single-trial characteri-
Hansen, K. (2012). Detrended fluctuation analysis: zation of neural rhythms: Potential and challenges.
A scale-free view on neuronal oscillations. NeuroImage, 206. doi: 10.1016/j.neuroimage.
Frontiers in Physiology, 3. doi: 103389/ 2019.116331
fphys.2012.00450 Kriegeskorte, N., Goebel, R., & Bandettini, P. (2006).
He, B. J., Zempel, J. M., Snyder, A. Z., & Raichle, M. Information-based functional brain mapping. Pro-
E. (2010). The temporal structures and functional ceedings of the National Academy of Sciences of the
significance of scale-free brain activity. Neuron, United States of America, 103(10), 3863–3868.
66(3), 353–369. Lachaux, J. P., Rodriguez, E., Martinerie, J., & Varela,
Hebart, M. N., & Baker, C. I. (2018). Deconstructing F. J. (1999). Measuring phase synchrony in brain
multivariate decoding for the study of brain func- signals. Human Brain Mapping, 8(4), 194–208.
tion. NeuroImage, 180, 4–18. Lendner, J. D., Helfrich, R. F., Mander, B. A., Romund-
Hefft, S., Brandt, A., Zwick, S., von Elverfeldt, D., stad, L., Lin, J. J., Walker, M. P., Larsson, P. G., &
Mader, I., Cordeiro, J., Trippel, M., Blumberg, J., & Knight, R. T. (2020). An electrophysiological
Schulze-Bonhage, A. (2013). Safety of hybrid elec- marker of arousal level in humans. Elife, 9. doi:
trodes for single-neuron recordings in humans. 10.7554/eLife.55092
Neurosurgery, 73(1), 78–85. Leszczyński, M., Barczak, A., Kajikawa, Y., Ulbert, I.,
Helfrich, R. F., Fiebelkorn, I. C., Szczepanski, S. M., Falchier, A. Y., Tal, I., … Schroeder, C. E. (2020).
Lin, J. J., Parvizi, J., Knight, R. T., & Kastner, S. Dissociation of broadband high-frequency activity
(2018). Neural mechanisms of sustained attention and neuronal firing in the neocortex. Science
are rhythmic. Neuron, 99(4), 854–865.. Advances, 6(33). doi: 10.1126/sciadv.abb0977
Helfrich, R. F., Herrmann, C. S., Engel, A. K., & Linkenkaer-Hansen, K., Nikouline, V. V., Palva, J. M.,
Schneider, T. R. (2015). Different coupling modes & Ilmoniemi, R. J. (2001). Long-range temporal
mediate cortical cross-frequency interactions. correlations and scaling behavior in human brain
NeuroImage, 140, 76–82. oscillations. Journal of Neuroscience, 21(4),
Helfrich, R. F., & Knight, R. T. (2019). Cognitive neu- 1370–1377.
rophysiology of the prefrontal cortex. Handbook Lobier, M., Siebenhühner, F., Palva, S., & Palva, J. M.
of Clinical Neurology, 163, 35–59. (2014). Phase transfer entropy: A novel
470 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

phase-based measure for directed connectivity in in the brain. International Journal of Psychophysi-
networks coupled by oscillatory interactions. Neu- ology, 18(1), 49–65.
roImage, 85(2), 853–872. Piai, V., Dahlslätt, K., & Maris, E. (2015). Statistically
Lopes da Silva, F. (2013). EEG and MEG: Relevance to comparing EEG/MEG waveforms through succes-
neuroscience. Neuron, 80(5), 1112–1128. sive significant univariate tests: How bad can it be?
Lundqvist, M., Herman, P., Warden, M. R., Brincat, S. Psychophysiology, 52(3), 440–443.
L., & Miller, E. K. (2018). Gamma and beta bursts Polich, J. (2007). Updating P300: An integrative
during working memory readout suggest roles in theory of P3a and P3b. Clinical Neurophysiology,
its volitional control. Nature Communications, 118(10), 2128–2148.
9(1). doi: 10.1038/s41467-017-02791-8 Prerau, M. J., Brown, R. E., Bianchi, M. T., Ellenbogen,
Lundqvist, M., Rose, J., Herman, P., Brincat, S. L., J. M., & Purdon, P. L. (2017). Sleep neurophysiologi-
Buschman, T. J., & Miller, E. K. (2016). Gamma and cal dynamics through the lens of multitaper spectral
beta bursts underlie working memory. Neuron, analysis. Physiology, 32(1), 60–92.
90(1), 152–164. Pritchard, W. S. (1992). The brain in fractal time:
Mante, V., Sussillo, D., Shenoy, K. V., & Newsome, W. 1/f-like power spectrum scaling of the human
T. (2013). Context-dependent computation by electroencephalogram. International Journal of
recurrent dynamics in prefrontal cortex. Nature, Neuroscience, 66(1-2), 119–129.
503, 78–84. Quian Quiroga, R., & Panzeri, S. (2009). Extracting
Maris, E., & Oostenveld, R. (2007). Nonparametric information from neuronal populations: Informa-
statistical testing of EEG- and MEG-data. Journal tion theory and decoding approaches. Nature
of Neuroscience Methods, 164(1), 177–190. Reviews: Neuroscience, 10(3), 173–185.
Miller, K. J., Sorensen, L. B., Ojemann, J. G., & den Quinn, A. J., Lopes-dos-Santos, V., Dupret, D., Nobre,
Nijs, M. (2009). Power-law scaling in the brain A. C., & Woolrich, M. W. (2021). EMD: Empirical
surface electric potential. PLoS Computational mode decomposition and Hilbert-Huang spectral
Biology, 5(12). doi: 10.1371/journal.pcbi.1000609 analyses in python. Journal of Open Source Soft-
Murray, J. D., Bernacchia, A., Roy, N. A., Constantinidis, ware, 6(59). doi: 10.21105/joss.02977
C., Romo, R., & Wang, X.-J. (2017). Stable popula- Raut, R. V., Snyder, A. Z., & Raichle, M. E. (2020).
tion coding for working memory coexists with het- Hierarchical dynamics as a macroscopic organizing
erogeneous neural dynamics in prefrontal cortex. principle of the human brain. Proceedings of the
Proceedings of the National Academy of Sciences of National Academy of Sciences of the United States
the United States of America, 114(2), 394–399. of America, 117(34), 20890–20897.
Nolte, G., Bai, O., Wheaton, L., Mari, Z., Vorbach, S., Ray, S., & Maunsell, J. H. R. (2011). Different origins
& Hallett, M. (2004). Identifying true brain interac- of gamma rhythm and high-gamma activity in
tion from EEG data using the imaginary part of macaque visual cortex. PLoS Biology, 9(4). doi:
coherency. Clinical Neurophysiology, 115(10), 10.1371/journal.pbio.1000610
2292–2307. Rich, E. L., & Wallis, J. D. (2017). Spatiotemporal
Nolte, G., Ziehe, A., Nikulin, V. V., Schlögl, A., dynamics of information encoding revealed in
Krämer, N., Brismar, T., & Müller, K.-R. (2008). orbitofrontal high-gamma. Nature Communica-
Robustly estimating the flow direction of informa- tions, 8(1). doi: 10.1038/s41467-017-01253-5
tion in complex physical systems. Physical Review Rigotti, M., Barak, O., Warden, M. R., Wang, X.-J.,
Letters, 100(23). doi: 10.1103/PhysRevLett.100. Daw, N. D., Miller, E. K., & Fusi, S. (2013). The
234101 importance of mixed selectivity in complex cogni-
Palva, J. M., Zhigalov, A., Hirvonen, J., Korhonen, O., tive tasks. Nature, 497, 585–590.
Linkenkaer-Hansen, K., & Palva, S. (2013). Neuronal Rutishauser, U., Schuman, E. M., & Mamelak, A. N.
long-range temporal correlations and avalanche (2006). Online detection and sorting of extracel-
dynamics are correlated with behavioral scaling laws. lularly recorded action potentials in human medial
Proceedings of the National Academy of Sciences of temporal lobe recordings, in vivo. Journal of Neu-
the United States of America, 110(9), 3585–3590. roscience Methods, 154(1–2), 204–224.
Panzeri, S., Macke, J. H., Gross, J., & Kayser, C. Saxena, S., & Cunningham, J. P. (2019). Towards the
(2015). Neural population coding: Combining neural population doctrine. Current Opinion in
insights from microscopic and mass signals. Trends Neurobiology, 55, 103–111.
in Cognitive Sciences, 19(3), 162–172. Seth, A. K., Barrett, A. B., & Barnett, L. (2015).
Parvizi, J., & Kastner, S. (2018). Promises and limita- Granger causality analysis in neuroscience and
tions of human intracranial electroencephalogra- neuroimaging. Journal of Neuroscience, 35(8),
phy. Nature Neuroscience, 21(4), 474–483. 3293–3297.
Pascual-Marqui, R. D., Michel, C. M., & Lehmann, D. Shannon, C. E., & Weaver, W. (1998). The mathemat-
(1994). Low resolution electromagnetic tomogra- ical theory of communication. Urbana, IL: Univer-
phy: A new method for localizing electrical activity sity of Illinois Press.
Neural Recordings and Time Series Analyses 471

Shenoy, K. V., Sahani, M., & Churchland, M. M. Voytek, B., Kramer, M. A., Case, J., Lepage, K. Q.,
(2013). Cortical control of arm movements: A Tempesta, Z. R., Knight, R. T., & Gazzaley, A.
dynamical systems perspective. Annual Review of (2015). Age-related changes in 1/f neural electro-
Neuroscience, 36(1), 337–359. physiological noise. Journal of Neuroscience,
Siegel, M., Donner, T. H., & Engel, A. K. (2012). Spec- 35(38), 13257–13265.
tral fingerprints of large-scale neuronal interac- Vyas, S., Golub, M. D., Sussillo, D., & Shenoy, K. V.
tions. Nature Reviews: Neuroscience, 13(2), (2020). Computation through neural population
121–134. dynamics. Annual Review of Neuroscience, 43,
Siegel, M., Warden, M. R., & Miller, E. K. (2009). 249–275.
Phase-dependent neuronal coding of objects in Wainio-Theberge, S., Wolff, A., & Northoff, G.
short-term memory. Proceedings of the National (2021). Dynamic relationships between spontane-
Academy of Sciences of the United States of ous and evoked electrophysiological activity.
America, 106(50), 21341–21346. Communications Biology, 4(1). doi: 10.1038/
Staresina, B. P., Bergmann, T. O., Bonnefond, M., van s42003-021-02240-9
der Meij, R., Jensen, O., Deuker, L., …Fell, J. Wainio-Theberge S, Wolff A, Gomez-Pilar J, Zhang J,
(2015). Hierarchical nesting of slow oscillations, Northoff G. (2022). Variability and task-respon-
spindles and ripples in the human hippocampus siveness of electrophysiological dynamics: Scale-
during sleep. Nature Neuroscience, 18(11), free stability and oscillatory flexibility. NeuroImage,
1679–1686. 256. doi: 10.1016/j.neuroimage.2022.119245
Stolk, A., Brinkman, L., Vansteensel, M. J., Aarn- Warden, M. R., & Miller, E. K. (2007). The representa-
outse, E., Leijten, F. S., Dijkerman, C. H., … Toni, I. tion of multiple objects in prefrontal neuronal
(2019). Electrocorticographic dissociation of alpha delay activity. Cerebral Cortex, 17(S1), 41–50.
and beta rhythmic activity in the human sensori- Warden, M. R., & Miller, E. K. (2010). Task-dependent
motor system. ELife, 8. doi: 10.7554/eLife.48065 changes in short-term memory in the prefrontal
Tagliazucchi, E., von Wegner, F., Morzelewski, A., cortex. Journal of Neuroscience, 30(47),
Brodbeck, V., Jahnke, K., & Laufs, H. (2013). 15801–15810.
Breakdown of long-range temporal dependence Watrous, A. J., & Buchanan, R. J. (2020). The oscil-
in default mode and attention networks during latory reconstruction algorithm adaptively identi-
deep sleep. Proceedings of the National Academy fies frequency bands to improve spectral
of Sciences of the United States of America, decomposition in human and rodent neural
110(38), 15419–15424. recordings. Journal of Neurophysiology, 124(6),
Tagliazucchi, E., Chialvo, D. R., Siniatchkin, M., 1914–1922.
Amico, E., Brichant, J. F., Bonhomme, V., … Lau- Weber, J., Solbakk, A.-K., Blenkmann, A. O., Llorens,
reys, S. (2016). Large-scale signatures of uncon- A., Funderud, I., Leske, S., … Helfrich, R. F. (2021).
sciousness are consistent with a departure from Population coding and oscillatory subspace syn-
critical dynamics. Journal of the Royal Society chronization integrate context into actions.
Interface, 13(114). doi: 10.1098/rsif.2015.1027 bioRxiv. doi: 10.1101/2021.12.17.473118
Van Veen, B. D., van Drongelen, W., Yuchtman, M., Wen, H., & Liu, Z. (2016). Separating fractal and
& Suzuki, A. (1997). Localization of brain electrical oscillatory components in the power spectrum of
activity via linearly constrained minimum variance neurophysiological signal. Brain Topography,
spatial filtering. IEEE Transactions on Bio-Medical 29(1), 13–26.
Engineering, 44(9), 867–880. Wolff, A., Berberian, N., Golesorkhi, M., Gomez-Pilar,
Varela, F., Lachaux, J. P., Rodriguez, E., & Martinerie, J., Zilio, F., & Northoff, G. (2022). Intrinsic neural
J. (2001). The Brainweb: Phase synchronization timescales: Temporal integration and segregation.
and large-scale integration. Nature Reviews: Neu- Trends in Cognitive Sciences, 26(2), 159–173.
roscience, 2(4), 229–239. Yuste, R. (2015). From the neuron doctrine to neural
Ville, D. V. D., Britz, J., & Michel, C. M. (2010). EEG networks. Nature Reviews: Neuroscience, 16(8),
microstate sequences in healthy humans at rest 487–497.
reveal scale-free dynamics. Proceedings of the Zhang, J, Huang, Z., Chen, Y., Zhang, J., Ghinda, D.,
National Academy of Sciences of the United States Nikolova, Y., …Northoff, G. (2018). Breakdown in
of America, 107(42), 18179–18184. the temporal and spatial organization of sponta-
Voytek, B., Canolty, R. T., Shestyuk, A., Crone, N. E., neous brain activity during general anesthesia.
Parvizi, J., & Knight, R. T. (2010). Shifts in gamma Human Brain Mapping, 39(5), 2035–2046.
phase-amplitude coupling frequency from theta to Zijlmans, M., Zweiphenning, W., & van Klink, N.
alpha over posterior cortex during visual tasks. (2019). Changing concepts in presurgical assess-
Frontiers in Human Neuroscience, 4. doi: 10.3389/ ment for epilepsy surgery. Nature Reviews:
fnhum.2010.00191 Neurology, 15(10), 594–606.
29
Advances in Psychophysical
Techniques
I r i n a G . S k o t n i k o v a , A l e x e i N . G u s e v, a n d
Alexandr M. Chernorizov

INTRODUCTION known as the study of observer’s signal detection


and discrimination. Psychophysics II is a study of
an individual’s quantitative estimation of supra-
Paradigms in Psychophysics threshold sensations, expressed in subjective units
of measurement.
Historically, in the mid-1900s, Psychophysics was
developed by German physicist Gustav Fechner as
the first experimental field of psychology, based
on a natural-science orientation. Fechner pro-
posed an objective method to study and measure PSYCHOPHYSICS-I: DISCREETNESS/
mental phenomena (instead of the earlier subjec- CONTINUITY PARADIGM
tive introspective method) beginning with the
elementary form (sensations). He assumed that
individuals can’t directly estimate their values Justification of Sensory Range
quantitatively and proposed an indirect way to Discreteness/Continuity Problem
measure them, in terms of the units of stimuli
physical values, and proposed “Psychophysics.” Fechner’s theory postulates sensory thresholds
In 1860, Gustav Fechner presented a magnitude reality, dividing all stimuli into felt and impercep-
of sensation as the sum of its just noticeable dif- tible. Thus, a range of sensations appears to be
ferences (jnd) over the starting point. In order to discrete. A subsequent sensation arises after a
determine them he introduced the concept of the previous one only when a stimulus value increase
sensation threshold measured in stimulus units. exceeds jnd. It was the first classical conception
The absolute threshold and the differential one within the paradigm of the sensory system dis-
(jnd) is the minimum value of a stimulus and a creteness. Fechner explained the smooth S-shaped
difference between two stimuli, the increase of form of the psychometric function by the thresh-
which causes a conscious feeling of this stimulus old fluctuation in time, while his opponents
and difference, while the decrease does not cause. (Müller, Jastrow, and Urban) associated it with the
Therefore, Fechner’s Psychophysics is called threshold absence, proposing a classical concep-
Objective one or “Psychophysics-I,” that became tion within the paradigm of the sensory range
Advances in Psychophysical Techniques 473

continuity (a continuous series of intermediate discrimination. Data obtained are presented as


degrees of clearness). Therefore, there is no empirical frequencies of one kind of response
threshold point on the psychometric curve, and its or another (Receiver Operating Characteristic
S-shaped form is due to random distribution of – ROC). While in threshold tasks a stimulus has
favorable/unfavorable non sensory variables. more values in different trials (5–7 in the Method
Fechner realized the idea of discreteness method- of Constants and an almost unlimited number in
ologically: he has developed the three experimen- the two other classic methods).
tal methods for measuring absolute and differential
thresholds. Yet, within the classical paradigm of
discreteness, no other methods have been sug-
gested, but the same Fechner methods are used
today. The discussion has begun on the sensory DISCRETE VERSIONS: NEURO-QUANTUM
range discreteness/continuity problem, which still THEORY, THEORIES OF HIGH THRESHOLD
remains one of the fundamental problems of AND OF 2/3 SENSORY STATES,
Psychophysics. CONCEPTION OF TWO THRESHOLDS

Neuro-Quantum Theory using psychometric func-


tion analysis describes the sensory effect as asso-
DEVELOPMENT OF SENSORY RANGE ciated with activity of hypothetical functional
DISCRETENESS/CONTINUITY PROBLEM units in the sensory system – neuro-quants (NQ),
if the incoming irritation reaches their threshold.
Depending on the stimulus value, a different
Continuity Version: Signal Detection number of NQ per unit of time is activated, which
Theory in Psychophysics determines the different sensation value and a
psychometric function rectilinear (but not
Experimental methods for sensation measurement S-shaped) form found experimentally by the
were developed in line with the three main theo- authors, but not others, since all noise suppression
retical approaches (Fechner, Thurstone, and is required.
Stevens). The fourth approach: psychophysical
Signal Detection Theory (SDT: McMillan and
Creelman, 2005) revised the previous psycho-
physical methods by providing a fundamental step
in understanding of sensory responses. The theory DISCRETE VERSIONS USING THE SDT
was developed for detecting a weak sound signal SENSORY AXIS METRIC AND ROC
against a background noise (as in radio engineer- ANALYSIS
ing initially) and was applied further to other
sensory modalities. In Classical Psychophysics, Though SDT describes continues sensory system
sensory impressions were understood as the only functioning, four threshold- (discrete) theories
reason of individual’s responses, on the basis of were developed using the SDT sensory axis metric
which the sensitivity value (as the reciprocal of (SD of noise sensory effects normal distribution)
the threshold value) was assessed. SDT has identi- and the recognition of false alarms as natural
fied the two response’s components: the observ- observer’s responses (in contrast to Fechner’s
er’s sensory sensitivity and decision making about Psychophysics). Within these four theories, the
the sensory impression character. The components same SDT three methods are used: Yes-No,
are studied and quantitatively assessed by separate Rating, and Forced Choice. Data obtained are
indices via the development of three experimental presented by ROC, as well.
methods. Therefore, the thresholds indices are not
characteristics of a “pure” sensitivity, but integral
measures of performance: sensitivity and decision
making. High-Threshold Theory
In SDT detection and discrimination tasks ran-
dom noise is considered to be either an external According to High-Threshold Theory, only a
or internal noise of a sensory system. During an signal, but not noise can cause sensation (i.e., the
experiment, stimulation is stationary and the same threshold is located above the average noise value,
for every observer. Only one pair of stimuli is which sensory effect cannot exceed the threshold).
given randomly: noise and signal for detection, Therefore, false alarms are not sensory-based
a pair of same and a pair of different stimuli for but guessing. ROC looks like a straight line.
474 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

In contrast, false alarms may have sensory nature to an observer’s criterion. It is similar to the Low-
within the remaining three theories using the SDT Threshold Theory, but describes not the sensory
methodology (see below). threshold (which is difficult to identify unambigu-
ously), but the threshold of the local (stable) com-
ponent of the orientation reaction (determined by
objective psychophysiological methods). The
Low-Threshold Theory threshold presence was proved not by ROC analy-
sis, but by α-rhythm depression. While the theo-
Low-Threshold Theory assumes a threshold exist- retical concept of the sensory threshold remains in
ence, below which sensory events are indistin- doubt, Mikhalevskaya’s concept of a threshold
guishable, and above, the sensory continuum is region (where the detection rate varies from 0 to
continuous. The decision-making mechanism 1) is objective one.
(including the criterion) is functioning only when
sensory effects exceed the threshold. If not, guess-
ing occurs, as in the High Threshold Theory. ROC
is a straight line for high false alarm rates (as in
the High Threshold Theory) and a part of an arc PSYCHOPHYSICS-II: SCALING
for low ones. METHODOLOGY

Thurstone’s Approach to Scaling


Theory of Two States
Theoretical framework
Theory of Two States of sensory system (detection Louis Thurstone’s approach was based on the
and non-detection) assumes a threshold between principle of an indirect estimation of the per-
them. There is guessing on a sensory basis if the ceived quality set in the instructions by a person
both states are equiprobable. ROC includes two via making comparative judgments about stim-
rectilinear segments: a shallow one for high false uli. Its main idea is very simple: when a person
alarm rates and a steeper one for low rates. compares two stimuli, the impact of each gener-
ates a sensory representation of some subjective
quality of a certain value (e.g., the attractiveness
of the advertised product) and, in order to
Theory of Three States ­determine the degree of their psychological dif-
In the Three States Theory, two possible states of ference, these values are subtracted from each
sensory system (detection and non-detection) other. The measurement task is to quantify this
determine an observer’s responses by their nature. difference.
The third state (uncertainty) has to be overcomed Developing Fechner’s postulate about the
by response strategy based on non-sensory infor- equality of sensory impressions of all jnd
mation. The three states are separated by two Thurstone emphasized the possibility of measur-
thresholds, so ROC includes three rectilinear seg- ing sensations s on a continuum of “equally fre-
ments and approach the arcuate SDT curve. quently noticed differences.” This idea allows to
Moreover, ROC can change its shape due to sen- create scaling procedure using the preference fre-
sory sensitivity variability. This idea is very fruit- quency of one stimulus to another as the basis of
ful and appeareds later in conceptions shown a subjective metric. Thurstone’s model describing
below. According experimental tests of the stimuli’ effects on a person and comparing them
Modern Threshold Theories and SDT, ROC’s is as follows:
experimental points scatter doesn’t permit to
prefer any theory. a. The scaled set of stimuli (A, B, … N) can be
ordered into a continuum by the magnitude
of a certain quality, and this quality does not
necessarily correspond to a certain physical
CONCEPTION OF TWO THRESHOLDS characteristic.
b. The impact of each stimulus causes the individual
Conception has been suggested of two successive to have a corresponding discriminal process (ψi).
thresholds in the detection process: the threshold A lot of these processes constitutes a psychologi-
of an involuntary orientation reaction and the cal continuum of the values of subjective repre-
threshold of an arbitrary response corresponding sentations or sensory magnitudes (ψA, ψB … ψN).
Advances in Psychophysical Techniques 475

Due to random fluctuations on the psychologi- 1/ 2


Ψ A −Ψ B = z A, B × (σ Ψ2 A + σ Ψ2 B − 2rΨAΨBσ ΨAσ ΨB ) .
cal continuum when a person is exposed to the
same stimulus, it will correspond not to one (29.3)
fixed value, but to a certain distribution of the
discriminal processes, which has the form of a It is Thurstone’s law of comparative judgments.
With its help, you can build a psychological scale
normal distribution.
of intervals, where the unit of measurement is the
c. The mean of this normal distribution ( Ψ i ) is SD value of a probability density of discriminat-
taken as the value of the impact of the i-th stimu- ing differences normal distribution. The above
lus on a person on a psychological scale, and its assumptions from Thurstone’s model are incor-
SD is considered as the discriminal dispersion porated in Figure 29.1.A: two hypothetical distri-
(σψi). butions of the discriminal processes of stimuli A
d. The presentation of a pair of stimuli i and j and B, ex., by taste or beauty. Figure 29.1B shows
the probability density distributions of discrimi-
causes, respectively, two discriminal processes –
nal differences (ΨA – ΨB) with a large number of
ψi and ψj. The difference between them (ψj − ψi)
comparative judgements about these stimuli.
is called the discriminal difference. When a pair
of stimuli is repeatedly presented, the corre-
sponding values of the discriminal differences
also form a normal distribution with a mean Stevens’ Approach to Scaling
equal to ( Ψ i − Ψ j ) and SD equal to
In the century after Fechner’s Psychophysics, a
different paradigm was established in parallel,
1/ 2
σ ( Ψ j − Ψ i ) = (σ Ψ2 j + σ Ψ2 i − 2rΨjΨiσ Ψjσ Ψi ) , (29.1) based on the possibility of direct subjective quan-
titative assessment of sensations values. Stanley
where σψi and σψj are the SDs of discriminal pro- Stevens in the mid-1900s (following his predeces-
cesses distributions corresponding to i-th and j-th sors – Plateau, Brentano, Thurstone) developed
stimuli, respectively, and rψjψi is the correlation direct methods for subjective sensory scales con-
between the values of these discriminal processes structing. They were based on observers’ estima-
that occurred when each pair was presented. tion of their increasing sensations order, or
It is assumed that if the discriminal difference distances or relationships between them, or on
(ΨA – ΨB) is > 0, then an assessment will follow assigning them numbers. Therefore, Stevens’
that stimulus B is greater (or preferable) than stim- Psychophysics is called “Subjective one” or
ulus A. And, accordingly, when (ΨA – ΨB) < 0, the “Psychophysics-II.”
opposite comparative assessment will be made. Stevens’ Psychophysics includes not only the
If the distributions of distinguishing processes development of fundamental ideas on the four
overlap (Figure 29.1A), this means that a person types of scales as levels of psychological measure-
makes comparative estimates not unambiguously, ments (the nominal scale, the order scale, the inter-
but with a certain probability. val or partition scale and the ratio scale), but also
The average value of this distribution is equal the creation of subjective scaling methods, called
to the difference in the scale values of stimuli direct scaling methods. Stevens’ methods allow to
A and B (i.e., an estimate of subjective distance construct metric ratio scales (i.e., to designate its
between these stimuli). This value can be found zero point and unit of measurement on the sensory
from the table of areas under the unit (standard) scale). They differ in the simplicity of procedures
normal curve. Knowing the proportion of judg- for obtaining scale values, since the value of the
ments “stimulus A is greater than stimulus B” measured feature on the scale is expressed directly
from the total number of estimates for a given pair in persons’ responses.
of stimuli, we can perform the so–called standard In contrast to indirect scaling methods (Fechner
z-transformation, “P→Z.” and Thurstone), Stevens’ approach was based on
In units of variance σ (ΨA – ΨB), this can be two assumptions: (1) the presence of persons’
written as: internal scales of measured psychological traits;
(2) and, as a result – the ability to make quantitative
judgments about their sensations directly. Indirect
   Ψ A −Ψ B = z A, B × σ (Ψ A −Ψ B ) , (29.2) scaling methods, on the contrary, are based on the
use of intermediate procedures and assumptions
where zA,B denotes the desired scale difference. about how to use behavioral or physiological reac-
Substituting this expression into equation tions to transform them to scale values, and, as a
(29.1), we get rule, the use of a mathematical model describing
476 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

ΨA ΨВ

Psychological scale values, Ψ

ΨВ –ΨA

Figure 29.1
A: Psychological continuum with two partially overlapping distributions of discriminal pro-
cesses, eliciting by multiple presentation of two stimuli. The numbers on the abscissa axis
indicate the magnitude of the stimulus effect.
B: A distribution of discriminal differences between the two discriminal processes (ΨA и ΨB)
resulting from comparative judgments made. 0 on the abscissa axis means the critical value
above which a person makes a judgment about the preference of stimulus B (the light grey
part of the distribution) and below which about the preference of stimulus A (dark grey part).

this transformation. Although direct scaling meth- phenomenon of false alarms ignoring (while
ods were mainly used to construct sensory scales “Modern” Psychophysics pays it great attention).
when the physical continuum of stimuli corre- However, Fechner’s Psychophysics remains the
sponding to sensations is known, Stevens believed Classical one.
that there are no fundamental limitations of this Sensations subjective values determined by
approach to construct a scale of any complicated Stevens’ direct scaling methods, are associated
psychological characteristic. with stimuli values by power dependence. Stevens
received it empirically and mathematically as
well: R = k (S – S0)n (Stevens’ psychophysical
Psychophysical Laws law), introducing a priority postulate (calling both
criticism and agreement) about the constancy of
Fechner has derived the psychophysical law of a jnd sensory equivalent relation regarding this
sensation value dependence on a stimulus value, equivalent initial value in the whole sensations
based on the Weber’s law about the constancy of range. Fechner’s law was confirmed in loudness
jnd increment regarding a stimulus initial value estimations, where logarithmic decibel units were
and a priori postulate about the equality of jnd introduced. Stevens’ law became widespread in
sensory equivalents values in the whole stimuli scaling tasks (Grondin and Laflamme, 2015).
range. It is the logarithmic function: R = k ln (S– The origins of the power function exponent have
S0), where R is the sensation value, S is the actual been shown to derive from the Tweedie probabil-
stimulus value, S0 is the absolute threshold. Thus, ity distribution including Gaussian, Poisson, and
you can calculate a sensation value for the actual Gamma ones. The exponent connections have
stimulus by measuring the absolute threshold. been revealed to many properties of sensory trans-
Fechner’s postulate is criticized reasonably for its duction (Wong, 2018). Mathematical modeling
groundlessness, and the theory – for the has confirmed initial nonlinear relations (power
Advances in Psychophysical Techniques 477

ones particularly) between stimulus values and dS / S = dR / R z , where 0 ≤ z ≤ 1,


receptor neuron responses (Fokin, 2021).
The laws have been developed expressing in where z reflects not an objective, but a subjective
other mathematical equations: exponential func- scale: individual’s psychological operations in
tion (Putter), tangential one (Zinner), arctgenen- stimulus assessing. Zabrodin’s equation describes
tial one, integral φ-gamma function, and others, signals discrimination (z = 0), their values estima-
which are not universal and have narrow appli- tion (z = 1), categorical estimates (0 < z < 1) and
cations. They do not contradict each other, since corresponds to some neurophysiological regu-
describe different sides of sensory and perceptive larities (Zabrodin, Lebedev, 2022). Contemporary
processes. The generalized psychophysical law scaling studies indicate that Zabrodin’s equation
describes both logarithmic and power functions, cannot cover all psychophysical functions.
as well as these and any other mathematical func- Zabrodin has developed a systematic-dynamic
tion between them. approach to sensory processes analysis and a
Zabrodin has built the sensory space (SP) theo- model of an adaptive ideal observer, including
retical model. SP structure is characterized by its other sensory process models as special cases. He
local and global regions and by borders – areas has suggested the general theory of psychophysi-
of adequate reflection. Local areas are determined cal foundations, which has no analogs today. It
in signal detection/discrimination tasks, thresh- has combined four main fields of Psychophysics:
olds measurement, and characterize the topology sensory sensitivity, decision-making, subjec-
of images in SP, its resolution. Global areas are tive scaling, psychophysical laws. However, this
determined in scaling tasks and characterize sub- experimentally theory was not utilized further
jective distances between supra threshold stimuli experimentally after the 1980s.
(SP metric). Topology and metric are connected
with integral-differential relations which identify
the following.
NEW METHODOLOGY IN
1 Internal agreement between Fechner’s and
PSYCHOPHYSICS AND COGNITIVE
Stevens’ laws: both of them are grounded on
Weber’s law about constancy of the jnd ratio PSYCHOLOGY OF ACTIVITY
to the stimulus initial value. The computational
basis of this law has been found further. It is In traditional Psychophysics a participant is con-
“time-intensity equivalence in discrimination,” sidered as just an object of experimenter’s influ-
ences, an ideal but not real observer, whose own
describing how the distribution of reaction times
activity and individual differences are not taken in
changes as a function of their absolute levels account. Quantitative relations are examined
(Pardo-Vazquez et al., 2019). Changes of Weber between external factors used (stimuli values and
fractions have been revealed between forced instructions) and responses indices obtained.
and unforced-choice tasks (allowing the “don’t However, these changes in indices were found in
know” response; Hatzfeld et al., 2018). dependence on the observer’s own variables: dif-
2 The main contradiction between Fechner’s and ferent kinds of individual activity or peculiarities
Stevens’ laws concerns only one item. Fechner’s (Fritsche et al., 2017; Grzeczkowski et al., 2017;
law equates the ratio: the differential threshold, Gurler et al., 2015; Siegel and Kelly, 2017; Wilson
i.e., stimulus jnd minimal increase (dS0) regarding et al., 2016) and confidence (Braun et al., 2017;
Schoenherr and Petrusic, 2015; Vickers and Lee,
the initial stimulus value S (dS0 / S) - to the sensa-
1998). In particular, the psychometric properties
tion jnd minimal increase (dR0): of confidence scales are described within three-
dimensional framework: whether information is
dS0 / S = k dR 0 , from here: R = a ln (S − S0 ) directly or indirectly scaled from the usual pri-
mary decision (about stimuli presented), the locus
Stevens’ law equates the differential threshold of confidence processing (following the primary
to the sensation jnd increase, regarding its initial decision or during it), and the source of informa-
value: tion used to obtain a confidence rating from the
secondary decision (Schoenherr, 2019).
Therefore, the analysis of response propor-
dS0 / S = k dR 0 / R, from here: R = k(S − S0 )n . tions obtained, a “machine-like approach” in
Psychophysics has become insufficient (Vickers
The generalization of these equations leads to and Lee, 1998), and the methodological neces-
the expression: sity have become clear to include the variables
478 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

named into psychophysical analysis. However, the discrimination, and it agrees with data in humans
observers’ own variables are examined separately (Skotnikova and Zhelankin, 2019).
from each other mostly in the studies mentioned. Relations has been found between ST differ-
In Russian Psychophysics, the new systematic ent characteristics and the both inter- and intra-
approach is added to the traditional paradigm. It individual factors affecting sensory performance.
develops the general theoretical and methodo- High activation, achievement motivation, emo-
logical idea of Russian psychology of a person tional stability, and low anxiety facilitated visual
as an active author of his/her mental life, behav- signal detection. Observers’ activation level,
ior, and performance. Subject-oriented research cognitive styles, motivational and volitional dis-
in Psychophysics (Bardin et al., 1993; Gusev, positions, self-regulation were revealed, which
2013; Skotnikova, 2011) considers the observer’s determined cognitive resources mobilization and
own variables as principal ones but not artifacts, strategies efficiency in auditory ST (Chekalina
studied via the integrative approach: participants’ and Gusev, 2011, Emelianova and Gusev, 2018;
individual activity investigation in sensory tasks Gusev and Utochkin, 2006; Pöppel et al., 2018;
(ST) performance. Quantitative psychophysical Volkova and Gusev, 2017). Theoretical useful-
analysis is included into this activity qualitative ness of a “functional organ” concept was shown,
research. which allowed to provide a systematic analysis of
The task is understood as a person’s goal given observers’ activity (Gusev, 2013).
in certain conditions. ST main characteristics are: Thus, the subject-oriented Psychophysics
stimuli (or their differences) low values; random of sensory tasks is the fundamental approach to
order and high (often forced) temp of stimuli pres- sensory signals detection/discrimination inves-
entation, which cause high information load to tigation, which allows expanding psychophysi-
the observer having limited mobility. Individual cal studies boundaries and reveals the role of
activity shows matching of person’s capabilities top-down mechanisms of sensory processes.
and ST conditions which actualize observer’s In cognitive psychology of activity, it presents
individual resources in uncertainty situations for the cultural-historical approach, accounting for
weak signals or small differences between signals human awareness and attention multiple phenom-
detection. A contribution of subjective factors may ena (Falikman and Asmolov, 2017).
cause up to 30% of psychophysical indices total
dispersions and is comparable with stimuli char-
acteristics contributions (Gusev, 2013).
The hierarchical structure of a participant’s
activity appears to be an internal determinant of METHODS OF PSYCHOPHYSICS-I
results obtained. This structure components are:
a task as an observer accepted it, his (her) inner Fechner’s Methods
states (activation, effort, confidence degree),
individual peculiarities (cognitive styles, extra- One of the main tasks of Psychophysics is the
version–introversion, neuroticism–emotional sta- study of minimum and maximum levels of a per-
bility), performance strategies determined by the son’s sensory abilities. In classical Psychophysics,
task and individual peculiarities. These compo- the absolute threshold (absolute limen – AL) is the
nents have been revealed experimentally in signal minimum value of the stimulus that causes a
detection and discrimination, where intensities barely noticeable sensation. AL is a measure of the
of visual (brightness) and auditory (loudness) lower limit of sensory sensitivity and character-
stimuli have been used as well as visual temporal izes a person’s ability to sense signals of very low
(durations) and spatial (line length) stimuli (i.e., value. Thus, the concept of sensory sensitivity is
quantitative” and “qualitative” ones, according defined as the inverse of the absolute threshold:
Stevens, 1957) during their simultaneous and suc- the higher the threshold, the lower the sensitivity,
cessive presentation (Gusev, 2013; Skotnikova, and vice versa. The difference threshold (differ-
2011). The mathematical model of decision mak- ence limen – DL) is the minimal difference
ing and confidence has been suggested for uni- between two stimuli in a certain physical param-
dimensional simple sensory stimuli discrimination eter value, which causes a barely noticeable dif-
(proved experimentally) and for complicated ference in sensations. The difference threshold
multi-attributed perceptual objects (Shendiapin, characterizes the individual discrimination ability
2018). Our approach is continued regarding ani- or difference sensitivity.
mals’ behavior: their individual activity (of differ- The range of perceived stimuli has its own upper
ent reptile’s species: grass snakes’ and slow-warm limit – the upper or terminal absolute threshold.
lizards’ moving in the maze) effects visual stimuli This is the maximum permissible stimulus value,
Advances in Psychophysical Techniques 479

at which it is still felt as a stimulus of a certain 1 N


modality. When it is exceeded, the sensation of AL = ∑ Li   (29.4)
N i =1
this modality passes into the sensation of another
modality (tactile, painful). where AL is the average absolute threshold, Li
In the Fechner’s work, three methods for meas- is the value of a single threshold in both ascend-
uring the absolute and difference thresholds were ing and descending series, N represents the total
proposed, which were called classical psycho- amount of series.
physical methods. Distinct from the modern psy- The regular presentation of stimuli and the cor-
chophysical methods, Fechner’s approach pays responding expectations of participants result in
special attention to stimulus variables, and do the appearance of so-called errors of habituation
not sufficiently take into account the influence of and errors of expectation, which are sources of the
subject’s own variables or so-called non-sensory accuracy of threshold measurements reducing.
factors, which are almost always included in psy-
chological measurements. Statistical indices are
accepted in these methods as threshold measures
of sensory task performance, since they are deter-
mined not only by subject’s sensitivity level, but DIFFERENCE THRESHOLDS MEASURING
also by non-sensory factors controlling a response
choice. In DL measuring stimuli are presented in pairs.
Simultaneously or sequentially with a variable
stimulus (Svar), the subject is presented with a
standard stimulus (Sst), which sets the initial
METHOD OF LIMITS stimulus value, relative to which DL value is
determined. The subject is usually allowed to give
three responses categories: “greater,” “less,”
The method of limits expresses a direct approach “equal” (or “I doubt”). The threshold is taken as
to psychological measurements, since it gives a the stimulus value corresponding to the middle of
threshold value estimation during the measure- the inter stimulus interval, where the response
ment itself. This method procedure most clearly category changes for the first time: from “greater”
reflects the understanding of the threshold as a to “equal” and from “equal” to “less” in the
barrier separating the stimulus series into the two descending series, and from “less” to “equal” and
classes: of sensed and non-sensed stimuli or their from “equal” to “greater” in the ascending series.
differences. Thus, when DL measuring, four threshold values
are estimated (two in each series). This is the
upper threshold (DLh) in the ascending and
descending series (DLh↑ and DLh↓) and the lower
Absolute Thresholds Measuring threshold (DLl) in the ascending and descending
In AL measuring each trial begins with an series (DLl↑ and DLl↓). In Figure 29.2, they are
“Attention” signal, after which a stimulus is pre- marked with dots in each series, which illustrate
sented for a short interval. As a rule, a subject is the rule of a threshold point setting in the descend-
allowed only two responses categories (ex., “Yes,” ing and ascending series.
“No”: “I see,” “I don’t see”). Stimuli presentation DLh and DLl are boundaries of IU (i.e., they
is carried out by a descending or an ascending limit that stimulus region where equality responses
series – it is the principal difference between this prevail). IU is that zone of stimuli that is limited
method and others. In the first case, a severity from above by a stimulus that on average barely
degree of a certain stimulus parameter, sensitivity differs from the standard one (Sst) as a larger one,
to which is measured, gradually decreases from a and from below by the stimulus that on average
maximum to a minimum, in the second, on the barely differs from the standard one as a smaller
contrary. The threshold in this series is taken as the one. IU contains two barely noticeable differences
stimulus value located in the middle of the interval (i.e., it is equal to two DLs):
in which the first change of the subject’s response
category has occurred. In the descending series, IU = Lh − Ll   (29.5)
the threshold for the sensation disappearance is
determined – L↓, in the ascending one – the thresh-
old for its appearance – L↑. The absolute threshold IU Lh − Ll
is taken as the arithmetic mean of all the thresh- DL = = (29.6)
olds of appearance and disappearance found: 2 2
480 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

CE = Sst − PSE   (29.7)

Many authors point out that the value of the SD (σ)


of adjustments made can also serve as a more reli-
able measure of difference sensitivity.

Absolute Thresholds Measuring


In AL measuring the observer adjusts the stimulus
magnitude that initially caused a distinct sensa-
tion, until she/he establishes such a value at which
she/he loses the feeling of the stimulus for the first
time. If the adjusting begins with an obviously
imperceptible stimulus value, then the subject
must find its value at which the sensation appears
Figure 29.2 Illustration of the rule for firstly. When the adjusting begins from a suprath-
determining Lh and Ll and the threshold reshold stimulus, the subject reproduces the mag-
indexes in the descending and ascending nitude of the stimulus at which the sensation
series based on the subject’s responses. disappears for the first time. The final value of AL
is estimated by averaging all separate trials per-
formed by the observer. Usually, the arithmetic
The stimulus located in the middle of the IU as the mean is used.
subjective equivalent of Sst is called the point of
subjective equality (PSE).

METHOD OF CONSTANT STIMULI


METHOD OF ADJUSTMENT
Method of constant stimuli consists in presenting
the subject with a limited number of stimuli that
Method of adjustment differs procedurally from remain unchanged throughout the entire session.
the other threshold methods in that the observer The result of the experiment is the subject’s
himself regulates the value of the stimulus varia- response frequencies, from which the threshold
ble parameter. Method of adjustment gives the values are calculated. Therefore, this method is a
lowest threshold values compared to other thresh- way of thresholds indirectly estimating, as
old methods. opposed to the methods of limits and adjustment.
The constant stimuli method has a reputation for
being the most accurate and reliable, since its
procedure eliminates errors of habituation and
Difference Thresholds Measuring expectation.
According to Fechner, this method was intended
primarily for measuring difference sensitivity. To
do this, the observer is presented with two stimuli
at the same time: the standard stimulus (Sst) and
the variable one (Svar), the value of which can be DIFFERENCE THRESHOLDS MEASURING
smoothly changed by the subject himself. An
observer’s task is to adjust the Svar to the Sst, no When measuring DL in preliminary trials, the
restrictions on the freedom of movement are intro- experimenter approximately determines the
duced during the adjustment process. threshold zone (i.e., the range of stimuli differ-
The arithmetic mean of several adjustments is ences, at the boundaries of which the observer
most often used as a statistical measure for evalu- almost always begins to feel the difference
ating the subject’s performance in the method of between the standard stimulus and the compared
adjustment. In the sense of the concepts intro- one). Then, within this zone, 5–9 stimuli are
duced above, this index is PSE. To estimate DL, selected that are compared with the Sst. The choice
the value of the PSE distance from the Sst is used: is made in such a way that the weakest among
Advances in Psychophysical Techniques 481

them causes the response “greater” in 5–10% of


cases, and the strongest in 90–95%. Stimuli are

z-scores of “greater” responses


presented in pairs simultaneously or sequentially
in random sequence (i.e., the individual cannot
guess neither the place of the Sst in the pair, nor the
Svar value). Usually, each pair of stimuli is repeated
20–200 times. Two (“greater,” “less”) or three
(“greater,” “less,” “equal”) responses categories
are used. In any case, the threshold calculation is
based on a relative frequency of various kinds of
judgments for stimuli each pair.
As the Svar increases, it increases values the pro- Values of comparison stimulus
portion of “greater” responses gradually increases
from 0 to 1. This proportion is convenient to use
when experimental results are presented in the Figure 29.4 Linear psychometric function
form of a special graph called the psychomet- in z-coordinates, obtained as a result of the
ric function (Figure 29.3). It has the S-shaped difference threshold measuring.
form (the ogive) of the normal distribution inte-
gral function, which is theoretically justified
and empirically confirmed. In z-coordinates, the The point of subjective equality is defined as the
psychometric function looks like a straight line median: PSE = Med.
(Figure 29.4). The constant error occurs in the case of the
In the method of constant stimuli, the usual sta- median mismatch regarding the Sst:
tistical indices are used: median (Med) or arithme-
tic mean (X), semi-quartile range (Q) or SD (σ), CE = Md − Sst   (29.9)
as well as the first, second and third quartiles (Q1,
Q2, and Q3). With the help of these indices, the In case of two responses categories, the differ-
following threshold indices are calculated accord- ence threshold is defined as a half of the IU and
ing to an empirical psychometric curve. corresponds to Q and is, in fact, a measure of the
The interval of uncertainty is estimated as the subject’s responses variability (of the psychomet-
interquartile range: (Q3 − Q1) ric curve slope):
IU = S0.75 − S0.25 .   (29.8)
S0.75 − S0.25
DL ( 2) = Q ( 2) =   (29.10)
2
When using a psychometric function con-
structed in z-coordinates, instead of quartile esti-
Proportion of “greater” responses

mates, the SD and the well-known formula are


used:

    σ = 1.483Q       (29.11)

The median of empirical frequencies distribution


is found as a value corresponding to σ = 0.

Values of comparison stimulus ABSOLUTE THRESHOLDS MEASURING

The procedure for AL measuring differs from DL


Figure 29.3 A typical psychometric func- measuring only in one point: in each sample one of
tion obtained by the method of constant several constant stimuli is presented, to which a
stimuli, as a result of the difference thresh- subject gives one of the two possible responses –
old measuring. The empirically found points “Yes “(I feel) or “No” (I don’t feel). According to
are superimposed on the S-shaped form responses frequencies empirically obtained for
curve (ogive). each of the constant stimuli, a psychometric curve
482 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Proportion of “yes” responses


ADAPTIVE THRESHOLD METHODS

Along with Fechner’s methods, adaptive thresh-


olds procedures have been developed in which an
order of stimuli presentation depends on the indi-
vidual’s responses. Such automated procedures
are usually implemented via computer or a techni-
cal device (such as the Bekesy audiometer).
One of adaptive procedures is the method of
limits variant, developed in 1962 by Cornsweet
and called the staircase method (also up-and-
Stimulus intensity down method). The essence of the method is that
as soon as there is a change in the response cate-
gory, there is immediately a corresponding change
Figure 29.5 Typical psychometric function, in the direction of the stimulus value change from
obtained in absolute threshold measuring a descending series to an ascending one. Thus, the
via the method of constant stimuli. The presentation of stimuli adjusts (adapts) to subject’s
empirically found points are superimposed responses, and stimuli values change occurs in a
on the S-shaped curve. fairly narrow near-threshold range (Figure 29.6).
In the Psychophysics literature, other adaptive
methods for measuring thresholds are described
too (e.g., threshold tracking method or forced
choice method – Geschheider, 1997).
is constructed (Figure 29.5). The 50%-point on this
curve (i.e., median is taken as the estimate of AL).
From classical threshold theory, the 50%-point METHODS BASED ON SIGNAL DETECTION
corresponds to the minimum stimulus value that
causes sensation only when there is a balance THEORY IN PSYCHOPHYSICS (SDT)
of favorable and unfavorable factors. The semi-
quartile range (Q) or the SD (σ) characterizes the Yes–No Method
threshold estimations reliability.
There is a problem of correlation between It is one of the most popular SDT methods for
Fechner’s three methods’ threshold indices. For sensory sensitivity measuring. Unlike classical
proprioception testing no significant correlations Psychophysics, we measure not the absolute or
have been found, possibly due to different cogni- difference threshold, but sensory sensitivity in
tive processes involved in the corresponding tasks signals detection or discrimination tasks. In this
performance (Yang et al., 2018). method, only two types of stimuli are used, which

Stimulus Number of trials


intensity 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
10 +
9 +
8 +
7 +
6 +
5 + + +
4 + + + + +
3 - - - -
2 - +
1 -

Figure 29.6 Recording of subject’s responses and changing stimuli values when measuring
the absolute threshold by the staircase method. Plus and minus signs correspond to the
“Yes” and “No” responses, respectively. As a result, AL corresponds to Stimulus 4.
Advances in Psychophysical Techniques 483

differ slightly in the value of some physical qual- d'


ity: one “significant” or “signal” – S (for example,
a weak tone of 1000 Hz on white noise back- N S
ground), and the other “empty” or “noise” – N
(white noise without tonal additive). Stimuli pres-
entation follows each other many times in a
random order at regular intervals, and each time
the observer answers responds “Yes” if he thinks
C Xs
there has been a signal (or a difference between Decision mak
compared signals), or “No” if he does not detect a
signal (or a difference between signals).
There are four possible <stimulus-response> Figure 29.7 The distributions of sensory
combinations, with two correct responses: “Yes” magnitudes probability densities under signal
(Correct signal Detection – Hit) and “No” (Correct (f(X/S)) and noise (f(X/N)) stimuli effects.
signal Rejection – CR) and two erroneous ones:
“Yes, there was a signal,” when only noise was (The abscissa is sensory magnitude, while
presented – False Alarm (FA) and “No” – signal ordinate axis is probability density. The light
Miss (Miss). grey area – False Alarms; The dark grey area –
Observer’s results in signal detection or dis- Misses – the symmetrical criterion position.)
crimination are usually presented in the form of
conditional probabilities scores: P(Hit), P(FA), (its strictness) is estimated as the likelihood ratio
P(Miss), P(CR). P(Hit) and P(FA) fully character- (Figures 29.8 and 29.9):
ize an observer’s performance, since P(Miss) and
P(CR) just complement them up to unity. ordinate of S distribution at criterion
β=
Stimuli effects are associated with their sensory ordinate of N distribution at criterion
representations randomly or stochastically. In one
of the SDT variants, this relationship is depicted as Each pair of empirical P(Hit) and P(FA) scores
two overlapping probability density functions of the can be matched with a point inside the unit square
normal distributions (Figure 29.7). The abscissa is (Figure 29.10). It represents observer’s use of
a hypothetical axis of sensory magnitudes that have various criteria, for example, from five different
appeared under appearance of S (right distribution payoff matrices. The curve obtained is called the
f(X/S)) and N (left distribution, f(X/N)). Receiver’s Operating Characteristic (ROC) curve.
Their overlap means that the sensory represen- P(Hit) and P(FA) change together and recip-
tations of S and N are similar to each other, and rocally (i.e., it is impossible to simultaneously
the observer cannot decide with 100% certainty increase one of them and decrease the other
each time which stimulus has been presented (or only by the criterion changing). T Only a pair
whether there has been a difference in a pair of of these probabilities and not each of them sepa-
stimuli). Further, in SDT, it is assumed that when rately, characterize the observers’ sensory ability
making a decision, the observer sets a strictly (Figure 29.10). The higher the observer’s sensory
defined rule of correspondence between sensa-
tions obtained and types of response (“Yes” and
“No”). If the current sensation value is above a
certain critical level or criterion, then the observer N S
says “Yes,” if under, the observer says “No.”
The criterion is a certain memory sensory
image or a sensory standard (point C on the
abscissa axis in Figure 29.7) with which each
sensory representation is compared. Its position
on the sensory magnitude axis may depend on a Xs
number of reasons: subjective weights of various C
β= / =
errors, knowledge of objective probabilities of S
and N presentation, “payments” and “penalties,”
for correct and erroneous responses respectively Figure 29.8 The liberal criterion position
(payoff matrix), etc. When using a liberal or a on the sensory magnitude axis.
conservative criterion, there is P(Hit) and P(FA)
increase or decrease respectively. The criterion (The designations the same as in Figure 29.7.
position uniquely determines P(Hit) and P(FA) The grey area of S-distribution – Hits, its
empirical scores. The theoretical criterion value light area – Misses.)
484 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

and P(FA), converted into units of SD according to


the normal distribution table.
N S
FORCED CHOICE METHOD (FCM)

FCM has an important advantage over the “Yes–


No” method, since its procedure stabilizes fluctua-
tions of the criterion position, as SDT states. It is
C
Xs achieved by the fact that when a stimulus trial is
β= / = t criterion presented, it always contains a signal stimulus along
with one or two non-signal stimuli. Let’s consider
this procedure on the example of Two Alternative
Figure 29.9 The conservative criterion Forced Choice (2AFC) as the most frequently used
position on the sensory magnitude’s axis. variant. In the 2AFC stimuli presentation is always
(The designations are the same as in Figure carried out in pairs, either sequentially or simultane-
29.8.) ously. One pair always contains S and N, and the
subject knows it, but he must decide himself, which
of the stimuli contains a signal, and which is non-
signal. The place of S and N in the stimulus pair
changes randomly. The experimenters agreed to call
one of the two variants of correct and erroneous
p(H) responses as Hit and FA. In all other respects, 2AFC
is no different from the “Yes–No” method.
If the trial includes 3, 4, or 6 empty stimuli, the
FCM variants are called 3AFC, 4AFC, or 6AFC,
respectively. Since the stimulus procedure of the
p(FA) FCM itself excludes a bias of observer’s responses
in favor of a strict or liberal criterion, a good meas-
ure of sensory sensitivity is the percentage of cor-
Figure 29.10 ROC curve in liner coordinates. rect responses P(C). As in the “Yes–No” method,
the index d’ is also used, calculated by the formula:
sensitivity, the more convex the ROC is and the
closer it is to the upper-left corner of the square d ' 2 AFC = 2d ' Yes − No
(Figure 29.11). On the diagonal corresponds to
the zero level of sensory sensitivity.
SDT introduces a special sensory sensitivity CONFIDENCE RATING METHOD (CRM)
measure d’, as the distance on the horizontal axis
(Figure 29.7) between the centers of the two dis- The CRM using is recommended when it is neces-
tributions f(X/S) and f(X/N), expressed in SD units. sary to build a ROC curve within one session,
Based on empirical data, d’ is calculated as: d’ = without changing either a payoff matrix or a priori
z(Hit) – z(FA), where: z(Hit) and z(FA) are P(Hit) probability of a signal stimulus presentation.

N
1.5 S
d’ =
=1 0.3
d’ d’
=
=0

Xs
d’

N S
p(H)

Xs
N S

Xs
p(FA)

Figure 29.11 A family of ROC curves in observers having different sensory sensitivity (at
left), and corresponding distributions of s and n sensory magnitudes (at right).
Advances in Psychophysical Techniques 485

The only CRM distinction from the “Yes–No” ADVANTAGES AND DISADVANTAGES OF
method is that after each trial the subject indicates CLASSICAL AND SDT-BASED METHODS
a degree of his confidence in the signal presence/
absence instead of responses “Yes” or “No.” As a
rule, a percentage scale is used: 100%, 75%, 50%, Classical psychophysical methods give integral
25%, or 0% confidence that there is a signal. performance indices, but not separate estimations
Suppose that the individual is given seven eval- of their components: sensory sensitivity and deci-
uation categories (from –3 = “exactly No” to +3 = sion-making criterion. Nevertheless, they are still
“exactly Yes”). Usually, the experiment is carried used in sensory measurements, where strictly
out without a payoff matrix or with a symmetric precise sensitivity estimation is not necessary.
payment matrix and with P(S) = P(N) = 0.5. The (1) In preliminary trials before main psychophysi-
results of such an experiment can be presented in cal research. (2) In applied massive measurements
the form of Table 29.1, whereby p and q are esti- for picking up different stimuli values for different
mates of an observer’s confidence in s and n detec- persons when estimating their individual sensory
tion, respectively. It is assumed that if the observer thresholds. As distinct, SDT–based methods can’t
is given seven categories (from complete confi- do it, since to all persons the same two stimuli are
dence of the S absence to complete confidence of presented, which may induce 100% detection/
the S presence), then, when making a decision, he discrimination in some of them, while 0% in some
is based on a value of a certain sensory quality (as others because of different individual sensitivity.
in the “Yes–No” experiment), but divides a sen- (3) Methods of limits and adjustment are economy
sory continuum not into two areas, and into seven and suitable in medicine for express sensory
areas. In fact, the confidence level sets the level of defects diagnostic. (4) Method of adjustment is
criterion strictness for signal detection. appropriate for performance strategies in uncer-
Based on these data, a fairly smooth six-point tainty studies since persons themselves manage
ROC curve can be constructed. Of course, in order stimuli values actively. (5) Method of constants is
to obtain such data, it is required to use a larger suitable for differential characteristic of threshold
number of trials than in the YNM. Mutual inter- regions obtaining by psychometric functions.
relations were found experimentally between indi- SDT-based methods are used to obtain pre-
ces obtained in the methods of confidence rating, cise separate indices of sensitivity and criterion.
lumen, and constants (Zabrodin and Shpagonova, Yes–No and Rating Methods are appropriate for
1988). Fifty years of SDT in Psychophysics and decision making dynamics primarily. Rating gives
of ROC analysis have been traced (Adams et al., the most smooth and stable ROC, but is more
2017). difficult for an observer who has keep and use

Table 29.1 The theoretical results of the CRM experiment


Stimuli Rating -3 -2 -1 0 1 2 3
categories

Ss p(-3) p(-2) p(-1) p(0) p(1) p(2) p(3)


Nn q(-3) q(-2) q(-1) q(0) q(1) q(2) q(3)

Note. According to data, six pairs of P(Hit) and P(FA) values can be calculated, and six criteria can be set on the sensory
effects axis (Table 29.2).

Table 29.2 P(Hit) and P(FA) calculating based on CRM data


Criterion position P(Hit) = P(FA) =

C3 p(3) p(3)
C2 p(2) + p(3) p(2) + p(3)
C1 p(1) + p(2) + p(3) p(1) + p(2) + p(3)
C0 p(0) + p(1) + p(2) + p(3) p(0) + p(1) + p(2) + p(3)
C-1 p(-1) + p(0) + p(1) + p(2) + p(3) p(-1) + p(0) + p(1) + p(2) + p(3)
C-2 p(-2) + p(-1) + p(0) + p(1) + p(2) + p(-2) + p(-1) + p(0) + p(1) + p(2) +
p(3) p(3)
486 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

several criteria simultaneously. According SDT, criterion was found. Line lengths discriminabil-
forced choice method estimates sensitivity most ity decreased in medium and low stress resist-
precisely, since criterion is rather stable. ance patients, while criterion decreased in the first
ones and increased in the last ones. But sensitivity
increased and criterion decreased in high stress
resistance patients (Zabrodina and Sorokin, 2019).
SENSORY SENSITIVITY AND DECISION-
MAKING: MUTUALLY INDEPENDENT IN
EVERY PERSON AND TASK?
METHODS OF PSYCHOPHYSICS II
According to SDT, sensory sensitivity (deter-
mined by stimuli values and an observer’s physi- Stevens’s Paradigm in Subjective
ological limits) and decision-making (determined Scaling: Ratio Scales
by stimuli priory probabilities, responses costs
and payoffs, instructions and feedback) are mutu- In all Stevens’s scaling methods, two types sub-
ally independent in every person and every task. jects’ responses organization are used: estimation
Nevertheless, the opposite data were obtained (by procedure, when they report about the sensations
Russian psychophysicists mostly). Instructions, values that have arisen in numerical form, and
induced a criterion shift, led to sensitivity decrease production procedure, in which they produce
or violations: in persons with mental disorders specified sensations values, adjusting correspond-
(because they were stressed: Matveeva, 1981), in ing stimuli values. Stevens developed the four
healthy, highly trained observers (Mikhalevskaya main methods for constructing sensory ratio
and Finkel, 1985; Zabrodin et al., 1984), in intro- scales: magnitude estimation, magnitude produc-
verts and extraverts (opposite dynamics in them; tion, ratio estimation and ratio production. Let’s
Zabrodin et al., 1981) – as a result of cortical consider two of them briefly as the most charac-
information processing primary phases violation teristic ones.
(judging by Evoked Potentials early components: Two modifications of ratio production method
Mikhalevskaya et al., 1988). Thus, non-sensory are known: the fractionation (or fractioning)
information led to criterion initial shifts and reac- method and the multiplication method. In the frac-
tively – to sensitivity changes. It points to system- tionation procedure, a subject is presented with
atic integration between them. Their correlated several standard stimuli (Sst) and selects compa-
dynamics have been described under observers’ rable stimuli (Sc), the subjective values of which
activation level changes. In vigilance tasks sensi- equal to a given part of the corresponding Sst val-
tivity decreased due to activation drop during ues. Usually, simple fractions (1/n = 1/2, 1/3, etc.)
monotonous work with low signal probabilities. are given. Most often, 1/n = 1/2 is used (i.e., divi-
The criterion increased when observers assimi- sion in half). When selecting a stimulus that is in a
lated these probabilities and decreased at high given relation to Sst, either estimation or reproduc-
probabilities, while both sensitivity and criterion tion procedures are used. In the first case, a direct
increased with increasing differences between estimation of the Sc to Sst ratio is made, and in the
signals (Parasuraman and Moulona, 1987). In second case, the Sc value is adjusted in accordance
monotony and fatigue sensitivity decreased, while with the ratio specified in the instruction. The mul-
criterion increased in the first case (to maintain tiplication procedure is opposite to the fractiona-
vigilance, weakening due to the monotonous tion one: the subject must select a comparable
work) and decreased in the second one (to com- stimulus that exceeds the standard stimulus by a
pensate “Yes” responses frequency decrease, given number of times.
caused by sensitivity decrease; Frishman, 1991). To obtain a scale using the magnitude estima-
In these works, and under pharmacies, activation tion method, an observer is presented with a fixed
level changes caused sensitivity changes, and set of supra threshold stimuli covering a fairly
they, in turn evoked the criterion changes. As a wide range of the measured characteristic. There
whole, non-sensory information induced criterion are two forms of this method: with a set mod-
shifts initially and sensitivity changes secondarily, ule or with a free module (otherwise it is called
while activation level changes caused sensitivity without a module). In the first case a subject is
changes, which subsequently led to criterion shifts presented with a standard stimulus and a certain
(considering this rubric, see Skotnikova, 2008). numerical value (e.g., 100), which is signified as
Under patients’ stress before a dental treatment, the corresponding sensation The subject should
individually different dynamics of sensitivity and relate all own estimates of the stimuli values with
Advances in Psychophysical Techniques 487

Thurstone’s law of comparative estimates is the


method of paired comparisons. The material for
constructing the scale is the frequency of com-
parative assessments of the type: “I prefer stimu-
lus j more than stimulus i.” It is fundamentally
important that this is a one-dimensional subjective
characteristic.
In each sample, the individual’s response about
the preferred stimulus in the pair is recorded.
Based on these comparisons, the frequency of
preference of one stimulus to another is deter-
mined for each pair. As a rule, each pair of a set
of n stimuli is presented for comparison several
times. Data processing consists in the transition
from the frequency matrix nxn to the probability
matrix. The element of this matrix - pA,B, is the
relative frequency of the number of preferences of
stimulus A to stimulus B to the total number of
Figure 29.12 The psychophysical loudness comparisons of these two stimuli. We pass from
function of 1000 Hz tone signal. the probability matrix to the difference matrix, tak-
ing into account that the value of the subjective
difference between the two stimuli is expressed in
this module. In the second case, no stimulus is units of the SD of the normal distribution. Thus,
declared as standard, and no restrictions are intro- a matrix is obtained, the element of which (zA,B)
duced when choosing numbers for responses. The in the framework of the Thurstone’s model is an
only requirement for the individual to try to use estimate of the subjective difference (ΨA – ΨB),
numbers that accurately express the magnitude of measured in units of the SD of the distribution of
the sensation caused by the stimulus. discriminable differences.
In the Stevens’ work, a number of ratio sensory In the original work of Thurstone five variants
scales were constructed: scales of loudness, sweet of the application of this law are considered. S
sensations, and so on (Geschheider, 1997). Some However, simplified version of Thurstone’s law or
of them are widely used as international standards its V version (rΨAΨB = 0 and σΨA = σΨB = σ) has
for assessing sensations intensities (e.g., the sone received the greatest application:
scale curves of equal loudness; Figure 29.12).
The abscissa axis is the signal intensity in deci- Ψ A −Ψ B = z ABσ   (29.12)
bels above the absolute threshold. The ordinate
axis is the subjective loudness estimates in sones. In 1958, American mathematical psychologist
One sone is defined as loudness of 1000 Hz at 40 (Warren Torgerson) proposed specific computa-
dB above the absolute threshold. For comparison, tional algorithms for implementing each of the
the straight dotted line is a function predicted by five variants of the law.
Fechner’s law.
A detailed description of the procedures for
ratio scales constructing and examples of sensory
scales can be found in (Geschheider, 1997). METHODS BASED ON DIRECT ESTIMATES

Other methods for interval scales constructing are


based on observers’ direct estimations of a certain
SUBJECTIVE SCALING PARADIGMS: feature scaled value. Partition scaling methods
PARTITION SCALES: MEASUREMENT OF assume that a person is able to divide a given con-
tinuum of stimuli into equal sensory distances.
PAIRED COMPARISONS Within the framework of this paradigm, two
groups of methods have been developed: equisec-
Above, we have already described Thurstone’s tion method and category scaling method
approach. Below we consider the method of (Gecsheider, 1997).
paired comparisons, which allow us to build a The equisection method requires an individual
scale of intervals. An empirical procedure for to divide a range of stimuli set for scaling by
constructing a scale of intervals based on equal sensory distances. Two slightly different
488 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

techniques are usually used for a subjective scale An alternative to the simultaneous solution is
obtaining. A subject is presented with the smallest the sequential solution procedure, when a sub-
and the largest stimuli from the set of N stimuli, ject is required to select only one stimulus from
and asked to choose N-1 stimuli in order to obtain a sequential series of stimuli samples that divides
equal sensory distances between them. This pro- the specified sensory interval in half.
cedure is called a simultaneous solution, since The categorical scaling method is also designed
all the scale values are evaluated simultaneously to measure sensory qualities on the interval scale.
by the observer. A similar procedure is illustrated In this method, a subject is presented with a vari-
in Figure 29.13. First, the individual is asked to ety of stimuli and is asked to match them with a
appraise for himself the magnitude of the per- certain number of categories. Usually numbers (1,
ceived difference between two extreme stimuli, 2, and 3) or descriptive adjectives (low, medium,
for example, 10 (smallest) and 100 (largest). Then and high) are used as categories. The method of
the participant is asked to set the intensities’ val- equal-appearing intervals is one of the most wide-
ues of the other three stimuli in such a way that the spread and simple procedures for categorical scal-
entire range of the perceived difference between ing. It is assumed that a person is able to take into
the extreme stimuli is divided by these three account the intervals between the boundaries of
stimuli into four subjectively equal distances. In the categories used, when he attributes stimuli to
our example, the values 1 and 5 denote sensations different categories. Based on this assumption, the
caused by extreme stimuli, 10 and 100, respec- categories associated with certain stimuli are con-
tively. The values of sensations 2, 3, and 4 cor- sidered as values on the interval scale. To obtain
respond to the stimuli selected by the subjects reliable scale values by this method, averaging of
themselves. As a result, four equal distances are a large number of judgments is required, which
obtained on the psychological axis: 1-2, 2-3, 3-4, is achieved by using a large number of individu-
4-5. On the stimulus magnitude axis, the values als or presenting a large number of stimuli to one
10, 20, 35, 55, and 100 correspond to the scale individual.
values obtained by this procedure ( 1, 2, 3, 4, 5:
Figure 29.13 at top). Based on these data, the psy-
chophysical function is constructed (Figure 29.13,
at bottom).
MULTIDIMENSIONAL SCALING (MDS)

Advantages of MDS
Subjective magnitude of the stimulus
MDS began its intensive development in the
middle of the twentieth century in the works of
American scientists (Torgerson, Shepard,
Adjustable
Kruskal). The MDS is a field of mathematical
Smallest Largest
psychology (psychometric). In the most general
form, MDS can be defined as a mathematical
The physical value of the stimulus tool designed to process data on pairwise simi-
larities, connections and relationships between
the analyzed objects (stimuli) in order to repre-
sent these objects as points in a certain coordi-
Subjective magnitude

nate space.
of the stimulus

The MDS methods are used to build a model


of human behavior when making judgments
about the similarity between objects of differ-
ent modality and complexity (colors, graphemes,
faces, etc.). The MDS is based on the assumption
that when comparing stimuli, a person (explic-
itly or implicitly) focuses on their basic charac-
The physical value of the stimulus teristics. The greater the divergence of stimuli
according to these characteristics (factors), the
higher the subjective measure of the difference
Figure 29.13 Equisection scale (at the between them. The process of assessing simi-
bottom) determined by a simultaneous solu- larities (or differences) between objects by an
tion (at top). ­individual can be represented in the form of a
Advances in Psychophysical Techniques 489

“black box,” the input and output of which are (“subjective space”). There objects, distinguished
formed by evaluated objects and their subjective by an individual, are graphically represented as
estimations, respectively. The task is to uncover certain points, and inter point distances depict
this black box. The MDS allows solving this “subjective differences” between the objects.3
problem by building a geometrical coordinate
C) Analysis of objects’ coordinate functions in the
space where objects are represented as certain
points, and inter points distances correlate with space (specifically, values of their projections on
subjective scores. Such a model is understood as the axes) is provided with the aim of identifying
a system of rules, guided by which it is possible the magnitude of contribution of each axe to
to generate the same results about objects’ simi- object’s characteristics. At this stage, it is impor-
larities that have been perceived by the subject. tant to identify the axes that make the greatest
The geometric representation of the objects in the contribution to the spread of coordinates in order
space of a small number of dimensions provides to minimize the dimensionality of the model.
a convenient visual analysis and interpretation of D) Usage the coordinate functions for interpretation
experimental data. of the space axes as some factors, determin-
The main task of MDS as a mathematical tech-
ing discrimination between objects. The axes
nique is to identify a structure of a studied set of
objects. Identification of the structure is under- of the model, which form its orthogonal basis,
stood as identification of a set of main ­factors, may be considered as the quantitative scales
determining objects’ differences, and description (metric scaling) or the ordinal ones (non-dimen-
of each object in terms of these factors. The pro- sional scaling), reflecting the contribution of
cedure for constructing the structure is based on some independent factors (the variables) to
the analysis of objective or subjective information objects’ discrimination. The measure of similar-
about the proximity between objects or informa- ity between two objects is the inverse of the
tion about preferences of a set of objects. distance between their corresponding points. The
MDS algorithm includes the following steps: closer the points are to each other, the higher
the measure of similarity between correspond-
A) The measurement of large (supraliminal) dif- ing objects (and the lower the measure of dif-
ferences1 between all pairs in a sample of n ference). Subjective evaluations of differences
objects. Values of “pair wise differences” may between stimuli represent the cumulative effect:
be obtained in psychophysical (Steven’s method (a) of the activity of all neural networks involved
of direct scaling) and psycho physiological (EEG, in stimulus physical characteristics encoding,
ERP)2 experiments in humans, in behavioral and (b) of electrophysiological pattern decoding
(methods of conditioned learning) and neuro- into a cognitive image. In order to understand
physiologic (registration of neuronal activity) the overall structure of the cognitive system,
experiments with animals. Methods of measuring it is necessary to distinguish contributions of
large supra threshold differences between stimuli separate links in this integral estimation of inter
in humans have a long tradition and are fully stimulus differences. In some cases, it is possible
detailed in handbooks on scaling (Torgerson,). to interpret objects’ coordinates as a reflection of
Electrophysiological techniques for such meas- the contributions into their characteristics, made
urements are less well developed. They provide by the neurophysiologic mechanisms (channels).
information on stimuli discrimination by large Thus, the geometric modeling, based on the MDS,
neural networks, recording evoked potentials in is not simply a technique for the formal descrip-
the cortex and in the sub cortical brain structures tion of psychological processes and states, but
of humans and animals, as well as by local area a special approach, a formal language that, like
networks, recording extracellular and intracel- artificial international language Esperanto, ena-
lular neuronal activity. At the foundation of these bles to integrate and analyze both psychological
measurements is a method first developed by phenomenology and the neurophysiologic data
Bongard for instantaneous exchange of stimuli, in one unified mathematical language. The MDS
which in the English-language literature has methods are actively used for big data of expert
been then called “stimuli exchange” or “silent information analysis and visualization not only in
substitution method (Izmailov et al., 2015). psychology, but also in various fields of sociol-
B) Analysis of the resulting matrix of pair wise differ- ogy, economics, history, geography, pedagogic,
ences (n x n) provided by the MDS methods aims anthropology, etc. (cf. Davison, 1991; Izmailov
of construction a geometrical coordinate space et al., 2005, 2015; Sokolov, 2013).
490 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

METRIC MULTIDIMENSIONAL SCALING The use of uniform metrics assumes that when
(mMDS) assessing similarities (or differences), a subject
equally takes all factors into account. One may
resort to a weighted metric, where a certain weight
Given the pair wise dissimilarity matrix D = (dxy), is attributed to each factor, when there is a reason
mMDS seeks the Euclidean space construction, to assert that the factors are not equal for the indi-
which allows using quantitative calculations for vidual and he takes them into account in different
representing differences between any pair of degrees. Different persons can take into account
objects in resulting matrix, as some distances different factors. Then each individual is charac-
between points in a geometrical model. This terized by his/her own set of the factor’s weights
requires that values of pair wise differences Wti. The weighted metric of Minkowski has the
between any objects x and y in an experimental form:
matrix must satisfy the axioms of a distance, that r
is, they must represent the values of a “distance p

function,” called “metric”:


d ijk = p
∑w i
t x jt − x kt
t =1

1 d (x, y) ≥ 0, Such a model is called “individual scaling” or a


2 d (x, y) = 0 if and only if x = y, “weighted factor model.” Geometrically, it is inter-
3 d (x, y) = d (y, x), preted as follows. Let there be a configuration of
4 d (x, z) ≤ d (x, y) + d (y, z). points in the coordinate space that reflects the per-
ception of some “average individual” in the group.
Euclidean models are constructed by orthogonal- In order to obtain the perception space of the
izing the initial matrices. Torgerson’s method is i-th subject, it is necessary to stretch the “average
based on orthogonal projection into a subspace, configuration” in the direction of those axes, for
formed by axes that are characterized by a signifi- which Wti > Wtav, and compress it in the direction
cant spread of the objects-points. The solution is of the axes, for which Wti < Wtav. Let’s consider
sought by factorizing the matrix of initial differ- the case when in the space of two factors for the
ences: its Eigen values and eigenvectors are calcu- average individual all objects-points lie on a cir-
lated that determine the axes orientation. The first cle. Then for an individual, which is characterized
axis is characterized by the maximum spread of by weights W1i = 2, W2i = 1, these stimuli will be
the points along it, the second axis is orthogonal to located on an ellipse elongated along the horizontal
the first one and is determined by the next largest axis, and for an individual, which is characterized
spread, the third axis is orthogonal to the plane of by weights W2i = 2, W1i = 1, they will be located on
the first two axes, etc. After determining the an ellipse elongated along the vertical axis.
dimensionality of the model, one needs to enter a Using this metric, the degree of adequacy of
metric in the coordinate space, the choice of which the model regarding the initial data is evaluated.
has a great influence on the modeling results and For this purpose, a quality criterion is introduced,
on interpretation of dataset. Minkowski’s metric called “stress,” which measures a degree of dis-
(cf. Billock et al., 2021) is usually used: crepancy between differences Djk in the matrix
and corresponding distances djk in the space.
r
p Different types of functional are used as stress cri-
d jk = p
∑x jt − x kt
terion (SD). In the simplest case it looks like this:
t =1

2
where r is the dimension of the space, djk is the SD = ∑( d jk − D jk )
distance between the points corresponding to the j<k

j-th and k-th objects; xjt and xrt are the values of the
projections of the j-th and k-th points on the t-th Another formal criterion for the adequacy of the
axis. The most common cases of this metric are model can be Pearson’s correlation coefficient
the Euclidean metric: between Djk values in the original matrix and djk
r
distances in the model (it should be high enough).
2 One more way to approximate the point-to-
d jk = ∑x jt − x kt
point distances is to calculate the scalar products
t =1
of the vectors, connecting the object-points with
and city-block metric (p = 1): the center of the space coordinate system:
r r

d jk = ∑ x jt − x kt b jk = ∑ x jt x kt
t =1 t =1
Advances in Psychophysical Techniques 491

In this case, the maximum values of the scalar dimensionality. Figure 29.14 represents the results
product correspond to the minimum values of the of the mMDS application in psychophysical
differences between evaluated objects. research of perception (subjective evaluation) of
One of the ways to increase the accuracy of face emotional expressions. In study by Izmailov
the formal solution is the increasing of the space et al. (2015), using the mMDS of the matrix of

Figure 29.14 Projection of schematic face-points on the three planes.


(A) X1X2, (B) X3X4, and (C) X1X3 of a four-dimension space derived through multidimensional
scaling of dissimilarities estimates.
492 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

paired differences between 25 facial stimuli, a The non-metric approach has some advantage
geometric model of schematic facial emotional over the metric one: it is not limited to orthogonal
expressions perception was constructed. In this (linear) design and allows getting a good represen-
model, the schematic faces were represented by tation of a set of objects in the space of a smaller
points in a four-dimensional space so that the number of dimensions.
Euclidian distances between points were propor-
tional to perceived differences between the emo-
tional expressions of the presented faces. Three
angles of the four-dimensional sphere are jux-
taposed with subjective characteristics of facial MDS VS FACTORY ANALYSIS
expressions called “emotional quality” (joy, sor-
row, sadness, etc.) (Figure 29.14, A), “emotional The MDS has some advantages over Factory
intensity (activity)” (Figure 29.14, B), and “emo- Analysis (FA), which also aims at finding latent
tional saturation (fulness)” (Figure 29.14, C). The variables (factors) underlying estimates of differ-
four Cartesian coordinates of this space were com- ences between objects. For example, the MDS can
prised of neural channels activation, coding mouth be applied to any types of distances or similarities,
and brow linear orientation. if the FA algorithms are based on the analysis of a
Point numbers correspond to numbers of face- matrix of normally distributed values of linear
stimuli (adapted from Izmailov et al., 2005, 2015). correlation coefficients. If the FA requires obtain-
Such structure of the emotional space is simi- ing estimates of objects compared for a certain set
lar to the structure of the color space, where the of features, then the MDS allows using the direct
color tone corresponds to the emotional tone, the ratings (subjective scaling) of differences.
brightness of the color corresponds to the intensity The correlation matrices, used in the FA, can be
of emotions, and the color saturation corresponds converted into difference matrices:
to the emotional saturation. The obtained data
allow shedding light on some general principles of D ij = (2 − 2 rij )1/ 2
encoding information in the visual system.
The difference measures Dij obtained in this case
do not violate the axioms of distances and can
be processed using mMDS methods (Davison,
NON-METRIC MULTIDIMENSIONAL 1991).
SCALING (nMDS)

If the original difference matrix is not an exact Notes


distance matrix in any metric space, then a non-
metric (or monotonic) approach can be used to 1  Estimates of the similarity and correlation
build the model. The essence of the approach between the analyzed objects can also be used as
consists of a multi-stage procedure for selecting input data for MDS. For brevity, the text refers to
such a sequence of numbers d̂ that would be{} the “inter stimulus differences.”
2  EEG – electroencephalogram; ERP – event-related
monotonically related to initial differences (de
Leeuw, 2017; Shepard, 1981). The functional of potential.
the following form is used as a stress criterion: 3  To facilitate comparative analysis, the terms “sub-
ject,” “subjective difference,” and “subjective
2 space” are used for both humans and experimen-
∑ (d jk − dˆ jk ) tal animals.
j<k
Sd̂ =
∑d 2
jk
j<k

Non-metric scaling is based on gradient proce- REFERENCES


dures for this functional minimizing. The dis-
tances between points are calculated, as a rule, Adams, R. D., Waddington, G., Steinberg, N. & Han,
using the Euclidean metric, which is not sensi- J. (2017). John Swets, ROC curves, and fifty years
tive to the axes rotation and the coordinate center of signal detection theory and Psychophysics. In K.
transferring. The quality of the solution does not Ueda, H. Ito, G. B. Remijn & Y. Nakajima (Eds.),
depend on the coordinate system direction, and for Fechner Day 2017. Proceedings of the 33rd Annual
a meaningful interpretation they must be oriented Meeting of the International Society for Psycho-
accordingly. physics (pp. 51–56). Fukuoka: Kyushu University.
Advances in Psychophysical Techniques 493

Bardin, K. V., Frishman, E. Z., & Skotnikova, I. G. differences in multisensory speech perception and
(1993). Quantitative data analysis in Psychophysics eye movements. Attention, Perception & Psycho-
in qualitative context. In R. Steyer & K. Wender physics, 77, 1333–1341.
(Eds.), Psychometric methodology (pp. 14–17). Gusev, A. N. (2013). Ot psikhofiziki «chistykh” osh-
Frankfurt: Fisher Verlag. chushcheniy k psikhofizike sensornykh zadach:
Billock, V. A., Kinney, M. J., Schnupp, J. W. H., & Mer- Sistemno-deyatel’nostnyy podkhod v psikhofizike
edith, M. A. (2021). A simple vector-like law for [from the Psychophysics of “pure” sensations to
perceptual information combination is also followed the Psychophysics of sensory tasks: A system-
by a class of cortical multisensory bimodal neurons. activity approach in Psychophysics]. Voprosy Psik-
Science, 24(6). doi: 10.1016/j.isci.2021.102527 hologii, 3, 143–155.
Braun, A., Urai, A. E., & Donner, T. H. (2017). Adap- Gusev, A. N., & Utochkin, I. S. (2006). Rol’ aktivatsii
tive history biases result from confidence-depend- sub”yekta v reshenii sensornykh zadach razlichnoy
ent accumulation of past decision variables biases slozhnosti: Resursnyy i funktsional’nyy podkhody.
perceptual choice. BioRxiv. doi: 10.1101/172049 Psikhologiya, 4, 21–31.
Chekalina, A. I, & Gusev, A. N. (2011). Osobennosti Hatzfeld, C., Kupnik, M., & Kattner, F. (2018). Com-
kognitivnogo stilya pri vypolnenii sensornykh parison of Weber fractions obtained in a simula-
zadach: Systemno-deyatelnosnyi podkhod k ana- tion study and a behavioral experiment using
lizu protsessov raspoznavaniya I obnaruzheniya unforced choice paradigms. In F. Muller, L. Lud-
signalov. Saarbrucken, Germany: LAP Lambert. wigs & M. Kupper (Eds.), Fechner Day, 2018. Pro-
Davison, M. L. (1991). Multidimensional scaling. ceedings of the 34th Annual Meeting of the
Malabar, FL: Krieger. International Society for Psychophysics (pp. 155–
de Leeuw, J. (2017). Shepard non-metric multidi- 160). Lüneburg, Germany: Leuphana University.
mensional scaling. Working paper. doi: 10.13140/ Izmailov, C. A.; Sokolov, E. N., & Korshunova, S. G.
RG.2.2.34864.56325 (2005). Multidimensional scaling of schematically
Emelianova, S. A., & Gusev, A. N. (2018). Self-control represented faces based on dissimilarity estimates
determines observer’s strategies in loudness dis- and evoked potentials of differences amplitudes.
crimination. In F. Muller, L. Ludwigs & M. Kupper Madrid: Universidad Complutense de Madrid.
(Eds.), Fechner Day, 2018. Proceedings of the 34th Izmailov, C. A., Polaynsky, V. B., & Chernorizov, A. M.
annual meeting of the international society for (2015). Human-model-neuron: spherical model of
Psychophysics (pp. 198–202). Lüneburg, Ger- signal discrimination in visual system. In C. Forsythe,
many: Leuphana University. M. Zotov, G. Radvansky & L. Tsvetkova (Eds.), Rus-
Falikman, M. V., & Asmolov, A. G (2017). Cognitive sian cognitive neuroscience: Historical and cultural
psychology of activity: Attention as a constructive context (pp. 181–210). New York: Create Space.
process. Revue Internationale du CRIRES, 4 (1), Macmillan, N. A., & Creelman, C. D. (2005). Detec-
54–62. tion theory: A user’s guide. Hillsdale, NJ: Erlbaum.
Fechner, G. T. (1966). Elements of Psychophysics. Mikhalevskaya, M. B., & Zabrodin, Yu. M. (2022).
N.Y: Holt, Rineart and Winston. What Objective Sensometrics can do for Psycho-
Fokin, S. I. (2021). Specifics of the neuron action physics // Natural Systems of Mind, 2(2),
potential frequency dependence on the intensity 110–119.
of the excitatory influence. Natural Systems of Pardo-Vazquez, J.L., Juan, R. Castiñeiras-de Saa, J.R.,
Mind, 1, 71–93. Valente, M., Damião I., Costa, T., Vicente, M.I.,
Fritsche, M., Mostert, P., & de Lange, F. P. (2017). …& Renart, A. (2019). The mechanistic founda-
Opposite effects of recent history on perception tion of Weber’s Law. In N. du Bois et al. (Eds.).
and decision. Current Biology, 27, 590–595. Fechner Day 2019. Proceedings of the 35th
Gescheider, G. A. (1997). Psychophysics: The funda- Annual Meeting of the International Society for
mentals (3rd ed.). Hillsdale, NJ: Erlbaum. Psychophysics (p. 35). Antalya, Turkey: Akdeniz
Grzeczkowski, L., Clarke, A. M., Francis, G., Mast, F. University.
W. & Herzog, M. H. (2017). About individual differ- Pöppel, E., Yang, T., Zhou, B., & Bao, Y. (2018). Time
ences in vision. Vision Research, 141, 282–292. windows in sensory processing as neural basis for
Green, D. M., & Swets, J. A. (1974). Signal detection cognitive functions. In F. Muller L. Ludwigs & M.
theory and psychophysics (2nd ed.). NY: Wiley. Kupper (Eds.), Fechner Day, 2018. Proceedings of
Grondin, S. (2016). Psychology of perception. Berlin: the 34th Annual Meeting of the International
Springer-Verlag. Society for Psychophysics (pp. 21–22). Lüneburg,
Grondin, S., & Laflamme, V. (2015). Stevens’ law for Germany: Leuphana University.
time. Attention, Perception and Psychophysics, 77, Shendyapin, V. M. (2018). Mathematical definition of
1044–1051. evidence in Bayesian model of decision making
Gurler, D., Doyle, N., Walker, E., Magnotti, J., & and confidence estimation in sensory tasks. In F.
Beauchamp, M. (2015). A link between individual Muller, L. Ludwigs & M. Kupper (Eds.), Fechner
494 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Day, 2018. Proceedings of the 34th annual meet- Sokolov, E. N. (2013). The psychophysiology of con-
ing of the International Society for Psychophysics sciousness. Oxford, UK: Oxford University Press.
(pp. 106–110). Lüneburg, Germany: Leuphana Stevens, S.S. (1956). On the psychophysical law //
University. Psychological Review, 64(1), 153–181.
Shepard, R. (1981). Mnogomernoe shkalirovanie i Vickers, D., & Lee, M. D. (1998). Dynamic models of
nemetricheskie predstavlenia. Normativnye i simple judgments: I. Properties of a self-regulating
deskriptivnye modeli priniatia reshenii. In Materials accumulator model. Nonlinear Dynamics, Psychol-
of the Soviet-American Symposium (pp. 84–97). ogy and Life Sciences, 2, 169–194.
Moscow: Nauka. Volkova, N. N., & Gusev, A. N. (2017). Do cognitive
Schoenherr, J. R., & Petrusic, W. M. (2015). Scaling styles affect psychophysical tasks performance?
internal representations of confidence: Effects of European Proceedings of Social and Behavioral
range, interval and number of response catego- Sciences, 6, 445–452.
ries. In S. Grondin & V. Laflamme (Eds.), Fechner Wilson, K. E., Lowe, M. X., Ruppel, J., Pratt, J., &
Day 2015. Proceedings of 31st annual meeting of Ferber, S. (2016). The scope of no return: Open-
the International Society for Psychophysics ness predicts the spatial distribution of Inhibition
(pp. 36–37). Québec: Laval University. of Return. Attention, Perception, & Psychophysics,
Schoenherr, J. R. (2019). Metacognitive assessments 78(1), 209–217.
of performance: The psychometric properties of Wong, W. (2018). On the origins of the power func-
confidence scales and confidence models (2019). tion exponent. In F. Muller, L. Ludwigs & M.
In N. du Bois, S. Arndt, E. Varlık Özsoy, S. Bayrak- Kupper (Eds.), Fechner Day, 2018. Proceedings of
tar, E. Gülbetekin & M. Elliott (Eds.), Fechner Day the 34th annual meeting of the International Soci-
2019. Proceedings of the 35th Annual Meeting of ety for Psychophysics (p. 105). Lüneburg, Ger-
the International Society for Psychophysics many: Leuphana University.
(pp. 73–80). Antalya, Turkey: Akdeniz University. Yang, N., Adams, R. D., Han, J., & Waddington, G.
Skotnikova, I. G. (2008). Problemy subject-orientiro- (2018). Fechner’s three experimental methods: Do
vannyi psikhofiziki [Problems of subject–oriented the results correlate? In F. Muller, L. Ludwigs & M.
Psychophysics]. Moscow: Institute of Psychology, Kupper (Eds.), Fechner Day, 2018. Proceedings of
Russian Academy of Sciences. the 34th annual meeting of the International Soci-
Skotnikova, I. G. (2011). Subject–oriented approach ety for Psychophysics (pp. 161–166). Lüneburg,
in psychophysics. In D. Algom, D. Zakay, E. Chajut, Germany: Leuphana University.
S. Shaki, Y. Mama & V. Shakuf (Eds.), Fechner Day, Zabrodin, Yu. M., & Lebedev, A. N. (2022). On the
2011. Proceedings of the 27th annual meeting of possible relationships between psychophysical
the International Society for Psychophysics (pp. laws. Natural Systems of Mind, 2(2), 110-119. doi:
485–490). Ra’anana, Israel: International Society 10.38098/nsom.
for Psychophysics. Zabrodin, Yu. M., & Shpagonova, N. G. (1988).
Skotnikova, I. G., & Zhelankin, R. V. (2019). Slow- Vzaimosvyaz’ pokazateley razlichnykh psikhofizich-
worm lizards’ decision making in different visual eskikh metodov [Interrelation of various psycho-
stimuli discrimination. In N. Du Bois, S. Arndt, E. V. physical methods indices]. Psikhologicheskii Zhurnal,
Özsoy, S. Bayraktar, E. Gülbetekin & M. A. Elliott 9(5), 129–136.
(Eds.), Fechner Day 2019. Proceedings of the 35th Zabrodina, T. A., & Sorokin, A. S. (2019). Visual sig-
annual meeting of the International Society for nals discrimination under stress. Book of Abstracts:
Psychophysics (pp. 54–60). Antalya, Turkey: Inter- XVI European Congress of Psychology (ECP 2019)
national Society for Psychophysics. (p. 1833). Moscow: Russian Psychological Society.
30
Revolutions in “Wet”
Neurobiology
John Bickle and David J. Parker

INTRODUCTION mammals, seems best characterized as a transition


from Kuhn’s pre-paradigm state into paradigm-
This chapter details three historical cases of “revo- governed normal science (although features of a
lution” in one branch of neuroscience. Kuhnian “crisis” are present). Our second exam-
Neuroscience is the scientific study of the nervous ple, the discovery of long-term depression and
system, from biophysics to behavior, and is com- mechanisms of metaplasticity seems best char-
posed of numerous fields and disciplines. acterized as progress in normal science under
Neurobiology and neurophysiology, branches that the paradigm of Hebbian synaptic plasticity
study the biology and function of nervous sys- (Hebb, 1949). Our third example, the develop-
tems, are conducted in wetlabs, where “wet” is the ment of intracellular recording technologies and
colloquial expression for the laboratory materials basic neurobiological insights generated, seems
necessary to investigate biological processes better to fit Bickle’s (2016) alternative to Kuhn’s
experimentally, and the infrastructure needed to account, of a scientific revolution driven primar-
properly use and handle them. Our specific focus ily by development of new research tools. While
is on cellular and molecular investigations. These our focus here is on historical episodes, our goals
typically manipulate cells and molecules in model are prospective.2 Revolutions recognized as such
organisms and measure outcomes.1 by neurobiologists come in a variety of forms, so
Neurobiologists routinely refer to numerous recognizing and pursuing potential revolutions in
episodes in their field’s history as “revolutions” development is not a matter of looking for one set
or “paradigm shifts,” using terminology intro- of features (such as Kuhn’s “anomalies,” “crisis
duced by Thomas Kuhn (1962). Parker (2018) science,” and “emerging new paradigms”).
outlined examples of neuroscientific revolutions
using Kuhn’s resources, and pointed out that it
isn’t clear that neurobiologists actually follow
Kuhn’s account. In this chapter we examine some ADULT NEUROGENESIS
historical episodes that neurobiologists routinely
recognize as revolutionary which appear not to fit Historically, it has been accepted knowledge that
within Kuhn’s “paradigm-shift” model. Our first no new neurons were generated in the adult brain
example, the discovery of neurogenesis in adult (Rakic 1985). Neurogenesis was considered
496 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

limited to the pre- or very early-postnatal period. Pasko Rakic.” According to Specter (2001), Rakic
During the first half of the twentieth century there had claimed that, “the cells Kaplan had detected
were sporadic reports of adult mammalian neuro- may look like neurons in New Mexico, but they
genesis, but little unequivocal evidence of mitotic don’t in New Haven.” In the paper published
figures characteristic of dividing cells in neurons. a year later, cited almost 700 times (PubMed,
Absence of neurogenesis was also consistent with June 17, 2021), Rakic (1985a) examined slices
the assumed fixity of the adult brain considered from adult rhesus monkey brains at various ages
necessary for stability of adult brain function using 3H-thymidine and claimed that neurogenesis
(Gross, 2000). only occurred in the prenatal or early postnatal
Evidence of adult mammalian neurogenesis first period. Rakic’s authority as one of the most influ-
appeared during the early 1960s. Joseph Altman ential neuroscientists in the world unfortunately
and colleagues used the then-new 3H-thymidine limited continuing work on adult neurogenesis
technique and autoradiography as a marker of and acceptance of data already reported (Gould,
DNA synthesis in adult rats and cats (Altman, 2007; Kaplan, 2001).
1962; Altman and Dias, 1965). 3H-thymidine is Acceptance of adult neurogenesis finally
incorporated into replicating DNA during the began with studies in songbirds in the 1980s by
“S phase” of cell division and thus labels new Fernando Nottebohm and colleagues (Nottebohm,
cells. But as 3H-thymidine labels any newly syn- 1996). They hypothesized that neurogenesis
thesized DNA (e.g., during DNA repair), cell divi- could underlie seasonal differences in the volume
sion must also be verified to confirm the presence of two mating song-production nuclei in male
of new cells (Nowakowski and Hayes, 2000). canaries, and using 3H-thymidine and ultras-
Altman found evidence of adult neurogenesis in tructural analyses showed neurogenesis in song-
the hippocampus dentate gyrus and olfactory bulb related circuits (Nottebohm, 1996). While this
(Altman, 2011). However, despite this evidence evidence for neurogenesis was readily accepted,
being reported in major scientific journals, neuro- the “no-new-neurons” view was maintained as
genesis was not generally accepted. For example, orthodoxy by the claim that this more primitive
in his textbook on developmental neurobiology neural system was not representative of mammals
Jacobson (1970) insisted that “there is no convinc- (Specter, 2001).
ing evidence of neuron production in the brains of By the mid-1990s the thymidine analogue
adult mammals.” Subsequently, there was no men- 5-bromo-3-deoxyuridine (bromodeoxyuridine,
tion of Altman’s work in Purves and Lichtman’s BrdU) was being used to label dividing cells to
(1985) textbook, while Kandel et al. (1991) stated investigate adult neurogenesis (Gould, 2007).
dogmatically that “Neurogenesis ceases early in Immunocytochemical detection of BrdU labeling
the development of the mammalian brain.” was faster and simpler to use than 3H-thymidine
Gross (2000) claimed that Altman’s evidence autoradiography (Altman, 2011). In addition, by
for adult neurogenesis was ignored because of the early 1990s antibodies that labeled neuron and
his junior status. Altman (2011) denied this as glial cell-specific proteins were available. The use
he was among the top 1000 most cited neurosci- of BrdU and cell markers was quickly adopted
entists between 1965 and 1978 (Garfield, 1981). into rodent and primate studies (Gould, 2007;
However, Altman eventually shifted his research Specter, 2001). Several labs replicated Altman’s
focus from neurogenesis to the embryologi- and Kaplan’s original data, showing experience-
cal development of the CNS in rats and humans dependent adult neurogenesis in the olfactory bulb,
(Altman, 2011). dentate gyrus, and neocortex of rats and primates.
In the late 1970s, Michael Kaplan also used Gage and his colleagues also found evidence of
3
H-thymidine labeling combined with electron adult neurogenesis in the hippocampus in post-
microscopy (Altman had used light microscopy) mortem brains of human cancer patients injected
and found evidence of neurogenesis in the adult with BrdU before death to visualize tumor growth
rat dentate gyrus and olfactory bulb, supporting (Eriksson et al., 1998).
Altman’s findings. Kaplan also showed neurogen- This growing evidence for adult neurogen-
esis in rat visual cortex and the subventricular zone esis through the late 1990s continued to be met
of adult macaque monkeys, suggesting adult neu- with skepticism. Rakic (2002) wrote, “The dis-
rogenesis in a primate. Kaplan also experienced covery of neurogenesis in the adult human den-
negative reactions to his work which prevented him tate gyrus is exciting … [but an] exception to
from challenging the “no-new-neurons” establish- the rule of ‘no new neurons’ that still applies to
ment. He later wrote about “the political death” most of the brain.” Rakic appealed to historical
of his project into neurogenesis (Kaplan, 2001). authorities, Kölliker (1896), and Ramón y Cajal
Kaplan mentioned that “one of the most fervent (1928) who had argued for the irreplaceability
supporters of the dogma of no neurogenesis was of neurons.3 Focused on the stability argument
Revolutions in “Wet” Neurobiology 497

against adult neurogenesis, Rakic (1985a) wrote Kuhn’s conditions of a genuine paradigm shift
that, “The addition of new ‘naïve’ neurons into requires detailed historical study. Evidence that
existing circuits would interfere with the retention adult neurogenesis extended beyond pre- and
of acquired experience and learned behavior that early post-natal development periods, negated the
is evolutionarily the most significant advantage of concept of the brain having its full complement
all primate species including human.” However, of neurons around the time of birth (incommen-
the increasing weight of evidence for adult mam- surate with the “no-new-neurons” view that had
malian neurogenesis, and the realization that the informed neuroscience throughout the twentieth
adult brain is not immutable (Buonomano and century). Indeed, Nowakowski and Hayes (2000)
Merzenich, 1998), eventually led to the accept- pointed out that evidence of adult neurogenesis
ance of adult neurogenesis by the early 2000s would require “re-evaluation of virtually all cur-
(Gould 2007). rent conceptual bases for understanding how
A recent study failed to find evidence of adult neuronal circuitries in neocortex develop and are
hippocampal neurogenesis in humans (Sorrells modified,” suggesting that they viewed neurogen-
et al., 2018), conflicting with the findings of esis as a potential Kuhnian revolution. Regarding
Boldrini et al. (2018). Despite having found evi- its broader impact, adult neurogenesis has been
dence for neurogenesis in the adult primate dentate suggested as being involved in memory formation
gyrus (Kornack and Rakic, 1999), Rakic’s ready (Kempermann 2008), potential neurological repair
acceptance of the negative findings by Sorrell et al. strategies (Grade and Götz, 2017), and as a new
(2018) illustrates how evidence can be evaluated target for psychopharmacology (Whalley, 2006).
differently by different investigators (Shen, 2018). Hence, the case for a Kuhnian paradigm shift
Nottebohm, suspecting a psychological factor at account of adult neurogenesis seems strong.
work, commented that, “I think Pasko Rakic single- However, this interpretation faces a difficult
handedly held the field of neurogenesis back by question. Which paradigm did adult neurogenesis
at least a decade” (Specter, 2001). Subsequently, replace? A “no new neurons” paradigm doesn’t
Altman (2011) remarked that, “Open criticism of a seem correct. For Kuhn a paradigm directs the
scientific claim that runs counter to an established scientific work conducted as “normal science,”
dogma is fully justified; indeed, such a claim must but absence of adult neurogenesis leaves no spe-
be subjected to a thorough scrutiny before the cific normal science to pursue and thus doesn’t
established paradigm is prematurely abandoned.” provide a paradigm to be replaced. Instead,
However, Gage, mentioned above as an early pro- acceptance of adult neurogenesis constitutes a
ponent of human adult neurogenesis, insisted that new paradigm emerging from a pre-paradigm
Sorrells et al. (2018) were “not really measuring stage, generating normal science research ques-
neurogenesis… Neurogenesis is a process, not an tions (Kuhnian “puzzles”) now needing answers.
event. They just took dead tissue and looked at it How are new neurons made? What determines the
at that moment in time” (Shen 2018; cf. Costandi, rate of neurogenesis? What is the purpose of adult
2012). In discussing the different methodological neurogenesis? How do new neurons integrate into
approaches of Sorrells et al. (2018), and Bolderini existing circuits without disturbing ongoing func-
et al. (2018), Kamperman et al. (2018) concluded tions? How can adult neurogenesis be harnessed
that, “there is currently no reason to abandon the clinically for regeneration and repair?
idea that adult-generated neurons make important For example, striatal neurogenesis is far greater
functional contributions to neural plasticity and in humans than in animal models, suggesting a
cognition across the human lifespan.” potential therapeutic application in striatal motor
Numerous features of this episode have a disorders (e.g., Parkinson’s and Huntington’s dis-
Kuhnian ring. The tone and content of debates ease). Changes in the striatum during evolution
about neurogenesis read like Kuhnian crisis sci- also suggests a role in human cognitive function.
ence. Appeals to authority, selective citing of evi- Degenerative diseases like Alzheimer’s disease
dence, attacks on individual scientists, and various exhibit reduced neurogenesis in the adult hip-
“sociological and psychological factors” appear to pocampus that may contribute to depression and
have dominated the debate about adult neurogen- anxiety often seen during the presymptomatic
esis. Adult neurogenesis has directly been referred stage (Bergmann et al., 2015). The utility of neu-
to as a “paradigm shift” by prominent neurosci- rogenesis in the pathological brain environment
entists. Charles Gross commented that, “we are will be questions for normal science conducted
now in the midst of a paradigm shift” (Kaplan, under this first paradigm.
2001), while Eric Kandel considered Fernando To understand a neural circuit requires iden-
Nottebohm’s work as having “led to one of the tifying neurons involved, their connections, and
great paradigm shifts of modern biology” (Specter, their properties. If neurons are constantly being
2001). But determining whether an episode meets added, then conventional wisdom is problematic.
498 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Plasticity has been an acknowledged challenge response), duration (short- or long-term) and
for this general understanding, but plasticity oper- mechanisms that provide flexibility to anatomi-
ates within certain bounds on existing cells and cally fixed circuits. But with this obvious utility
is an extension of conventional circuit analyses comes the need of ensuring stability, allowing
(Selverston, 1980). Having to consider circuit- changes to occur without disrupting ongoing or
ries that add new components is a different issue. established functions (Abraham and Robins,
So perhaps adult mammalian neurogenesis can 2005).5 Failure to satisfy this demand in artificial
be seen as an example of a Kuhnian revolution systems can lead to “catastrophic interference”
for some fields (e.g., hippocampal research or (French 1999), where all stored information is lost
research into neural repair), while marking a tran- when the number of stored patterns exceeds a
sition from pre-paradigmatic science to a normal critical threshold (∼0.14N, where N is the number
science-generating paradigm for neuroscience of network components; Srivastava et al., 2014).
more broadly.4 Long-term potentiation (LTP) has dominated
Clearly any attempt to harness neurogenesis neuroscientific research on synaptic plasticity
will require understanding how and why it occurs. for nearly five decades, not least because of its
This will require analyses of behavioral effects claimed link to learning and memory (Bliss et al.,
that may influence the natural rate of neurogen- 2018; Morris, 2003; Silva et al., 2014; cf. Jeffrey,
esis and the integration of neurons into existing 1997; Keith and Rudy, 1990; McEachern and
circuits, as well as ways it can be manipulated Shaw, 1999; Parker, 2019; Queenan et al., 2017).
in animal models of pathology and evidence that LTP relates to Hebb’s rule (Hebb 1949), although
neurons generated by these manipulations in path- the rule was suggested before Hebb (Berlucci and
ological conditions have integrated into circuits in Buchtel, 2008). Hebb’s rule holds that correlated
a functionally-relevant way. For example, in some firing of pre- and postsynaptic neurons strength-
studies learning can enhance neurogenesis, but in ens the “weight” of the connection between them.
other contexts it can inhibit it, so the assumption Hebb’s rule is not universally neurobiologically
that neurogenesis is necessarily beneficial is over- true; for example, in the cerebellum predicted
simplified. These analyses will require multiple Hebbian plasticity (Marr, 1969) turned out the
approaches that include molecular cell biological be anti-Hebbian (Feldman, 2012). But Hebbian
approaches of cell phenotypes and differentiation, effects have been shown at many synapses across
electrophysiological or other ways of monitoring the CNS. So much wetlab research has focused
or manipulating function (including imaging to on investigating Hebbian synaptic plasticity
determine the activity of cells and optogenetics to that it seems appropriate to call it a paradigm of
manipulate their function). Clearly, there needs to late-twentieth-century neurobiology.
be a move away from qualitative analyses show- A well-known shortcoming of Hebbian plastic-
ing that neurogenesis occurs to more quantitative ity is that it has no mechanism to prevent a positive
analyses that relate neurogenesis to function and feedback effect in which potentiated connections
behavior. This will also require a reconsideration could exceed the plasticity threshold and continu-
of traditional views. Understanding of neurogen- ally increase in strength, a synaptic version of the
esis in the hippocampus and striatum will depend “Matthew effect” that obviously fails to address
on our knowledge of the structure and function of the stability-plasticity dilemma. This means that
these regions. Evidence of neurogenesis necessar- Hebbian synaptic mechanisms can only be one
ily forces revision of our previous understanding part of any putative storage mechanism, as syn-
of these brain areas, and their broader functions apses that follow a Hebbian rule have potential to
(Abrous and Wojtowicz 2015). grow in strength without limit (assuming graded
rather than binary changes; Petersen et al., 1998;
cf. Enoki et al., 2009), and thus erase any salient
changes at synapses.
Although these concerns came to be focused
HEBBIAN SYNAPTIC PLASTICITY on LTP in the hippocampus and its putative role
BEYOND LTP in memory (Bear, 1995), they were initially raised
in relation to Hebbian changes in development
Synaptic plasticity is a ubiquitous feature of all of the visual cortex (Bear et al., 1997). Von der
nervous systems. Under some conditions the Malsburg (1973) proposed rate-based network
strength or efficiency of transmission at all (chem- models of development of the visual cortex that
ical) synapses appears to be modifiable by neu- included synaptic plasticity using Hebbian synap-
ronal activity. Synaptic plasticity covers a tic potentiation and competition between cells to
multitude of effects that differ in terms of direc- account for the formation of orientation selectivity
tion (potentiation or depression of postsynaptic and ocular dominance maps. He hypothesized that
Revolutions in “Wet” Neurobiology 499

potentiation was proportional to the product of potentiated, and when it did not exceed θm the syn-
presynaptic and postsynaptic activity and coupled apse was depressed. They added the novel sugges-
it with a normalization of the total synaptic weight tion that θm was also modifiable: when a synapse
to a cell so that the total synaptic input remained is potentiated θm shifts to the right, and when it
constant. This generated stability by preventing is depressed θm shifts to the left, and thus poten-
excessive growth of the excitatory input, and spec- tial positive feedback cycles of potentiation and
ificity as cells would become insensitive to stimuli depression are prevented. This threshold modifi-
they had not been trained on. Although his pro- cation is akin to a homeostatic effect that keeps
posed mechanism was Hebbian, von der Malsberg synaptic strengths within certain bounds, using a
(1973) did not think that it related to then recently time-dependent average of the postsynaptic activ-
identified hippocampal LTP, because the reported ity to determine the homeostatic sliding thresh-
duration of LTP (Bliss and Gardner-Medwin, old that in turn influences subsequent synaptic
1973) was insufficient for long-term changes asso- modification.
ciated with visual cortex development. At first glance, the BCM model may not seem
Stent (1973) addressed the plasticity-stability to be a major change to views of synaptic plastic-
issue by predicting the need for bi-directional syn- ity held over the last four decades, but some con-
aptic plasticity, both potentiation and depression. siderations reveal its significant implications and
For the latter, he hypothesized an anti-Hebbian impacts. An obvious influence is that it inspired
process such that, “when a presynaptic neuron the search for LTD and bidirectional synaptic
repeatedly fails to excite a postsynaptic cell, meta- plasticity (Bear, 1995; Dudek and Bear, 1992).
bolic change takes place in one or both cells so Bienenstock et al. (1982) stated that “in order to
that the presynaptic neuron is less effective in acti- actually use Hebb’s principle, one must state con-
vating the postsynaptic cell.” He suggested that ditions for synaptic decrease as specific as those
this feature could account for the specificity of for synaptic increase: if synapses are allowed only
connections formed during development by spa- to increase, all synapses will eventually saturate;
tial competition, an increase in the strength of a no information will be stored and no selectivity
connection to a region resulting in the weakening will develop.” Bear (1995) claimed that the BCM
of other connections. Stent’s influence on theo- model was the inspiration for his experimental
retical and computational modelling over the fol- search for LTD, in the hippocampus (Dudek and
lowing decade was extensive (Bienenstock et al., Bear, 1992), and in the visual cortex (Kirkwood
1982; Cooper et al., 1979; Sejnowski, 1977) but et al., 1996). Evidence quickly emerged that the
seemed to have been forgotten in later reviews of same basic mechanisms involved in LTP were
long-term depression (LTD; Bear, 1995; Bear and also involved in LTD, with the rate of calcium
Malenka, 1994). Following Stent’s (1973) work, ion (Ca2+) influx through activated N-methyl-
Sejnowski (1977) asserted that, “unless some D-aspartate receptors (NMDARs) being one
means exist for weakening the plastic synapse, key determinant of which effect occurred. The
continual random coincidences inexorably push higher rate of postsynaptic Ca2+ influx generating
it to maximum strength.” Sejnowski suggested LTP activates calmodulin kinase II (CaMKII) to
that strengthening would occur through Hebbian increase the number of excitatory AMPA recep-
long-term facilitation when there is correlation tors in the postsynaptic density, and the lower
between pre- and postsynaptic neuron activity, rate of Ca2+ influx generates LTD by activating
but that depression would occur when there is no postsynaptic phosphatases to internalize AMPA
correlated firing of neurons. receptors (Purves et al., 2018). Identifying these
Cooper et al. (1979) also followed Stent in phosphatases and other substrates remains an area
suggesting a plasticity threshold above which of active research.
inputs potentiate and below which they depress. A second influential feature of the BCM
This gives some selectivity as neurons will only model is that it highlights that a plasticity mech-
respond to potentiated inputs, but as the thresh- anism cannot act in isolation; to preserve ongo-
old was fixed there would still be a potential for ing functions a plastic change must be integrated
runaway cycles of potentiation and depression. A and coordinated with other changes. While the
fixed plasticity threshold could also not account sliding plasticity threshold has been the main
for monocular or binocular deprivation effects on influence of the BCM model, Bienenstock et al.
visual cortex development (Bear et al., 1987). To (1982) highlighted other aspects that influenced
account quantitatively for selective increases and subsequent research. Exposure to a single input
decreases in inputs in the visual cortex, the BCM (e.g., a vertical line) can lead to a visual cortex
(Bienenstock et al., 1982) model suggested that neuron responding preferentially to that input,
when an input evoked a postsynaptic response but it will be less sharply tuned to the input than
that exceeded a threshold θm the synapse was if all possible orientations had been presented.
500 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

This is because temporal competition between argument it is obviously better if we can explain
incoming patterns and the activation of inhibitory why different reported effects occurred. If syn-
synapses is required for maximum stimulus selec- apses in different studies started out in different
tivity (Bienenstock et al., 1982). The BCM model functional states depending on prior activities,
thus moved investigations of synaptic plasticity metaplasticity suggests that these difference could
beyond the traditional focus on single effects on produce different functional outcomes under the
single cells by highlighting the distributed nature same experimental conditions (Parker, 2015). This
of synaptic modification: the need to sample mul- feature might even account for the still unresolved
tiple inputs, need for inhibition, as well as need presynaptic versus postsynaptic mechanisms of
to modify plasticity thresholds (Bear et al., 1997). LTP debate, as under certain conditions a synapse
A third influence of the BCM model has may be biased towards a pre- or a postsynaptic
been on attempts to provide a common frame- plasticity mechanism (Edwards, 1995). In terms
work for synaptic plasticity. Plasticity phenom- of translation of basic neurobiological results,
ena are diverse, with differences in the duration an appreciation of context-dependent effects has
(various short and long-term effects), direc- major significance. Any intervention that targets
tion (potentiation or depression), and underly- synapses, whether an educational approach or a
ing mechanisms (e.g., diverse second messenger psychopharmacological approach to treat psy-
pathways involved in LTP and LTD; Sanes and chological disorders (with notoriously variable
Lichtman, 1999). The same effects can also be effects; Ninnemann, 2012) or improve function
evoked by different stimulus protocols. Thus, in in the normal brain (e.g., cognitive enhancement;
common with the neuronal code, long-term syn- Sahakian et al., 2015), will need to be tailored to
aptic plasticity rules typically follow either a rate specific contexts if we are to promote beneficial
code such as that used in conventional LTP studies effects (or more importantly eliminate deleteri-
where the direction of plasticity reflects frequency ous ones). This context specificity at the level of
of presynaptic action potentials; or a timing code synaptic plasticity is not typically considered in
where plasticity depends on relative timing of pre- the systematic application of drugs, but in light
synaptic and postsynaptic activity (spike timing- of the implication of metaplasticity/homeostatic
dependent plasticity, STDP; Caporale and Dan, plasticity motivated by the BMC model, perhaps
2008). Plurality of plasticity mechanisms presum- it should become so.
ably allow nervous systems to respond flexibly, Thus, the BCM model has had a revolutionary
but they seem to leave us adrift in a sea of effects impact on wetlab work on synaptic plasticity. But
and mechanisms. There is obvious utility in find- is it an exemplar instance of a scientific revolution
ing a unifying basis for these diverse effects. The involving a paradigm shift? The BCM model was
BCM model is proving useful in this search, by explicitly developed as, “a general mathematical
inspiring attempts to find links between STDP and framework” (Bienenstock et al. 1982), and “a new
BCM learning rules (Izhikevich and Desai, 2003; mathematical form for synaptic modification”
Keck et al., 2017; Pfister and Gerstner, 2006). whose “theoretical conclusions are consistent
Arguably the most influential aspect of the with experimental data.” Indeed, it seems to have
BCM model is that it highlights that synaptic guided many wetlab experimental advances on
changes are context-dependent. This idea has the nature and mechanisms of synaptic plasticity,
motivated the fields of metaplasticity and homeo- especially on LTD.
static plasticity, which have become major areas of From the perspective of neurobiology, a strict
research over the last three decades (Abraham and Kuhnian account of scientific revolutions is open
Richter-Levin, 2018). Numerous examples, from to criticism that goes beyond more familiar criti-
model organisms to the human cortex, have shown cisms from the physical sciences (e.g., Kuhn’s use
that the history of activity at a synapse, even of a few spectacular examples). Neuroscientists
when it doesn’t leave any apparent alteration, can seem to use the term “paradigm” with appar-
modify the magnitude and direction of subsequent ently agreed-upon meaning, and also frequently
plasticity. A key component in metaplasticity and the term “paradigm shift” (although perhaps too
homeostatic plasticity research has been the BCM readily). So knowingly or unknowingly, they are
model’s sliding potentiation threshold, and this referring to Kuhn. But neuroscientists’ use of the
has significant implications. First, a sliding thresh- term “revolutionary” seems to be the crux, because
old might account for the variability often reported it can be applied to many episodes in many ways
in plasticity experiments (for variability in LTP (e.g., for distinct pragmatic or communicative
experiments alone, see Edwards, 1995). A lack of purposes). Some episodes that neurobiologists
consistent effects across labs when the same phe- recognize as revolutionary in their field seem to be
nomenon is studied in the same way is confusing, developments within Kuhnian normal science, and
but instead of acquiescing in disagreement and this analysis seems to apply to the BCM model.
Revolutions in “Wet” Neurobiology 501

Considerations in the 1970s about Hebbian plas- of pulses had no effect when presented at 10 Hz
ticity in the developing visual cortex inspired and produced LTP when presented at 60 Hz. And
the BCM model. Hebbian plasticity introduced the LTD induced by this new stimulation proce-
an immediate problem, a Kuhnian “puzzle,” the dure was reversibly blocked by NMDAR antago-
need for depressive plasticity at potentiated syn- nists, suggesting that the same basic mechanisms
apses to prevent runaway cycles of potentiation. were at work in both LTP and LTD induction,
Mark Bear’s laboratory was prominent in con- namely postsynaptic Ca2+ influx through acti-
firming LTD as a neurobiological phenomenon vated NMDARs, only at different rates to initiate
and revealing its mechanisms, and they stressed different second messenger effects (cf. Bear and
the theoretical BCM model’s guidance for their Malenka, 1994). This too is normal science: the
groundbreaking experimental work. Dudek and novel use of conventional research tools to exam-
Bear (1993), who first reported homosynaptic LTD ine a feature of plasticity known to be necessary
in the hippocampus, began by describing the BCM under the Hebbian paradigm (Hebb, 1949).
model and its shifting modification threshold, and Many aspects of normal science associated with
reported that they had “tested this theoretical pre- metaplastic changes present challenges for future
diction in the Schaffer collateral-CA1 pathway in research. Metaplasticity has obvious implications
rat hippocampus slices.” Dudek and Bear (1993) for our understanding of learning and memory
compared their findings “with the theoretical form beyond traditional Hebbian views that see suprath-
of modification that inspired it.” There was no reshold stimuli acting as a switch that evokes a
anomaly, and no crisis ensued from recognizing learning-evoked change, to a focus on understand-
this limitation on strictly Hebbian plasticity, and in ing the state-dependent metaplastic influences
this episode a central component of the paradigm, involved in processing these inputs. This may, for
LTP, got incorporated as the complementary bi- example, account for the variability between stud-
directional process alongside LTD. These aspects ies that has dogged the LTP field and generated
all reflect Kuhnian normal rather than revolution- controversy over the mechanisms (e.g., presynap-
ary science. Kuhn emphasized the importance of tic or postsynaptic locus for maintenance) that led
normal science for scientific progress; it was in no to the complaint that those outside were “mock-
way a pejorative label for him. The BCM model is ing” the field (Nicoll 2017). State-dependency
an example in neurobiology that developments in that evokes these metaplastic changes could offer
normal science can have significant implications a solution (suggested early in the pre- versus post-
to our understanding of synaptic plasticity, and for synaptic debate; Edwards 1995). Understanding
basic and translational neuroscience those were these metaplastic phenomena will require know-
indeed revolutionary. ing the molecular and cellular changes that under-
Instead of new research tools qua physical lie state-dependent influences, which can be
devices driving the discovery of LTD, new elec- examined using conventional molecular and elec-
trophysiological stimulation procedures were trophysiological or imaging approaches in isolated
key. Stanton and Sejnowski (1989), who first tissue (e.g., hippocampal slices). Knowing why
reported associative LTD in hippocampus slices these effects occur will require a link to system-
(where LTP induced in some synapses produces level effects (e.g., circadian or other intrinsic
depression in others), achieved their results by mechanisms) and behavior (how activity or sen-
focusing on a “new stimulus variable,” the time sory inputs evoke changes). This will necessitate
interval between bursts, to deliver “in-phase” and approaches that allow us to link behavioral effects
“out-of-phase” test inputs to slices. A low fre- to molecular and cellular changes, which is now
quency test input that was negatively correlated the major question facing neuroscience.
with a high frequency conditioning input reli- While research into metaplasticity has been
ably generated LTD, as did activating presynap- ongoing now for three decades, its mechanisms of
tic terminals while the postsynaptic terminal was action still remain poorly understood. Given the
hyperpolarized, although replicating associative focus on translations based on claimed understand-
LTD across laboratories proved “experimentally ing of memory, either in the context of educational
elusive” (Bear and Malenka, 1994). The more approaches or through more direct pharmacologi-
reliable and reproducible experimental findings cal interventions (cognitive enhancement), insight
of Bear and colleagues also resulted from a new into state-dependent metaplastic factors that influ-
stimulation procedure, the use of several hundred ence these changes is essential to ensure that
stimuli delivered at low frequencies (1 to 3Hz). interventions are efficacious and safe. Misguided
The depressive effect of this novel conditioning educational policies have adversely impacted on
stimulus was specific to synapses receiving it generations of schoolchildren, but potential dan-
(hence “homosynaptic” LTD) and depended on gers of pharmacological interventions that claim
frequency of the stimulation, as the same number “enhancement” based on unknown mechanisms
502 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

of action are far greater, given the modest ben- technique that proves useful. A popular belief is
efits that result from these invasive approaches. that sharp glass micropipettes were introduced by
Remarkably, Bostrom and Sandberg (2009) have Ralph Gerard and his students Judith Graham and
claimed that cognitive enhancement could solve Gilbert Ling in the 1940s (Graham and Gerard,
societal problems by making people “smarter, 1946; Ling and Gerard, 1949). But micropipettes
wiser, or more creative,” and that “the societal had been used throughout the 1920s and 1930s,
benefits of effective cognitive enhancement may with technology introduced at the start of the
even turn out to be so large and unequivocal that twentieth century by Marshall Barber (Chambers,
it would be Pareto optimal to subsidize enhance- 1918; Bretag, 2017). Barber had improved cell
ment for the poor just as the state now subsidizes manipulation approaches used by nineteenth-cen-
education.” Taking breaks or a nap significantly tury embryologists and applied them to bacteriol-
improves cognitive performance (Sievertsen ogy, making micropipettes to isolate a bacterium
et al., 2016; Smith-Coggins et al., 2006), so might for injection into an animal. His techniques for
we investigate whether an organizational (i.e., microinjection and nuclear transfer remain in use
allowing such breaks or naps) rather than a phar- today (Korzh and Strahle, 2002). While the new
macological approach affords better cognitive micropipettes were not used originally for electro-
performance outcomes? physiology, several of the people responsible for
this further development and application learned
about them directly from Barber (Bretag, 2017).
Initial electrophysiological approaches using
Barber’s micropipettes began with the work of
INTRACELLULAR RECORDING Frederick Pratt in 1917 and Ida Henrietta Hyde in
1921 (Bretag, 2017). They used glass pipettes to
Intracellular recording has been a keystone of apply graded extracellular electrical stimulation
electrophysiology for decades. It allows measure- to frog muscle and to protozoa Vorticella, respec-
ment and control of the voltage or current across a tively. Pratt claimed that an even narrower capil-
cell membrane, typically using a glass micropi- lary pipette would enable application of chemicals
pette pulled to a fine tip and placed through (or for to a restricted area of a cell’s surface, a technique
patch clamping, sealed onto) the membrane. that later came to be developed as iontophoresis.
While extracellular electrodes can record action Initial attempts at measuring cellular potentials
potentials, they cannot easily monitor subthresh- began in the early 1920s. This required advances
old events and membrane properties. Few today in micropipette manufacture and also in associated
might judge intracellular recording as revolution- equipment. Tibor Péterfi developed a microman-
ary science, but it should be more widely appreci- ipulator to guide precise movements of micropi-
ated as one of the major technical advances in the pettes that was manufactured commercially by Carl
history of neuroscience (Bretag, 1983). In this Zeiss, and Robert Chambers developed a widely
section we will sketch the historical development used micromanipulator that was manufactured
of these techniques and make the case for this commercially by Leitz (Bretag, 2017). Electronic
extended episode counting as a revolution in recording and amplification also advanced at this
wetlab neuroscience. time (Gasser and Newcomer, 1921). The string
Electrophysiology arguably dates back to the galvanometer had been a staple recording device
end of the eighteenth century and Luigi Galvani’s since the early nineteenth century, but its iner-
demonstration of “animal electricity” in the frog tia severely limited recording of rapid signals. It
leg. Over the first half of the nineteenth century, was replaced in this period by technology based
animal electricity replaced the previous view of on the cathode ray oscillograph invented by Karl
animal spirits; constituting an incommensurate Ferdinand Braun, modified to give the sensitivity
paradigm shift characteristic of a genuine Kuhnian and speed needed to display small voltage signals
scientific revolution (Parker, 2018). For the next (Bretag, 2017). These advances were essential for
several decades nervous system activity was development of intracellular recordings, and on
investigated using extracellular recordings by, their own allowed new aspects of cellular activity
among others, Helmhotz, du Bois-Reymond and to be detected, including the Nobel prize winning
Bernstein. Animal electricity has been described work by Erlanger et al. (1924) on the composition
as “the single most important discovery in the of the peripheral nerve compound extracellular
exploration of nervous mechanisms” (Brazier, action potential.
1964). These developments triggered rapid advances.
The development of intracellular record- Florey (1966) claimed that by 1925 Péterfi was
ing techniques passed through several stages of using fine-tipped micropipette electrodes to
advances and false starts, as expected for any novel measure small membrane potentials in Amoeba.
Revolutions in “Wet” Neurobiology 503

Taylor and Whitaker (1926) described small and The rapid (sub-millisecond) membrane current-
variable membrane potentials in sea urchin eggs voltage properties that were responsible for the
and highlighted the need for better micropipettes. action potential still could not be measured using
In 1927 they developed what would become the the available techniques, which led to develop-
standard intracellular glass micropipette electrode ment of the voltage clamp (Cole, 1949; Marmont,
filled with saturated potassium chloride (KCl) 1949). Hodgkin and Huxley modified Cole’s and
to reduce the electrode resistance, connected to Marmont’s original design into a two-electrode
electrical circuits with nonpolarizable silver chlo- set-up, one to record voltage and one to supply
ride (Ag/AgCl2) junctions. Péterfi’s technique current via a feedback circuit to “clamp” the mem-
of hand-pulling glass micropipettes was subse- brane of the squid giant axon at the desired volt-
quently improved by Gicklhorn and by Umrath. age. Four publications that appeared in the Journal
The resulting sharp-tipped capillary glass micro- of Physiology in 1952, the first with Bernard Katz
pipettes allowed reliable intracellular potentials to as third author, detailed their painstaking wetlab
be first measured in plant cells in the late 1920s experimental work. Hodgkin and Huxley used
and early 1930s (cf. Bretag 2017). The first pub- their physiological data to develop their quan-
lished account of intracellular recordings from titative theory of the action potential based on
animal cells seems to have been by Hogg et al. voltage-dependent sodium and potassium ionic
(1934) from embryonic rat heart cells in tissue currents (Hodgkin and Huxley, 1952).
culture (Hoyle, 1983). All of these investigations took advantage of
After the plant cell studies, the introduction of the large diameter of the squid giant axon, and
another new research tool, the squid giant axon, thus were not generally applicable to all neurons.
initiated the next major advances in neurophysi- Smaller cells became amenable to intracellular
ology. It was identified as an axon, not a blood recordings with micropipette developments made
vessel, by Young (1936). Soon after, Kenneth in the lab of Ralph Gerard. Florey (1966) claimed
Cole and Howard Curtis switched from working that Gerard learned micropipette techniques after
on algae to the squid giant axon (Cole and Curtis, visiting Umrath in 1937 (Bretag, 2017), but Stuart
1939), and Alan Hodgkin switched from extra- and Brownstone (2016) claim that Umrath had lit-
cellular recordings from frog and crab periph- tle influence as Gerard didn’t cite the plant cell
eral nerve fibres to the squid giant axon (Huxley, research. In 1940 Gerard tasked his Ph.D. student
2002). The large diameter (up to 1000μm) of the Judith Graham with improving glass micropi-
axon meant that micropipettes were not needed, pettes.6 They ultimately achieved tip diameters
as intracellular glass cannulae of about 100μm of 2–5μm and were able to measure resting mem-
diameter could be inserted longitudinally down brane potentials in frog skeletal muscle (Graham
the axon without damage to the fiber (although and Gerard, 1946), although producing “obvi-
the resting potential was more depolarized than ous damage” (Ling and Gerard, 1949). Gilbert
that measured later with less invasive micropipette Ling next used a stable heat source to consist-
techniques; Brown, 2019). This, together with the ently obtain pipettes with tips of less than 0.5μm
availability of relatively drift-free DC amplifiers diameter, a key step in reducing cell damage and
with sufficiently high input impedances and fre- recording lower and more stable resting potentials.
quency responses, and cathode ray oscilloscopes, Nastuk and Hodgkin (1950) used Ling’s tech-
allowed analyses of the action potential and its nique for preparing micropipettes to record resting
propagation (Cole and Curtis, 1939; Huxley, potentials and action potentials from mammalian
2002). Importantly, these recordings showed that muscle fibres. The latter had not been accom-
that the action potential overshot zero millivolts. plished by Graham and Gerard (1946), or Ling
Earlier suggestions of this overshoot in embry- and Gerard (1949), as in addition to the need for
onic rat cardiac myocytes and frog skeletal muscle fine-tipped micropipettes, the task required an
fibres had been considered experimental artefacts amplifier input stage with an improved frequency
(Cole, 1968), the prevailing view being that dur- response. A micropipette behaves electrically as
ing an action potential the membrane potential a shunt capacitance with a series resistance that
decayed to zero due to a temporary breakdown slows the time constant of the recording system
of the membrane resistance that made the mem- and distorts rapid signals, resulting in smaller,
brane permeable to all ions. Work with these more prolonged action potentials. Nastuk and Hodgkin
sophisticated electronic devices and the squid reduced capacitance of the input stage of the
giant axon quickly showed that the overshooting recording system using a lead shield and, as Taylor
action potential reflected a selective increase in and Woodward (1927) had originally done, by
sodium permeability (Hodgkin and Katz, 1949), filling their micropipettes with 3M KCl to lower
contradicting earlier claims (also using intracellu- their resistance. Woodbury (1952) independently
lar recordings) by Curtis and Cole (1942). improved the recording of action potentials by
504 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

modifying the input stage to electronically com- and neuronal interactions, could be examined
pensate for these effects in his recording of action directly using advances in micropipette elec-
potentials from the frog sciatic nerve. trodes. These mechanisms are so fundamental to
Intracellular recording from vertebrate CNS our knowledge of the nervous system that they
neurons was first achieved by John Eccles, are taught now to high school/pre-university stu-
Lawrence Brock, and John Coombs in New dents. Hartline et al. (1952) wrote that the intra-
Zealand (Brock et al., 1952), and in parallel by cellular recording technique “offers great promise
J. Walter Woodbury and Harry Patton in the US of resolving some of the difficulties that we have
(Stuart and Brownstone, 2011). Eccles recog- encountered in identifying electrical processes.”
nized the utility of the tapering tips and small Eyzaguirre and Kuffler (1955) highlighted how
tip diameters that the Ling and Gerard technique intracellular studies had overturned simplistic
generated for impaling small spinal neurons, and assumptions about the activity of sensory recep-
in 1951 with Brock and Coombs, he began intra- tors; and Hagiwara and Bullock (1957) empha-
cellular recordings in the cat spinal cord. Eccles sized that these techniques offered the possibility
was already experienced in extracellular record- to examine subthreshold events and evaluate ele-
ings of motor neuron responses to stimulation of mentary processes of neuronal integration. Data
dorsal and ventral roots. Intracellular recordings gathered using these new techniques removed
were not a major departure from these techniques, the need for extensive theoretical speculation and
but modifications to amplifiers, oscilloscopes, assumptions about aspects of neuronal activities
and stimulators were required. This work aimed that couldn’t be tested with extracellular elec-
to examine “the mode of origin of the so-called trodes, and led to explanations of effects that pre-
synaptic potentials” – both “excitatory synaptic viously had to be couched in uncertainty (e.g., the
action” and “the mode of action of an inhibitory graded relationship between nerve response and
impulse” (Brock et al., 1952). These recordings muscle response; Lucas 1909). In summarizing
were instrumental in the ultimate acceptance and the squid work up to that point, and with an eye to
development of chemical neurotransmission in the the future of cardiac muscle analyses, Cole (1957)
CNS (although Eccles was previously a staunch wrote that it offered a “completely new level of
advocate of the electrical transmission view; analysis and understanding.” This is familiar ter-
Parker, 2018). minology about scientific revolutions.
Improvements to achieve sub-μm tip micro- In the early 1950s, glass micropipettes began
pipettes and in amplification resulted in a rapid to be pulled by mechano-electrical pullers rather
accumulation of intracellular recordings data, than by hand (Alexander and Nastuk, 1953). These
including from frog muscle fibres (Ling and technological developments yielded micropipettes
Gerard, 1949; Nastuk and Hodgkin, 1950); frog with reliable characteristics and specific features
heart muscle; frog and mammalian nerve fibers (length, taper, and tip diameter). Subsequent
(Woodbury and Woodbury, 1950); mammalian developments in micropipettes included double-
heart muscle (Draper and Weidmann, 1951); frog barreled micropipettes to both record voltage and
dorsal root ganglion cells (Svaetichin, 1951); and inject current (Brock and McIntyre, 1953); the use
from the electric organ of the electric ray Torpedo of a single electrode for stimulation and record-
(Stuart and Brownstone, 2011). Intracellular ing by using a Wheatstone bridge circuit to allow
recordings were also made in invertebrates, includ- current to be passed through the recording micro-
ing photoreceptors in the Limulus eye; motor electrode (Araki and Otani, 1955); drug applica-
neurons controlling sound production in insects; tion delivered iontophoretically to specific sites
neurons innervating stomach muscles in cray- in microscopic quantities using single (Nastuk,
fish and the Aplysia abdominal ganglion (Stuart 1951) or multi-barreled micro-pipettes (Curtis
and Brownstone, 2011); sensory neurons of lob- and Eccles, 1958); development of new cell stains
ster and crayfish (Eyzaguirre and Kuffler, 1955); for better analysis of the morphology of recorded
and several types of neurons forming the lobster cells; development of the discontinuous cur-
cardiac ganglion circuit (Hagiwara and Bullock, rent clamp to allow current injection and voltage
1957). Paired recordings from the presynaptic and recording through the same electrode (Brennecke
postsynaptic neurons in the squid giant axon syn- and Lindemann, 1971); single electrode voltage
apse seem to have first been reported by Hagiwara clamps (Merickel and Gray, 1980); and the patch
and Tasaki (1958), and over subsequent decades clamp to record currents flowing through single
this technique provided insight into chemical syn- ionchannels by sealing the micropipette tip onto
aptic transmission and neuronal interactions in the surface of the membrane (Neher and Sakmann,
circuits. 1976; Hamill et al., 1981).7
Mechanisms, ranging from those of the action While some claim that intracellular recordings
potential to those of chemical neurotransmission cannot tell us much about complex behaviors and
Revolutions in “Wet” Neurobiology 505

cognition because these involve cell populations and disadvantages of both approaches, and for
rather than single cells (although single neuron recording from cells deep within tissue, intracel-
activity can reflect behaviorally significant effects lular recording remains the technique of choice.
in the mammalian brain; Houweling and Brecht, This will be especially important as we move from
2008), intracellular recording has been key to our tissue slices to study effects in more intact sys-
mechanistic understanding of the nervous system. tems (e.g., to examine metaplastic interactions).
The mechanisms of action potential signaling and Appealing to methods that are fashionable or
the final acceptance of chemical transmission are promoted over others can lead to the “Law of the
obvious examples. Beyond these, the identifica- Instrument,” where we use tools that may not be
tion of GABA-mediated inhibitory transmission appropriate for all problems and we start to look
(Otsuka et al., 1966), electrical synaptic transmis- for questions to address using the technique rather
sion at gap junctions (Furshpan and Potter 1959), than using techniques to address unresolved ques-
electrically mediated inhibition (Furshpan and tions. Clearly, different tools do different jobs and
Furukawa, 1962), and mixed electrical and chemi- should be selected on the basis of the question that
cal synapses (Martin and Pilar, 1963) were all dis- is being asked. At each stage in the century-long
covered from intracellular recordings. Phenomena development of intracellular recording devices
that go beyond single cells have been discov- and techniques, the limitations of tools and tech-
ered from the use of intracellular recordings, for niques and the need for adaptation has changed,
example the demonstrations of lateral inhibition alongside the questions that these evolving tools
in the horseshoe crab eye (Hartline et al., 1953); have been used to address.
presynaptic inhibition (Dudel and Kuffler, 1961); The revolution in wet neurobiology gener-
and key features of sensory and other processes, ated by intracellular recording capabilities fits
as well as various functional groupings between Bickle’s (2016) account of revolutions driven
neurons (Getting, 1989). Individual neurons and by tool development. This entire history, which
synapses form the alphabet of neuronal activities, we’ve only sketched in broad outlines, is a series
the building-blocks or motifs that characterize of events that match Bickle’s meta-scientifically
neuronal circuit activities underlying not just sen- derived model. At each stage existing technolo-
sory and motor processes, but also cognitive func- gies (or lack thereof, in the earlier stages) created
tions. Neuronal properties determine signals and motivating problems for new tools. Extracellular
are to cognitive representations what letters are recordings couldn’t measure subthreshold volt-
to phonemes and phonemes are to semantics. We ages effectively; glass micropipette electrodes
need to understand the syntax, the computations with large diameter tips could not measure from
performed on or by these building-blocks, in order smaller neurons or neuronal processes. Solving
to understand how representations are generated these problems and developing the new elec-
and transformed. Intracellular recordings are still trophysiological tools—microelectrodes, tissue
valuable in investigating these processes. preparations, micro-drivers, amplifiers, and elec-
Despite newer techniques like functional tronic recording equipment—required practical
imaging and optogenetics leading some to claim ingenuity and engineering solutions. For scien-
that “the photon will progressively replace the tists involved in the specific research endeavours
electron” (Scanziani and Hauser, 2009), electro- in which these tools developed, first-phase hook
physiology shows no signs of being replaced in experiments confirmed their experimental valid-
mainstream wetlab neurobiology, at present or in ity and utility. The publications reporting these
the foreseeable future. While imaging and optoge- experiments meet Bickle’s (2016) conditions.
netics have obvious advantages in the assaying and They were typically published in top journals
manipulation of populations of cells, a limitation of the field; they used the target experimental
with the low throughput of electrophysiology, the preparations of research interest (the squid giant
resolution of single cellular and synaptic effects axon, smaller myelinated neurons in mammalian
using electrophysiology remains unrivalled. CNS); and they addressed exactly the phenomena
Examining cellular excitability and the underlying of then-current research interests. Second-phase
ionic mechanisms is still best undertaken using hook experiments brought these tools to the atten-
electrophysiology. This is as true today as it was tion and use of a broader group of researchers,
for the historical analyses of the squid giant axon extending the scope of the tools’ impacts. And
by Hodgkin and Huxley (1952). Analyses of sub- the best-confirmed theories that we now possess
threshold events and inhibition imaging lags far across all of neuroscience—the mechanisms of the
behind electrophysiology in terms of temporal action potential, chemical neurotransmission, and
resolution and signal to noise ratio. While intra- activities in circuits of neurons—followed from
cellular recording has been replaced by whole cell the development and ingenious uses of these tools
recording to some extent, there are advantages on wetlab benches. ¡Viva la revolución!
506 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Notes Altman, J. (1962). Are new neurons formed in the


brains of adult mammals? Science, 135,
1  We adopt this focus because of our professional 1127–1128.
training and interests, Bickle in the philosophy of Altman, J. (2011). The discovery of adult mammalian
cellular and molecular neurobiology, Parker in neurogenesis. In T. Seki, K. Sawamoto, J. Parent &
neurophysiology. A. Alvarez-Buylla (Eds.), Neurogenesis in the adult
2  Regarding the prospective use of historical brain I: Neurobiology (pp. 3–46). Tokyo: Springer.
episodes to guide methodology in ongoing neu- Altman, J., & Das, G. D. (1965). Autoradiographic
roscience research – see Bickle (2022). and histological evidence of postnatal hippocam-
3  Ramó y Cajal (1928) had written, “In the adult pal neurogenesis in rats. Journal of Comparative
centers, the nerve paths are something fixed, Neurology, 124, 319–336.
ended, and immutable. Everything may die, noth- Araki, T., & Otani, T. (1955). Response of single
ing may be regenerated”; he also added, “It is for motoneurons to direct stimulation in toad’s spinal
the science of the future to change, if possible, cord. Journal of Neurophysiology, 18, 472–485.
this harsh decree.” Bear, M. F. (1995). Mechanism for a sliding synaptic
4  A case can also be made for adult mammalian modification threshold. Neuron, 15, 1–4.
neurogenesis being a new research tool-driven Bear, M. F., & Malenka, R. C. (1994). Synaptic plastic-
revolution in the sense of Bickle (2016). The tool ity: LTP and LTD. Current Opinion in Neurobiolol-
Altman and colleagues used, 3H-thymidine, was ogy, 4, 389–399.
introduced into genetic studies in 1957 and first Bear, M. F., Cooper, L. N., & Ebner, F. N. (1987). A
used in mammals in 1958. BrdU and immunocy- physiological basis for a theory of synapse modifi-
tochemistry tools were developing alongside their cation. Science, 237, 42–48.
uses in adult neurogenesis studies. Bergmann, O., Spalding, K. L., & Frisen, J. (2015).
5  This issue was one of the principal arguments Adult neurogenesis in humans. Cold Spring Harbor
against neurogenesis (see previous section). Perspectives in Biology, 7. doi: 10.1101/cshper-
6  Gerard’s interest in the nervous system led to spect.a018994
formation of the “Axonologist group” (Bretag Berlucchi, G., & Buchtel, H. (2008). Neuronal plastic-
2017). Hodgkin (1976) remarked that this group ity: Historical roots and evolution of meaning.
was dominant in physiology, and that they were Experimental Brain Research, 192, 307–319.
skeptical of the membrane theory and local cir- Bickle, J. (2016). Revolutions in neuroscience: tool
cuit theory of conduction originally outlined by development. Frontiers in Systems Neuroscience,
plant physiologists (the latter being work that 10. doi: 10.3389/fnsys.2016.00024
Hodgkin admired – see Bretag 2017). W.O. Fenn Bickle, J. (2022). Tinkering in the lab. In J. Bickle, C. F.
had stated that, “Axonologists were the impor- Craver, & A.-S. Barwich (Eds.), The tools of neuro-
tant people, and almost strutted through the cor- science experiment: Philosophical and scientific
ridors, being very conscious that they alone were perspectives (pp. 13–36). New York: Routledge.
in the frontiers of physiological discovery” (Mar- Bienenstock, E., Cooper, L., & Munro, P. (1982).
shall 1983). Theory for the development of neuron selectivity:
7  Bickle (2021) discusses some detailed episodes in Orientation specificity and binocular interaction in
the history of development of the patch clamp, visual cortex. Journal of Neuroscience, 2, 32–48.
as well as the metal microelectrode to withstand Bliss, T. V. P., & Gardner-Medwin, A. R. (1973). Long-
longer recording sessions. lasting potentiation of synaptic transmission in the
dentate area of the unanaesthetized rabbit follow-
ing stimulation of the perforant path. Journal of
Physiology, 232(2), 357–374.
Bliss, T. V. P., Collingridge, G. L., Morris, R. G. M., &
REFERENCES Reymann, K. G. (2018). Long-term potentiation in
the hippocampus: Discovery, mechanisms and
Abraham, W. C., & Richter-Levin, G. (2018), From function. Neuroforum, 24, A103–A120.
synaptic metaplasticity to behavioral metaplastic- Boldrini, M., Fulmore, C. A., Tartt, A. N., Simeon, L.
ity. Neurobiology of Learning and Memory: Behav- R., Pavlova, I., Poposka, V., … Mann, J. J. (2018).
ioral Metaplasticity, 154, 1–4. Human hippocampal neurogenesis persists
Abraham, W. C., & Robbins, A. (2005). Memory throughout aging. Cell Stem Cell, 22(4), 589–599.
retention: The synaptic stability versus plasticity Brazier, M. (1964). The electrical activity of the nerv-
dilemma. Trends in Neuroscience, 28(2), 73–78. ous system. Science, 146, 1423–1428.
Alexander, J. T., & W. L. Nastuk. (1953). An instru- Brennecke, R., & Lindemann, B. (1971). A chopped-
ment for the production of microelectrodes used current clamp for current injection and recording of
in electrophysiological studies. Review of Scientific membrane polarization with single electrodes of
Instruments, 24, 528–531. changing resistance. TI-TJ Life Science, 1, 53–58.
Revolutions in “Wet” Neurobiology 507

Bretag, A. (2017). The glass micropipette: A history Dudel, J., & Kuffler, S. W. (1961). Presynaptic inhibi-
of its inventors and users to 1950. Journal of tion at the crayfish neuromuscular junction.
General Physiology, 149, 417–430. Journal of Physiology, 155, 543–562.
Brock, L. G., & McIntyre, A. K. (1953). Responses of Edwards, F. (1995). LTP: A structural model to explain
motor neurons to stimulation by internal microe- the inconsistencies. Trends in Neuroscience, 18,
lectrodes. Proceedings of the University of Otago 250–255.
Medical School, 31, 19–20. Enoki, R., Hu, Y-l., Hamilton, D., & Fine, A. (2009).
Brock, L., Coombs, J., & Eccles, J. (1952). The record- Expression of long-term plasticity at individual
ing of potentials from motoneurones with an synapses in hippocampus is graded, bidirectional,
intracellular electrode. Journal of Physiology, 117, and mainly presynaptic: Optical quantal analysis.
431–460. Neuron, 62, 242–253.
Brown, A. (2019). The sodium hypothesis of action Eriksson, P. S., Perfilieva, E., Bjork-Eriksson, T., Alborn,
potential generation: A classic updated or an act A-M., Nordborg, C., Peterson, D. A., & Gage, F. H.
of electrophysiological sacrilege? Physiology News, (1998). Neurogenesis in the adult human
114, 32–35. hippocampus. Nature Medicine, 4, 1313–1317.
Buonomano, D., & Merzenich, M. (1998). Cortical Erlanger, J., & Gasser, H. S. (1924). The compound
plasticity: From synapses to maps. Annual Reviews nature of the action current of nerve as disclosed
in Neuroscience, 21, 149–186. by the cathode ray oscillograph. American Journal
Caporale, N., & Dan, Y. (2008). Spike timing-depend- of Physiology-Legacy Content, 70, 624–666.
ent plasticity: A Hebbian learning rule. Annual Eyzaguirre, C., & Kuffler, S. W. (1955). Processes of
Reviews in Neuroscience, 31, 25–46. excitation in the dendrites and in the soma of
Chambers, R. (1917). The microvivisection method. single isolated sensory nerve cells of the lobster
Biological Bulletin, 34, 121–136. and crayfish. Journal of General Physiology, 39,
Cole, K. S. (1968). Membranes, ions and impulses: A 87–119.
chapter of classical biophysics. Berkeley: University Feldman, D. (2012). The spike-timing dependence of
of California Press. plasticity. Neuron, 75, 556–571.
Cole, K. S. (1949). Dynamic electrical characteristics Florey, E. (1966). An introduction to general and
of the squid axon membrane. Archives des Sci- comparative animal physiology. Philadelphia:
ences Physiologiques, 3, 253–258 Saunders.
Cole, K. S. (1957). Beyond membrane potentials. French, R. (1999). Catastrophic forgetting in connec-
Annals of the New York Academy of Sciences, 65, tionist networks. Trends in Cognitive Science, 3,
658–662. 128–135.
Cole, K. S., & Curtis, H. J. (1939). Electric impedance Furshpan, E. J., & Potter, D. D. (1959). Transmission
of the squid giant axon during activity. Journal of at the giant motor synapses of the crayfish.
General Physiology, 22, 649–670. Journal of Physiology, 145, 289–325.
Cooper, L. N., Lieberman, F., & Oja, E. (1979). A Furshpan, E. J., & Furukawa, T. (1962). Intracellular
theory for the coordinate. acquisition and loss of and extracellular responses of the several regions
neuron specificity in visual cortex. Biological Cyber- of the mauthner cell of the goldfish. Journal of
netics, 33, 9–28. Neurophysiology, 25, 732–771.
Costandi, M. (2012). The mystery of the missing brain Gage, F. H. (2002). Neurogenesis in the adult brain.
cells. New Scientist. https://www.newscientist. Journal of Neuroscience, 22, 612–613.
com/article/mg21328521-600-the-mystery-of-the- Garfield, E. (1981). The 1,000 contemporary scien-
missing-brain-cells/ (retrieved November 19, 2021) tists most-cited 1965–1978. Part I. The basic list
Curtis, H., & Cole, K. (1942). Membrane resting and and introduction. Current Contents, 41, 5–14.
action potentials from the squid giant axon. Jour- Gasser, H. S., & Newcomer, H. S. (1921). Physiologi-
nal of Cellular and Comparative Physiology, 19, cal action currents in the phrenic nerve: An appli-
135–144. cation of the thermionic vacuum tube to nerve
Curtis, D. R., & Eccles, R. M. (1958). The excitation of physiology. American Journal of Physiology-Leg-
Renshaw cells by pharmacological agents applied acy Content, 57, 1–26.
electrophoretically. Journal of Physiology, 141, Getting, P. (1989). Emerging principles governing the
435–445. operation of neural networks. Annual Review of
Draper, M. H., & Weidmann, S. (1951). Cardiac rest- Neuroscience, 12, 185–204.
ing and action potentials recorded with an intra- Gould, E. (2007). How widespread is adult neurogen-
cellular electrode. Journal of Physiology, 115, esis in mammals? Nature Reviews: Neuroscience,
74–94. 8, 481–488.
Dudek, S. M., & Bear, M. F. (1993). Bidirectional long- Grade, S., & Gotz, M. (2017). Neuronal replacement
term modification of synaptic effectiveness in the therapy: Previous achievements and challenges
adult and immature hippocampus. Journal of ahead. NPJ Regenerative Medicine, 2(29).
Neuroscience, 13, 2910–2918. doi: 10.1038/s41536-017-0033-0
508 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Graham, J., & R. W. Gerard. (1946). Membrane Keck, T., Hubener, M., & Bonhoeffer, T. (2017). Inter-
potentials and excitation of impaled single muscle actions between synaptic homeostatic mecha-
fibers. Journal of Cellular and Comparative nisms: An attempt to reconcile BCM theory,
Physiology, 28, 99–117. synaptic scaling, and changing excitation/inhibi-
Gross, C. G. (2000). Neurogenesis in the adult brain: tion balance. Current Opinion in Neurobiology:
Death of a dogma. Nature Reviews: Neuroscience, Neurobiology of Learning and Plasticity, 43,
1, 67–73. 87–93.
Hagiwara, S., & Bullock, T. H. (1957). Intracellular Keith, J., & Rudy, J. (1990). Why NMDA-receptor-
potentials in pacemaker and integrative neurons dependent long-term potentiation may not be a
of the lobster cardiac ganglion. Journal of Cellular mechanism of learning and memory: Reappraisal
and Comparative Physiology, 50, 25–47. of the NMDA-receptor blockade strategy. Psycho-
Hagiwara, S., & Tasaki, I. (1958). A study on the biology, 18, 251–257.
mechanism of impulse transmission across the Kempermann, G., Gage, F. H., Aigner, L., Song, H.,
giant synapse of the squid. Journal of Physiology, Curtis, M. A., Thuret, S., … Frisén, J. (2018).
143, 114–137. Human adult neurogenesis: Evidence and remain-
Hamill, O. P., Marty, A., Neher, E., Sakmann, B., & ing questions. Cell Stem Cell, 23, 25–30.
Sigworth, F. J. (1981). Improved patch-clamp tech- Kirkwood, A., Rioult, M. G., & Bear, M. F. (1996).
niques for high-resolution current recording from Experience-dependent modification of synaptic
cells and cell-free membrane patches. Pflugers plasticity in visual cortex. Nature, 381, 526–528.
Archiv: European Journal of Physiology, 391, Kölliker, A. (1896). Handbuch der gewebelehre des
85–100. menschen. Leipzig, Germany: Englmann.
Hartline, H. K., Wagner, H. G., & Macnichol, E. F. Kornack, D. R., & Rakic, P. (1999). Continuation of
(1952). The peripheral origin of nervous activity in neurogenesis in the hippocampus of the adult
the visual system. Cold Spring Harbor Symposium macaque monkey. Proceedings of the National
on Quantitative Biology, 17, 125–141. Academy of Sciences of the United States of
Hebb, D. (1949). The organization of behavior: A America, 96, 5768–5773.
neuropsychological theory. New York: Wiley. Korzh, V., & Strahle, U. (2002). Marshall Barber and
Hodgkin, A. (1976). Chance and design in electro- the century of microinjection: From cloning of
physiology: An informal account of certain experi- bacteria to cloning of everything. Differentiation,
ments on nerve carried out between 1934 and 70, 221–226.
1952. Journal of Physiology, 263, 1–21. Kuhn, T. (1962). The structure of scientific revolu-
Hodgkin, A., & Huxley, A. (1952). A quantitative tions. Chicago: University of Chicago Press.
description of membrane current and its applica- Ling, G., & Gerard, R. W. (1949). The normal mem-
tion to conduction and excitation in nerve. Journal brane potential of frog sartorius fibers. Journal of
of Physiology, 117, 500–544. Cellular and Comparative Physiology, 34,
Houweling, A., & Brecht, M. (2008). Behavioural 383–396.
report of single neuron stimulation in somatosen- Lucas, K. (1909). On the refractory period of muscle
sory cortex. Nature, 451, 65–68. and nerve. Journal of Physiology, 39, 331–340.
Hoyle, G. (1983). Origins of intracellular microelec- Marmont, G. (1949). Studies on the axon membrane.
trodes. Trends in Neuroscience, 6, 163. doi: I: A new method. Journal of Cellular and Com-
10.1016/0166-2236(83)90077-2 parative Physiology, 34, 351–382.
Huxley, A. F. (2002). Hodgkin and the action poten- Marr, D. (1969). A theory of cerebellar cortex. Journal
tial 1935–1952. Journal of Physiology, 538(1)2. of Physiology, 202, 437–470.
doi: 10.1113/jphysiol.2001.014118 Marshall, L. (1983). The fecundity of aggregates: The
Izhikevich, E. M., & Desai, N. S. (2003). Relating STDP axonologists at Washington University,
to BCM. Neural Computation, 15, 1511–1523. 1922–1942. Perspectives in Biology and Medicine,
Jeffrey, K. (1997). LTP and spatial learning: Where to 26, 613–636.
next? Hippocampus, 7, 95–110. Martin, A.R., & Pilar, G. (1963). Dual mode of synap-
Jacobson, M. (1970). Developmental neurobiology. tic transmission in the avian ciliary ganglion. Jour-
New York: Holt, Rinehart & Winston. nal of Physiology, 168, 443–463.
Kandel, E. R., Schwartz, J. H., & Jessell, T. M. (1991). McEachern, J., & Shaw, C. (1999) The plasticity-
Principles of neural science (3rd ed.). East Norwalk, pathology continuum: Defining a role for the LTP
CT: Appleton & Lange. phenomenon. Journal of Neuroscience Research,
Kaplan, M. S. (2001). Environment complexity stimu- 58, 42–61.
lates visual cortex neurogenesis: Death of a dogma Merickel, M., & Gray, R. (1980). Investigation of
and a research career. Trends in Neuroscience, 24, burst generation by the electrically coupled cyber-
617–620. chron network in the snail Helisoma using a
Revolutions in “Wet” Neurobiology 509

single-electrode voltage clamp. Journal of Neuro- Queenan, B. N., Ryan, T. J., Gazzaniga, M., & Gallis-
biology, 11, 73–102. tel, C. R. (2017). On the research of time past: the
Morris, R. G. M. (2003). Long-term potentiation and hunt for the substrate of memory. Annals of the
memory. Philosophical Transactions of the Royal New York Academy of Sciences, 1396, 108–125.
Society of London Series B: Biological Sciences, Rakic, P. (1985a). Limits of neurogenesis in primates.
358, 643–647. Science, 227, 1054–1056.
Nastuk, W. L. (1951). Membrane potential changes Rakic, P. (1985b). DNA synthesis and cell division in
at a single end plate produced by acetylcholine. the adult primate brain. Annals of the New York
Federation Proceedings, 10, 96. Academy of Sciences, 457, 193–211.
Nastuk, W. L., & Hodgkin, A. L. (1950). The electrical Rakic, P. (1998). Young neurons for old brains?
activity of single muscle fibers. Journal of Cellular Nature Neuroscience, 1, 645–647.
and Comparative Physiology, 35, 39–73. Rakic, P. (2002). Neurogenesis in adult primate neo-
Neher, E., & Sakmann, B. (1976). Single-channel cur- cortex: An evaluation of the evidence. Nature
rents recorded from membrane of denervated frog Reviews: Neuroscience, 3, 65–71.
muscle fibres. Nature, 260, 799–802. Ramón y Cajal, S. (1928). Degeneration and regen-
Ninnemann, K. M. (2012). Variability in the efficacy eration of the nervous system. New York: Oxford
of psychopharmaceuticals: Contributions from University Press.
pharmacogenomics, ethnopsychopharmacology, Sahakian, B. J., Bruhl, A. B., Cook, J., Killikelly, C.,
and psychological and psychiatric anthropologies. Savulich, G., Piercy, T., … Jones, P. B. (2015). The
Culture, Medicine and Psychiatry, 36, 10–25. impact of neuroscience on society: Cognitive
Nottebohm, F. (1996). The King Solomon lectures in enhancement in neuropsychiatric disorders and in
neuroethology: A white canary on Mount healthy people. Philosophical Transactions of the
Acropolis. Journal of Comparative Physiology A, Royal Society B: Biological Sciences, 370. doi:
179, 149–156. 10.1098/rstb.2014.0214
Nowakowski, R. S., & Hayes, N. L. (2000). New neu- Sanes, J., & Lichtman, J. (1999). Can molecules
rons: Extraordinary evidence or extraordinary con- explain long-term potentiation? Nature Neurosci-
clusion? Science, 288, 771–771. ence, 2, 597–604.
Otsuka M., Iversen L. L., Hall Z. W., & Kravitz E. A. Scanziani, M., & Hausser, M. (2009). Electrophysiol-
(1966). Release of gamma-aminobutyric acid from ogy in the age of light. Nature, 461, 930–939.
inhibitory nerves of lobster. Proceedings of the Sejnowski, T. J. (1977). Statistical constraints on syn-
National Academy of Sciences of the United States aptic plasticity. Journal of Theoretical Biology, 69,
of America, 56, 1110–1115. 385–389.
Parker, D. (2015). Synaptic variability introduces Selverston, A. (1980). Are central pattern generators
state-dependent modulation of excitatory spinal understandable? Behavior and Brain Sciences, 3,
cord synapses. Neural Plasticity, 2015. doi: 535–571.
10.1155/2015/512156 Shen, H. (2018). Does the adult brain really grow
Parker D, (2018). Kuhnian revolutions in neurosci- new neurons? Scientific American. https://www.
ence: The role of tool development. Biology & scientificamerican.com/article/does-the-adult-
Philosophy 33. doi: 10.1007/s10539-018- brain-really-grow-new-neurons/ (retrieved Novem-
9628-0 ber 19, 2021)
Parker, D. (2019). Psychoneural reduction: A perspec- Silva, N. A., Sousa, N., Reis, R. L., & Salgado, A. J.
tive from neural circuits. Biology & Philosophy, 34. (2014). From basics to clinical: A comprehensive
doi: 10.1007/s10539-019-9697-8 review on spinal cord injury. Progress in Neurobiol-
Petersen, C. C., Malenka, R. C., Nicoll, R. A., & Hop- ogy, 114, 25–57.
field, J. J. (1998) All-or-none potentiation at CA3- Sorrells, S. F., Paredes, M. F., Cebrian-Silla, A., Sand-
CA1 synapses. Proceedings of the National oval, K., Qi, D., Kelley, K. W., … Alvarez-Buylla, A,
Academy of Sciences of the United States of (2018). Human hippocampal neurogenesis drops
America, 95, 4732–4737. sharply in children to undetectable levels in adults.
Pfister, J.-P., & Gerstner, W. (2006). Triplets of spikes Nature, 555, 377–381.
in a model of spike timing-dependent plasticity. Specter, M. (2001). Rethinking the brain. In The New
Journal of Neuroscience, 26, 9673–9682. Yorker, (July 23, pp. 42–53). New York: Advance
Purves, D., Augustine, G. J., Fitzpatrick, D., Hall, W. Publications. www.newyorker.com/magazine/
C., LaMantia, A.-S., Mooney, R. D., Platt, M. L., & 2001/07/23/rethinking-the-brain (retrieved
White, L. E. (2018). (Eds.), Neuroscience (6th ed.). November 19, 2021)
New York: Oxford University Press. Srivastava, V., Sampath, S., & Parker, D. (2014). Over-
Purves, D., & Lichtman, J. W. (1985). Principles of coming catastrophic interference in connectionist
neural development. Sunderland, MA: Sinauer networks using Gram-Schmidt Orthogonalization.
Associates Inc. PLoS One, 9(9). doi: 10.1371/journal.pone.0105619
510 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Stanton, P.K., & Sejnowski, T.J. (1989). Associative outside medium. Carnegie Institution of Washing-
long-term depression in the hippocampus ton Year Book, 25, 248–249.
induced by Hebbian covariance. Nature, 339, Taylor, C.V., & Whitaker, D. M. (1927). Potentiometric
215–218. determinations in the protoplasm and cell-sap of
Stent, G. S. (1973). A physiological mechanism for Nitella. Protoplasma, 3. doi: 10.1007/BF02057000
Hebb’s postulate of learning. Proceedings of the Udeigwe, L.C., Munro, P.W., and Ermentrout, G.B.
National Academy of Sciences of the United States (2017). Emergent dynamical properties of the
of America, 70, 997–1001. BCM learning rule. Journal of Mathematical Neu-
Stuart, D.G., & Brownstone, R.M. (2011). The begin- roscience, 7(2). doi: 10.1186/s13408-017-0044-6
ning of intracellular recording in spinal neurons: von der Malsburg, C, (1973), Self-organization of
Facts, reflections, and speculations. Brain Research, orientation sensitive cells in the striate cortex.
1409, 62–92. Kybernetik, 14, 85–100.
Svaetichin, G. (1951). Electrophysiological investiga- Whalley, K, (2006), Homing in on the target of anti-
tions of single ganglion cells. Acta Physiologica depressants. Nature Reviews: Neuroscience, 7,
Scandinavica, 24, 1–57. 504–505.
Svensson, E., Aspergis-Schoute, J., Burnstock, G., Woodbury, J.W. (1952). Direct membrane resting
Nusbaum, M., Parker, D., & Schioth, H. (2019). and action potentials from single myelinated nerve
General principles of neuronal co-transmission: fibers. Journal of Cellular and Comparative Physiol-
Insights from multiple model systems. Frontiers in ogy, 39, 323–340.
Neural Circuits, 21. doi: 10.3389/fncir.2018.00117 Woodbury, J. W., & Woodbury L. A. (1950). Mem-
Taylor, C. V., & Whitaker, D. M. (1926). A measurable brane resting and action potentials of excitable
potential difference between the cell interior and tissues. Federation Proceedings, 9, 139–140.
31
Human Brain Project and Beyond
J o r g e F. M e j í a s , K a t r i n A m u n t s , J a n G . B j a a l i e ,
Sander M. Bohté, Alain Destexhe, Lars Muckli,
Pier S. Paolucci, Martin J. Pearson,
and Cyriel M. A. Pennartz

INTRODUCTION A core element is that HBP’s scientific ques-


tions are addressed with the help of a digital
Funded as a European Flagship project under the research infrastructure – EBRAINS (European
program for Future and Emerging Technologies Brain Research Infrastructure), delivering a range
(FET), the Human Brain Project (HBP) to date of computing and data-processing technologies
has run from 2013 until 2023. It aims to achieve a that can be used by empirical and theoretical
better understanding of how the human brain is neuroscientists. This chapter focuses on cogni-
structured and how it functions. Also, it translates tive and systems-level research conducted within
fundamental knowledge about the brain into novel the HBP, and how this is supported by its digital
methods, technology, and medicine to treat brain research infrastructure. In addition, we highlight
disorders. Through a multidisciplinary approach, differences and commonalities with other large-
it addresses brain structure and function at multi- scale international brain initiatives, and how these
ple scales of organization, ranging from the relate to, and collaborate with, each other. The
molecular and cellular levels to the network and key rationale behind the HBP approach is that
systems-level, culminating in studies on complex advances in understanding brain systems’ func-
behavior, cognition, and consciousness. Not all of tioning and cognition are facilitated by combined
these can be addressed equally well by research theory-driven empirical research and computer
on human brains, as this is mostly restricted to modeling. This leads to predictions that can be
post-mortem examination, non-invasive brain tested in subsequent cycles of neural measure-
imaging, behavioral testing, and intracranial phys- ments, analysis, modeling, and theory. Throughout
iological recordings (e.g., from epileptic patients). this innovative methodology tackling cognition
Thus, complementary research on animal models and systems complexity, the use of computational
and in silico research on brain simulations is tools for data curation, analysis, atlasing, mod-
required. HBP supports these empirical and mod- eling, simulation, hardware implementations and
eling approaches with strong theoretical research, robotics lies is central. Hence the HBP approach
as well as ethical and philosophical reflection may be captured as “Computing the Brain.”
addressing the significance of the research for In this chapter we focus on a subset of HBP
society, medicine, and ultimately, the neural basis projects that cohere thematically and illustrate use
of the human mind. of EBRAINS’ computational resources. We begin
512 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

with studies into dynamic states characteristic of FAIR principles (to make data findable, accessi-
the various phases of wakefulness and sleep that ble, interoperable, and reusable); (ii) anatomical
the human brain passes through in the day-night atlases of the human and rodent brain, sup-
cycle. These studies illustrate multiscale research porting the mapping of electrophysiological
as the alternation between various sleep and awake recordings and imaging onto neural structures;
states, expressed at the level of single-neuron (iii) simulation engines for building brain and neu-
dynamics, local and system-wide networks, inter- ral-network models, to study both brain dynamics
linking with macroscopically manifested changes and cognitive processing; (iv) the neurorobotics
such as in behavior. Transition from deep sleep to platform for implementing computation-intensive
wakefulness is marked by a return of our percep- robotic simulations, (v) neuromorphic comput-
tual capacity: the ability to process sensory stimuli ing services for emulation of neuronal networks
originating outside the brain, and transform these in brain-like hardware; (vi) high-performance
sensory inputs into a consciously experienced sen- computing resources, including a European net-
sation of stimuli embedded in a spatiotemporal work of computers (FENIX) to support all of
context (Pennartz, 2015). Here we emphasize how these activities. Finally, we pay attention to other
empirical, modeling, and theoretical researchers large-scale brain initiatives, including the BRAIN
collaborate to underpin perception as a process initiative in the USA, the Brain/MINDS initiative
of actively constructing latent representations in in Japan, the Korea Brain Initiative (KBRI), the
corticothalamic brain systems, involving the con- China Brain Project, the Canadian Brain Research
textualization of individual sensory inputs and the Strategy (CBRS), the Australian Brain Alliance,
simulation of predictive cortical hierarchies. and others. We also review the efforts to coordi-
Next, we show how sensory signals originat- nate resources and goals of many brain initiatives
ing from multiple modalities, such as vision and via the International Brain Initiative (IB), and the
touch, can be combined to produce multisensory importance of international collaboration among
integration, and how this capacity can be utilized these initiatives to achieve worldwide progress in
for cognitive abilities such as episodic memory data standardization, ethical policies, and accessi-
and spatial navigation in space. Departing from bility of digital tools to move forward to a more
neurophysiological and behavioral findings, this globalized access to resources for brain research.
approach illustrates how robotics – via physical
implementation and in silico simulation – provides
a key instrument to test whether computer models
of multisensory prediction enhance behavioral
performance of artificial agents. With or without
SYSTEMS-LEVEL MODELS OF
robotics, computer models are becoming increas- BRAIN STATES
ingly sophisticated, not only in their resemblance
to brain physiology, but also in their representa- First, we illustrate HBP’s multiscale approach by
tional and cognitive capacities. The final neurosci- a brief overview of computational models of brain
entific topic we touch on concerns the neural basis states, such as wakefulness and slow-wave sleep.
of consciousness. On the one hand, we highlight Pioneering experiments (Massimini et al., 2005)
consciousness as a particular kind of brain state, have demonstrated that the same stimulus, deliv-
reaching back to the differentiation between sleep ered to the same brain in different states, can lead
and wakeful state dynamics. On the other hand, to very different responses. These experiments not
we suggest how the ‘contents’ aspect of con- only showed that the response to a given stimulus
sciousness can be addressed: the question of, how is “state-dependent,” but it was also found that in
conscious representational content can be realized the awake – but not sleeping brain, the response
in neural substrates, and why complex network spreads in the whole brain according to complex
operations can give rise to subjective experience propagation patterns. The complexity of the
at all. spread of this evoked response is thought to be a
Following up on the scientific questions HBP measure of the consciousness level of the
addresses in Cognitive and Systems Neuroscience, individual.
we summarize how services provided by the In parallel, progress in micro-electrode tech-
EBRAINS research infrastructure act to sup- niques, have enabled the recording of unit activity
port HBP research. Again, we do not provide an in the human brain with unprecedented precision.
in-depth overview, but emphasize the toolkits of Human multi-electrode recordings showed that
primary importance to the neuroscientific topics “regular-spiking” (RS) cells and “fast-spiking”
covered here. Briefly, we highlight the following (FS) cells can be discriminated (putative pyrami-
services: (i) storage, curation, and analysis of data dal cells and interneurons, respectively Dehghani
in neuroscientific and clinical domains, applying et al., 2016; Peyrache et al., 2012). The high
Human Brain Project and Beyond 513

density of the recordings allowed us to demon- the same stimulus was very different in the two
strate that many of the RS cells are excitatory, states. The model was able to capture this differ-
while many FS cells are inhibitory (Peyrache ence in responsiveness. It also captured the fine
et al., 2012). These data provided, for the first details of the timecourse of the response, which
time, the possibility of characterizing excitatory was the first time such a level of realism was
and inhibitory populations in humans, during obtained for a conductance-based model (di Volo
wake and sleep states, as well as during epilep- et al., 2019).
tic seizures (Dehghani et al., 2016). Such data are To scale-up the adaptive mean-field model to
invaluable for building computational models. simulate large-scale networks, we first studied
Motivated by these experiments, we next inves- the “mesoscale” dynamics of propagating waves
tigated models of spiking networks composed found in the primary visual cortex of the awake
of RS and FS cells. The simplest model display- monkey (Muller et al., 2014). These propagating
ing RS and FS cells is the Adaptive Exponential waves were found across large cortical distances
(AdEx) integrate-and-fire model (Brette and (several millimeters up to centimeters), and mod-
Gerstner, 2005). We studied AdEx networks of eling such phenomena at the cellular level would
RS and FS cells, and determined their firing require simulating millions of neurons. However,
characteristics based on human recordings from using mean-field models, one can simulate the
these cell types. Such networks, if endowed with visual cortex at the same resolution as imag-
conductance-based synaptic interactions and ing studies, where one “pixel” of the camera is
spike-frequency adaptation, can display two typi- equivalent to hundreds to thousands of neurons,
cal network states seen experimentally: asynchro- and create a network of mean-field units. This
nous-irregular (AI) states, in which all cells fire network was calibrated using propagating waves.
irregularly, as typically seen in wakefulness, and Because such waves can be quantified in spatial
Up/Down states, where the dynamics oscillates extent and in propagation speed, it was possible
between periods of AI-like activity (Up state) and to constrain the connectivity between the mean-
silent periods (Down state), as seen in slow-wave field units to reproduce the propagating waves as
sleep in humans (Goldman et al., 2019). measured experimentally (Zerlaut et al., 2018). It
To scale up these spiking network models to was found later that the same mean-field model
large brain areas, we have conceived population- was able to capture the interaction between two
level models using mean-field techniques. Here, propagating waves, and in particular their sub-
our motivation was to describe, as realistically linearity (Chemla et al., 2019). Thus, adaptive
as possible, the dynamics of RS-FS populations, mean-field models, if connected appropriately,
and obtain mean-field models representing one account for mesoscale phenomena in the awake
small brain area (e.g., a cortical column). A large monkey brain. Next, it is integrated at the whole-
number of mean-field units can then be con- brain scale.
nected according to the structure of large-scale The integration of mean-field models at the
networks, up to the whole brain (Goldman et al., whole human brain scale was done recently (and
2020, 2022). Because it is important that such is still in progress in the HBP). Using The Virtual
mean-field models capture the dynamics of spik- Brain simulator in EBRAINS, it is possible to
ing networks, we needed to use a formalism that reconstruct a large network of mean-field units,
allowed us to consider conductance-based synap- connected according to the human connectome,
tic interactions. We have used a Master Equation thus forming a whole-brain model (Sanz-Leon
formalism (El Boustani and Destexhe, 2009) with et al., 2015). The resolution of this network can
which conductance-based mean-field models are be adjusted from a few tens of nodes (where one
possible. Adapting this formalism to AdEx net- mean-field unit represents one brain area), up to
works (Zerlaut et al., 2018) and later including hundreds of thousands of nodes (one mean-field
spike-frequency adaptation (di Volo et al., 2019) unit representing the size of a cortical column).
yielded mean-field models that can reproduce the Recent results (Goldman et al., 2020, 2022) show
dynamics of AI states and Up/Down states as seen that integrating the adaptive mean-field model can
in the spiking networks. This “Adaptive mean- yield a whole-brain model reproducing previous
field” model was able to simulate, at the popula- observations (Massimini et al., 2005). Figure 31.1
tion level, the different states exhibited by AdEx illustrates the response obtained in two differ-
spiking networks. ent brain states, asynchronous and synchronized
An important test of the accuracy of the adap- slow-waves with Up/Down states. As in the exper-
tive mean-field was to simulate state-dependent iments, the stimulus evokes complex patterns
responsiveness. This was done by considering net- spreading across the whole brain, but only in the
works in different states, such that the response to asynchronous state.
514 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 31.1 Integration of mean-field models to yield whole-brain models of the human
brain.
A. Scheme of integration of mean-field units (nine squares), each representing a population
of neurons, and connected using excitatory and inhibitory connections.
B. Whole-brain model formed by mean-field units (left) connected according to the human
connectome (RS: regular spiking, FS: fast spiking, W: adaptation variable).
C. Whole-brain simulations of response to stimulus in visual cortex (arrow) during slow-wave
dynamics.
D. Same simulation when the simulated brain activity is made asynchronous (wake-like). In
this case, the firing-rate increase evoked by the stimulus is much larger than during slow
waves.
Source: Adapted from Goldman et al., 2020, 2022.

COMPUTATIONAL MODELS OF BRAIN Hopfield, 1982; Wilson and Cowan, 1972). The
DYNAMICS AND THEIR RELATIONSHIP extension to computational models describing the
dynamics of brain activity at larger spatial scales,
WITH COGNITION or even at the whole-brain level, occurred after
the popularization of neuroimaging techniques,
Besides their use to understand brain states, as researchers started to use structural connec-
large-scale (or systems-level) models may be tomes as a basis for computational models to
used to uncover perceptual and cognitive func- simulate human brain dynamics (Deco et al.,
tions in the brain. In the early years of computa- 2008, 2011; Friston et al., 2003). More recently,
tional modeling, the field had a strong focus on large initiatives such as the Human Brain Project,
‘microscopic’ models of small, homogeneous the Human Connectome Project and the Allen
neural populations (Amit and Brunel, 1997; Institute for Brain Science facilitated to extend
Human Brain Project and Beyond 515

the focus on large-scale modeling to other species mechanistic. In contrast, models of small neural
such as rodents (Foster et al., 2021; Gămănuţ circuits or local brain regions have traditionally
et al., 2018; Knox et al., 2018) and nonhuman provided a very useful way to test and explore
primates (Markov et al., 2013, 2014), and to neural mechanisms underlying cognition, includ-
include further biophysical details in the models. ing functions like perception, working mem-
With large amounts of curated data becoming ory and decision making (Compte et al., 2000;
available in the HBP (and importantly, following Meijer et al., 2020; Wang, 1999, 2002; Wong and
open access guidelines), researchers have started Wang, 2006). New modeling work combining the
to constrain their models with data more often, description of accurate large-scale neural dynam-
leading to a new generation of models which are ics with cognitive functionalities has only started
more accurate and provide descriptions which are to emerge. For example, data-constrained models
closer to the real brain. Computational models of large-scale cortical networks have been recently
constrained by data at different scales have been used to explain the emergence of working mem-
able to explain observed neural dynamics across ory-related patterns of persistent activity across
multiple spatial and temporal levels of descrip- the cortex (Mejías and Wang, 2022). Rather than
tion. For example, computational models of the a simple extension, this work predicted the exist-
macaque cortical network have been constrained ence of a gradient of ‘counterstream inhibition’
using tract-tracing connectivity data which con- along the cortical hierarchy, a prediction compat-
tains information about the cortical layers involved ible with predictive coding architectures.
in each long-range projection between area-to- More complex cognitive and perceptual func-
area pairs (Chaudhuri et al., 2015; Jaramillo et al., tions are still missing a large-scale modeling
2019; Joglekar et al., 2018; Mejías and Wang, description properly rooted in experimental data.
2022; Mejías et al., 2016). When such level of Recent work within the Human Brain Project is
biological detail is considered, the resulting model advancing in the exploration of such questions.
reproduces neural activity at the intra-laminar For example, the challenge of combining realistic
level (e.g., enhancement of gamma rhythms by neural dynamics with informative cognitive archi-
visual input), inter-laminar level (power-phase tectures in a common theoretical framework can be
coupling between gamma and alpha rhythms achieved by developing multiple models in parallel.
across layers), inter-areal level (gamma and alpha Each model may have a different balance between
rhythms mediating feedforward and feedback cognitive capacities and biophysical plausibil-
interactions), and large-scale level (emergence of ity, and keeping these models coherent with each
so-called functional hierarchies, or hierarchical other would ensure that they remain in a similar
relationships in the activity patterns of brain areas theoretical framework despite differences in imple-
(Mejías et al., 2016)). Similar data-constrained mentation. A promising example is a suite of neu-
models of macaque networks have shed light on robiologically realistic models of predictive coding,
previously unrecognized problems in our classi- for which models combining cognitive capabilities
cal understanding of large-scale communication with biophysically realistic aspects are currently in
(Chaudhuri et al., 2015; Joglekar et al., 2018) and development (Dora et al., 2021). We are construct-
pointed toward a proper balance between excita- ing, in parallel, predictive coding models based on
tion and inhibition as the key factor to achieve neurophysiologically detailed spiking networks (to
an optimal communication across distant brain reproduce biophysical signatures of predictive cod-
regions, for example during conscious perception. ing) and models based on more abstract networks
In other animal models, data-constrained models and rate dynamics (but with greater capacity for
of the mouse brain have been able to reproduce complex cognitive computations). Such models
and explain multiple neural signatures linked constitute a common ground to study further per-
to visual perception and processing, providing ceptual functionalities, such as multisensory inte-
insights in fundamental mechanisms of visual gration and conscious perception.
processing and brain communication, such as the
emergence of orientation selectivity and other vis-
ual computations (Billeh et al., 2020; Knox et al.,
2018; Lindeman et al., 2021).
There are, however, important milestones
PERCEPTION, CONTEXTUALIZATION, AND
which data-constrained models of large-scale ALIGNMENT IN HUMAN BRAIN MAPPING
brain networks still need to reach. Most notably,
large-scale models have been primarily focused Progress in large-scale modeling of cognition and
on neural dynamics and electrophysiological perception depends on its interaction with empiri-
signatures, with contributions to cognitive func- cal data acquisition in both human and animal
tions being descriptive and qualitative rather than models. This is illustrated next by the
516 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

contextualization of sensory processing. Human maps of neurons responding to the visual field.
cognition rests upon the association of our ongo- A series of interconnected visual areas have dif-
ing mental activity with our subjective perceptions ferent response criteria, for example, in higher
of sensory inputs. Large scale cortical circuitry cortical areas, receptive fields process a larger
reflects this synergy in densely interconnected space in the visual field (Wandell et al., 2007).
processing streams sending information to and When these higher areas project top-down to
from our sensory brain areas, and between dedi- earlier levels in the cortical hierarchy, the spread-
cated functional areas. In these streams, neuronal ing out of neuronal projections reaches a wider
mechanisms and computations must provide spa- field than the small, locally tuned receptive fields
tiotemporal context to the incoming stream of selective for feedforward sensory inputs. In these
sensory information, with precisely tuned levels “feedback receptive fields,” we can read out the
of conscious awareness, and while undergoing top-down contextual information. In instances of
shifts in neuromodulatory states. Increasing visual occlusion, there is no feedforward stimula-
empirical evidence from rodent recordings shows tion, offering insight into how the brain infers its
that pyramidal cortical neurons have two function- inputs using contextual guesses that influence vis-
ally separate computational compartments ual information processing (Muckli et al., 2015).
(Larkum, 2013). This knowledge motivates inves- We study the spatial specificity and information
tigations at larger spatial scales, using multiscale content of this top-down contextual information
biological data across humans, monkeys and flow in humans using functional brain imaging
rodents to understand the neuronal principles (Morgan et al., 2019; Revina et al., 2018), and
underlying this contextualization. Along this theo- its dynamics in animal models at different spatial
retical theme, advances in human brain imaging and temporal scales. We can align the cortical net-
technologies and the opportunity to constrain works of these vastly different sizes of brains in
neuronal data with models and machine learning humans, monkeys and rodents by aligning them
approaches is driving a more mechanistic under- to a shared functional coordinate system based on
standing of neuronal information processing a retinotopic grid system. We can then draw com-
underlying human cognitive function, at the same parisons between species on a functionally aligned
time delivering innovative testable hypotheses for reference system and establish the content of top-
systems neuroscience (Phillips, 2017). down communication, the layer-specific neuronal
At psychological levels of description, count- compartments and implementation of contextual
less examples can be found of how our brains use computations, all of which we can then extract to
context to fulfill behavioural goals (Bar, 2004). inspire neuronal network models.
Take, in visual perception for example, the simple
act of grasping a cup (Figure 31.2). Your hand cov-
ers part of the cup, but we can readily understand
this image; we see discontinued contours of the
cup as part of the larger context, and we under-
SPIKING NEURAL NETWORK MODELS OF
stand that shadows cast neither belong to the hand PERCEPTION AND PREDICTION
nor to the mug. Our brains make hundreds of such
inferences every day, and now we are beginning Thus far we have mainly surveyed large-scale
to understand the neuronal operations that allow dynamic models of the brain, but these were
for these contextual computations (Phillips et al., specified at a mean-field level, leaving open the
2015). A compelling neurobiological description question how models may be extended to spiking
of contextual processing that is emerging entails neural networks that perform perceptual and cog-
two integration sites in pyramidal neurons, one nitive tasks. Deep artificial neural networks have
at the soma, processing driving inputs, and one made great strides both in AI and as models of
in the apical tuft dendrites computing contextual visual perception in the brain (Güçlü and Gerven,
modulatory signals. These two sites are computa- 2015; Kriegeskorte, 2015): activations in a trained
tionally distinct and both sites together determine deep feedforward neural network map better to
a neuron’s output. One operation of the input to brain measures as compared to hand-crafted
the apical integration site is to amplify the neu- models (Kriegeskorte, 2015). Moreover, neural
ron’s response to its driving input in a context- networks that include types of recurrency
dependent manner, where context refers to activity (Kubilius et al., 2018) are able to explain certain
in other brain areas (Phillips, 2017). response dynamics in the ventral visual stream.
One empirical approach for testing contextual Still, artificial neural networks are at best bio-
cortical processing utilizes a simple retinotopic inspired, and there is much debate regarding the
principle. Sensory visual information is processed degree to which current successes relate to the
topographically by retinotopically organized functioning in the brain (Jacob et al., 2021).
Human Brain Project and Beyond 517

Figure 31.2 The brain uses top-down knowledge to make inferences about noisy or incom-
plete bottom-up sensory inputs, such as when objects are occluded.
Both information streams combine in individual pyramidal neurons with cell bodies in corti-
cal layer 5. Feedforward sensory inputs arrive to basal dendrites at the somatic region, and
feedback inputs arrive to the tuft dendrites in layer 1 and contextualise transmission of
information in the feedforward stream. Contextual inputs to superficial layers in human pri-
mary visual cortex can be studied using ultra high-resolution fMRI that offers a spatial scale
to approximately resolve cortical layers and columns.
Source: From Muckli et al., 2015.

At a lower level, artificial neurons are abstract have been suggested as multiplexing learning
models as compared to biological neurons, also signals and representational information (Payeur
in manners that ignore aspects that many argue et al., 2021; Shadmehr, 2018). In applications
are integral to the functioning of the brain. This like computer vision, latency coding, where the
includes in particular (i) the fact that biological relative timing of spikes encodes analogue val-
neurons communicate via spikes (Figure 31.3), ues, has been demonstrated to enable the map-
(ii) the spatial extent of biological neurons; and ping of deep neural networks to equally powerful
(iii) the diversity of influences mediated by vari- deep spiking neural networks (Stöckl and Maass,
ous neurotransmitters. For instance, the spatial 2021; Tavanaei et al., 2019). When spike timing is
extent of pyramidal neurons combined with the regulated relative to network oscillations such as
selective targeting of feedback connections is theta and gamma rhythms, phasic coding can be
suggested to play a key role in the generation of deployed as a mechanism of information transmis-
consciousness. sion, complementary to firing-rate coding (Huxter
The pulsed nature of communication between et al., 2003; Pennartz, 2009; Siegel et al., 2009).
biological, spiking neurons contrasts with the Rate-coding by single spiking neurons can be
standard “rate-based” abstraction of neuronal com- implemented by considering not the spikes but the
munication in artificial neurons, where the rate- effect of spikes on the membrane potential, like the
coding paradigm is typically presumed to model a resultant postsynaptic potential (PSP). Although
form of population coding (i.e., the instantaneous the postsynaptic response of an impinging spike
activity in a group of neurons can be read out as can be highly diverse, as the neurotransmitter
an approximately real-valued activation value by released may elicit activate multiple receptor sub-
downstream neurons; Gerstner and Kistler, 2002). types and involve multiple synapses, in simplified
However, the sparse and event-based activation spiking neuron models like the leaky-integrate-
of spiking neurons allows much richer dynamics. and-fire neuron the effect of impinging spikes is
The exact timing of the pulse can communicate typically reduced to the weighted inhibitory and
more information, expressed as bits per spike, excitatory responses to the neuron’s membrane
compared to rate-coding models (Nemenman potential, in the form of postsynaptic potentials.
et al., 2004). The timing of individual spikes may Thus, interpreting neural activations enables
enable the multiplexing of selective informa- the conversion of deep artificial neural networks
tion streams between brain areas (Fries, 2005; to deep spiking neural networks (Rueckauer et al.,
Singer and Gray, 1995), and complex spike pat- 2017; Zambrano et al., 2019), and to powerful
terns like bursts and complex spikes in cerebellum approximate supervised learning methods like
518 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 31.3 Biological neurons communicate via spikes.


(a) In simplified spiking neuron models, a spike traveling from one neuron to another
contributes a change in the target’s membrane potential in the form of a weighted
(with weight wij) inhibitory (negative) or excitatory (positive) postsynaptic potential (PSP),
denoted by a kernel ε(t).
(b) Schematic model of a simplified spiking neuron: impinging spikes at times ti cause
(weighted) postsynaptic potentials ε(t) that are added linearly to the membrane potential
u(t). A non-linear function f(u(t)) then determines when the target neuron emits a spike (tj)
that then influences downstream neurons k through synaptic weight wjk.
(c) In a simple model, f(u(t)) is determined by a threshold ϑ where a spike is emitted at time
tj when the potential induced by the sum of PSPs crosses the threshold from below. A refrac-
tory response is then subtracted from the membrane potential to reset the state of the
spiking neuron.

surrogate gradient learning (Bohte, 2011; Neftci by a rise and fall time. Effectively, an impinging
et al., 2019). Surrogate gradient approaches to spike will affect the membrane potential at the
supervised learning have resulted in competitive target neuron in a characteristic manner. Various
recurrent spiking neural networks for temporal studies suggest that this time-course can be inter-
tasks like ECG-wave and gesture recognition preted as a form of temporal prediction, either in
(Yin et al., 2020), while also allowing investi- populations of neurons encoding time-varying
gations into neural circuit dynamics (Keijser variables (Boerlin and Denève, 2011), or com-
and Sprekeler, 2020; Perez-Nieves et al., 2021). bined with synaptic short-term plasticity (Pfister
Biologically plausible reinforcement learning et al., 2010).
can furthermore train spiking neural networks Predictive coding posits that the brain con-
on and cognitive tasks like forced-alternative- stantly generates predictions of sensory input
choice (Frémaux et al., 2013; Karamanis et al., and adjusts its internal model of the environment
2018). Together, these studies demonstrate the based on discrepancies between predictions and
computational power of single spiking neurons actual input (Dayan et al., 1995; Rao and Ballard,
as units of neural computation, where spiking 1999). In such models, deeper layers generate
neural networks map to corresponding biological increasingly abstract top-down hypotheses about
structures. states of the environment, which is compared
A different interpretation of the effect of spikes against bottom-up input; the observed errors then
on target neurons focuses on the particular shape influence higher-layers to adjust their hypothesis
of the typical postsynaptic potential, characterized such that these errors are minimized.
Human Brain Project and Beyond 519

That real neurons are spiking neurons is an agents moving through the world (Bermudez-
obvious but mostly ignored constraint on pre- Contreras et al., 2020; Byrne et al., 2007). For
dictive coding models. Not only do inter-neural robotics the problem of autonomously operating
connections have to follow Dale’s law (that is, within novel environments is also an area of
neurons release the same type of neuro-transmit- extensive research with examples of interdiscipli-
ters to all their respective targets); such networks nary work generating useful insights (Milford and
also have to operate in a time-continuous man- Wyeth, 2008).
ner, where neural state progresses following the The central problem of navigation is how to
dynamical spiking neuron equations, and with combine information from noisy and intermittent
inherent non-linear responses. At present, these sensory observations to inform a useful estimate
issues are still open problems, with neurobiologi- of place and orientation within the environment,
cally realistic models of predictive coding still often referred to as pose in robotics. A distinc-
defined in rate-based standard artificial neural tion is made between allothetic and idiothetic
networks (Dora et al., 2021). The energy cost of sensory cues. “Allothetic” refers to external glob-
computational simulations is such that dedicated ally anchored landmarks; idiothetic refers to those
hardware in the form of neuromorphic chips, describing the motion of the animal or robot. For
like BrainScales and SpiNNaker, are crucial for example, the vestibular system in mammals is
sustainable scaling to network sizes relevant for an ever-present source of idiothetic information,
studying learning in large-scale brain structures. however, it cannot be relied upon for long peri-
Thus, developing effective and trainable net- ods of time due to accumulative error through
works of spiking neurons is an important step in integration. Similarly, the integration of odometry
increasing our understanding of how the brain or inertial measurements of self-motion in robots
generates perception and cognition at a mecha- suffers from drift. This error can be reduced by
nistic level, forcing researchers to explicitly con- calibrating against allothetic sensory cues that
sider aspects like temporal dynamics and time are anchored to the environment, however, these
scales while at the same time adhering to biologi- are less frequently available and are also subject
cal constraints like Dale’s law. Recent develop- to aliasing and other forms of perceptual uncer-
ments in learning paradigms have now opened up tainty. Such perceptual uncertainty can be reduced
novel venues for study, enabling function-guided through the integration of information from dif-
approaches that extend beyond hand-crafted ferent sensory modalities, so called sensor fusion
neural circuit models both in realism and in func- in robotics (Khaleghi et al., 2013). However, the
tionality (Zenke et al., 2021). problem still remains that associating allothetic
sensory cues to places implied through the inte-
gration of noisy idiothetic cues will lead to an
ever-increasing mismatch between an agent’s
perceived location and its ground truth pose. In
MULTISENSORY INTEGRATION AND robotics this problem has been addressed by incor-
SPATIAL NAVIGATION, FROM BRAIN porating mechanisms that recognize and exploit
loop-closure events, in other words, recalling that
TO ROBOTICS the robot is currently in a place that it has visited
in the past when it was less uncertain of its pose.
Whether models are spike-based or not, eventu- This prior can then be used to correct for accumu-
ally their practical effectiveness must meet the lative errors in current self-estimate of pose and
constraints of physical embodiment and environ- the corresponding relationships associated to other
mental ecology. Efforts for bridging models to allothetic cues observed from the start of the loop
embodied agents such as robots are illustrated (Durrant-Whyte and Bailey, 2006). This iterative
here for multisensory integration and spatial navi- process of exploration and consolidation builds
gation. Most animals have the ability to recall toward a stable representation of the environment
places that they have previously visited to navi- that can be used to compensate for idiothetic cue
gate their environment efficiently in their hunt for drift and, ultimately, used for route planning and
food and to find their way safely home. The neural goal directed behaviour.
mechanisms that underlie this ability in mammals In biology, recordings from place cells in rats that
have been extensively investigated with much are moved between familiar environments show
research focused on place, motion, head direction, that stable representations are maintained between
and boundary sensitive cells (Hafting et al., 2005). distinct environments (Alme et al., 2014). This cor-
These findings have inspired many computational relation between ensemble place cell activity and
models to emulate how these cells respond and previously explored environments implies that
interact in response to simulated or physical they maintain a trace memory of spatial contexts.
520 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

How these memories are formed through the con- about the HBP approach to this topic, in relation
solidation of multisensory experience has not been to the section above. Above we illustrated how
explicitly identified, however, in the Human Brain different brain states, including slow-wave sleep
Project we have been exploring the idea of episodic and asynchronous wakeful states can be modeled
memory formation through a process of predictive by mean-field approaches. This characterization
sensory reconstruction. We have collected multi- of brain states is closely linked to the experimen-
sensory data sets (visual-tactile) sampled from a tal demonstration of Up and Down states at the
biomimetic, whiskered mobile robot “WhiskEye” cellular level (Steriade et al., 2001) and has been
as it moves through and interacts with a bounded linked to the search for a complexity measure of
environment. These data sets have been used to consciousness.
train a multisensory deep predictive coding network Following the Integrated Information Theory
we have called MultiPredNet (Pearson et al., 2021) of consciousness (Haun et al., 2017; Massimini
inspired by work referred to above. The activity of et al., 2005; Tononi et al., 2016), it was found that
the multisensory top layer in the network drives pre- the Lempel-Ziv complexity of spatiotemporally
dictions of the activity in lower levels (i.e., toward unfolding cortical EEG responses elicited by a
the sensory input). These top-down predictions are transcranial magnetic stimulation (TMS) pulse is
compared to the bottom-up activity between layers significantly higher in wakeful than slow-wave
(originating from the ground truth sensory input) sleep states, and this finding was essentially repro-
and used to modify network weights to give better duced for the anesthetized state (Ferrarelli et al.,
predictions in the future. We have found that pre- 2010). In the HBP, this TMS approach has been
dictions can be inferred from one modality in the successfully applied to stratify patient groups suf-
absence of another (i.e., we can reconstruct a visual fering from Disorders of Consciousness (DoCs)
impression of the view through the presentation with different degrees of severity (Casarotto et al.,
of only touch). This implies that information from 2016). Although these findings might be taken to
visual cues have been associated with tactile cues confirm IIT, caution in the interpretation of results
within the multisensory latent representation space. is in place here, because the TMS results are in line
This approach is interesting to both neurosci- with a key role of Up and Down states regulating
ence and to robotics for three reasons; firstly, local the spatiotemporal propagation and reverberation
Hebbian like learning rules applied in parallel of cortical responses, whereas IIT’s cornerstones
throughout this network model has a greater affin- of Information Integration and Differentiation
ity to biological neuronal learning mechanisms. have been derived from the properties of phe-
Secondly, information from the two disparate sen- nomenal experience, which are not addressed by
sory modalities have been associated without any the electrophysiological TMS approach (Olcese
handcrafted intervention (i.e., this is a model-free et al., 2018).
learning approach to sensor fusion). Finally, the We have already introduced an alternative, rep-
observation that the form of the joint latent repre- resentation-oriented way of thinking about con-
sentation space of the trained network relates to the sciousness. This approach originates from the idea
actual pose of the robot implies that the network that conscious perception reflects a spatiotempo-
has formed a rudimentary map of the environment rally wide inference on the causes of the sensory
without explicit reference to conventional refer- inputs the brain receives continuously (Marcel,
ence frames or metrics. To explore this observa- 1983; Pennartz, 2015, 2022). All that the brain
tion further we have developed a simulated model receives as fast, discrete inputs informing it about
of the WhiskEye for use within the Neurorobotics the state of the environment and the subject’s own
platform of EBRAINS such that data sets can be body, comes in the form of action potential trains
collected during longer duration experiments not along the afferent nerves and pathways from the
subject to breakages or battery replacement. This spinal cord. For conscious perception to arise,
model is also available as an open-source project the brain needs to make sense of these myriads
to further encourage open science and collabora- of spikes trains, which it does by internally con-
tion of ideas. structing a model of the world, including the sub-
ject’s body. This line of thinking entails that we
do not consciously experience external reality
directly (in contrast to “Direct Realism,” assuming
that we do experience it directly) but rather a best-
REPRESENTATIONS AND guess representation of reality (even though our
CONSCIOUSNESS subjective intuition holds that we do seem to expe-
rience the world directly (Lehar, 2003; Pennartz,
The subject of consciousness is sufficiently broad 2015, 2018).
and deep to deserve a chapter in its own right, Computationally, this constructive process is
hence we can only summarize what is specific approached by predictive coding, in which the
Human Brain Project and Beyond 521

higher structures (in a sensory cortical hierarchy) The pluriformity of frameworks considered in the
forge predictive representations, which are sent HBP is not conceived of as a problem per se, as
down to lower structures to produce a compari- it reflects the current diversity of theories in the
son with bottom-up sensory inputs, resulting in a overall field of consciousness research. Instead,
prediction error used to improve the inference and the HBP stimulates initiatives to compare different
enable the network to learn (Dayan et al., 1995; approaches, develop adversarial experiments that
Rao and Ballard, 1999). Within the HBP, differ- discriminate between contrasting predictions from
ent neural implementations of predictive cod- the various theories, and integrate insights from
ing are pursued, such as those based on different different frameworks. For instance, predictive
cell populations encoding representations versus coding principles may be combined with elements
errors (Dora et al., 2021; Pennartz et al., 2019a) or from global neuronal workspace theory (Dehaene
on distinct somatodendritic compartments of cor- and Changeux, 2011; Whyte and Smith, 2021) and
tical pyramidal cells having different functions in studies into information integration-differentiation
bottom-up versus top-down, contextual feedback in feedforward-feedback cortical architectures.
(cf. Suzuki and Larkum, 2020).
Likewise, the HBP offers space to develop and
test different theories of how conscious experi-
ence may arise from constructs that are more WORKFLOWS IN THE HUMAN BRAIN
complex than the basic implementations of pre-
dictive coding, which are recognized to be insuf-
PROJECT: FROM PHYSIOLOGICAL DATA
ficient to explain consciousness on their own. TO MULTISCALE MODELS
According to Active Inference theory (Friston
et al., 2020; Hohwy and Seth, 2020), it is particu- We next take a closer look at the facilities and
larly the generation of actions that drives predic- tools developed in the HBP to produce, test and
tions on the sensory consequences of movement. run computational models for cognitive and sys-
Here, (conscious) perception is considered a tems neuroscience. The quantitative exploration
form of belief updating, in which the uncer- of the relationships within the brain’s architecture,
tainty about the causes of sensations is reduced. the rich repertoire of dynamics it expresses, and
Thus, an external object is only consciously seen its cognitive functions supports the creation of
when the subject actively looks at it. In contrast, theoretical and data-driven models that aim to
Neurorepresentationalism (Pennartz, 2015, 2022) capture the essential mechanisms underlying the
does not assume an essential role for motor action, capabilities of the brain and its pathologies. The
but characterizes conscious experience as an effectiveness of this effort is being demonstrated
immersive multimodal, situational survey of the through the reproduction of observable activity
individual’s environment and body state, which and cognitive capabilities of the brain in large-
can be built on complex sensory inference alone. scale simulations. The acquisition, demonstration
What makes representations ‘conscious’ in this and application of this knowledge can be sup-
framework is not such much the activity in senso- ported through the creation of Workflows dedicated
rimotor loops necessarily involving the body, but to acquisition, storage, analysis, and comparison
rather the upscaling of predictive activity across of experimental acquisitions and simulations.
the spatial domain (to account for the situational First, experimental data, models, analysis tools
aspect of conscious experience) and the domain and simulation results should be Findable: it must
of multimodal integration (to account for distinct be easy to search and locate the sets of informa-
sense modalities, or “qualia,” which we experi- tion relevant for each investigation. An essen-
ence as being distinct and rich in their totality). tial instrument is the inclusion of machine- and
Computational efforts to expand basic predictive human-readable Metadata (i.e., data that provide
coding models to deep networks and architectures information about the nature of each piece of
with more extensive representational scope and stored information). Second, information must be
cognitive abilities are ongoing (e.g., Dora et al., Accessible: after identification it must be possible
2021; Mannella et al., 2021). to get proper authentication and authorization,
Importantly, the ethical and philosophical retrieve the dataset and use it. Third, interoperable
investigation of the implications of these pluri- data, models and tools need to be incrementally
form approaches to consciousness is progress- integrated with novel contributions and users need
ing and has been yielding new perspectives, for to interoperate by subjecting the data to further
instance on ways to tackle the Hard Problem applications and workflows for analysis, compari-
(Pennartz, 2015), assessing consciousness in DoC son, storage, and processing. The fourth target is
patients (Farisco et al., 2017; Sanz et al., 2021), that information must be incrementally Reusable.
but also on consciousness in animals and artifi- The quality of the information and the complete-
cial intelligence devices (Pennartz et al., 2019b). ness of the accompanying metadata are essential
522 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

and must be certified by appropriate Curation experimental data and to data-driven and theoreti-
steps. Among the purposes of the HBP project is cally derived models of thalamo-cortical systems,
the creation of a research infrastructure platform to assess their similarity (e.g., using Wasserstein
(EBRAINS) that adopts such FAIR (Findable, (Panaretos and Zemel, 2019) or Kullback-Leibler
Accessible, Interoperable, Reproducible) method- metrics or automatic estimates of the number and
ology to integrate the knowledge body produced parameters of modes in multi-gaussian multi-
by HBP and by the research community at large. variate distributions). It also supports methods
A number of interoperable workflows are to infer from the experimental recordings data-
under development. A first example is the set driven models of the subject-specific brain under
of workflows to create and operate rodent and investigation (Capone et al., 2021). The pipeline is
human Brain Atlases. Brain Atlases provide sys- composed of modular Blocks, Stages and Cycles
tems to integrate neuroscientific knowledge in a organized at incremental levels according to work-
spatial reference framework. Information about flow Description Charts that specify the nature of
the shape, location, variability, structure, connec- experimental recordings, simulation models or
tivity, and activity of brain regions is collected in simulation outputs to be analyzed and compared.
common coordinate spaces, from multiple sub- Looking at the global future of neuroscience,
jects and, ideally, multiple species. Moreover, the the creation of solid foundations and method-
HBP creates workflows dedicated to more specific ologies for neuroscience workflows to be made
topics. As an example, we mention the HBP available the EBRAINS research infrastructure
workflow dedicated to the study of slow waves, is among the cornerstones on which incremental,
brain state transitions, their cognitive functions, large-scale cooperative endeavors in cognitive
and complexity in the framework of the activities and systems neuroscience across the next decades
dedicated to the investigation of networks under- can rely.
lying cognition and consciousness. The aim of this
workflow (Figure 31.4) is to integrate multi-scale,
multi-methodology experimental data, models,
and simulation outputs in a reproducible and
adaptable pipeline dedicated to analyzing slow MULTI-LEVEL HUMAN BRAIN ATLAS OF
wave activity (expressed under natural sleep, path- THE HUMAN BRAIN PROJECT
ological states or anaesthesia), in relation to the
transitions to higher complexity states and finally A main service supplied by HBP’s research infra-
to the specificities of awake regimes. structure EBRAINS for Cognitive and Systems
The goal of the slow-wave activity (SWA) Neuroscience is the multi-level human brain atlas.
workflow is the study of cognitive functions of This atlas is a tool to explore brain complexity at
NREM and REM sleep during incremental cycles multiple spatial sales and link it with cognitive
of awake learning and sleep, including the creation function and behavior. The atlas integrates state-
of data-driven and theoretical models that repro- of-the-art maps of the human brain in a common
duce and explain the mechanisms at work during 3D space. It combines in vivo neuroimaging data
sleep, and how they produce beneficial effects (e.g., functional MRI and DTI) to study networks
on awake cognition. In spiking neural network with microstructurally defined areas of the Julich-
models, a combination of dominant depression Brain Atlas, data on receptor architecture, gene
and selective potentiation of synaptic strengths expression, or the axonal architecture. The atlas
optimizes the post-sleep energetic working point allows to zoom into microscopical details of brain
and at the same time creates novel associations organization. Its siibra tool allows to extract data
between similar memories that improve post-sleep in an automated way using Python. The atlas
cognitive performances (Capone et al., 2019; provides an increasing range of maps, data, and
Golosio et al., 2021). This workflow supports the accompanying tools to serve as a resource for
characterization of the complex spatiotemporal clinical research targeting manipulations to allevi-
structure of cortical slow waves that travel along ate brain disorders, neural interfaces, brain mod-
the cortical surface during the different stages of eling, AI research, and cognitive neuroimaging.
physiological sleep and pathological sleep-like Starting from Korbinian Brodmann’s idea in
states and at different levels of anaesthesia. The 1909 of structure-function relationships at the
workflow estimates the probability density func- level of cortical areas, evidence has been pro-
tions of wave directions, speeds, and wave fre- vided that the cortical architecture (including its
quencies from experimental data or simulation cytoarchitecture) is more differentiated than his
outputs. Furthermore, it includes a set of data map suggests (Amunts and Zilles, 2015). Key in
analysis algorithms that can be applied to both developing new maps of the human brain is to
multi-scale, multi-species, multi-methodology formulate quantitative, reproducible criteria for
Human Brain Project and Beyond 523

Figure 31.4 Example of scientific workflow diagram: Characterization of spatio-temporal


features of cortical slow wave activity.
The workflow includes algorithmic blocks that support a quantitative and reproducible
comparison among experimental data acquired with multiple methodologies and/or the
outputs of different kinds of simulation models. Systematic curation of experimental data,
models, signals, and analysis outputs is essential to support HBP’s FAIR methodology.
Abbreviations: SWA, slow-wave activity; AW: awake state; TMS, transcranial magnetic
stimulation; ECoG, electrocorticography.

defining a cortical area, to consider novel prepara- different labs while addressing different levels of
tions, neuroimaging, and optical methods, as well brain organization.
as 2D & 3D quantitative measures. The large size The Julich Brain Atlas with its cytoarchitec-
of the human brain with its billions of nerve cells, tonic probability maps serves as microstructural
forming complex networks, implies to collect reference of the atlas. It is based on mapping
and analyze large amounts of data, making high- serial, cell-body-stained sections in a sample of
performance computing necessary. Deep learning 10 postmortem human brains in order to take into
has become a valuable tool to support brain map- account intersubject variability of brain organiza-
ping, and to derive features describing its micro- tion (Amunts et al., 2020). The maps have been 3D
structure. To integrate the different aspects of the reconstructed for each postmortem brain, and then
cortical architecture such as its cytoarchitecture, superimposed to a common reference space, and
connectivity, molecular, and genetic maps into a probability maps have been calculated. Different
common spatial reference system, and to investi- templates are being supported by the atlas, to
gate their role in cognitive functions and behavior, address the needs of the neuroimaging commu-
a multimodal human brain atlas is mandatory. The nity. The cytoarchitectonic maps are linked with
atlas developed in the HBP offers an increasing a comprehensive map of DTI-based fibre tracts in
number of high-quality and well-annotated data healthy subjects or patients (Ji et al., 2019), and
resulting from different experiments, coming from functional parcellations schemes based on fMRI,
524 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

which provide insights into the cognitive dimen- Going more deeply means connecting anatomic
sion of brain parcellation (Pinho et al., 2020, 2021). scales, and going to the next step in terms of detail.
The BigBrain (Amunts et al., 2013) allows us With its partners in Canada, the HBP team aims
to zoom into brain organization even to a resolu- to develop an atlas with 1-micrometer resolution
tion of 20 micrometers. It is created from thou- (Hiball; https://bigbrainproject.org/hiball.html). On
sands of microscopically imaged brain sections, this level, not only the cellular architecture, but also
stitched together into a 3D whole-brain volume. It the fine-scale detail of cellular morphology is vis-
allows us to anchor volumes of interest from opti- ible. Here, the mapping is increasingly sped up by
cal methods going down to the nanometer scale deep-learning methods (Kiwitz et al., 2020). This
(e.g., light-sheet imaging, using the VoluBA tool is relevant for cognitive research and brain medi-
of EBRAINS). The BigBrain is also a resource cine, because important details of connectivity,
to study cognition, and a specialized toolbox for example, only become visible at such resolu-
“BigBrainWarp” has been developed to link it tion. The perforant path is an example – it projects
with multi-modal neuroimaging data, for studying to the cornu ammonis and subiculum, and plays a
resting state, functional networks (Paquola et al., major role in learning and memory but also in the
2021). The BigBrain is also a tool to study neuro- pathogenesis of Alzheimer’s. Microscopical studies
imaging results obtained with high-field MRI, as based on 3D-PLI have shown that it does not form a
shown in Figure 31.2, to study visual processing uniform tract but rather a multi-component system
at laminar level. that originates in the entorhinal cortex with many
On the genetic level, the Jugex-tool allows to thin bundles (Zeineh et al., 2017). These data are
link cytoarchitecture and gene expression, inte- part of the HBP atlas, and accessible to the research
grating tissue transcriptomic data from the Allen community via EBRAINS (https://ebrains.eu/).
Brain Atlas and cytoarchitectonic segregation of The HBP brought major developments in cre-
the Julich Brain Atlas (Bludau et al., 2018). So ating the first truly multi-level and multi-modal
far, the molecular level of receptor maps has been atlases of the human brain. In EBRAINS, users can
integrated in a number of areas (Goulas et al., navigate and find well-curated, high-quality data
2021). Linking the molecular fingerprint of an with the help of the EBRAINS Knowledge Graph,
area to other structural and functional modalities or explore the same data visually in the EBRAINS
thus comes into reach. Area-to-area connectivity atlas viewer. Future uses for surgery and brain
is integrated on multiple scales, with Polarized stimulation in Parkinson’s disease, or other brain
Light Imaging and DTI data sources (Caspers and implants are on the horizon. In the HBP, existing
Axer, 2019) and connectivity profiles are being new connections have also been made to brain
computed on the fly for brain areas. Clinical data modeling communities. Of note here is ongoing
is being integrated from the HBP Human intracer- work on the Virtual Big Brain Epilepsy project
ebral EEG data database. (Hashemi et al., 2020) and the creation of cogni-
In future work, the aim is to both go more tive architectures for robotics control. Moreover,
broadly and more deeply into human brain atlas- individualized brain models of patients and their
ing, and to establish the multi-modal HBP atlas simulation play an increasing role in medicine.
further as an impactful community platform serv- The Virtual Brain (www.humanbrainproject.
ing basic science, medicine, and technology devel- eu/en/medicine/the-virtual-brain/) is a technol-
opment. Going more broadly means connecting ogy that has pioneered progress in this area. It
more modalities such a coming from immuno- may serve as an in-silico platform for clinical
histological studies revealing cell types and their hypothesis testing, improved diagnosis and devel-
distribution or spatial connectomics, to make brain opment of novel interventions (Jirsa et al., 2017).
models ever more comprehensive. For cognitive The general entry point for EBRAINS atlas ser-
neuroscience, enhanced precision of studies of vices is the EBRAINS web portal, which provides
structure-function relations is made possible. The information about the atlases and the most impor-
atlas framework allows to link the scales, and con- tant related tools. (see https://ebrains.eu/service/
nects different communities of neuroscience. A human-brain-atlas/)
major frontier of brain research is understanding
and interspecies differences, for instance between
mammalian and avian brains (Stacho et al., 2020). HUMAN BRAIN PROJECT –
Such comparative work has not only importance
in basic neuroscience, but has concrete conse-
INTERNATIONAL CONTEXT:
quences in the field of neurological and psychiat- COLLABORATIVE BRAIN INITIATIVES
ric disorders, considering that evolution has a role
in several brain disorders and models designed to The second decade of this century saw a remark-
replicate human brain diseases are not “perfect” able growth in large brain initiatives around the
(Brune and Schiefenhovel, 2019). world. HBP and the US BRAIN (United States
Human Brain Project and Beyond 525

Figure 31.5 The multi-level human brain atlas of the HBP includes BigBrain (Amunts et al.,
2013) as a template space at microscopical resolution. Different modalities such as cortical
layers, the extent of cortical areas or receptor fingerprints of different neurotransmitter
systems (polar plot) are available.

Brain Research through Advancing Innovative declaration also covered engagement with citizens,
Neurotechnologies), both launched in 2013, were patients, and relevant communities to understand
the first in a wave of new initiatives (Kandel et al., their concerns and communicate the opportunities
2013), later followed by Brain/MINDS (Brain and challenges arising from brain research. These
Mapping by Integrated Neurotechnologies for commitments were fully compatible with the mis-
Disease Studies) in Japan and several other initia- sions of the HBP and the actions that the project
tives. In 2017, the initiatives from Europe, the had initiated and later pursued with increasing
United States, Japan, Korea, and Australia, have intensity, in the areas of international collabora-
agreed on a Declaration of intent to collaborate tion, education, outreach, community building,
and join forces through the International Brain capacities for shared research data, neuro-ethics,
Initiative (IBI). The primary motivation was the and data governance (e.g., Amunts et al., 2016,
common recognition that “no single initiative can 2019; Bjerke et al., 2018; Salles et al., 2019).
tackle the challenge to measure, map, image, Neuroethics and data sharing are two pri-
model, simulate, understand, imitate, diagnose mary examples of overlapping areas of interest
and heal the brain.” The new organization (IBI) between the HBP and the other brain initiatives.
was formed one year later, with two additional Neuroethics addresses questions of relevance for
initiatives in China and in Canada (Adams et al., neuroscience in general and of primary impor-
2020). Details about the constituent initiatives are tance for cognitive neuroscience. Neuroethics
included in Table 31.1. research relates to understanding of the nature of
The declaration to instigate the IBI included a the self and identity, the existence and meaning of
series of commitments. In brief, the commitments free will, and defining the role of reason in human
were centered around leveraging and aligning behaviour. The IBI has been active in aligning
efforts to maximize efficiency, working with key efforts in the field of neuroethics, as exempli-
stakeholders, exploring and implementing data fied by a list of neuroethics questions serving as
sharing and standardization mechanisms, and col- a guidance for ethical research, developed by the
laborating in the fields of neuro-ethics and respon- Neuroethics working group of the IBI (Amadio
sible data stewardship and privacy protection. The et al., 2018; also see Box 29.1).
526 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Table 31.1 List of large-scale neuroscience projects included in the International


Brain Initiative
Project/initiative Country Focus Starting year

Human Brain Project European Union Development of tools and software for data analysis, 2013
modeling, cognitive neuroscience and neuromorphics
US BRAIN Initiative USA Development of innovative brain technologies 2013
Brain/MINDS Japan Developing of brain mapping technologies for human and 2014
other animals (marmoset)
Australian Brain Alliance Australia “Cracking the brain’s code,” developing neural interfaces and 2016
brain-inspired computing
Korean Brain Initiative South Korea Advancing brain mapping, AI-related R&D, multidisciplinary 2016
research and personalized medicine
China Brain Project China Advancing research in cognitive function, diagnosis of brain 2016
disorders and brain-inspired AI
Canadian Brain Canada Establishing open, transdisciplinary and collaborative research 2017
Research Strategy in brain science and brain-inspired AI

Note. All the projects listed above share common principles and complementary goals (including topics in neurosci-
ence, cognitive science, medicine, AI, and novel technologies), and engage in a framework of cooperation and knowledge
dissemination.

Box 29.1 Neuroethical questions guiding ethical research in the International Brain
Initiatives (from Amadio et al., 2018)

Q1. What is the potential impact of a model or neuroscientific account of disease on individuals,
communities, and society?
Q2. What are the ethical standards of biological material and data collection and how do local standards
compare to those of global collaborators?
Q3. What is the moral significance of neural systems that are under development in neuroscience research
laboratories?
Q4. How could brain interventions impact or reduce autonomy?
Q5. In which contexts might a neuroscientific technology/innovation be used or deployed?

SUMMARY AND CONCLUSIONS within the areas of data governance (Eke et al.,
2021), findability of data across projects and
Neuroscience is in the midst of a revolution as to assuring interoperability and training of a new
how the field manages compiled research data researchers prepared for the next generation solu-
within a new era of international collaborative tions for data management. New training material
sharing, driven by relatively recently introduced is disseminated through the International
technical/technological opportunities enabling the Neuroinformatics Coordinating Facility ( INCF)
making of large amounts of data widely available Training Space (https://training.incf.org/).
in combination with computing resources. All The Human Brain Project (HBP) has defined
brain initiatives around the world are facing the sharing of open data, models, and tools, as crit-
equivalent implementation challenges. ical for reaching its goal of integrating brain sci-
Consequently, the International Brain Initiative ence across research modalities and spatial scales.
(IBI) has initiated a new working group in relation The HBP launched the European Brain Research
to maintaining data standards and sharing aimed Infrastructure (EBRAINS) in 2019, and since the
at facilitating discovery, harmonization, and use of beginning of the project has been very active in
data across brain projects and initiatives globally. developing solutions for data governance, data
The working group was tasked to deliver guidance sharing, and standards, and training of research-
to the international neuroscience community ers. Services for FAIR data, open to the broader
Human Brain Project and Beyond 527

research community, aimed both at researchers Amunts, K., Mohlberg, H., Bludau, S., & Zilles, K.
producing data and those using data for analysis (2020). Julich-Brain: A 3D probabilistic atlas of the
and modeling, are embedded within EBRAINS. human brain’s cytoarchitecture. Science, 369,
The future development and these and other ser- 988–992.
vices in EBRAINS is influencing, and is influ- Amunts, K., & Zilles, K. (2015). Architectonic map-
enced by the activities and results achieved by the ping of the human brain beyond Brodmann.
several other brain initiatives all around the world. Neuron, 88(6), 1086–1107.
Finally, the HBP has been a major driver of neu- Bar, M. (2004). Visual objects in context. Nature
roethics research, as well as ethics related actions Reviews: Neuroscience, 5(8), 617–629.
also involving societal engagement. The HBP has Bermudez-Contreras, E., Clark, B. J., & Wilber, A.
placed a strong emphasis on ethics management (2020). The neuroscience of spatial navigation and
and ethics rapporteur functions at the project level. the relationship to artificial intelligence. Frontiers
Data sharing is another common area of priority in Computational Neuroscience, 14. doi: 10.3389/
within both both the HBP and the IBI. Data shar- fncom.2020.00063
ing is a prerequisite for large scale data integration Billeh, Y. N., Cai, B., Gratiy, S. L., Dai, K., Iyer, R.,
across modalities and addresses fundamental chal- Gouwens, N. W., … Arkhipov, A. (2020). System-
lenges within the fields of neuroscientific research atic integration of structural and functional data
reproducibility and replicability. into multi-scale models of mouse primary visual
cortex. Neuron, 106(3), 388–403.
Bjerke, I. E., Øvsthus, M., Papp, E. A., Yates, S. C.,
Silvestri, L., Fiorilli, J., … Bjaalie, J. G. (2018). Data
integration through brain atlasing: Human brain
REFERENCES project tools and strategies. European Psychiatry,
50, 70–76.
Adams, A., Albin, S., Amunts, K., Asakawa, T., Ber- Bludau, S., Mühleisen, T. W., Eickhoff, S. B., Hawry-
nard, A., Bjaalie, J. G., … Zheng, J. (2020). Inter- lycz, M. J., Cichon, S., & Amunts, K. (2018). Inte-
national Brain Initiative: An innovative framework gration of transcriptomic and cytoarchitectonic
for coordinated global brain research efforts. data implicates a role for MAOA and TAC1 in the
Neuron, 105(2), 212–216. limbic-cortical network. Brain Structure and Func-
Alme, C. B., Miao, C., Jezek, K., Treves, A., Moser, E. tion, 223(5), 2335–2342.
I., & Moser, M.-B. (2014). Place cells in the hip- Boerlin, M., & Denève, S. (2011). Spike-based popu-
pocampus: Eleven maps for eleven rooms. Pro- lation coding and working memory. PLoS Compu-
ceedings of the National Academy of Sciences of tational Biology, 7(2). doi: 10.1371/journal.pcbi.
the United States of America, 111(52), 1001080
18428–18435. Bohte, S. M. (2011). Error-backpropagation in net-
Amadio, J., Bi, G.-Q., Boshears, P. F., Carter, A., works of fractionally predictive spiking neurons.
Devor, A., Doya, K., … Singh, I. (2018). Neuroeth- In T. Honkela, W. Duch, M. Girolami & S. Kaski
ics questions to guide ethical research in the inter- (Eds.), Artificial neural networks and machine
national brain initiatives. Neuron, 100(1), 19–36. learning: ICANN 2011 (pp. 60-68). Berlin:
Amit, D. J., & Brunel, N. (1997). Model of global Springer.
spontaneous activity and local structured activity Brette, R., & Gerstner, W. (2005). Adaptive exponen-
during delay periods in the cerebral cortex. Cere- tial integrate-and-fire model as an effective
bral Cortex, 7(3), 237–252. description of neuronal activity. Journal of Neuro-
Amunts, K., Ebell, C., Muller, J., Telefont, M., Knoll, physiology, 94(5), 3637–3642.
A., & Lippert, T. (2016). The Human Brain Project: Brune, M., & Schiefenhovel, W. (2019). In M. Brüne
Creating a European research infrastructure to & W. Schiefenhövel (Eds.), Oxford handbook of
decode the human brain. Neuron, 92(3), evolutionary medicine. Oxford, UK: Oxford
574–581. University Press.
Amunts, K., Knoll, A. C., Lippert, T., Pennartz, C. M. Byrne, P., Becker, S., & Burgess, N. (2007). Remem-
A., Ryvlin, P., Destexhe, A., … Bjaalie, J. G. (2019). bering the past and imaging the future: A neural
The Human Brain Project: Synergy between neuro- model of spatial memory and imagery. Psychologi-
science, computing, informatics, and brain- cal Review, 114(2), 340–375.
inspired technologies. PLoS Biology, 17(7). doi: Capone, C., De Luca, C., De Bonis, G., Pastorelli, E.,
10.1371/journal.pbio.3000344 Mascaro, A. L. A., Resta, F., Pavone, F., & Paolucci,
Amunts, K., Lepage, C., Borgeat, L., Mohlberg, H., P. S. (2021). Simulations approaching data: Corti-
Dickscheid, T., Rousseau, M.-É., … Evans, A. C. cal slow waves in inferred models of the whole
(2013). BigBrain: An ultrahigh-resolution 3D hemisphere of mouse. http://arxiv.org/abs/
human brain model. Science, 340, 1472–1475. 2104.07445 (retrieved Jan. 6, 2022)
528 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Capone, C., Pastorelli, E., Golosio, B., & Paolucci, P. S. along the visual cortical hierarchy. Frontiers in Com-
(2019). Sleep-like slow oscillations improve visual putational Neuroscience, 15. doi: 10.3389/
classification through synaptic homeostasis and fncom.2021.666131
memory association in a thalamo-cortical model. Durrant-Whyte, H., & Bailey, T. (2006). Simultaneous
Scientific Reports, 9(1). doi: 10.1038/ localization and mapping: Part I. IEEE Robotics
s41598-019-45525-0 Automation Magazine, 13(2), 99–110.
Casarotto, S., Comanducci, A., Rosanova, M., Sar- Eke, D., Bernard, A., Bjaalie, J. G., Chavarriaga, R.,
asso, S., Fecchio, M., Napolitani, M., … Massimini, Hanakawa, T., Hannan, A., … Pestilli, F. (2021).
M. (2016). Stratification of unresponsive patients International data governance for neuroscience.
by an independently validated index of brain com- PsyArXiv. doi: 10.31234/osf.io/esz9b
plexity. Annals of Neurology, 80(5), 718–729. El Boustani, S., & Destexhe, A. (2009). A master
Caspers, S., & Axer, M. (2019). Decoding the micro- equation formalism for macroscopic modelling of
structural correlate of diffusion MRI. NMR in Bio- asynchronous irregular activity states. Neural Com-
medicine, 32(4). doi: 10.1002/nbm.3779 putation, 21(1), 46–100.
Chaudhuri, R., Knoblauch, K., Gariel, M.-A., Ken- Farisco, M., Laureys, S., & Evers, K. (2017). The intrin-
nedy, H., & Wang, X.-J. (2015). A large-scale cir- sic activity of the brain and its relation to levels and
cuit mechanism for hierarchical dynamical disorders of consciousness. Mind and Matter,
processing in the primate cortex. Neuron, 88(2), 15(2), 197–219.
419–431. Ferrarelli, F., Peterson, M. J., Sarasso, S., Riedner, B.
Chemla, S., Reynaud, A., Volo, M. di, Zerlaut, Y., Per- A., Murphy, M. J., Benca, R. M., … Tononi, G.
rinet, L., Destexhe, A., & Chavane, F. (2019). Sup- (2010). Thalamic dysfunction in schizophrenia
pressive traveling waves shape representations of suggested by whole-night deficits in slow and fast
illusory motion in primary visual cortex of awake spindles. American Journal of Psychiatry, 167(11),
primate. Journal of Neuroscience, 39(22), 1339–1348.
4282–4298. Foster, N. N., Barry, J., Korobkova, L., Garcia, L., Gao,
Compte, A., Brunel, N., Goldman-Rakic, P. S., & L., Becerra, M., … Dong, H.-W. (2021). The mouse
Wang, X.-J. (2000). Synaptic mechanisms and cortico–basal ganglia–thalamic network. Nature,
network dynamics underlying spatial working 598, 188–194.
memory in a cortical network model. Cerebral Frémaux, N., Sprekeler, H., & Gerstner, W. (2013).
Cortex, 10(9), 910–923. Reinforcement learning using a continuous time
Dayan, P., Hinton, G. E., Neal, R. M., & Zemel, R. S. actor-critic framework with spiking neurons. PLoS
(1995). The Helmholtz machine. Neural Computa- Computational Biology, 9(4). doi: 10.1371/journal.
tion, 7(5), 889–904. pcbi.1003024
Deco, G., Jirsa, V. K., & McIntosh, A. R. (2011). Fries, P. (2005). A mechanism for cognitive dynamics:
Emerging concepts for the dynamical organization Neuronal communication through neuronal coher-
of resting-state activity in the brain. Nature ence. Trends in Cognitive Sciences, 9(10), 474–480.
Reviews: Neuroscience, 12(1), 43–56. Friston, K. J., Harrison, L., & Penny, W. (2003).
Deco, G., Jirsa, V. K., Robinson, P. A., Breakspear, M., Dynamic causal modelling. NeuroImage, 19(4),
& Friston, K. (2008). The dynamic brain: From spik- 1273–1302.
ing neurons to neural masses and cortical fields. Gămănuţ, R., Kennedy, H., Toroczkai, Z., Ercsey-
PLoS Computational Biology, 4(8). doi: 10.1371/ Ravasz, M., Van Essen, D. C., Knoblauch, K., &
journal.pcbi.1000092 Burkhalter, A. (2018). The mouse cortical connec-
Dehaene, S., & Changeux, J.-P. (2011). Experimental tome, characterized by an ultra-dense cortical
and theoretical approaches to conscious process- graph, maintains specificity by distinct connectivity
ing. Neuron, 70(2), 200–227. profiles. Neuron, 97(3), 698–715.
Dehghani, N., Peyrache, A., Telenczuk, B., Le Van Gerstner, W., & Kistler, W. M. (2002). In W. Gerstner
Quyen, M., Halgren, E., Cash, S. S., … Destexhe, & W. Kistler (Eds.), Spiking neuron models: Single
A. (2016). Dynamic balance of excitation and inhi- neurons, populations, plasticity. Cambridge, UK:
bition in human and monkey neocortex. Scientific Cambridge University Press.
Reports, 6(1). doi: 10.1038/srep23176 Goldman, J. S., Kusch, L., Yalcinkaya, B. H., Depanne-
di Volo, M., Romagnoni, A., Capone, C., & Destexhe, maecker, D., Nghiem, T.-A. E., Jirsa, V., & Destexhe,
A. (2019). Biologically realistic mean-field models A. (2020). Brain-scale emergence of slow-wave
of conductance-based networks of spiking neu- synchrony and highly responsive asynchronous
rons with adaptation. Neural Computation, 31(4), states based on biologically realistic population
653–680. models simulated in the virtual brain. BioRxiv. doi:
Dora, S., Bohte, S. M., & Pennartz, C. M. A. (2021). 10.1101/2020.12.28.424574
Deep gated Hebbian predictive coding accounts for Goldman, J.S., Kusch, L., Aquilue, D., Yalcinkaya,
emergence of complex neural response properties B.H., Depannemaecker, D., Ancourt, K., Nghiem,
Human Brain Project and Beyond 529

T-A., Jirsa, V., & Destexhe, A. (2022). A compre- feedback pathways in cognitive computations.
hensive neural simulation of slow-wave sleep and Neuron, 101(2), 321–336.
highly responsive wakefulness dynamics. Frontiers Ji, E., Guevara, P., Guevara, M., Grigis, A., Labra, N.,
in Computational Neuroscience 16: 1058957. doi: Sarrazin, S., … Houenou, J. (2019). Increased and
10.3389/fncom.2022.1058957 decreased superficial white matter structural con-
Goldman, J. S., Tort-Colet, N., di Volo, M., Susin, E., nectivity in schizophrenia and bipolar disorder.
Bouté, J., Dali, M., … Destexhe, A. (2019). Bridg- Schizophrenia Bulletin, 45(6), 1367–1378.
ing single neuron dynamics to global brain states. Jirsa, V. K., Proix, T., Perdikis, D., Woodman, M. M.,
Frontiers in Systems Neuroscience, 13. doi: Wang, H., Gonzalez-Martinez, J., … Bartolomei, F.
10.3389/fnsys.2019.00075 (2017). The virtual epileptic patient: Individualized
Golosio, B., Luca, C. D., Capone, C., Pastorelli, E., whole-brain models of epilepsy spread. NeuroIm-
Stegel, G., Tiddia, G., … Paolucci, P. S. (2021). age, 145, 377–388.
Thalamo-cortical spiking model of incremental Joglekar, M. R., Mejias, J. F., Yang, G. R., & Wang,
learning combining perception, context and X.-J. (2018). Inter-areal balanced amplification
NREM-sleep. PLoS Computational Biology, 17(6). enhances signal propagation in a large-scale cir-
doi: 10.1371/journal.pcbi.1009045 cuit model of the primate cortex. Neuron, 98(1),
Goulas, A., Changeux, J.-P., Wagstyl, K., Amunts, K., 222–234.
Palomero-Gallagher, N., & Hilgetag, C. C. (2021). Kandel, E. R., Markram, H., Matthews, P. M., Yuste,
The natural axis of transmitter receptor distribu- R., & Koch, C. (2013). Neuroscience thinks big
tion in the human cerebral cortex. Proceedings of (and collaboratively). Nature Reviews: Neurosci-
the National Academy of Sciences of the United ence, 14(9), 659–664.
States of America, 118(3). doi: 10.1073/ Karamanis, M., Zambrano, D., & Bohté, S. (2018).
pnas.2020574118 Continuous-time spike-based reinforcement learn-
Güçlü, U., & Gerven, M. A. J. van. (2015). Deep ing for working memory tasks. In V. Kůrková, Y.
neural networks reveal a gradient in the complex- Manolopoulos, B. Hammer, L. Iliadis & I. Maglogi-
ity of neural representations across the ventral annis (Eds.), Artificial neural networks and machine
stream. Journal of Neuroscience, 35(27), learning: ICANN 2018 (pp. 250-262). Cham, Swit-
10005–10014. zerland: Springer.
Hafting, T., Fyhn, M., Molden, S., Moser, M.-B., & Keijser, J., & Sprekeler, H. (2020). Interneuron diversity
Moser, E. I. (2005). Microstructure of a spatial map is required for compartment-specific feedback inhi-
in the entorhinal cortex. Nature, 436, 801–806. bition. BioRxiv. doi: 10.1101/2020.11.17.386920
Hashemi, M., Vattikonda, A. N., Sip, V., Guye, M., Khaleghi, B., Khamis, A., Karray, F. O., & Razavi, S. N.
Bartolomei, F., Woodman, M. M., & Jirsa, V. K. (2013). Multisensor data fusion: A review of the
(2020). The Bayesian virtual epileptic patient: A state-of-the-art. Information Fusion, 14(1),
probabilistic framework designed to infer the spa- 28–44.
tial map of epileptogenicity in a personalized Kiwitz, K., Schiffer, C., Spitzer, H., Dickscheid, T., &
large-scale brain model of epilepsy spread. Neuro- Amunts, K. (2020). Deep learning networks reflect
Image, 217. doi: 10.1016/j. cytoarchitectonic features used in brain mapping.
neuroimage.2020.116839 Scientific Reports, 10(1). doi: 10.1038/
Haun, A. M., Tononi, G., Koch, C., & Tsuchiya, N. s41598-020-78638-y
(2017). Are we underestimating the richness of Knox, J. E., Harris, K. D., Graddis, N., Whitesell, J. D.,
visual experience? Neuroscience of Consciousness, Zeng, H., Harris, J. A., … Mihalas, S. (2018). High-
2017(1). doi: 10.1093/nc/niw023 resolution data-driven model of the mouse con-
Hopfield, J. J. (1982). Neural networks and physical nectome. Network Neuroscience, 3(1), 217–236.
systems with emergent collective computational Kriegeskorte, N. (2015). Deep neural networks: A
abilities. Proceedings of the National Academy of new framework for modeling biological vision and
Sciences of the United States of America, 79(8), brain information processing. Annual Review of
2554–2558. Vision Science, 1, 417–446.
Huxter, J., Burgess, N., & O’Keefe, J. (2003). Inde- Kubilius, J., Schrimpf, M., Nayebi, A., Bear, D.,
pendent rate and temporal coding in hippocampal Yamins, D. L. K., & DiCarlo, J. J. (2018). CORnet:
pyramidal cells. Nature, 425, 828–832. Modeling the neural mechanisms of core object
Jacob, G., Pramod, R. T., Katti, H., & Arun, S. P. recognition. BioRxiv. doi: 10.1101/408385
(2021). Qualitative similarities and differences in Larkum, M. (2013). A cellular mechanism for cortical
visual object representations between brains and associations: An organizing principle for the cere-
deep networks. Nature Communications, 12(1). bral cortex. Trends in Neurosciences, 36(3),
doi: 10.1038/s41467-021-22078-3 141–151.
Jaramillo, J., Mejias, J. F., & Wang, X.-J. (2019). Lehar, S. (2003). Gestalt isomorphism and the pri-
Engagement of pulvino-cortical feedforward and macy of subjective conscious experience: A Gestalt
530 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Bubble model. Behavioral and Brain Sciences, drawings. Journal of Neuroscience, 39(47),
26(4), 375–444. 9410–9423.
Lindeman, S., Hong, S., Kros, L., Mejias, J. F., Muckli, L., De Martino, F., Vizioli, L., Petro, L. S.,
Romano, V., Oostenveld, R., … Zeeuw, C. I. D. Smith, F. W., Ugurbil, K., … Yacoub, E. (2015).
(2021). Cerebellar Purkinje cells can differentially Contextual feedback to superficial layers of V1.
modulate coherence between sensory and motor Current Biology, 25(20), 2690–2695.
cortex depending on region and behavior. Pro- Muller, L., Reynaud, A., Chavane, F., & Destexhe, A.
ceedings of the National Academy of Sciences of (2014). The stimulus-evoked population response
the United States of America, 118(2). doi: in visual cortex of awake monkey is a propagating
10.1073/pnas.2015292118 wave. Nature Communications, 5(1). doi: 10.1038/
Mannella, F., Maggiore, F., Baltieri, M., & Pezzulo, G. ncomms4675
(2021). Active inference through whiskers. Neural Neftci, E. O., Mostafa, H., & Zenke, F. (2019). Surro-
Networks, 144, 428–437. gate gradient learning in spiking neural networks:
Marcel, A. J. (1983). Conscious and unconscious Bringing the power of gradient-based optimiza-
perception: An approach to the relations between tion to spiking neural networks. IEEE Signal Pro-
phenomenal experience and perceptual processes. cessing Magazine, 36(6), 51–63.
Cognitive Psychology, 15(2), 238–300. Nemenman, I., Bialek, W., & de Ruyter van Ste-
Markov, N. T., Ercsey-Ravasz, M. M., Ribeiro Gomes, veninck, R. (2004). Entropy and information in
A. R., Lamy, C., Magrou, L., Vezoli, J., … Kennedy, neural spike trains: Progress on the sampling prob-
H. (2014). A weighted and directed interareal con- lem. Physical Review E, 69(5). doi: 10.1103/
nectivity matrix for macaque cerebral cortex. Cer- PhysRevE.69.056111
ebral Cortex, 24(1), 17–36. Olcese, U., Oude Lohuis, M. N., & Pennartz, C. M. A.
Markov, Nikola T., Ercsey-Ravasz, M., Essen, D. C. V., (2018). Sensory processing across conscious and
Knoblauch, K., Toroczkai, Z., & Kennedy, H. nonconscious brain states: From single neurons to
(2013). Cortical high-density counterstream archi- distributed networks for inferential representation.
tectures. Science, 342. doi: 10.1126/science. Frontiers in Systems Neuroscience, 12. doi:
1238406 10.3389/fnsys.2018.00049
Markov, Nikola T., Vezoli, J., Chameau, P., Falchier, Panaretos, V. M., & Zemel, Y. (2019). Statistical
A., Quilodran, R., Huissoud, C., … Kennedy, H. aspects of Wasserstein distances. Annual Review
(2014). Anatomy of hierarchy: Feedforward and of Statistics and Its Application, 6(1), 405–431.
feedback pathways in macaque visual cortex. Jour- Paquola, C., Royer, J., Lewis, L. B., Lepage, C., Gla-
nal of Comparative Neurology, 522(1), 225–259. tard, T., Wagstyl, K., … Bernhardt, B. (2021). The
Massimini, M., Ferrarelli, F., Huber, R., Esser, S. K., BigBrainWarp toolbox for integration of BigBrain
Singh, H., & Tononi, G. (2005). Breakdown of 3D histology with multimodal neuroimaging.
cortical effective connectivity during sleep. Sci- ELife, 10. doi: 10.7554/eLife.70119
ence, 309, 2228–2232. Payeur, A., Guerguiev, J., Zenke, F., Richards, B. A., &
Meijer, G. T., Marchesi, P., Mejias, J. F., Montijn, J. S., Naud, R. (2021). Burst-dependent synaptic plastic-
Lansink, C. S., & Pennartz, C. M. A. (2020). Neural ity can coordinate learning in hierarchical circuits.
correlates of multisensory detection behavior: Nature Neuroscience, 24(7), 1010–1019.
Comparison of primary and higher-order visual Pearson, M., Dora, S., Struckmeier, O., Knowles, T.,
cortex. Cell Reports, 31(6). doi: 10.1016/j. Mitchinson, B., Tiwari, K., … Pennartz, C. M. A.
celrep.2020.107636 (2021). Multimodal representation learning for
Mejías, J. F., Murray, J. D., Kennedy, H., & Wang, X.- place recognition using deep Hebbian predictive
J. (2016). Feedforward and feedback frequency- coding. Frontiers in Robotics and AI. doi: 10.3389/
dependent interactions in a large-scale laminar frobt.2021.732023
network of the primate cortex. Science Advances, Pennartz, C. M. A. (2009). Identification and integra-
2(11). doi: 10.1126/sciadv.1601335 tion of sensory modalities: Neural basis and rela-
Mejías, J. F., & Wang, X.-J. (2022). Mechanisms of tion to consciousness. Consciousness and
distributed working memory in a large-scale net- Cognition, 18(3), 718–739.
work of macaque neocortex. eLife, 11. doi: Pennartz, C. M. A. (2015). The brain’s representa-
10.7554/eLife72136 tional power: On consciousness and the integra-
Milford, M. J., & Wyeth, G. F. (2008). Mapping a tion of modalities. Cambridge, MA: MIT Press.
suburb with a single camera using a biologically Pennartz, C. M. A. (2018). Consciousness, represen-
inspired SLAM system. IEEE Transactions on Robot- tation, action: The importance of being goal-
ics, 24(5), 1038–1053. directed. Trends in Cognitive Sciences, 22(2),
Morgan, A. T., Petro, L. S., & Muckli, L. (2019). Scene 137–153.
representations conveyed by cortical feedback to Pennartz, C. M. A. (2022). What is neurorepresenta-
early visual cortex can be described by line tionalism? From neural activity and predictive
Human Brain Project and Beyond 531

processing to multi-level representations and con- Salles, A., Bjaalie, J. G., Evers, K., Farisco, M., Fother-
sciousness. Behavioural Brain Research, 432. doi: gill, B. T., Guerrero, M., … Amunts, K. (2019). The
10.1016/j.bbr.2022.113969 Human Brain Project: Responsible brain research
Pennartz, C. M. A., Dora, S., Muckli, L., & Lorteije, J. for the benefit of society. Neuron, 101(3),
A. M. (2019a). Towards a unified view on path- 380–384.
ways and functions of neural recurrent processing. Sanz, L. R. D., Thibaut, A., Edlow, B. L., Laureys, S.,
Trends in Neurosciences, 42(9), 589–603. & Gosseries, O. (2021). Update on neuroimaging
Pennartz, C. M. A., Farisco, M., & Evers, K. (2019b). in disorders of consciousness. Current Opinion in
Indicators and criteria of consciousness in animals Neurology, 34(4), 488–496.
and intelligent machines: An inside-out approach. Sanz-Leon, P., Knock, S. A., Spiegler, A., & Jirsa, V. K.
Frontiers in Systems Neuroscience, 13. doi: (2015). Mathematical framework for large-scale
10.3389/fnsys.2019.00025 brain network modeling in the virtual brain. Neu-
Perez-Nieves, N., Leung, V. C. H., Dragotti, P. L., & roImage, 111, 385–430.
Goodman, D. F. M. (2021). Neural heterogeneity Shadmehr, R. (2018). How the cerebellum learns to
promotes robust learning. Nature Communica- build a sequence. ELife, 7. doi: 10.7554/
tions, 12(1). doi: 10.1038/s41467-021-26022-3 eLife.40660
Peyrache, A., Dehghani, N., Eskandar, E. N., Madsen, Siegel, M., Warden, M. R., & Miller, E. K. (2009).
J. R., Anderson, W. S., Donoghue, J. A., … Phase-dependent neuronal coding of objects in
Destexhe, A. (2012). Spatiotemporal dynamics of short-term memory. Proceedings of the National
neocortical excitation and inhibition during human Academy of Sciences of the United States of
sleep. Proceedings of the National Academy of America, 106(50), 21341–21346.
Sciences of the United States of America, 109(5), Singer, W., & Gray, C. (1995). Visual feature integra-
1731–1736. tion and the temporal correlation hypothesis.
Pfister, J.-P., Dayan, P., & Lengyel, M. (2010). Syn- Annual Review of Neuroscience, 18, 555–586.
apses with short-term plasticity are optimal esti- Stacho, M., Herold, C., Rook, N., Wagner, H., Axer,
mators of presynaptic membrane potentials. M., Amunts, K., & Güntürkün, O. (2020). A
Nature Neuroscience, 13(10), 1271–1275. cortex-like canonical circuit in the avian fore-
Phillips, W. A., Clark, A., & Silverstein, S. M. (2015). brain. Science, 369. doi: 10.1126/science.
On the functions, mechanisms, and malfunctions abc5534
of intracortical contextual modulation. Neurosci- Steriade, M., Timofeev, I., & Grenier, F. (2001). Natu-
ence & Biobehavioral Reviews, 52, 1–20. ral waking and sleep states: A view from inside
Phillips, W. A. (2017). Cognitive functions of intracel- neocortical neurons. Journal of Neurophysiology,
lular mechanisms for contextual amplification. 85(5), 1969–1985.
Brain and Cognition, 112, 39–53. Stöckl, C., & Maass, W. (2021). Optimized spiking
Pinho, A. L., Amadon, A., Fabre, M., Dohmatob, E., neurons can classify images with high accuracy
Denghien, I., Torre, J. J., … Thirion, B. (2021). through temporal coding with two spikes. Nature
Subject-specific segregation of functional territo- Machine Intelligence, 3(3), 230–238.
ries based on deep phenotyping. Human Brain Suzuki, M., & Larkum, M. E. (2020). General anes-
Mapping, 42(4), 841–870. thesia decouples cortical pyramidal neurons. Cell,
Pinho, A. L., Amadon, A., Gauthier, B., Clairis, N., 180(4), 666-676.
Knops, A., Genon, S., … Thirion, B. (2020). Indi- Tavanaei, A., Ghodrati, M., Kheradpisheh, S. R.,
vidual Brain Charting dataset extension, second Masquelier, T., & Maida, A. (2019). Deep learning
release of high-resolution fMRI data for cognitive in spiking neural networks. Neural Networks, 111,
mapping. Scientific Data, 7(1). doi: 10.1038/ 47–63.
s41597-020-00670-4 Tononi, G., Boly, M., Massimini, M., & Koch, C.
Rao, R. P. N., & Ballard, D. H. (1999). Predictive (2016). Integrated information theory: From con-
coding in the visual cortex: A functional interpreta- sciousness to its physical substrate. Nature
tion of some extra-classical receptive-field effects. Reviews: Neuroscience, 17(7), 450–461.
Nature Neuroscience, 2(1), 79–87. Wandell, B. A., Dumoulin, S. O., & Brewer, A. A.
Revina, Y., Petro, L. S., & Muckli, L. (2018). Cortical (2007). Visual field maps in human cortex. Neuron,
feedback signals generalise across different spatial 56(2), 366–383.
frequencies of feedforward inputs. NeuroImage, Wang, X.-J. (1999). Synaptic basis of cortical persis-
180, 280–290. tent activity: The importance of NMDA receptors
Rueckauer, B., Lungu, I.-A., Hu, Y., Pfeiffer, M., & Liu, to working memory. Journal of Neuroscience,
S.-C. (2017). Conversion of continuous-valued 19(21), 9587–9603.
deep networks to efficient event-driven networks Wang, X.-J. (2002). Probabilistic decision making by
for image classification. Frontiers in Neuroscience, slow reverberation in cortical circuits. Neuron,
112. doi: 10.3389/fnins.2017.00682 36(5), 955–968.
532 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Whyte, C. J., & Smith, R. (2021). The predictive spiking neural networks. Frontiers in Neurosci-
global neuronal workspace: A formal active infer- ence, 12. doi: 10.3389/fnins.2018.00987
ence model of visual consciousness. Progress in Zeineh, M. M., Palomero-Gallagher, N., Axer, M.,
Neurobiology, 199. doi: 10.1016/j.pneurobio. Gräβel, D., Goubran, M., Wree, A., … Zilles, K.
2020.101918 (2017). Direct visualization and mapping of the
Wilson, H. R., & Cowan, J. D. (1972). Excitatory and spatial course of fiber tracts at microscopic resolu-
inhibitory interactions in localized populations of tion in the human hippocampus. Cerebral Cortex,
model neurons. Biophysical Journal, 12(1), 1–24. 27(3), 1779–1794.
Wong, K.-F., & Wang, X.-J. (2006). A recurrent network Zenke, F., Bohté, S. M., Clopath, C., Comşa, I. M.,
mechanism of time integration in perceptual deci- Göltz, J., Maass, W., … Goodman, D. F. M. (2021).
sions. Journal of Neuroscience, 26(4), 1314–1328. Visualizing a joint future of neuroscience and neu-
Yin, B., Corradi, F., & Bohté, S. M. (2020). Effective romorphic engineering. Neuron, 109(4),
and efficient computation with multiple-timescale 571–575.
spiking recurrent neural networks. Paper presented Zerlaut, Y., Chemla, S., Chavane, F., & Destexhe, A.
at international conference on neuromorphic sys- (2018). Modeling mesoscopic cortical dynamics
tems. New York: Association for Computing using a mean-field model of conductance-based
Machinery. doi: 10.1145/3407197.3407225 networks of adaptive exponential integrate-and-
Zambrano, D., Nusselder, R., Scholte, H. S., & Bohté, fire neurons. Journal of Computational Neurosci-
S. M. (2019). Sparse computation in adaptive ence, 44(1), 45–61.
32
Advances in Brain Imaging
S i c o n g Tu , W i l l i a m H u y n h , a n d
Matthew C. Kiernan

INTRODUCTION MEASURES OF FUNCTIONAL


BRAIN ACTIVITY
The advancement of cognitive and systems neuro-
science is intricately tied to our ability to capture At the most basic level, consciousness can be
in-vivo brain function in real-time. This can be viewed as the result of billions of neurons firing
achieved through various non-invasive functional rapidly. While non-invasive in-vivo measurement of
neuroimaging methods, including functional mag- individual neuron activity remains a vision of the
netic resonance imaging (fMRI), magnetoencepha- future, current neuroimaging methods provide
lography (MEG), and positron emission increasingly powerful non-invasive methods for
tomography (PET). Traditionally, the goal of func- inferring brain function of neuronal populations
tional neuroimaging has been to localize cognitive through various modalities, the most prevalent being
processes by tracking changes in brain activity fMRI, MEG, and PET. Crucially, each method
through controlled experimental paradigms. By records brain activity in a fundamentally different
modulating brain activity across specific task-based manner, resulting in unique strengths and weak-
conditions, associated neural substrates underlying nesses for inferring underlying brain function.
cognition can be localized and quantified. More
recently, the trend has shifted towards interpreting
neural activity at the systems level across the whole
brain, namely how brain networks facilitate the
mind’s functional states. In contrast to task-based
FUNCTIONAL MAGNETIC RESONANCE
approaches, connectivity of the functional connec- IMAGING (FMRI)
tome is typically studied while the brain is in a
resting-state. In doing so we can map the functional Functional Magnetic Resonance Imaging (fMRI)
organization of neural connections by investigating is a measure of blood oxygenation level-dependent
the association in spontaneous fluctuation between (BOLD) contrast of the brain’s microvasculature.
widespread regions throughout the brain. This chap- Hemoglobin in blood is the body’s primary oxygen-
ter covers advances in localization and connectivity- transport for sustaining metabolic function. The
based brain imaging and current methodology in release of oxygen results in the formation of
fMRI, MEG, and PET. deoxyhemoglobin, a paramagnetic molecule that
534 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

provides a naturally occurring BOLD contrast strength of fMRI relates to the ease of implemen-
which can be accentuated through application of tation and acquisition of data using conventional
gradient-echo MR sequences. The seminal paper MRI scanners and head coil. As such, fMRI can be
by Ogawa et al. (1990) demonstrated that the integrated as part of a seamless research protocol
BOLD contrast can be used to generate real-time with multi-modal weighted sequences (T1, T2, dif-
maps that reflects induced physiological changes fusion, spectroscopy). This facet has made fMRI a
associated with metabolic demand and blood flow. core component of large-scale open access research
Using a standard 3 Tesla (3T) MRI scanner, fMRI initiatives for mapping out the brain’s connectome
allows for very high spatial resolution (∼1-2mm) (Van Essen et al., 2013) and biological factors that
and signal depth, allowing activity to be recorded influence brain structure and function (Miller et al.,
from all cortical and subcortical regions of the 2016), and led to its rapid development and wide-
brain. This can be further enhanced to sub-­ spread application in neuroscience research. Over
millimeter spatial resolution when scanning at the past decade ultra-high field fMRI acquired at
ultra-high field strengths of 7T and above (Kemper 7T has emerged, but this remains a developing
et al., 2018). Change in blood oxygenation is, area constrained by scanner accessibility. Only in
however, a relatively slow biological process, thus the past year has the first 7T MRI scanner been
the temporal resolution of fMRI is typically lim- approved for clinical practice.
ited to differentiating changes in brain activity An early comparison study of the sensitiv-
around one second or more apart. ity of standard 3T and 7T fMRI for detecting
Despite its relatively recent development, fMRI BOLD changes, elicited through simple repetitive
has emerged as the most widely employed func- hand opening/closing, demonstrated a significant
tional brain imaging modality in neuroscience and increase in mean peak signal of the hand knob
provides a good balance between spatial resolution, region of the motor cortex (Beisteiner et al., 2011).
temporal resolution, and signal depth. The greatest The increase in signal sensitivity is the primary

Figure 32.1 Characteristics of the fMRI signal. The BOLD contrast represents the ratio of local
oxygenated (Hb) and deoxygenated (dHb) blood, and typically demonstrates a 3-stage stimulus
evoked hemodynamic response (initial dip, overshoot, and undershoot). Hb = hemoglobin;
dHB = deoxyhemoglobin.
Advances in Brain Imaging 535

benefit of moving to a higher field strength and has research in functional brain networks with pro-
opened the door for more fine-grained localization gressive advances made in both data acquisition
of functional brain changes by reducing voxel and analysis techniques in recent years.
sizes to sub-millimeter resolution. For exam-
ple, advancing our understanding of the laminar
organization of sensory processing (Lawrence
et al., 2019). It is, however, necessary to highlight
that significant technical and practical challenges MAGNETOENCEPHALOGRAPHY (MEG)
currently exist for fMRI acquisition at 7T (and
beyond) besides scanner accessibility. From a MEG is a measure of the magnetic induction gener-
technical standpoint, inhomogeneity of the applied ated by dendritic post-synaptic potential of the
magnetic fields (B0, B1) is significantly more pro- brain’s neuronal activity. Unlike fMRI and PET,
nounced across the brain, leading to greater signal MEG directly measures the net magnetic field
loss in the basal temporal lobe and cerebellum, changes induced by intracellular postsynaptic cur-
as well as increased geometric distortion. From rent flows (excitatory/inhibitory) and does not rely
a practical standpoint, even healthy young adults on secondary effects induced by brain activity. The
report significant subjective discomfort within measured pattern of electrical potential change
a 7T magnetic field resulting in vertigo, and to a resembles that of traditional electroencephalogram
lesser degree, nausea (Rauschenberg et al., 2014). (EEG), as demonstrated by Cohen (1972). MEG
Current non-ferromagnetic medical devices are measures changes in magnetic field which travels at
also not made to be compliant beyond 3T. This the speed of light and suffers minimal spatial blur-
poses challenges for the application of ultra-high ring from passing through distortive tissue (i.e.,
field MRI at both ends of the age spectrum, in skull). This allows for excellent temporal (<1ms) and
early developmental as well as neurodegenerative spatial (∼2-6mm) resolution, with high sensitivity in
research. Technical improvements addressing field the order of 100 femtoTesla (1013 orders smaller than
inhomogeneity are available using dielectric pads a 1.5T MRI scanner). The strength of magnetic field
(passive modulation), or through more advanced changes does, however, decrease rapidly with dis-
solutions such as parallel transmit radiofrequency tance, making recording of neural activity from
coil arrays (active modulation). Participant scan- deep-gray brain structures (i.e., thalamus, hippocam-
ning discomfort remains a challenge, which can pus, amygdala) challenging to implement. Another
significantly impact the quality of acquired data. consideration is that anatomical localization is not
In contrast, MRI at 3T is a mature technology inherent to the acquired MEG signal and requires
that is widely accessible and the current staple for inverse mapping from sensors to source.

Figure 32.2 Characteristics of the MEG signal. Neuronal activity detected by MEG represents
the sum of local magnetic field changes generated by synaptic transmission. Magnetic fields
occur perpendicular to the synaptic potential in accordance with Maxwell’s right-hand rule.
Recorded signal primarily represents local post-synaptic activity. Pre-synaptic potential is
extremely short-lived and biphasic, such that spiking activity would be cancelled out unless
perfectly synchronized.
536 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

MEG was first introduced in 1972 as a highly including perception/memory encoding, and
sensitive magnetic field-based alternative to tra- modulation of attentional states (Lopes da Silva,
ditional EEG for directly measuring the brain’s 2013). Progressive improvements in whole-head
neuronal activity (Cohen, 1972). The seminal coverage, spatial resolution, and prospective head
study by Cohen utilized a novel superconducting position monitoring and correction have also led
quantum interference device (SQUID) to demon- to the direct translation of MEG as a clinical tool
strate that MEG recordings could be directly taken with accepted practice guidelines, notably in epi-
without noise averaging. Modern commercial lepsy for the localization of epileptic foci (Bagic
MEG systems have since undergone substantial et al., 2011) and presurgical functional mapping
technological advances, capable of housing over (Burgess et al., 2011) (albeit EEG remains the
300 SQUID sensors to provide multi-channel standard clinical approach). This is attributed to
whole-brain measurement of neuronal activity at the higher accuracy of source localization of MEG
a temporal resolution of <1ms, sufficient to track over conventional EEG, which would require inva-
even the fastest neuronal processes. Cryogenic sive intracranial monitoring to achieve the same
temperatures are required for the operation of level of accuracy. In principle, MEG is primarily
SQUID sensors, which are housed within a liquid sensitive to cortical sources arranged in a tangen-
helium-filled Dewar surrounding a circular recess tial orientation to the skull with the converse being
to accommodate the participant’s head, typically true for EEG (i.e., sulcal bank and gyral crown,
adjustable for a supine or seated position. In direct respectively). In practice, it is rare to see corti-
contrast to MRI, MEG is a passive and contactless cal activity picked up through EEG but absent on
measurement technique that is silent to operate. MEG, due to its higher sensitivity. Progressive
While MEG is considered less demanding on par- methodological advances in source localization
ticipants relative to the acquisition of other func- and artifact removal in the analysis of MEG data
tional imaging measures (fMRI, PET), whereby have also led to its increasing application beyond
the hardware forms an enclosed space around the the study of more fundamental localized neuro-
whole head, participants are still required to main- physiological phenomena to whole-brain activity
tain a motionless head position and will typically at the network level (Brookes et al., 2011).
have several secondary sensors placed on both
wrists (electrocardiogram) and close to the eyes
(electrooculogram) to control for physiological
artifacts during analysis. Furthermore, comple-
mentary structural MRI is typically also acquired POSITRON EMISSION TOMOGRAPHY (PET)
(independently) for more precise anatomical
co-localization. PET is a measure of metabolic brain activity by
MEG sensors are extremely sensitive to mag- monitoring positron annihilation associated with
netic field changes and capture neural evoked the decay of radiolabeled tracers. Since the incep-
fields as small as 10−14T (the earth’s magnetic field tion of PET imaging in the early 1950s, it has
is approximately 10−4T) and requires installation shown significant clinical application, in particu-
within a heavily magnetic shielded room to pre- lar for localizing and differentiating brain tumors
vent signal contamination from external sources. (Sweet, 1951). PET is a technique for counting
An early combined MEG and subdural EEG study radioisotope decay. Reconstructed images can
by Oishi et al. (2002) demonstrated 3–4cm2 of cor- provide an averaged static image or dynamic
tex is required to be activated for an identifiable image of regional tracer uptake throughout the
spike on MEG recordings. Relative to MRI, MEG brain. Radiolabeled tracers are introduced through
installations are more limited, primarily located in a bolus injection, and comprise a positron emit-
specialized settings, which has been a key factor ting isotope (11C, 13N, 15O, 18F) attached to a mol-
restricting its more widespread adoption clinically ecule of interest (e.g., glucose) to synthesize a
and for research. A second barrier is the relative radioisotope, such as the widely used (18)
lack of standardization in study protocols and F-fluorodeoxyglucose (FDG-PET). Over time, the
analytical methods, which has been a focal point injected tracer accumulates in brain regions of
for the increasing push toward open-source fMRI high molecular affinity and levels can be quanti-
brain connectivity research (Esteban et al., 2019). fied by measuring the resulting photon annihila-
This is despite MEG being a better suited tech- tion associated with positron emission decay. The
nique for directly capturing the underlying tempo- spatial resolution of PET is relatively poor com-
ral characteristics of localized neuronal processes. pared to MRI/MEG and is impacted by both hard-
MEG has provided significant contributions to ware and chemical elements. Resolution will vary
current understanding of the dynamic role of brain depending on the type of tracer used, but typically
oscillations in task-evoked cognitive processes, varies between 4mm and 10mm.
Advances in Brain Imaging 537

Figure 32.3 Characteristics of the PET signal. PET counts the number of positron annihilation
events resulting from the decay of proton-rich radionuclides, resulting in the release of a
positron (e+) and neutrino (v). The emitted positron travels through tissue giving up kinetic
energy by Coulomb interactions with electrons (e−), large deviations in direction occur with
each interaction. Continual loss of kinetic energy ultimately results in a collision leading to
an annihilation event, emitting two anti-parallel 511 keV photons (γ ) detected by PET array
sensors. Localization of the annihilation event is made along the estimated line of response.
P = proton; N = neutron; v = neutrino; e+ = positron; e− = electron.

PET emerged in the 1950s, but its expanded PET installations are now common in clini-
application occurred alongside the development cal settings due to the widespread adoption of
of complementary radiopharmaceutical tracers, FDG-PET as a marker for diagnosis and staging
namely FDG, in the late 1970s. In contrast to in oncology and neurological disorders (Brown
fMRI and MEG, a core component of PET is the et al., 2014; Chen, 2007). However, accessibility
use of radiotracers administered via bolus injec- remains a problem due to high scanning costs,
tion prior to scanning. Methodological advances which can be double that of MRI. The design of
are tied not only to scanning hardware and data conventional PET systems remains fundamen-
analysis, but also progressive radiopharmaceutical tally unchanged (Cho et al., 1976) with sensors
development of novel pre/post-synaptic markers arranged in a cylindrical array for the detection of
for molecular targets of interest (e.g., neurotrans- positron annihilation events along a 180-degree
mitter systems, metabolism, inflammation, pro- line of response. Over the past two decades, the
teinopathies). The development, generational type of sensors used within the array has, however,
improvements, and validation of radiotracers is an increasingly transitioned from traditional bismuth
extensive field that falls outside the scope of the germanate (BGO) scintillators coupled to photo-
current chapter. Radiotracer reviews are plentiful, multiplier tubes, to the more recently developed
but typically disease and/or mechanism specific lutetium-based scintillators read out by silicon
(cf. van Waarde et al., 2021). photomultipliers (SiPMs). SiPMs not only provide
538 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

a high light-collecting efficiency and comparable ADVANCES IN FMRI


stopping power to BGO, but also significantly
improved coincident timing resolution (200–
400ps), allowing time-of-flight (TOF) PET imag- Multiband Acquisition
ing for more precise signal localization (Surti,
2015). Currently, lutetium-based scintillators are Virtually all fMRI acquisitions employ echo
found in all modern state-of-the-art PET scanners planar imaging (EPI) (Mansfield, 1977) for spatial
offered by mainstream commercial manufacturers. encoding of the MR image due to its fast scan
PET imaging is intrinsically complementary time (∼3 seconds for whole brain coverage) and
with MRI (albeit primarily used in combination BOLD sensitivity (Ogawa et al., 1990). Ideally,
with computed tomography [CT]), yielding addi- only localized intensity changes relating to neu-
tional insights of specific molecular mechanisms ronal activity should be detected. In practice, the
impacting structural, functional, and connectivity image-to-image signal is contaminated by multi-
brain changes. This has been complemented by ple sources, namely system instability, subject
recent hardware advances leading to the devel- movement, and physiological motion (Hu et al.,
opment of hybrid PET-MRI scanners for simul- 1995). Improving the temporal resolution (the rate
taneous image acquisition (Catana, 2017). An at which sequential brain images are acquired) is
advantage of integrated PET-MRI systems is the critical for reducing temporal instabilities.
improved spatial correspondence between PET Conventional fMRI data is acquired slice-by-slice,
and MR images due to the inherent co-registration meaning that whole brain data is acquired sequen-
carried out during data acquisition. Quantification tially in a slab like manner. Such an approach
of PET tracer tissue uptake can be more reliably poses clear limitations for the degree of accelera-
calculated by leveraging the higher spatial reso- tion possible for acquisition. Over the past decade,
lution of structural MRI images (1mm) to reduce simultaneous multi-slice (or multiband) EPI
partial volume effects (i.e., tissue heterogeneity sequences have been increasingly adopted as a
due to a mixture of gray and white matter phar- novel approach to improve temporal resolution to
macokinetics), a significant limitation affect- achieve 1s and even sub-second whole brain cov-
ing the reliability of quantifying tracer uptake erage (Feinberg et al., 2010).
in subcortical brain structures (Brown et al., Multiband acquisition leverages modern MRI
2014; Catana et al., 2012; Jucaite et al., 2012). hardware to simultaneously measure signal
The combined application of PET and MRI has from multiple detector coils simultaneously and
been widely employed across neurodegenerative separate overlapping images into their separate
conditions in support of the “network-opathy” slices (un-aliasing) through multi-channel radi-
hypothesis for progressive propagation of dis- ofrequency head coils (32 channels and higher;
ease pathology in the brain (Seeley et al., 2009). Larkman et al., 2001). The degree of multiband
Studies in Alzheimer’s disease have consistently acceleration implemented can be as low as 2 (i.e.,
demonstrated an association between altered con- acquiring 2 simultaneous brain slices) to 8, as
nectivity of large scale rsfMRI brain networks implemented by large scale neuroimaging initia-
and underlying disease burden as reflected by tives (Human Connectome Project; UK Biobank;
PET tracers of b-amyloid (Drzezga et al., 2011), Miller et al., 2016; Van Essen et al., 2013). The
and more recently, tau (Franzmeier et al., 2020) primary benefit of implementing multiband accel-
proteinopathies. Relative to fMRI, PET imaging eration is the reduction in repetition time (TR),
data of brain function is both temporally and spa- which is the amount of time taken to acquire a
tially limited. Functional connectivity between single whole-brain volume. This results in two
brain structures through standalone PET imaging main advantages for the acquired data: (i) wider
(i.e., metabolic connectivity) can only be derived range of signal sampling to identify physiological
through significant correlations at the group level, noise, (ii) greater number of sampled time-points
not dynamic signal fluctuations at the subject level for improved statistical power (i.e., a multiband
(fMRI, MEG). Methodological advances in PET factor of 8 is an 8-fold increase in number of col-
imaging are ongoing, aimed at improving data lected volumes for a given acquisition period). As
reconstruction for localization while optimizing with all MRI techniques, employing multiband
for spatial and temporal resolution. EPI sequences does come at a cost. A shorter
Each of the imaging modalities outlined above TR results in brain tissue being excited in a more
can be applied to examine changes in local and rapid succession allowing less recovery time. This
global “brain activity.” Their underlying princi- translates into reduced gray and white matter tis-
ples, however, vary greatly and so too does their sue contrast, which can be addressed with the
inherent strengths and weaknesses which impacts additional acquisition of a single-band reference
experimental design and interpretation of results. volume. Multiband data also suffers from more
Advances in Brain Imaging 539

Figure 32.4 Simultaneous multi-slice (multiband) acquisition. Data is acquired from multiple
slices simultaneously rather than sequential slice-by-slice acquisition.

artifacts relative to conventional data. This is removal of signal drift and high frequency noise,
exacerbated at higher multiband acceleration fac- and volume deletion. Despite the significant data
tors (i.e., 8) but allows fMRI data to be acquired reduction imposed by these steps it is clear that
at 2mm isotropic spatial resolution with ∼1s TR at systematic but spurious correlations still arise
3T field strength (Miller et al., 2016; Van Essen throughout the brain (Power et al., 2012). An
et al., 2013). In contrast, conventional fMRI data approach that has a long theoretical history but is
is acquired at 2.5–3.5mm isotropic voxels with only recently starting to gain popularity in fMRI
∼3s TR. More recently, it has been shown that high acquisition is the application of multi-echo EPI
multiband acceleration also poses challenges for sequences.
quantifying functional connectivity. Noise ampli- Echo time (TE) is the time gap between the
fication from multiband acceleration creates large excitation of brain tissue and read out of the result-
spatial biases (particularly at 8 and higher) which ing signal. The optimal echo time varies accord-
can lead to an underestimation of subcortical con- ing to tissue type, biological process of interest,
nectivity (Risk et al., 2021). Notably, multiband and scanner field strength. For the BOLD effect
acceleration factors of 3-4 provide significant in gray matter at 3T, 30ms is typically used.
benefits to the spatial and temporal resolution of Conventional EPI sequences use a single echo
fMRI data (2.5mm isotropic; ∼1s TR) and argu- time, however, multiple echo times (i.e., additional
ably a more balanced tradeoff between spatial signal read outs at shorter/longer delays resulting
biases and effect size when examining the correla- in n>1 volumes per TR) is proposed as a method
tion in subcortical-cortical (e.g., thalamocortical) to separate BOLD from non-BOLD signal to bet-
connectivity. ter clean fMRI data to improve statistical power
(Kundu et al., 2017). This follows the observa-
tion of TE-dependent scaling of the percent signal
change associated with the BOLD signal compo-
nent across both task (Lombardo et al., 2016) and
MULTI-ECHO ACQUISITION resting-sate (Kundu et al., 2013) fMRI. In contrast,
non-BOLD components show TE-independence
As previously raised, the observed BOLD signal remaining relatively similar across varying TE.
from fMRI data is impacted to varying degrees by The penalty associated with employing multi-
artifacts relating to subject and physiological echo acquisition is, understandably, an increase
motion, and hardware. The control and removal of in TR (to accommodate multiple-signal read outs)
structured noise from fMRI data is a critical relative to single-echo acquisition, regardless of
aspect of the analysis pipeline. To date, this has whether it is paired with multiband acquisition.
largely been addressed during the preprocessing Whether multi-echo acquisition is a better choice
stage involving regression of structured noise, overall depends on how well artifact removal from
frequency-restricting temporal filtering for the fMRI data can be achieved.
540 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

INDEPENDENT COMPONENTS of changes in the spatial organization of systems-


ANALYSIS (ICA) level dysfunction across clinical populations, such
as major depressive disorder (Hamilton et al.,
2015) and Alzheimer’s disease (Greicius et al.,
The estimation and localization of the spatial dis- 2004). The independent nature of components
tribution of BOLD changes at the voxel level has identified by ICA, however, can be a disadvantage
been fundamental to modern cognitive neurosci- to understanding the “true” network structure of
ence. Traditionally, fMRI has been used in combi- underlying brain networks. For example, in data
nation with tightly controlled task-based paradigms sets with very little noise, partially overlapping
to investigate hypotheses for identifying localized components would be spatially correlated and
functional contributions of specific brain struc- therefore not allowed as independent components.
tures in cognitive processing. This has led to the This has a significant impact for evaluating the
application of signal processing approaches to functional role of key subcortical structures (e.g.,
decoding localized mental representations, such thalamus) that play an active role in the relay and
as multivariate pattern analysis, whereby machine modulation of brain signaling to widespread corti-
learning is used to train a classifier to recognize cal regions. It is also difficult to disentangle inter/
dynamic patterns of BOLD change, most interest- intra-hemispheric in brain networks differences
ingly in the memory literature (Chen et al., 2017; given the strong correlation between correspond-
Polyn et al., 2005). Increasingly, however, a ing brain regions across hemispheres.
­systems-level approach is being adopted as we
recognize it is the communication, modulation,
and integration of sensory and higher order neural
signals across interconnected networks of brain
structures that underlies everyday cognition and NODE BASED ANALYSIS
behavior, and their dysfunction. In combination
with large-scale brain imaging initiatives (Miller While ICA is a powerful technique for comparing
et al., 2016; Van Essen et al., 2013) that have made the spatial organization of large-scale brain net-
high quality neuroimaging data sets publicly works, it does not allow investigation of the
available, data-driven approaches for mapping strength of functional connections between two
functional brain networks and their connectivity, regions or their comparison at a group level. For
such as independent components analysis (ICA), these questions node-based connectivity modeling
has pervaded the neuroscience literature. is a widespread approach increasingly employed
ICA is a model free data analysis approach in cognitive and systems neuroscience. The pri-
for decomposing a multivariate signal, such as mary difference between voxel-based approaches
fMRI acquired while the brain is in a “resting- (e.g., ICA) and node-based analysis of rsfMRI
state” (rsfMRI), into independent components relates to spatial scale. Node-based analysis
with unique sets of structured features that can be requires a-priori definition of “nodes” as different
applied during pre-processing to identify noise, brain regions of interest. These nodes can: (i)
as well as post-processing to identify large scale cover the whole brain and be defined by brain
brain networks. This follows the assumption that, atlas parcelations (structural/functional), (ii) cover
at rest, brain regions that share a similar function limited brain regions of a specific system of inter-
demonstrate similar dynamics in spontaneous neu- est, (iii) be used in conjunction with seed-based
ronal activity. ICA results in the decomposition of correlation maps to define regions connected with
unique spatial and/or temporal components (i.e., a specific brain structure. In essence a node
no correlation or higher order statistical relation- assigns labels to groups of voxels considered
ship) that are maximally independent of each other functionally homogenous. For each node, the
based on a cost function, such as measuring nega- functional connectivity (i.e., similarity in the
tive entropy, to determine the best set of linear com- rsfMRI timecourse) between all other nodes is
ponents that are maximally non-­Gaussian from the calculated as “edges” to generate a network
full rsfMRI data. The advantage of using ICA over matrix of pair-wise connections that can be com-
traditional seed-based (hypothesis driven) correla- pared at the group level. How an edge is defined
tion analysis is estimation of the full (e.g., spatial) varies greatly according to the research question
structure of a brain network. Rather than being and directly impacts interpretation of connectivity.
sensitive to the functional connectivity pattern of At its most basic form an edge between each pair
a specified seed region, ICA simultaneously char- of nodes can be binary (i.e., 0 or 1) to simply
acterizes multiple independent resting-state brain represent whether a connection is present or
networks, as captured by the data set. ICA derived absent, for a given statistical threshold. A range
brain network maps have advanced understanding between +1 and -1 is more typically used to pro-
Advances in Brain Imaging 541

solving this problem requires a-­priori knowledge


of the currents and magnetic field, while consider-
ing volume geometry and properties of the
medium. This is, however, an ill-posed problem as
the measured magnetic fields could be caused by
an infinite number of neuronal sources, or at least
more than available sensors. As such, there is no
unique solution. This is reflected by the absence
of a general model for source reconstruction with
Figure 32.5 Data-driven parcellation multiple approaches employed in the literature.
of rsfMRI data captures the whole-brain Overall, reconstruction models demonstrate good
spatial structure of large-scale brain consistency but differ in the degree of complexity
of the modeled source and imposed physiological
networks. Number of independent brain
assumptions, which impacts the spatial extent of
networks identified from a dataset is source images and subsequent statistical inference
variable depending on analysis parameters. (Samuelsson et al., 2021).
Parcellation masks are publicly available The equivalent current dipole (ECD) method is
and were generated from a data set of 1000 the simplest and most widely used estimation tech-
healthy young adults (Yeo et al., 2011). nique due to its relatively high accuracy and prac-
ticality, particularly in a clinical setting (Burgess
et al., 2011). Dipole fitting assumes that a small
vide more meaningful information, namely number of point-like ECDs can describe the spa-
the strength and relationship (correlation or anti-­ tial average of all evoked currents within a local-
correlation, respectively) of the connection ized area of the brain. Each dipole is characterized
between brain regions. Node-based analysis has by six parameters that describes its position, ori-
played an important role in mapping the func- entation, and strength within the head volume.
tional connectome and topographical organization Comparisons are performed between measured
of the human brain (Yeo et al., 2011). The primary and modeled field distributions using a non-linear
limitation of node-based analysis is the a-priori iterative algorithm to determine parameters of
definition of nodes that maintain the same shape best fit according to the measured data. At each
and size throughout downstream analysis stages iterative step, parameters are adjusted to minimize
when gradient-based boundaries are arguably measured and modeled field differences based on
more appropriate for parcellating cortical brain a cost function. Different conductor models can be
regions (Glasser et al., 2016). If the spatial bound- applied to approximate head volume, including a
aries of nodes do not accurately reflect the under- single homogenous sphere or ellipsoid, multiple
lying functional organization of the brain it is spheres fitted according to head curvature (bound-
difficult to reproduce connectivity patterns present ary elements), or in combination with structural
at the voxel scale (Craddock et al., 2012). MRI. The ECD method is not a perfect representa-
tion of all brain activity but provides a good esti-
mation of early cortical activity that involves only
a few square centimeters of cortex.
An increasingly popular alternative technique
ADVANCES IN MEG for extracting the origins of a MEG signal is to use
beamforming. Beamforming approaches extract
Source Localization signal from a prespecified spatial location through
the use of an adaptive spatial filter (Woolrich et al.,
A central problem that lacks a general solution in 2011). An optimal spatial filter estimates the neu-
MEG is source localization. Localization of the ral activation at a given location independent (i.e.,
MEG signal requires inverse mapping from sen- uncorrelated) of those generated elsewhere in the
sors to source as the acquired raw data does not brain. In essence, beamformers do not attempt to
hold inherent spatial information. Every source explain the complete measured field, rather what
estimation procedure faces the same “inverse is the unique contribution of a single position to
problem,” that is the forward modeling of the the field. A distinct advantage of beamforming is
estimated current distribution within the brain its ability to suppress artifactual signal (e.g., car-
from the net observed magnetic field measured diac interference) whose spatial profile cannot be
externally (as an input) into three-dimensional modeled by a dipolar source within the head. This,
space (i.e., along the cortical surface or within the however, does raise a fundamental problem with
brain volume; McMackin et al., 2019). Conceptually, beamforming approaches as correlated sources
542 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 32.6 Source localization using MEG and structural MRI. (A) MRI geometry is used
for building biophysical models of the brain to account for differences in tissue properties.
(B) Localized neuronal activity can be projected between sensor and source space through
inverse and forward modeling (McMackin et al., 2019).

cancel each other out depending on the correlation ARTIFACT REMOVAL


coefficient (i.e., spatially separate but temporally
correlated sources are suppressed), with fully cor-
related sources being undetectable. Beamforming The high sensitivity of MEG is a double-edged
is therefore not appropriate for certain experimen- sword and, like all functional brain imaging meas-
tal MEG paradigms, for example signal locali- ures, is highly susceptible to a variety of physio-
zation in bilateral auditory cortices following logical and environmental induced artifacts.
binaural stimuli (Wipf et al., 2010). Suppression Artifactual sources present in MEG arise from the
of correlated sources may not be a physiologically same sources as those present in EEG, including
justifiable approach for studying brain activity, movement, external electrical apparatus, as well
however, beamformer approaches have increas- as cardiac and muscle interference. The control
ingly demonstrated sufficient robustness for deriv- and removal of artifacts is not trivial and exacer-
ing large scale resting-state brain networks from bated by elevated physiological responses, such as
MEG (Brookes et al., 2011). MEG derived resting- heartbeat and eyeblink, the two most dominant
state brain networks show a remarkably similar sources of noise. Control measures currently applied
spatial structure to rsfMRI and have been used as during data acquisition typically include compensa-
evidence of a neural oscillatory basis for BOLD tory real-time noise cancelation through active field
functional connectivity (Brookes et al., 2011). coils to counteract residual environmental
Advances in Brain Imaging 543

magnetic interference, simultaneous secondary NETWORK CONNECTIVITY


physiological recordings of eyeblink (electroocu-
logram) and heartbeat (electrocardiogram), and The derivation of functional brain networks and
continuous head position monitoring. their statistical comparisons are not unique across
Physiological artifacts resulting from oculomo- functional brain imaging modalities. Large-scale
tor muscle movement during routine blinking, and brain networks in MEG can be generated using
electrical current induced magnetic fields associ- the same data-driven ICA approach used in fMRI
ated with a heartbeat, are two inherent sources of with considerable overlapping spatial structure of
noise present in the recorded MEG timecourse correlated spontaneous fluctuations in neuronal
(Hari, 2011). For spontaneous eyeblink, the arti- activity at rest (Brookes et al., 2011), or using a
fact timecourse corresponds with eyelid motion node-based hypothesis driven approach (Stam
and demonstrates a structured pattern, on average et al., 2009). However, the type of signal charac-
120ms after initiation and after returning to a fully teristics and post-processing steps applied to the
open state. Cardiac related artifacts demonstrate a acquired data to measure the degree of functional
relatively less structured pattern and are observable connectivity between brain regions differs signifi-
to varying degrees in MEG recordings resembling cantly across imaging modalities. The MEG
epileptic spikes and slow waves (Hari, 2011). The signal primarily represents the net magnetic field
timecourse corresponds to the propagation of heart changes generated by postsynaptic currents of
muscle electrical activity passing from the atria pyramidal neurons but is in essence a compound
through the ventricles as measured by electrocar- signal, comprising multiple sources superim-
diogram (i.e., P wave, QRS complex, T wave). posed. Given its extremely high temporal resolu-
Various approaches are suggested for denoising tion, each sensor captures the oscillatory activity
physiological artifacts from MEG recordings, of local neuronal populations, such that functional
the most straightforward (and long-standing) connectivity is determined by how closely neu-
approach being epoch rejection, which involves ronal populations oscillate in synchrony (i.e.,
discarding raw data within specific time-windows amplitude and phase-coupling). This follows the
from the continuous data with high artifactual influential “communication through coherence”
contamination. This is a time-consuming process hypothesis (Fries, 2005), which proposes a mech-
that is highly operator-dependent and can result in anistic principle whereby coherently oscillating
significant data loss in cases of widespread epoch neuronal groups reach peak inter-communication
rejection. Alternatively, regression of artifacts by efficiency if they share a similar timecourse for
projecting secondary physiological measures (i.e., maximum/minimum spiking threshold for send-
electrooculogram) is a widely applied approach ing and receiving a barrage of action potentials. It
for denoising the data, particularly for subtract- is important to note that synchrony does not
ing ocular related signal from the recorded data. reflect real information exchange, rather it reflects
More recently, development has focused on blind the relative conditions best suited to allow effi-
source separation methods, specifically con- cient physiological information exchange between
strained blind source separation (cBSS), to allow different neuronal populations. Neuronal oscilla-
for more automated and objective denoising tions are measured in a range from around 0.05Hz
using ICA (Escudero et al., 2011). As described to 500Hz and commonly classified into five fre-
earlier in the chapter, ICA is a model free data- quency bands (i.e., δ [2–4], θ [4–7], α [8–12], β
driven approach for decomposing a multivariate [12–30], γ [30–80]) that demonstrate marked top-
source signal into its independent linear compo- ological differences in neuronal networks. High
nents. Blind source separation methods isolate frequency bands are characteristic of smaller spa-
artifactual signal sources contaminating the full tial scale brain networks, and low frequency bands
timecourse, however, visual inspection is still for large-scale networks (Brookes et al., 2011;
required for identification. A direct extension Florin and Baillet, 2015). Coupled oscillations are
to blind source separation is cBSS, which intro- also not limited to within the brain, but also
duces the use of statistical metrics (e.g., kurtosis, extend between brain and body (cardiac, gastric,
entropy) to define criteria for the identification of muscle) activity rhythms (Klimesch, 2018).
artifactual source components. This can be aided For estimating functional connectivity between
by the inclusion of simultaneous secondary phys- two regions with MEG, two types of measures are
iological recordings to improve ocular and car- commonly used, coherence (for linear relation-
diac artifact identification (Dammers et al., 2008; ships), and phase locking (for non-linear relation-
Klados et al., 2011), which have been shown to ships). Coherence measures the degree of similarity
be practical for artifact removal from real world between two timeseries and is the equivalent of a
MEG recordings (Escudero et al., 2011). cross-correlation function for the frequency domain,
544 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 32.7. Characteristics of high and low synchronous signals in MEG. Neural oscillations
(i.e., brain waves) are sinusoidal, following a repeating rhythmic pattern demonstrating
frequency fluctuations in terms of amplitude and phase. Complex changes over time can be
depicted as changing points on a 2D cartesian co-ordinate system to visualize the spectral
representation of individual MEG signals, or the complex conjugation of the frequency
of two signals (i.e., cross-spectral density [CSD]). Connectivity can be conceptualized as
the synchronicity of frequency fluctuations of MEG signals over time, represented by the
consistency of CSD across observations. Coherence is calculated as the weighted sum of CSD
across observations as a measure of connectivity. Phase locking value is conceptually similar,
but only considers the consistency of phase differences, disregarding amplitude correlations
of the MEG signal.

representing a normalized coefficient between 0 estimating functional connectivity in MEG is not


and 1 that quantifies the amount of variance that clear cut when they in fact provide complementary
can be explained between two signals. Yet, recorded information on the dynamic nature of brain func-
neuronal activity more closely resembles noisy tion. In particular, their combined application better
deterministic patterns of chaotic activity, particu- encapsulate linear and non-linear interdependen-
larly for sensory neural systems, which may actu- cies of cortical dysfunction in patient populations
ally contribute to enhance functionality (Freeman, (Sakkalis et al., 2009).
1987). The intricate complexity of feed-forward
and feed-back mechanisms in the brain means that
activity at the level of neural synapses is non-linear
with high variability in short term dynamics associ-
ated with neuromodulation processes. To this end, ADVANCES IN PET
phase locking provides a non-linear measure of
the temporal relationship between two timeseries Time-of-Flight (TOF) PET
independent of their signal amplitude and is argu-
ably better suited for measuring functional connec- PET is a radioisotope decay counting technique
tivity with MEG. In practice, however, non-linear whereby the signal is produced by the time-­
measures are highly susceptible to noise and dem- coincidence detection of two 511-keV photons
onstrate less robust performance relative to linear emitted almost back-to-back along a line of response
measures in real world application. The case for (LOR) and picked up by surrounding sensor arrays.
using coherence or phase locking measures for best The role of the sensor is to capture the trajectory and
Advances in Brain Imaging 545

timing of released photons to reconstruct the origin Acquisition protocols used for PET imaging
of the emission along the LOR. In theory, the differ- varies greatly depending on application. The data
ence in arrival time of the emitted pair of photons acquired by PET imaging represents the detection
can be used to precisely localize where along the counts of coincident timed positron annihilation
LOR emission occurred, otherwise known as TOF events over time. Statistical accuracy is primar-
PET, a concept considered since the early days of ily determined by the number of acquired counts,
PET imaging. In practice, however, traditional BGO which in turn is constrained by administered tracer
scintillators cannot resolve the time difference in radiation dose and desired temporal resolution.
detection necessary for the precision of TOF meas- Clinical protocols (primarily for FDG) favor brief
urements (< 2ns). Instead, emission location is scans (∼10–20min) after a set post-injection delay,
reconstructed by collecting all possible LORs meas- pre-determined by validated intervals for greatest
ured around an object and a uniform probability of sensitivity depending on tissue and/or region of
the event location is assumed along the full LOR interest, to reconstruct “static” images averaging
length. Noise correlations from different emission all event counts. The primary benefit of a static
events significantly increases the uncertainty of imaging approach is fast data acquisition with low
localization. Early alternatives to BGO detectors noise to generate a spatial distribution map of radi-
with significantly greater timing gain were consid- oactivity in the brain (with no temporal informa-
ered (BaF2; CsF) but ultimately rejected due to poor tion) that can be used for qualitative assessment or
mechanical properties (i.e., low stopping power, simple dimensionless region of interest standard
light output, attenuation) resulting in overall worse uptake value (SUV) comparisons (i.e., “normal”
spatial resolution due to reduced detection sensitiv- or “abnormal”). Static PET imaging is simple to
ity that offset any effective gains provided by TOF acquire, standardize, interpret, and provides clini-
(Lewellen, 1998). This changed with the develop- cally meaningful biological information. A sig-
ment of SiPMs and led to the reintroduction of TOF nificant confound for such an approach, however,
PET in the late 2000s. With TOF PET, the emission is the presence of inter-individual differences in
point along a LOR can be directly determined (with tracer delivery or flow effects resulting in variable
a degree of uncertainty) by measuring the difference values that can lower qualitative and quantitative
in arrival times of the emitted photons. The tech- tracer sensitivity (Gallezot et al., 2020).
nique has been shown to significantly improve An alternative approach is “dynamic” imaging
image quality and/or reduce image acquisition time whereby a time series of images are acquired over
for low-count clinical imaging applications (Surti, an extended duration (∼60min) to better capture
2015). The use of increasingly fast timing detectors the time-activity curve of tracer concentration.
is now an integral component of modern PET imag- Dynamic imaging is necessary for kinetic mod-
ing as developments such as SiPMs have shown that eling and extraction of quantitative features hid-
increased timing performance does not need to come den from visual inspection and semi-quantitative
at a trade-off with sensitivity and spatial resolution. clinical parameters (i.e., SUV). Dynamic PET also
allows for more advanced research applications
such as investigating real-time neurotransmitter
receptor release/occupancy (Wey et al., 2014).
This comes at the cost of significantly longer
KINETIC MODELING scanning time, increased motion susceptibility,
and noisier data, requiring more advanced models
The goal of PET imaging is to measure a selected for reconstruction, but in most cases significantly
in-vivo physiological parameter of interest (e.g., improves sensitivity and specificity of focal tracer
glucose metabolism, enzyme activity, receptor/ uptake (Gallezot et al., 2020). The increased noise
transporter concentration, hypoxia). Injected radi- contained within individual voxels of dynamic
otracers are distributed through arterial blood to PET images is one of the main limitations that
allow binding with tissue in brain regions of high needs to be addressed with image reconstruction
molecular affinity. Direct measurements of radi- algorithms (e.g., Gaussian smoothing for spatial
otracer concentration from raw PET images are, smoothing; principal components analysis for
however, not representative (or even directly pro- temporal smoothing) to facilitate clinical adoption
portional) to underlying physiological function beyond research purposes. A variety of models
(Gallezot et al., 2020). Reconstruction algorithms can be used for estimating the time-varying tissue
are required to accurately model confounding concentration of a tracer, broadly categorized into
kinetic parameters, notably, tracer concentration in three categories: compartmental models, graphi-
blood plasma, motion (head, cardiac, respiratory), cal analyses, and spectral analyses (Gallezot
and noise, for meaningful interpretation of quantita- et al., 2020). Compartmental models are the
tive tracer uptake values. most widely used approach, defining one or more
546 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 32.8 Standard uptake value images of serotonin radiotracer (11)C-AFM. Averaged
uptake across brain regions at different post-injection time periods, over time tracer clears
from regions with low binding affinity. Kinetic modeling can be applied to calculate and
separate flow effects associated with tracer delivery to better identify sites of focal uptake,
such as the raphe nucleus, a key midbrain site of serotonergic fibers (Gallezot et al., 2020).

compartments, each with an associated kinetic rate of oxygen for functional brain imaging that has
flow rate, to account for any individual state (i.e., been phased out by fMRI, given its higher spatial
physical environment: plasma, tissue; binding sta- and temporal resolution, and lack of radiation
tus: free, specific, non-specific; chemical form: exposure. Nevertheless, 15O-PET has the distinct
pure tracer, metabolite) a tracer can assume. advantage of being able to separate and account for
The primary consideration for deciding between confounding vascular effects (blood flow/volume)
a static versus dynamic protocol is acquisition associated with brain oxygenation, and potentially
time and repeated blood sampling (although see better suited as a reliable inter-­individual physio-
Gallezot et al., 2020). A dynamic acquisition is logical marker of underlying brain “activation,”
always required for quantitative PET metrics and particularly in the context of longitudinal measure-
can also be averaged over specific time frames to ment, which remains a significant challenge for
generate a “static” weighted average if required. fMRI. It is notable that the “default mode network,”
a large-scale brain network continuously engaged
in the absence of external stimuli or conscious
engagement and widely popularized by rsfMRI
studies in cognition and neurological disorders
METABOLIC CONNECTIVITY (Buckner et al., 2008), was originally conceptual-
ized and characterized through 15O-PET imaging
Functional connectivity, or more accurately meta- (Raichle et al., 2001; Shulman et al., 1997).
bolic connectivity, derived from PET is fundamen- As mentioned earlier, the analysis methods used
tally quite different from fMRI and MEG. The to analyze network connectivity is not unique to
limited temporal resolution of conventional static specific functional brain imaging modalities and
PET imaging results in images with a steady-state the same holds true for PET imaging data, which
signal, typically resulting in a single representative draws on general signal processing approaches
image per subject, a stark contrast to the dynamic such as data-driven principal components analy-
signal variation present in the rich MRI/MEG data sis and ICA (Pagani et al., 2016; Tripathi et al.,
timeseries. Consequently, connectivity between 2016), and node-based approaches such as inter-
two regions depends on the significance of their regional correlation analysis and sparse inverse
correlation across subjects at a group level (i.e., not covariance estimation (Arthuis et al., 2015; Zou
spatial patterns of temporal activation per subject). et al., 2015). The bulk of connectivity stud-
The derived connectivity from PET does, however, ies published in the literature is based on static
provide a more specific indicator of the physiologi- FDG-PET, which typically relies on intensity-
cal relationship between brain regions. For exam- based analysis approaches, following image
ple, (15)oxygen (15O) labeled PET imaging is an reconstruction, similar to structural MRI. The
alternative method for quantifying the metabolic advantage of assessing network connectivity with
Advances in Brain Imaging 547

FDG-PET is the additional inferences that can be (ii) exploring regional neurotransmission/neurovas-
drawn, for example, between the reorganization cular coupling, supporting a divergent association
of local and long-distance energy consumption for experienced as compared with observed pain
and pathogenesis of disease in neurodegenerative (Karjalainen et al., 2017; Wey et al., 2014), and a
disorders (Sala et al., 2017). Of greater interest dissociation for placebo analgesic drug paradigms
are increasing studies employing dynamic PET/ (Linnman et al., 2018). The equivalent simultane-
MRI connectivity to advance our understanding ous acquisition of MEG and fMRI data is not tech-
of the physiological basis for network changes nically possible (except for EEG/fMRI). However,
observed in rsfMRI (Passow et al., 2015; Tomasi the analysis of their combined data, collected
et al., 2017). Traditionally, dynamic PET data through separate experimental sessions under simi-
is used to extract kinetic parameters, but with lar task conditions, is possible and can be per-
respect to the association of time-varying effects formed in a common domain through techniques
with rsfMRI, more subtle variations in the tracer such as representational similarity analysis (Cichy
timecourse are of interest. There does remain a and Oliva, 2020; Kriegeskorte and Kievit, 2013).
significant difference in the timescale between The framework only considers the timing of neural
PET and fMRI (minutes vs. seconds), leading to activity from MEG, and space from fMRI, estab-
reported mismatches between outcome measures lishing a representational link between spatial and
of metabolic and functional connectivity patterns temporal domains for a given study condition. Such
that requires methodological advances in time- an approach constrains the experimental design,
varying kinetic modeling to resolve (Tomasi et al., including the optimization of the same experimen-
2017). Development of dynamic PET/MRI meth- tal condition for both MEG and fMRI, ensuring a
odology opens new avenues of research inquiry, high signal can be detected within each imaging
such as pharmacological modulation of neural modality. A fundamental limitation is that the
receptor occupancy to assess neurovascular cou- recorded neural activity is not concurrent and origi-
pling mechanisms with hemodynamic change nates from separate recordings which will include
across brain networks, and cognition. Growing nuisance variability. Nevertheless, the synthesis of
developments in this area are a good example of MEG and fMRI data for reducing their respective
how methodological advances in functional brain spatial and temporal limitations has resulted in a
imaging goes hand-in-hand with advancing cogni- number of well-constructed paradigms examining
tive and systems neuroscience. underlying spatiotemporal dynamics of object,
scene, and higher order attention processing (Cichy
et al., 2014; Hebart et al., 2018; Henriksson et al.,
2019). Development of novel analytical frame-
works for multi-modal imaging are ongoing, aided
FUTURE DIRECTIONS through open-source datasets of combined fMRI,
MEG, and PET imaging data (Jamadar et al., 2020;
Multimodal Brain Imaging Schoffelen et al., 2019).

Multimodal brain imaging is increasingly recog-


nized as a necessary step for advancing cognitive
and systems neuroscience to overcome the inherent
limitations underlying traditional functional brain MAGNETIC RESONANCE SPECTROSCOPY
imaging techniques. The impact of hardware devel-
opments such as hybrid PET/MRI are already evi- While not covered in this chapter, another func-
dent, with studies advancing current understanding tional MRI modality that has significant potential
of neuroreceptor function across dopamine, seroto- for characterizing the neuromodulatory influences
nin, and opioid primary neurotransmitter systems of biological mechanisms underlying cognition,
in the context of behavioral modulation (Sander is magnetic resonance spectroscopy. MRS is a
et al., 2020). Studies have employed this approach non-invasive technique for quantifying tissue
for examining neural mechanisms of pain percep- metabolite concentrations derived through differ-
tion through simultaneous, rather than separate, ences in the resonance frequency exerted by the
data acquisition (Karjalainen et al., 2017; Linnman nucleus structure of constituent molecules includ-
et al., 2018; Wey et al., 2014). The simultaneous ing hydrogen (1H), phosphorus (31P), fluorine
nature of the data acquisition allowed accurate co- (19F), and carbon (13C). 1H-MRS is by far the most
localization of PET and fMRI signal change in a prevalent form of MRS reported in the literature
common domain for: (i) characterization of an and can be acquired using a conventional head
extended cortical/subcortical network of pain pro- coil as an additional sequence in a standard MR
cessing structures (Wey et al., 2014), and protocol. Clinically, it is widely used for the
548 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

identification, grading, and evaluation of tumour the increasing application of machine learning
subtypes to inform diagnosis and treatment. In based image classification algorithms, specifically
research, it has been widely used to quantify deep learning neural networks, being developed for
abnormalities in steady-state metabolite concen- neuroimaging (Ronneberger et al., 2015). In
trations in patient populations to infer pathomech- essence, neural networks specify an architecture for
anisms of disease, and also in combination with extracting features contained within a dataset that is
task-based fMRI studies of basic visual and refined and modeled with increasing complexity
motor, and higher order cognitive processing, to through successive “layers” to learn, and conse-
infer the influence of localized excitation-­ quently, allow automated recognition and classifica-
inhibition balance on neuronal activity, although tion of prospective data, according to modeled
with variable findings (Kiemes et al., 2021). features. To date, neural networks have demon-
A key consideration for brain MRS is relatively strated remarkable accuracy and, more importantly,
low concentrations of the metabolites of great- generalizability for structural neuroimaging, facili-
est interest in regard to cognition (i.e., 1H-MRS: tating precise segmentation of brain structures
glutamate/glutamine [Glu/Gln], γ-aminobutyric (Manjon and Coupe, 2016), and automated disease
acid [GABA], myoinositol [mIns], glutathione classification of neurodegenerative disorders based
[GSH]; 31P-MRS: nicotinamide adenine dinu- on atrophy patterns (Basaia et al., 2019; Eshaghi
cleotide ratio [NAD+/NADH]). Depending on the et al., 2021), that have led to their widespread adop-
metabolite(s) of interest and anatomical location, tion in research and promising clinical adoption.
a high number of signal averages are required for The successes of neural networks in structural neu-
detection, or use of spectral edited sequences to roimaging are in part due to the relatively standard-
resolve the overlapping nature of their signal (i.e., ized acquisition protocols, limited confounding
GABA) from more concentrated metabolites. factors impacting signal intensity, and simplicity of
Consequently, single voxel spectroscopy is the tissue contrast and anatomical morphology as mod-
most feasible means of MRS acquisition (∼5–10 eled features. While neural networks have also been
min), with additional voxels acquired as needed applied for similar application across functional
through additional sequences. Whole-brain 1H- brain imaging modalities, generalizability across
MRS is possible but currently impractical due independent data sets remains low but presents a
to high acquisition time (>20 min), high motion promising direction for future development.
susceptibility, and most importantly, only reliable
for quantifying high concentration metabolites
(i.e., N-acetylaspartate) (Stagg et al., 2013). The
shift to ultra-high field 7T MRI holds significant SUMMARY AND CONCLUSIONS
benefits for MRS and improves not only the sig-
nal to noise ratio, but also increases the chemical In the current chapter we have highlighted techni-
shift dispersion allowing further spectral separa- cal advances, practical considerations, and inter-
tion of overlapping metabolite signals (i.e., Glu/ pretability of current implementations of fMRI,
Gln; NAD+/NADH) that can only be measured MEG, and PET brain imaging to assess neural
as a pooled concentration at 3T. This can be fur- function and connectivity. Despite the distinct con-
ther paired with functional MRS, a conceptual ceptual differences that underlie each functional
framework whereby dynamic metabolite change imaging modality, methodological advances are
is assessed across block conditions of a behavio- uniformly aimed at improving signal localization
ral paradigm (Stanley and Raz, 2018). Increased while controlling for artifacts and noise. While this
accuracy to resolve the relative concentrations of is always best handled at the acquisition stage, a
individual metabolites and robust assessment of key part of preprocessing any dynamic brain imag-
their dynamic change will undoubtedly generate ing dataset is cleaning the data. ICA-based meth-
advances in current understanding of changes in ods are widely used given their robustness for
metabolic brain states and their impact on neu- identifying and removing structured noise, particu-
ronal circuits and cognition. larly for known physiological noise signatures,
whereby individual noise components are identi-
fied and regressed resulting in a “cleaned” signal.
Currently, fMRI strikes the best balance between
accessibility, ease of implementation, sensitivity of
MACHINE LEARNING spatial localization, and capture of dynamic changes
in brain function, but is fundamentally limited
On a final note, a noteworthy area that has seen a by slow, delayed, changes in blood oxygenation.
rapid resurgence of recent development, and holds When we consider the multitude and physical dis-
significant potential for functional brain imaging, is tance covered by concurrent synaptic transmission
Advances in Brain Imaging 549

occurring within a 1s time frame throughout the Guideline 1: Recording and analysis of spontaneous
brain, the limitations of fMRI to deliver systems- cerebral activity. Journal of Clinical Neurophysiology,
level interpretation of brain function is clear when 28(4), 348–354.
used in isolation. In contrast, MEG directly meas- Basaia, S., Agosta, F., Wagner, L., Canu, E., Magnani,
ures neuronal activity with exceptional temporal G., Santangelo, R., & Filippi, M. (2019). Auto-
resolution to best capture dynamic fluctuations in mated classification of Alzheimer’s disease and
whole-brain function, while PET provides the best mild cognitive impairment using a single MRI and
specificity of neural mechanisms underlying brain deep neural networks. NeuroImage: Clinical, 21.
function. Multi-modal functional brain imaging is doi: 10.1016/j.nicl.2018.101645
critical to overcome inherent limitations of indi- Beisteiner, R., Robinson, S., Wurnig, M., Hilbert, M.,
vidual functional neuroimaging modalities and is Merksa, K., Rath, J., … Geissler, A. (2011). Clinical
an increasingly common approach in neuroscience, fMRI: evidence for a 7T benefit over 3T. NeuroIm-
with MRI serving as an independent or embedded age, 57(3), 1015–1021.
complementary measure for MEG and PET, respec- Brookes, M. J., Woolrich, M., Luckhoo, H., Price, D.,
tively. In regard to deriving functional brain net- Hale, J. R., Stephenson, M. C., … Morris, P. G.
works, while there is considerable overlap between (2011). Investigating the electrophysiological basis
fMRI and MEG, the timescale difference with PET of resting state networks using magnetoencepha-
remains a barrier that needs to be overcome. lography. Proceedings of the National Academy of
Sciences of the United States of America, 108(40),
16783–16788.
Brown, R. K., Bohnen, N. I., Wong, K. K., Minoshima,
ABBREVIATIONS S., & Frey, K. A. (2014). Brain PET in suspected
dementia: Patterns of altered FDG metabolism.
Radiographics, 34(3), 684–701.
15O – (15)Oxygen Buckner, R. L., Andrews-Hanna, J. R., & Schacter, D. L.
BOLD - Blood Oxygenation Level-Dependent (2008). The brain’s default network: anatomy,
cBSS – Constrained Blind Source Separation function, and relevance to disease. Annals of the
CSD – Cross-spectral Density New York Academy of Science, 1124, 1–38.
CT – Computed Tomography Burgess, R. C., Funke, M. E., Bowyer, S. M., Lewine,
EEG - Electroencephalogram J. D., Kirsch, H. E., Bagic, A. I., & Committee, A. C.
EPI – Echo Planar Imaging P. G. (2011). American Clinical Magnetoencepha-
fMRI – Functional Magnetic Resonance Imaging lography Society Clinical Practice Guideline 2: Pre-
surgical functional brain mapping using magnetic
FDG – Fluorodeoxyglucose
evoked fields. Journal of Clinical Neurophysiology,
ICA – Independent Components Analysis
28(4), 355–361.
LOR – Line of Response Catana, C. (2017). Principles of simultaneous PET/MR
MEG – Magnetoencephalography imaging. Magnetic Resonance Imaging Clinics of
MRS – Magnetic Resonance Spectroscopy North America, 25(2), 231–243.
PET – Positron Emission Tomography Catana, C., Drzezga, A., Heiss, W. D., & Rosen,
SiPMs - Silicon Photomultipliers B. R. (2012). PET/MRI for neurologic applications.
SQUID - Superconducting Quantum Interference Device Journal of Nuclear Medicine, 53(12),
TE – Time Echo 1916–1925.
TOF – Time of Flight Chen, J., Leong, Y. C., Honey, C. J., Yong, C. H.,
Norman, K. A., & Hasson, U. (2017). Shared
TR – Time Repeat
memories reveal shared structure in neural activity
across individuals. Nature Neuroscience, 20(1),
115–125.
Chen, W. (2007). Clinical applications of PET in brain
REFERENCES tumors. Journal of Nuclear Medicine, 48(9),
1468–1481.
Arthuis, M., Micoulaud-Franchi, J. A., Bartolomei, F., Cichy, R. M., & Oliva, A. (2020). A M/EEG-fMRI
McGonigal, A., & Guedj, E. (2015). Resting cortical fusion primer: Resolving human brain responses in
PET metabolic changes in psychogenic non-epileptic space and time. Neuron, 107, 772–781.
seizures (PNES). Journal of Neurology, Neurosur- Cichy, R. M., Pantazis, D., & Oliva, A. (2014). Resolv-
gery, and Psychiatry, 86(10), 1106–1112. ing human object recognition in space and time.
Bagic, A. I., Knowlton, R. C., Rose, D. F., Ebersole, J. S., Nature Neuroscience, 17, 455–462.
& Committee, A. C. P. G. (2011). American Clinical Cho, Z. H., Chan, J. K., & Eriksson, L. (1976). Circular
Magnetoencephalography Society Clinical Practice Ring Transverse Axial Positron Camera for
550 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

3-Dimensional Reconstruction of Radionuclides Gallezot, J., Lu, Y., Naganawa, M., & Carson, R. E.
Distribution. IEEE Transactions on Nuclear Science, (2020). Parametric imaging with PET and SPECT.
23(1), 613–622. IEEE Transactions on Radiation and Plasma Medical
Cohen, D. (1972). Magnetoencephalography: Detec- Sciences, 4(1), 1–23.
tion of the brain’s electrical activity with a supercon- Glasser, M. F., Coalson, T. S., Robinson, E. C., Hacker,
ducting magnetometer. Science, 175, 664–666. C. D., Harwell, J., Yacoub, E., … Van Essen, D. C.
Craddock, R. C., James, G. A., Holtzheimer, P. E., 3rd, (2016). A multi-modal parcellation of human cer-
Hu, X. P., & Mayberg, H. S. (2012). A whole brain ebral cortex. Nature, 536, 171–178.
fMRI atlas generated via spatially constrained Greicius, M. D., Srivastava, G., Reiss, A. L., & Menon, V.
spectral clustering. Human Brain Mapping, 33(8), (2004). Default-mode network activity distin-
1914–1928. guishes Alzheimer’s disease from healthy aging:
Dammers, J., Schiek, M., Boers, F., Silex, C., Zvyagint- Evidence from functional MRI. Proceedings of the
sev, M., Pietrzyk, U., & Mathiak, K. (2008). Integra- National Academy of Sciences of the United States
tion of amplitude and phase statistics for complete of America, 101(13), 4637–4642.
artifact removal in independent components of Hamilton, J. P., Farmer, M., Fogelman, P., & Gotlib, I. H.
neuromagnetic recordings. IEEE Transactions on (2015). Depressive rumination, the default-mode
Biomedical Engineering, 55(10), 2353–2362. network, and the dark matter of clinical neurosci-
Drzezga, A., Becker, J. A., Van Dijk, K. R., Sreenivasan, ence. Biological Psychiatry, 78(4), 224–230.
A., Talukdar, T., Sullivan, C., … Sperling, R. A. Hari, R. (2011). Magnetoencephalography:
(2011). Neuronal dysfunction and disconnection of Methods and applications. In D. L. Schomer & F. H.
cortical hubs in non-demented subjects with ele- Lopes da Silva (Eds.), Niedermeyer’s electroen-
vated amyloid burden. Brain, 134(6), 1635–1646. cephalography: Basic principles, clinical applica-
Escudero, J., Hornero, R., Abasolo, D., & Fernandez, tions, and related fields (pp. 865–900). Amsterdam:
A. (2011). Quantitative evaluation of artifact Elsevier.
removal in real magnetoencephalogram signals Hebart, M. N., Bankson, B. B., Harel, A., Baker, C. I.,
with blind source separation. Annals of Biomedical & Cichy, R. M. (2018). The representational
Engineering, 39(8), 2274–2286. dynamics of task and object processing in humans.
Eshaghi, A., Young, A. L., Wijeratne, P. A., Prados, F., eLife, 7. doi: 10.7554/eLife.32816
Arnold, D. L., Narayanan, S., … Ciccarelli, O. (2021). Henriksson, L., Mur, M., & Kriegeskorte, N. (2019).
Identifying multiple sclerosis subtypes using unsuper- Rapid invariant encoding of scene layout in human
vised machine learning and MRI data. Nature Com- OPA. Neuron, 103, 161–171.
munications, 12. doi: 10.1038/s41467-021-22265-2 Hu, X., Le, T. H., Parrish, T., & Erhard, P. (1995). Retro-
Esteban, O., Markiewicz, C. J., Blair, R. W., Moodie, spective estimation and correction of physiological
C. A., Isik, A. I., Erramuzpe, A., … Gorgolewski, fluctuation in functional MRI. Magnetic Resonance
K. J. (2019). fMRIPrep: A robust preprocessing Medicine, 34(2), 201–212.
pipeline for functional MRI. Nature Methods, Jamadar, S. D., Ward, P. G. D., Close, T. G., Fornito, A.,
16(1), 111–116. Premaratne, M., O’Brien, K., … Egan, G. F. (2020).
Feinberg, D. A., Moeller, S., Smith, S. M., Auerbach, Simultaneous BOLD-fMRI and constant infusion
E., Ramanna, S., Gunther, M., … Yacoub, E. (2010). FDG-PET data of the resting human brain. Scientific
Multiplexed echo planar imaging for sub-second Data, 7. doi: 10.1038/s41597-020-00699-5
whole brain FMRI and fast diffusion imaging. PLoS Jucaite, A., Cselenyi, Z., Arvidsson, A., Ahlberg, G.,
One, 5(12). doi: 10.1371/journal.pone.0015710 Julin, P., Varnas, K., … Farde, L. (2012). Kinetic
Florin, E., & Baillet, S. (2015). The brain’s resting-state analysis and test-retest variability of the radioligand
activity is shaped by synchronized cross-frequency [11C](R)-PK11195 binding to TSPO in the human
coupling of neural oscillations. NeuroImage, 111, brain: A PET study in control subjects. EJNMMI
26–35. Research, 2. doi: 10.1186/2191-219X-2-15
Franzmeier, N., Neitzel, J., Rubinski, A., Smith, R., Karjalainen, T., Karlsson, H. K., Lahnakoski, J. M.,
Strandberg, O., Ossenkoppele, R., … Alzheimer’s Glerean, E., Nuutila, P., Jaaskelainen, I. P., … Num-
Disease Neuroimaging Initiative. (2020). Functional menmaa, L. (2017). Dissociable roles of cerebral
brain architecture is associated with the rate of tau μ-opioid and type 2 dopamine receptors in vicari-
accumulation in Alzheimer’s disease. Nature Com- ous pain: A combined PET-fMRI study. Cerebral
munications, 11. doi: 10.1038/s41467-019-14159-1 Cortex, 27(8), 4257–4266.
Freeman, W. J. (1987). Simulation of chaotic EEG Kemper, V. G., De Martino, F., Emmerling, T. C.,
patterns with a dynamic model of the olfactory Yacoub, E., & Goebel, R. (2018). High resolution
system. Biological Cybernetics, 56(2–3), 139–150. data analysis strategies for mesoscale human func-
Fries, P. (2005). A mechanism for cognitive dynamics: tional MRI at 7 and 9.4T. NeuroImage, 164, 48–58.
Neuronal communication through neuronal coher- Kiemes, A., Davies, C., Kempton, M. J., Lukow, P. B.,
ence. Trends in Cognitive Sciences, 9(10), 474–480. Bennallick, C., Stone, J. M., & Modinos, G. (2021).
Advances in Brain Imaging 551

A systematic review with meta-analysis of multi- McMackin, R., Muthuraman, M., Groppa, S.,
modal 1H-MRS-fMRI studies. Frontiers in Psychia- Babiloni, C., Taylor, J. P., Kiernan, M. C., … Hardi-
try, 12. doi: 10.3389/fpsyt.2021.644315 man, O. (2019). Measuring network disruption in
Klados, M. A., Papadelis, C., Braun, C., & Bamidis, neurodegenerative diseases: New approaches
P. D. (2011). REG-ICA: A hybrid methodology using signal analysis. Journal of Neurology, Neuro-
combining blind source separation and regression surgery, and Psychiatry, 90(9), 1011–1020.
techniques for the rejection of ocular artifacts. Miller, K. L., Alfaro-Almagro, F., Bangerter, N. K.,
Biomedical Signal Processing and Control, 6(3), Thomas, D. L., Yacoub, E., Xu, J., … Smith, S. M.
291–300. (2016). Multimodal population brain imaging in
Klimesch, W. (2018). The frequency architecture of the UK Biobank prospective epidemiological study.
brain and brain body oscillations: An analysis. Nature Neuroscience, 19(11), 1523–1536.
European Journal of Neuroscience, 48(7), Ogawa, S., Lee, T. M., Kay, A. R., & Tank, D. W.
2431–2453. (1990). Brain magnetic resonance imaging with
Kriegeskorte, N., & Kievit, R. A. (2013). Representa- contrast dependent on blood oxygenation. Pro-
tional geometry: Integrating cognition, computa- ceedings of the National Academy of Sciences of
tion, and the brain. Trends in Cognitive Sciences, the United States of America, 87(24), 9868–9872.
17, 401–412. Oishi, M., Otsubo, H., Kameyama, S., Morota, N.,
Kundu, P., Brenowitz, N. D., Voon, V., Worbe, Y., Masuda, H., Kitayama, M., & Tanaka, R. (2002).
Vértes, P. E., Inati, S. J., … Bullmore, E. T. (2013). Epileptic spikes: Magnetoencephalography versus
Integrated strategy for improving functional con- simultaneous electrocorticography. Epilepsia,
nectivity mapping using multiecho fMRI. Proceed- 43(11), 1390–1395.
ings of the National Academy of Sciences of the Pagani, M., Öberg, J., De Carli, F., Calvo, A., Moglia,
United States of America, 110(40), 16187–16192. C., Canosa, A., … Chiò, A. (2016). Metabolic
Kundu, P., Voon, V., Balchandani, P., Lombardo, spatial connectivity in amyotrophic lateral sclerosis
M. V., Poser, B. A., & Bandettini, P. A. (2017). as revealed by independent component analysis.
Multi-echo fMRI: A review of applications in fMRI Human Brain Mapping, 37(3), 942–953.
denoising and analysis of BOLD signals. NeuroIm- Passow, S., Specht, K., Adamsen, T. C., Biermann,
age, 154, 59–80. M., Brekke, N., Craven, A. R., … Hugdahl, K.
Larkman, D. J., Hajnal, J. V., Herlihy, A. H., Coutts, (2015). Default-mode network functional connec-
G. A., Young, I. R., & Ehnholm, G. (2001). Use of tivity is closely related to metabolic activity. Human
multicoil arrays for separation of signal from mul- Brain Mapping, 36(6), 2027–2038.
tiple slices simultaneously excited. Journal of Mag- Polyn, S. M., Natu, V. S., Cohen, J. D., & Norman,
netic Resonance Imaging, 13(2), 313–317. K. A. (2005). Category-specific cortical activity
Lawrence, S. J. D., Formisano, E., Muckli, L., & de precedes retrieval during memory search. Science,
Lange, F. P. (2019). Laminar fMRI: Applications for 310, 1963–1966.
cognitive neuroscience. NeuroImage, 197, Power, J. D., Barnes, K. A., Snyder, A. Z., Schlaggar,
785–791. B. L., & Petersen, S. E. (2012). Spurious but sys-
Lewellen, T. K. (1998). Time-of-flight PET. Seminars in tematic correlations in functional connectivity MRI
Nuclear Medicine, 28(3), 268–275. networks arise from subject motion. NeuroImage,
Linnman, C., Catana, C., Petkov, M. P., Chonde, 59(3), 2142–2154.
D. B., Becerra, L., Hooker, J., & Borsook, D. (2018). Raichle, M. E., MacLeod, A. M., Snyder, A. Z.,
Molecular and functional PET-fMRI measures of Powers, W. J., Gusnard, D. A., & Shulman, G. L.
placebo analgesia in episodic migraine: Preliminary (2001). A default mode of brain function. Pro-
findings. NeuroImage: Clinical, 17, 680–690. ceedings of the National Academy of Sciences of
Lombardo, M. V., Auyeung, B., Holt, R. J., Waldman, the United States of America, 98(2), 676–682.
J., Ruigrok, A. N. V., Mooney, N., … Kundu, P. Rauschenberg, J., Nagel, A. M., Ladd, S. C., They-
(2016). Improving effect size estimation and statis- sohn, J. M., Ladd, M. E., Moller, H. E., … Semmler,
tical power with multi-echo fMRI and its impact on W. (2014). Multicenter study of subjective accept-
understanding the neural systems supporting ance during magnetic resonance imaging at 7 and
mentalizing. NeuroImage, 142, 55–66. 9.4 T. Investigative Radiology, 49(5), 249–259.
Lopes da Silva, F. (2013). EEG and MEG: Relevance to Risk, B. B., Murden, R. J., Wu, J., Nebel, M. B., Venkata-
neuroscience. Neuron, 80(5), 1112–1128. raman, A., Zhang, Z., & Qiu, D. (2021). Which multi-
Manjon, J. V., & Coupe, P. (2016). volBrain: An online band factor should you choose for your resting-state
MRI brain volumetry system. Frontiers in Neuroin- fMRI study? NeuroImage, 234, 117965.
formatics, 10. doi: 10.3389/fninf.2016.00030 Ronneberger, O., Fischer, P., & Brox, T. (2015). U-Net:
Mansfield, P. (1977). Multi-planar image formation Convolutional networks for biomedical image
using NMR spin echoes. Journal of Physics C: Solid segmentation. Paper presented at the Interna-
State Physics, 10(3), L55–L58. tional Conference on Medical Image Computing
552 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

and Computer-Assisted Intervention. doi: 10.1007/ cognitive neuroscience and psychiatry research. Fron-
978-3-319-24574-4_28 tiers in Psychiatry, 9. doi: 10.3389/fpsyt.2018.00076
Sakkalis, V., Doru Giurc Neanu, C., Xanthopoulos, P., Surti, S. (2015). Update on time-of-flight PET imag-
Zervakis, M. E., Tsiaras, V., Yang, Y., … Micheloyan- ing. Journal of Nuclear Medicine, 56(1), 98–105.
nis, S. (2009). Assessment of linear and nonlinear Sweet, W. H. (1951). The uses of nuclear disintegration
synchronization measures for analyzing EEG in a in the diagnosis and treatment of brain tumor. New
mild epileptic paradigm. IEEE Transactions on Infor- England Journal of Medicine, 245(23), 875–878.
mation Technology in Biomedicine, 13(4), 433–441. Tomasi, D. G., Shokri-Kojori, E., Wiers, C. E., Kim,
Sala, A., Caminiti, S. P., Presotto, L., Premi, E., Pilotto, S. W., Demiral, Ş. B., Cabrera, E. A., & Volkow,
A., Turrone, R., … Perani, D. (2017). Altered brain N. D. (2017). Dynamic brain glucose metabolism
metabolic connectivity at multiscale level in early identifies anti-correlated cortical-cerebellar net-
Parkinson’s disease. Scientific Reports, 7(1). doi: works at rest. Journal of Cerebral Blood Flow &
10.1038/s41598-017-04102-z Metabolism, 37(12), 3659–3670.
Samuelsson, J. G., Peled, N., Mamashli, F., Ahve- Tripathi, M., Tang, C. C., Feigin, A., De Lucia, I.,
ninen, J., & Hamalainen, M. S. (2021). Spatial Nazem, A., Dhawan, V., & Eidelberg, D. (2016).
fidelity of MEG/EEG source estimates: A general Automated differential diagnosis of early Parkinson-
evaluation approach. NeuroImage, 224. doi: ism using metabolic brain networks: A validation
10.1016/j.neuroimage.2020.117430 study. Journal of Nuclear Medicine, 57(1), 60–66.
Sander, C. Y., Hansen, H. D., & Wey, H. Y. (2020). Van Essen, D. C., Smith, S. M., Barch, D. M., Behrens,
Advances in simultaneous PET/MR for imaging T. E., Yacoub, E., Ugurbil, K., & Consortium, W. U.-M.
neuroreceptor function. Journal of Cerebral Blood H. (2013). The WU-Minn Human Connectome Pro-
Flow & Metabolism, 40(6), 1148–1166. ject: An overview. NeuroImage, 80, 62–79.
Schoffelen, J., Oostenveld, R., Lam, N. H. L., Udden, van Waarde, A., Marcolini, S., de Deyn, P. P., & Dierckx, R.
J., Hulten, A., Hagoort, P. (2019). A 204-subject (2021). PET agents in dementia: An overview. Semi-
multimodal neuroimaging dataset to study lan- nars in Nuclear Medicine, 51(3), 196–229.
guage processing. Scientific Data, 6. doi: 10.1038/ Wey, H. Y., Catana, C., Hooker, J. M., Dougherty,
s41597-019-0020-y D. D., Knudsen, G. M., Wang, D. J., … Kong, J.
Seeley, W. W., Crawford, R. K., Zhou, J., Miller, B. L., (2014). Simultaneous fMRI-PET of the opioidergic
& Greicius, M. D. (2009). Neurodegenerative dis- pain system in human brain. NeuroImage, 102(2),
eases target large-scale human brain networks. 275–282.
Neuron, 62(1), 42–52. Wipf, D. P., Owen, J. P., Attias, H. T., Sekihara, K., &
Shulman, G. L., Fiez, J. A., Corbetta, M., Buckner, Nagarajan, S. S. (2010). Robust Bayesian estima-
R. L., Miezin, F. M., Raichle, M. E., & Petersen, S. tion of the location, orientation, and time course
E. (1997). Common blood flow changes across of multiple correlated neural sources using MEG.
visual tasks: II. Decreases in cerebral cortex. Jour- NeuroImage, 49(1), 641–655.
nal of Cognitive Neuroscience, 9(5), 648–663. Woolrich, M., Hunt, L., Groves, A., & Barnes, G.
Stagg, C. J., Knight, S., Talbot, K., Jenkinson, M., (2011). MEG beamforming using Bayesian PCA for
Maudsley, A. A., & Turner, M. (2013). Whole-brain adaptive data covariance matrix regularization.
magnetic resonance spectroscopic imaging meas- NeuroImage, 57(4), 1466–1479.
ures are related to disability in ALS. Neurology, 80, Yeo, B. T., Krienen, F. M., Sepulcre, J., Sabuncu, M. R.,
610–615. Lashkari, D., Hollinshead, M., … Buckner, R. L.
Stam, C. J., de Haan, W., Daffertshofer, A., Jones, (2011). The organization of the human cerebral
B. F., Manshanden, I., van Cappellen van Walsum, cortex estimated by intrinsic functional connectivity.
A. M., … Scheltens, P. (2009). Graph theoretical Journal of Neurophysiology, 106(3), 1125–1165.
analysis of magnetoencephalographic functional Zou, N., Chetelat, G., Baydogan, M. G., Li, J., Fischer,
connectivity in Alzheimer’s disease. Brain, 132(1), F. U., Titov, D., … Yakushev, I. (2015). Metabolic
213–224. connectivity as index of verbal working memory.
Stanely, J. A., & Raz, N. (2018). Functional magnetic Journal of Cerebral Blood Flow & Metabolism,
resonance spectroscopy: The “new” MRS for 35(7), 1122–1126.
33
Novel Approaches to Large-Scale
Data in Neuroimaging
M a t t h e w T h a r p , S h a n n o n L . R i s a c h e r,
Paula Bice, Paul M. Thompson,
A n d r e w J . S a y k i n , a n d M e i c h e n Yu

INTRODUCTION Structural magnetic resonance imaging (sMRI)


is rooted in the spatial domain and can be captured
Numerous techniques have been proposed to with different imaging sequence variations that are
advance data applications in neuroimaging, resulting sensitive to different tissue properties such as T1,
in some uncertainty as to which techniques are best T2, T2*, and FLAIR (Symms et al., 2004). One
suited to address the wide variety of unique chal- advantage of sMRI is the ability to image subcor-
lenges. While an exhaustive overview of available tical structures, such as the thalamus (Su et al.,
techniques is beyond the scope of this chapter, we 2019), where post-processing methods, including
highlight some promising novel neuroimaging voxel-based morphometry (VBM), surface-based
approaches that may be particularly crucial for future morphometry (SBM), and MR-relaxometry enable
large-scale data analyses. In particular, we address different views of structural brain features (Martin
novel approaches with neuroimaging modalities, et al., 2015). More recently, the MP2RAGEME
machine learning (ML), and deep learning (DL) as sequence has been developed, allowing T1, T2*
well as considering statistical advances and tech- and quantitative susceptibility maps (QSMs) to be
niques for managing high dimensionality data via acquired simultaneously (Caan et al., 2019).
source separation. Given the specificity of much of Functional MRI (fMRI) acquires blood-oxygen-
the neuroscientific and neuroimaging terminology, a level-dependent (BOLD) signals to provide a
list of briefly-defined key terms is presented below. noisy indication of underlying neural activity at
low spatiotemporal resolution. Atlases such as the
Yeo-17 (Yeo et al., 2011) are commonly used to
represent fMRI acquisitions, providing a set of
regions of interest (ROIs) that may be used for
NEUROIMAGING MODALITIES network analyses of functional connectivity (FC).
Most commonly, FC is assessed in a static manner
Before addressing large-scale data analysis with large time windows, although dynamic FC
approaches in neuroimaging, it is helpful to pro- approaches that utilize a sliding time window are
vide an overview of commonly used neuroimag- becoming increasingly popular (Hutchison et al.,
ing modalities. In this section we will introduce 2013). Recently, ultrafast fMRI has been explored
each neuroimaging modality while highlighting with mice to address spatiotemporal limitations of
novel developments. conventional fMRI (Gil et al., 2021).
554 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Table 33.1 Key terms and brief definitions


Key Terms Definitions

Autoencoder Deep learning model which learns data encodings via dimensionality reduction
with neural networks
Bayesian network Machine learning method which uses graphical models to represent variables and
dependencies as directed acyclic graphs
Canonical correlation analysis Source separation method which finds the maximum correlation of different linear
combinations of two vectors
Convolutional neural network Deep learning method typically used with images which uses convolutions to
extract patterns from feature maps
Data harmonization A set of statistical methods which can be used to reduce site/scanner effects from
neuroimaging measurements
Deep belief network Deep learning method which uses multiple layers of other deep learning models
such as restricted Boltzmann machines or autoencoders
Dynamic causal modeling Statistical method used for causal inference by comparing information between
Bayesian models
Generalized additive model Statistical method which uses generalized linear models to relate a response
variable to several predictor variables
Generative adversarial network Deep learning method which generates new data by placing two neural networks
in competition with one another
Independent component analysis Source separation method which separates a signal into subcomponents by
identifying maximal statistical independence
Markov model Machine learning method which uses stochastic models to represent variables and
dependencies graphically
Meta/mega-analysis Statistical method which pools data from multiple sites to improve statistical power
Multi-layer perceptron Deep learning method which uses multiple layers of perceptrons guided by
activation functions for pattern extraction
Principal component analysis Source separation method which separates a signal into subcomponents by
identifying maximal orthogonality
Recurrent neural network Deep learning method where outputs from nodes connect to previous nodes
cyclically to extract patterns recurrently
Residual neural network Deep learning method which uniquely includes skip connections to allow one
extraction layer skip over subsequent layers
Support vector machine Supervised machine learning method which uses a training dataset as a model for
assigning categories to data

Diffusion weighted imaging (DWI) is a MR approach, position-orientation adaptive smoothing


technique that measures the displacement of water (POAS), has been proposed as an alternative trac-
molecules across tissue, allowing white matter pro- tography approach (Yang et al., 2020).
jections in the brain to be detected. Diffusion tensor Perfusion weighted imaging (PWI) can be used
imaging (DTI) is commonly used as an extension of to study blood flow throughout the brain. Popular
DWI to indicate the directionality of image voxels PWI techniques include arterial spin labeling
via tensors, including fractional anisotropy (FA), a (ASL) and dynamic susceptibility contrast imag-
metric used to quantify the magnitude of direction- ing (DSC) (Bergamino et al., 2020). Another
ality of individual DTI tensors. Additional exten- modality, functional near-infrared spectroscopy
sions of DWI exist including neurite orientation (fNIRS), can monitor changes in blood flow by
dispersion and density imaging (NODDI) (Zhang measuring cerebral hemoglobin concentrations
et al., 2012), fiber-based analysis (FBA) (Pecheva (Hoshi and Michael, 2005). This modality pro-
et al., 2018), constrained spherical deconvolution vides opportunities for naturalistic imaging set-
(CSD) (Tournier et al., 2008), informed CSD (Roine tings as it can acquire measures in non-laboratory
et al., 2015), deterministic tractography (Alexander, environments (Pinti et al., 2020).
2010), and probabilistic tractography (Sarwar Techniques such as positron emission tomogra-
et al., 2019). A recent novel DWI post-processing phy (PET) and single photon emission computed
Novel Approaches to Large-Scale Data in Neuroimaging 555

tomography (SPECT) can be used to acquire the information theoretic Kolmogorov complexity
images of radioactive tracers within the brain. metric (Wachinger et al., 2019).
Both techniques are limited by spatial resolution, Some significant advancements for statistical
but their promise is exemplified by their abil- analyses can be found within multi-institutional
ity to measure highly specific brain processes. collaborations, which are becoming commonplace
For example, with PET, radiotracers have been to enhance statistical power via dataset integration
developed for imaging neurotransmitter activ- from multiple imaging sites. Specifically, methods
ity, amyloid plaques, neurofibrillary tangles, and related to data harmonization, meta-analysis, and
intracellular signals (Hooker and Carson, 2019). mega-analysis have brought promise to the reliabil-
Finally, techniques such as electroencepha- ity of future statistical analyses in neuroimaging.
lography (EEG) and magnetoencephalography For example, the ComBat technique (Johnson et al.,
(MEG) can be used to measure electrical signals 2007) is a popular method for modeling and remov-
within the brain. Commonly, EEG/MEG are uti- ing site-specific effects of multi-site neuroimaging
lized for studies of brain connectivity, making studies from DTI (Fortin et al., 2017), sMRI (Fortin
them useful for studying connectivity-related dis- et al., 2018), or fMRI (Yu et al., 2018), and longitu-
orders at higher temporal resolution than fMRI (da dinal ComBat has recently been developed for lon-
Cruz et al., 2020; Nentwich et al., 2020). gitudinal multi-scanner data (Beer et al., 2020). In
addition, ComBat-GAM has been proposed for use
in generalized additive models (GAMs) (Pomponio
et al., 2020). A recent study compared the perfor-
mance of multiple data harmonization techniques
STATISTICAL ADVANCES and found that the use of ComBat-GAM for har-
monizing multisite cortical thickness data can more
With increasing amounts of data, several statisti- effectively minimize confounds and increase statis-
cal challenges have arisen in neuroimaging includ- tical power than other techniques (Sun et al., 2021).
ing difficulties due to effect sizes (Reddan et al., Nonetheless, some limitations to the ComBat meth-
2017), increased vulnerability to confounding odology have been discussed, such as an inability
variables (Smith and Nichols, 2018), underreport- to preserve effect sizes from sites with several non-
ing of statistical power calculations (Szucs and linear scanner effects and the means by which small
Ioannidis, 2020), and concerns about the reliabil- pre-processing differences may lead to different
ity of statistical significance tests (Hupé, 2015). harmonization outcomes (Cetin-Karayumak et al.,
Other issues related to causal inference and statis- 2020). ComBat also requires spatial registration
tical methodologies for multi-site meta-analyses to a population template, although an alternative
have also garnered significant interest. In this approach, known as multisite image harmoniza-
section we will further introduce these statistical tion by cumulative distribution function alignment
challenges and discuss novel approaches toward (mica), has been proposed to address this issue
addressing them. by comparing several scans on a single subject
Earlier methods for neuroimaging analyses (Wrobel et al., 2020). Similarly, Neuroharmony,
have been criticized, as the statistical tests were which enables scanner bias reduction via DL with-
performed on thousands of voxels, thus increas- out reference to a prior population, has also been
ing susceptibility to selection bias (Reddan et al., suggested (Garcia-Dias et al., 2020). Other novel
2017). To address this issue, many studies have approaches to data harmonization have been pro-
used multiple comparisons corrections such as the posed in a DL context, including a dual generative
false discovery rate (FDR); however, these cor- adversarial network (GAN) approach (Zhong et al.,
rections are also prone to bias (Hupé, 2015). One 2020), an attention-guided deep domain adaptation
solution to this problem is to reduce the number (AD2A) approach (Guan et al., 2021), and a con-
of statistical tests required for a given study via volutional neural network (CNN) approach called
dimensionality reduction techniques. DeepHarmony (Dewey et al., 2019). GAN methods
Another concern for future statistical analyses is can also address biases in training ML methods on
causal inference, specifically regarding the misinter- multisite data, extending beyond scanner protocols
pretation of directionality with causality in graphi- to adjust for age, sex, and other demographic effects
cal models (Lindquist and Sobel, 2011). To address more completely (Zhao et al., 2020). When apply-
this, various causal inference methods have been ing highly nonlinear methods such as DL to mul-
proposed such as Bayesian methods (Cao et al., tisite data, it can be extremely difficult to remove
2019), dynamic causal modeling (DCM) (Moran confounders, and predictive models can lock onto
et al., 2013), and Granger causality analysis (Seth features that are coincidentally related to the out-
et al., 2015). In addition, another study developed a come, leading to poor generalization performance.
method to quantify the extent of confounding using GANs provide an attractive and novel solution to
556 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

this problem when typical linear regression meth- used in different noise or dimensionality reduction
ods are insufficient (Dinsdale et al., 2021; Dockès contexts to separate a signal of interest (SOI) from
et al., 2021). Lastly, various additional harmoniza- acquired data. These techniques are especially
tion approaches may yet be important for future important for neuroimaging where numerous
statistical analyses such as adaptive linear map- SOIs may be masked by noise or higher dimen-
ping of rotational invariant spherical harmonics sional representations. BSS methods can be cate-
(LinearRISH) developed for dMRI (Ning et al., gorized into four overarching categories: single
2020), diffusion parametric map harmonization data unidimensional (SDU), multiple dataset unidi-
(DPMH) (Pinto et al., 2020), and nonlinear distri- mensional (MDU), single dataset multidimensional
bution mapping for amyloid-PET harmonization (SDM), and multiple dataset multidimensional
(Properzi et al., 2019). (MDM) (Silva et al., 2016).
Besides data harmonization, meta-analytic, When analyzing data from a single modal-
and mega-analytic methods have increased the ity, oftentimes a single dataset unidimensional
reliability of statistical analyses. Meta-analytic approach is needed. Principal component anal-
methods refer to multi-site endeavors for coordi- ysis (PCA) is a popular SDU approach, and
nating data acquisition, processing, and analysis to different versions have been proposed for neuro-
ensure methodological consistency between sites, imaging contexts such as sparse PCA (SPCA) (De
enabling data pooling for larger sample sizes. Pierrefeu et al., 2017), sparse principal component
Notably, the Enhancing Neuroimaging Genetics of mediation (SPCM) (Zhao et al., 2020), and
through Meta-Analysis (ENIGMA) consortium shared component analysis (De Cheveigné, 2021).
has used both mega- and meta-analytic practices Independent component analysis (ICA) is another
extensively to discover relationships between SDU approach with several variations used for
neuroimaging and genetics, yielding the largest neuroimaging, including Infomax (Fedorov et al.,
MRI and DTI studies to date of over 15 psychi- 2019), FastICA (Ge et al., 2016), probabilistic
atric, neurological, and developmental disorders ICA (PICA) (Sarraf et al., 2014), and longitudinal
(Thompson et al., 2020). The use of standardized ICA (Wang and Guo, 2019).
analysis pipelines for MRI, DTI, fMRI, and most In some cases, researchers may desire a uni-
recently EEG (Smit et al., 2018) has led to well dimensional representation of multiple datasets.
powered studies relating brain metrics to common Canonical correlation analysis (CCA) can be used
and rare genetic variations (Grasby et al., 2020; to identify correlations between datasets, and a
Van Der Meer et al., 2020) and even epigenetic popular version known as multiset CCA (mCCA)
variation (Jia et al., 2019) by pooling data from has been applied to data fusion in neuroimaging
over 45 countries worldwide. Most commonly, (Correa et al., 2010). In addition, mCCA with
coordinate-based meta-analysis (CBMA) meth- reference (MCCAR) has also been proposed
ods have been used to enable comparisons within for data fusion with joint ICA (jICA) (Qi et al.,
standard stereotaxic spaces between studies (Fox 2016). Likewise, group ICA (GICA) (Calhoun
et al., 2014), and toolboxes such as the CluB tool- et al., 2009) and parallel ICA (pICA) (Laforce Jr
box (Berlingeri et al., 2019) and the ENIGMA et al., 2014) can be used for MDU purposes along
toolbox (Lariviere et al., 2020) have made it easier with additional ICA-based methods such as ten-
to integrate meta-analytic methods and to test asso- sor ICA (TICA) (Koush et al., 2019), linked ICA
ciations between neuroimaging and molecular, (LICA) (Groves et al., 2011), hierarchical group
transcriptomic, or histologic data in other bioin- PICA (Guo and Tang, 2013), and group informa-
formatics databases such as the Allen Brain Atlas. tion guided ICA (GIG-ICA) (Du and Fan, 2013).
Beyond meta-analysis, another technique known Some additional approaches worth considering
as mega-analysis pools raw data from multiple for MDU contexts include non-Gaussian meth-
sites instead of combining pre-­processed data. ods such as non-Gaussian component analysis
This technique has been adopted by the ENIGMA (NGCA) (Diederichs et al., 2010), and simultane-
consortium for the investigation of various brain ous NGCA (SING) (Risk and Gaynanova, 2020).
disorders (Jalbrzikowski et al., 2021; Koshiyama In other instances, a multidimensional rep-
et al., 2020; Wang et al., 2020). resentation of a single dataset may be desir-
able. In these cases, independent vector analysis
(IVA) methods such as Laplacian IVA (IVA-L),
Gaussian IVA (IVA-G) (Anderson et al., 2011)
or subspace analysis methods such as independ-
SOURCE SEPARATION ent subspace analysis (ISA) (Ma et al., 2010),
adaptive independent subspace analysis (AISA)
Source separation, or blind source separation (Ke et al., 2019), and stationary subspace analy-
(BSS), refers to a set of techniques that may be sis (SSA) (Khan et al., 2016) may be useful. In
Novel Approaches to Large-Scale Data in Neuroimaging 557

addition, a technique based on statistical physics used to predict psychometric relationships with
known as energy landscape analysis (Ezaki et al., multimodal data (Rasero et al., 2021), and several
2017) has been proposed for a SDM neuroimag- variations of group LASSO have been proposed
ing context. for neuroimaging applications to analyze sub-
Finally, MDM approaches may be crucial for groups of data (Beer et al., 2019; Liu et al., 2018,
future complex neuroimaging analyses where 2019). Another penalized least squares method,
a multidimensional representation of multiple the elastic net (ENET), has been used with the
datasets is needed, as with multi-omics applica- total variation (TV) penalty in a few novel varia-
tions (Subramanian et al., 2020). Some MDM tions for neuroimaging applications (Hadj-Selem
approaches suitable for neuroimaging applica- et al., 2018; Pietrosanu et al., 2021), and some
tions include multi-dataset ISA (MISA) (Silva other novel ENET-based techniques have been
et al., 2020) and joint ISA (JISA) (Lahat and proposed including a combined ENET and mul-
Jutten, 2016). In addition, a technique known as tiple kernel learning (MKL) approach (Mourao-
Laplacian eigenmaps for group ICA decompo- Miranda et al., 2012), a sparse partial correlation
sition (LEICA) can extract group-level spatial with ENET (SPC-EN) approach (Ryali et al.,
maps (Liu et al., 2018), and the kernel machine 2012), and a combined group LASSO and ENET
by detecting higher order interactions (KMDHOI) approach (Guo et al., 2018).
(Alam et al., 2018) incorporates an infinite-­ Another popular technique is the support vec-
dimensional Hilbert space, which can be used for tor machine (SVM), which has been used to
modeling higher order interactions such as epi- classify various diseases (Steardo Jr et al., 2020;
static factors between genetics and imaging data. Zhou et al., 2018). Notably, one study combined
MKL with the SVM for psychosis classification
(Squarcina et al., 2017), and ensemble SVM tech-
niques have been proposed to combine multiple
SVMs for enhanced classification (Bi et al., 2018;
MACHINE LEARNING Sørensen et al., 2018). SVMs have also been com-
bined with methods such as mutual information
Machine learning (ML) methods in neuroimaging (MI) (Fan and Chou, 2016) or recursive feature
are becoming increasingly popular, and several elimination (RFE) for enhanced feature selection
novel approaches with fundamental ML applica- (Richhariya et al., 2020). In addition, several neu-
tions, including regression-based methods, cluster roimaging applications have been proposed using
analysis, and graphical models, have been pro- support vector regression (SVR) (Shi et al., 2018;
posed in recent years. While in depth explanations Singer et al., 2019; Sperber et al., 2019) and the
of individual ML methods are beyond the scope of relevance vector machine (RVM) (Lin et al., 2021;
this chapter, we will briefly highlight some prom- Meyer et al., 2019).
ising approaches. Graphical models including Bayesian networks
Several variations of regression methods have or Markov models have also shown promise as
been proposed in recent years. Notably, robust meaningful ML applications for neuroimaging.
iteratively weighted least squares (IRLS) has been For example, dynamic Bayesian networks have
proposed as an alternative to ordinary least squares been used to jointly model structure-function
(OLS) for fMRI analysis (Wager et al., 2005), and relationships between DTI and fMRI (Dang et al.
the sandwich estimator (SwE) has been proposed 2017) and to assess connectivity with fMRI (Dang
to expand the OLS model for longitudinal appli- et al., 2017). Likewise, several novel approaches
cations (Guillaume et al., 2014). Penalized least have employed Markov models (Ficiarà et al.,
squares methods (Bunea et al., 2011), which intro- 2021; Zhang et al., 2019), with the hidden Markov
duce a regression penalty for bias minimization, model (HMM) being commonly utilized (Jamaloo
have also been proposed. Ridge regression (RR) et al., 2020; Tao et al., 2021; Tessadori et al.,
is a penalized least squares method that has been 2021). In addition, extensions of the HMM have
used for predictive modeling with connectomes been proposed for neuroimaging contexts such as
(Gao et al., 2019), and a kernelized RR (KRR) the hidden semi-Markov model (HSMM) (Fiecas
method has been used for fMRI pattern prediction et al., 2021) and semiparametric HMMs (Kang
(Chu et al., 2011). Other RR techniques include et al., 2019), and Markov random fields (MRFs)
Bayesian RR (BRR) for biological age predic- have been proposed as undirected Markov mod-
tion (Ball et al., 2021; Roudbar et al., 2021) and els to evaluate the spread of AD (Dyrba et al.,
Bayesian Rician regression for use with DWI data 2018). Furthermore, the hidden MRF (HMRF) has
(Wegmann et al., 2017). The least absolute shrink- also been used for many neuroimaging applica-
age and selection operator (LASSO), another tions (Górriz et al., 2018; Guerrout et al., 2020;
penalized least squares method, has also been Lukashenko and Chernyaev, 2020).
558 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Lastly, the random forest (RF) model is a 2021; Legarreta et al., 2021; Pinaya et al., 2019),
hybrid ML model that uses an ensemble classifi- and some additional variations of the AE model
cation approach known as bagging. An advantage have been proposed for neuroimaging applications
of the RF is its versatility, which is exemplified including the sparse AE (Almuqhim and Saeed,
by a wide range of recently proposed neuroimag- 2021), modified k-sparse AE (σKSA) (Bhatkoti
ing applications (Cordova et al., 2020; Dimitriadis and Paul, 2016), stacked AE (Ferri et al., 2021),
et al., 2021; Kamarajan et al., 2020; McKinley denoising AE (DAE) (Duffy et al., 2020), stacked
et al., 2017; Zhao et al., 2017; Zhu et al., 2018). denoising AE (SDAE) (Zheng-Lin et al., 2018),
randomized denoising AE (rDAE) (Ithapu et al.,
2014), variational AE (VAE) (Kim et al., 2021),
and the discriminative margin-sensitive AE
(MSAE) (Zhang et al., 2020).
DEEP LEARNING DBNs use several layers of separate simplis-
tic two-layer DL models known as restricted
Deep learning (DL) techniques are a more com- Boltzmann machines (RBMs). A few recent stud-
plex subset of ML techniques that use several ies have reported novel neuroimaging applica-
iterations of pattern-finding algorithms to extract tions with DBNs (Huang et al., 2020; Latha and
meaningful information from data. While an Kavitha, 2019; Zeng et al., 2021), and some novel
exhaustive overview of DL applications in neuro- variations of the DBN framework include the
imaging is beyond the scope of this chapter, we group LASSO sparse DBN (GLS-DBN) (Shen
will review some promising approaches. et al., 2019), the neural architecture search with
Specifically, we will address novel approaches particle swarm optimization DBN (NAS-DBN)
with multi-layer perceptrons (MLPs), autoencod- (Qiang et al., 2020; Ren and Liu, 2021), and the
ers (AEs), deep belief networks (DBNs), and volumetric sparse DBN (VS-DBN) (Dong et al.,
CNNs. Some additional approaches that are not 2019).
covered due to limited space but may be of inter- Finally, convolutional neural networks (CNNs)
est to the reader include residual neural networks have been used in a wide range of neuroimaging
(ResNets) (Sun et al., 2021; Zhang et al., 2021), applications, with several noteworthy approaches
recurrent neural networks (RNNs) (Nguyen et al., being used in AD prediction (Jo et al., 2019; Jo
2020; Thomas et al., 2019), and generative adver- et al., 2020; Yang et al., 2020; Zhang et al., 2021),
sarial networks (GANs) (Liu et al., 2020; Yan hippocampal segmentation (Liu et al., 2020;
Zhao et al., 2020). Nobakht et al., 2021), white matter hyperintensity
MLPs are fundamental DL models that use segmentation (Rudie et al., 2019; Umapathy et al.,
multiple layers of perceptrons guided by activa- 2021), and brain connectomics (Khosla et al.,
tion functions. In recent years, several neuroim- 2019; Phang et al., 2019).
aging studies have utilized MLPs for various
applications (Yun et al., 2019; Javed et al., 2021;
Karami et al., 2020), and some novel MLP frame-
works have been proposed including the moving
local window MLP (MLW-MLP) used with MR SUMMARY AND CONCLUSIONS
electrical properties tomography (MREPT) (Lee
et al., 2021) and the multi-head self-attention In this chapter we have provided a brief overview
model (MSAM) used for temporal lobe epilepsy of novel large-scale data applications in neuroim-
classification (Gu et al., 2020). In addition, a nota- aging while discussing advances with neuroimag-
ble study proposed a method for integrating DL ing modalities, statistics and novel techniques
into clinical practice by using MLPs to enhance with BSS, ML, and DL. Nonetheless, data science
stereotactic navigation in neurosurgical proce- applications that use neuroimaging are expansive
dures (Leuthardt et al., 2018). and several other approaches, including data man-
Autoencoders (AEs) are similar to MLPs with agement, computational advances, and data fusion
hidden layers that contain fewer nodes than the have not been covered. As neuroimaging evolves,
input and output layers. Many notable applica- numerous additional large-scale data applications
tions have employed AEs (Akramifard et al., are expected to emerge.
Novel Approaches to Large-Scale Data in Neuroimaging 559

Table 33.2 Acronyms with their definitions


Acronym Definition Acronym Definition

AD Alzheimer’s disease HSMM Hidden semi-Markov model


AD2A Attention-guided deep domain adaptation ICA Independent component analysis
AE Autoencoder IRLS Robust iteratively reweighted least squares
AISA Adaptive independent subspace analysis ISA Independent subspace analysis
ALE Activation likelihood estimation IVA Independent vector analysis
ASL Arterial spin labeling IVA-G Gaussian independent vector analysis
BHICP Bayesian hierarchical independent cluster IVA-L Laplacian independent vector analysis
process modeling jICA Joint independent component analysis
BRR Bayesian ridge regression JISA Joint independent subspace analysis
BSS Blind source separation KMDHOI Kernel machine by detecting higher order
CBMA Coordinate-based meta-analysis interactions
CCA Canonical correlation analysis KRR Kernelized ridge regression
CNN Convolutional neural network LASSO Least absolute shrinkage and selection
CSD Constrained spherical deconvolution operator
DAE Denoising autoencoder LEICA Laplacian eigenmaps for group independent
DBN Deep belief network component analysis decomposition
DCM Dynamic causal modeling LICA Linked independent component analysis
DL Deep learning LinearRISH Adaptive linear mapping of rotational
invariant spherical harmonics
DPMH Diffusion parametric map harmonization
LR Logistic regression
DSC Dynamic susceptibility contrast imaging
mCCA Multiset canonical correlation analysis
DTI Diffusion tensor imaging
MCCAR Multiset canonical correlation analysis with
DWI Diffusion weighted imaging
reference
EEG Electroencephalography
MDM Multiple dataset multidimensional
EIC Eigen clustering
MDU Multiple dataset unidimensional
ENET Elastic net
MEG Magnetoencephalography
ENIGMA Enhancing Neuroimaging Genetics through
MI Mutual information
Meta-analysis
MICA Multisite image harmonization by cumulative
ES-SDM Effect size signed differential mapping
distribution function alignment
FA Fractional anisotropy
MISA Multi-dataset independent subspace
FBA Fixel based analysis, fiber based analysis analysis
FDR False discovery rate MKDA Multilevel kernel density analysis
fMRI Functional magnetic resonance imaging MKL Multiple kernel learning
fNIRS Functional near-infrared spectroscopy ML Machine learning
GAM Generalized additive models MLP Multi-layer perceptron
GAN Generative adversarial network MLW-MLP Moving local window multilayer perceptron
GICA Group independent component analysis MREPT Magnetic resonance electrical properties
GIG-ICA Group information guided independent tomography
component analysis MRF Markov random fields
GIPCA Generalized integrative principal MSAE Discriminative margin-sensitive autoencoder
component analysis
MSAM Multi-head self-attention model
GLS-DBN Group least absolute shrinkage selection
MWK-Means Minkowski-weighted K-means
operator sparse deep belief network
NAS-DBN Neural architecture search deep belief
GMM Gaussian mixture modeling
network
HMM Hidden Markov modeling
NGCA Non-Gaussian component analysis
HMRF Hidden Markov random field
NODDI Neurite orientation dispersion and density
HPGRF Hierarchical Poisson/Gamma random field imaging
modeling
(continued)
560 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Table 33.2 Continued


Acronym Definition Acronym Definition

OLS Ordinary least squares SDU Single dataset unidimensional


PCA Principal component analysis SING Simultaneous non-Gaussian component
PET Positron emission tomography analysis
PICA Probabilistic independent component sMRI Structural magnetic resonance imaging
analysis SOI Signal of interest
pICA Parallel independent component analysis SPCA Sparse principal component analysis
POAS Position-orientation adaptive smoothing SPC-EN Sparse partial correlation with elastic net
PWI Perfusion weighted imaging SPCM Sparse principal component of mediation
QSM Quantitative susceptibility maps SPECT Single photon emission computed
RBM Restricted Boltzmann machine tomography
rDAE Randomized denoising autoencoder SSA Stationary subspace analysis
ResNets Residual neural networks SVM Support vector machine
RF Random forest SVR Support vector regression
RFE Recursive feature elimination TC-GICA Temporal-concatenation group independent
component analysis
RNN Recurrent neural networks
TICA Tensor independent component analysis
RR Ridge regression
TV Total variation
RVM Relevance vector machine
VAE Variational autoencoder
SampEn Sample entropy
VBM Voxel-based morphometry
SBM Surface-based morphometry
VS-DBN Volumetric sparse deep belief network
SDAE Stacked denoising autoencoder
σKSA Modified k-sparse autoencoder
SDM Single dataset multidimensional

ACKNOWLEDGMENTS and applications (pp. 383–395). Oxford, UK:


Oxford University Press.
Almuqhim, F., & Saeed, F. (2021). ASD-SAENet: A
The preparation of this chapter was supported in part
sparse autoencoder, and deep-neural network
by U.S. National Institutes of Health grants: R01
model for detecting autism spectrum disorder (ASD)
AG197711, P30 AG10133, P30 AG072976,
using fMRI data. Frontiers in Computational Neuro-
U01AG072177, U01AG024904, R01 CA129769,
science, 15. doi: 10.3389/fncom.2021.654315
R01 AG057739, R01 LM013463, R01 AG068193,
Anderson, M., Adali, T., & Li, X.-L. (2011). Joint blind
R01 AG061788, K01 AG049050, and U01 AG068057.
source separation with multivariate Gaussian
model: Algorithms and performance analysis. IEEE
Transactions on signal processing, 60(4),
1672–1683.
REFERENCES Ball, G., Kelly, C. E., Beare, R., & Seal, M. L. (2021).
Individual variation underlying brain age estimates
Akramifard, H., Balafar, M. A., Razavi, S. N., & Ramli, in typical development. NeuroImage, 235. doi:
A. R. (2021). Early detection of Alzheimer’s disease 10.1016/j.neuroimage.2021.118036
based on clinical trials, three-dimensional imaging Beer, J. C., Aizenstein, H. J., Anderson, S. J., & Krafty,
data, and personal information using autoencoders. R. T. (2019). Incorporating prior information with
Journal of Medical Signals and Sensors, 11(2), fused sparse group lasso: Application to prediction
120–130. of clinical measures from neuroimages. Biometrics,
Alam, M. A., Lin, H. Y., Deng, H. W., Calhoun, V. D., 75(4), 1299–1309.
& Wang, Y. P. (2018). A kernel machine method Beer, J. C., Tustison, N. J., Cook, P. A., Davatzikos, C.,
for detecting higher order interactions in multi- Sheline, Y. I., Shinohara, R. T., … Alzheimer’s Dis-
modal datasets: Application to schizophrenia. ease Neuroimaging Initiative. (2020). Longitudinal
Journal of Neuroscience Methods, 309, 161–174. combat: A method for harmonizing longitudinal
Alexander, A. L. (2010). Deterministic white matter multi-scanner imaging data. NeuroImage, 220.
tractography. Diffusion MRI: Theory, methods, doi: 10.1016/j.neuroimage.2020.117129
Novel Approaches to Large-Scale Data in Neuroimaging 561

Bergamino, M., Nespodzany, A., Baxter, L. C., da Cruz, J. R., Favrod, O., Roinishvili, M., Chkonia, E.,
Burke, A., Caselli, R. J., Sabbagh, M. N., … Brand, A., Mohr, C., … Herzog, M. H. (2020). EEG
Stokes, A. M. (2020). Preliminary assessment of microstates are a candidate endophenotype for
intravoxel incoherent motion diffusion-weighted schizophrenia. Nature Communications, 11(1).
MRI (IVIM-DWI) metrics in Alzheimer’s disease. doi: 10.1038/s41467-020-16914-1
Journal of Magnetic Resonance Imaging, 52(6), Dang, S., Chaudhury, S., Lall, B., & Roy, P. K. (2017).
1811–1826. The dynamic programming high-order dynamic
Berlingeri, M., Devoto, F., Gasparini, F., Saibene, A., Bayesian networks learning for identifying effec-
Corchs, S. E., Clemente, L., … Borghese, N. A. tive connectivity in human brain from fMRI. Jour-
(2019). Clustering the brain with “CluB”: A new nal of Neuroscience Methods, 285, 33–44.
toolbox for quantitative meta-analysis of neuroim- Dang, S., Chaudhury, S., Lall, B., & Roy, P. K. (2017).
aging data. Frontiers in Neuroscience, 13. doi: Tractography-based score for learning effective
10.3389/fnins.2019.01037 connectivity from multimodal imaging data using
Bhatkoti, P., & Paul, M. (2016). Early diagnosis of Alz- dynamic Bayesian networks. IEEE Transactions on
heimer’s disease: A multi-class deep learning frame- Biomedical Engineering, 65(5), 1057–1068.
work with modified k-sparse autoencoder De Cheveigné, A. (2021). Shared component analy-
classification. Paper presented at International Con- sis. NeuroImage, 226. doi: 10.1016/j.neuroimage.
ference on Image and Vision Computing New Zea- 2020.117614
land (IVCNZ). doi: 10.1109/IVCNZ.2016.7804459 De Pierrefeu, A., Löfstedt, T., Hadj-Selem, F., Dubois,
Bi, X., Shu, Q., Sun, Q., & Xu, Q. (2018). Random M., Jardri, R., Fovet, T., … Duchesnay, E. (2017).
support vector machine cluster analysis of resting- Structured sparse principal components analysis
state fMRI in Alzheimer’s disease. PLoS One, 13(3). with the TV-elastic net penalty. IEEE Transactions
doi: 10.1371/journal.pone.0194479 on Medical Imaging, 37(2), 396–407.
Bunea, F., She, Y., Ombao, H., Gongvatana, A., Dewey, B. E., Zhao, C., Reinhold, J. C., Carass, A.,
Devlin, K., & Cohen, R. (2011). Penalized least Fitzgerald, K. C., Sotirchos, E. S., … Calabresi, P. A.
squares regression methods and applications to (2019). DeepHarmony: A deep learning approach
neuroimaging. NeuroImage, 55(4), 1519–1527. to contrast harmonization across scanner changes.
Caan, M. W., Bazin, P. L., Marques, J. P., de Hollander, Magnetic Resonance Imaging, 64, 160–170.
G., Dumoulin, S. O., & van der Zwaag, W. (2019). Diederichs, E., Juditsky, A., Spokoiny, V., & Schutte,
MP2RAGEME: T1, T2*, and QSM mapping in one C. (2010). Sparse non-Gaussian component analy-
sequence at 7 Tesla. Human Brain Mapping, 40(6), sis. IEEE Transactions on Information Theory,
1786–1798. 56(6), 3033–3047.
Calhoun, V. D., Liu, J., & Adalı, T. (2009). A review of Dimitriadis, S. I., Salis, C. I., & Liparas, D. (2021). A
group ICA for fMRI data and ICA for joint inference sleep disorder detection model based on EEG
of imaging, genetic, and ERP data. NeuroImage, cross-frequency coupling and random forest.
45(1), S163–S172. medRxiv. doi: 10.1101/2020.06.10.20126268
Cao, Y., Summerfield, C., Park, H., Giordano, B. L., & Dinsdale, N. K., Jenkinson, M., & Namburete, A. I.
Kayser, C. (2019). Causal inference in the multi- (2021). Deep learning-based unlearning of dataset
sensory brain. Neuron, 102(5), 1076–1087. bias for MRI harmonisation and confound removal.
Cetin-Karayumak, S., Stegmayer, K., Walther, S., NeuroImage, 228. doi: 10.1016/j.
Szeszko, P. R., Crow, T., James, A., … Rathi, Y. neuroimage.2020.117689
(2020). Exploring the limits of ComBat method for Dockès, J. m., Varoquaux, G., & Poline, J.-B. (2021).
multi-site diffusion MRI harmonization. bioRxiv. Preventing dataset shift from breaking machine-
doi: 10.1101/2020.11.20.390120 learning biomarkers. GigaScience, 10(9). doi:
Chu, C., Ni, Y., Tan, G., Saunders, C. J., & Ashburner, J. 10.1093/gigascience/giab055
(2011). Kernel regression for fMRI pattern prediction. Dong, Q., Ge, F., Ning, Q., Zhao, Y., Lv, J., Huang, H.,
NeuroImage, 56(2), 662–673. … Liu, T. (2019). Modeling hierarchical brain net-
Cordova, M., Shada, K., Demeter, D. V., Doyle, O., works via volumetric sparse deep belief network.
Miranda-Dominguez, O., Perrone, A., … Lang- IEEE Transactions on Biomedical Engineering,
horst, B. (2020). Heterogeneity of executive func- 67(6), 1739–1748.
tion revealed by a functional random forest Du, Y., & Fan, Y. (2013). Group information guided
approach across ADHD and ASD. NeuroImage: ICA for fMRI data analysis. NeuroImage, 69,
Clinical, 26. doi: 10.1016/j.nicl.2020.102245 157–197.
Correa, N. M., Eichele, T., Adalı, T., Li, Y.-O., & Cal- Duffy, B. A., Toga, A. W., & Kim, H. (2020). Gradient
houn, V. D. (2010). Multi-set canonical correlation artifact correction for simultaneous EEG-fMRI using
analysis for the fusion of concurrent single trial denoising autoencoders. Paper presented at IEEE
ERP and functional MRI. NeuroImage, 50(4), 17th International Symposium on Biomedical Imag-
1438–1445. ing (ISBI). doi: 10.1109/ISBI45749.2020.9098447
562 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Dyrba, M., Grothe, M. J., Mohammadi, A., Binder, measures. NeuroImage, 201. doi: 10.1016/j.
H., Kirste, T., Teipel, S. J., & Alzheimer’s Disease neuroimage.2019.116038
Neuroimaging Initiative (2018). Comparison of dif- Garcia-Dias, R., Scarpazza, C., Baecker, L., Vieira, S.,
ferent hypotheses regarding the spread of Alzhei- Pinaya, W. H., Corvin, A., … McDonald, C. (2020).
mer’s disease using Markov random fields and Neuroharmony: A new tool for harmonizing volu-
multimodal imaging. Journal of Alzheimer’s Dis- metric MRI data from unseen scanners. NeuroIm-
ease, 65(3), 731–746. age, 220. doi: 10.1016/j.neuroimage.2020.117127
Ezaki, T., Watanabe, T., Ohzeki, M., & Masuda, N. Ge, R., Wang, Y., Zhang, J., Yao, L., Zhang, H., &
(2017). Energy landscape analysis of neuroimag- Long, Z. (2016). Improved FastICA algorithm in
ing data. Philosophical Transactions of the Royal fMRI data analysis using the sparsity property of
Society A: Mathematical, Physical, and Engineer- the sources. Journal of Neuroscience Methods,
ing Sciences, 375. doi: 10.1098/rsta.2016.0287 263, 103–114.
Fan, M., & Chou, C.-A. (2016). Exploring stability- Gil, R., Fernandes, F. F., & Shemesh, N. (2021).
based voxel selection methods in MVPA using ­Neuroplasticity-driven timing modulations revealed
cognitive neuroimaging data: A comprehensive by ultrafast functional magnetic resonance imag-
study. Brain Informatics, 3(3), 193–203. ing. NeuroImage, 225. doi: 10.1016/
Fedorov, A., Hjelm, R. D., Abrol, A., Fu, Z., Du, Y., j.neuroimage.2020.117446
Plis, S., & Calhoun, V. D. (2019). Prediction of Górriz, J., Gómez, J., Ramírez, J., Martinez-Murcia, F.,
progression to Alzheimer’s disease with deep info- Castillo-Barnes, D., Salas-Gonzalez, D., & Segovia,
max. Paper presented at IEEE EMBS International F. (2018). Segmentation of molecular neuroimages
Conference on Biomedical and Health Informatics using hidden Markov random fields in order to
(BHI). doi: 10.1109/BHI.2019.8834630 improve the assisted diagnosis of neurodegenera-
Ferri, R., Babiloni, C., Karami, V., Triggiani, A. I., tive diseases. Paper presented at IEEE Nuclear Sci-
Carducci, F., Noce, G., … Amenta, F. (2021). ence Symposium and Medical Imaging Conference
Stacked autoencoders as new models for an accu- Proceedings (NSS/MIC). doi: 10.1109/
rate Alzheimer’s disease classification support NSSMIC.2018.8824521
using resting-state EEG and MRI measurements. Grasby, K. L., Jahanshad, N., Painter, J. N., Colodro-
Clinical Neurophysiology, 132(1), 232–245. Conde, L., Bralten, J., Hibar, D. P., … McMahon,
Ficiarà, E., Crespi, V., Gadewar, S. P., Thomopoulos, M. A. B. (2020). The genetic architecture of the
S. I., Boyd, J., Thompson, P. M., … Pizzagalli, F. human cerebral cortex. Science, 367. doi: 10.1126/
(2021). Predicting progression from mild cognitive science.aay6690
impairment to Alzheimer’s disease using MRI- Groves, A. R., Beckmann, C. F., Smith, S. M., & Wool-
based cortical features and a two-state Markov rich, M. W. (2011). Linked independent compo-
model. Paper presented at IEEE 18th International nent analysis for multimodal data fusion.
Symposium on Biomedical Imaging (ISBI). doi: NeuroImage, 54(3), 2198–2217.
10.1109/ISBI48211.2021.9434143 Gu, P., Wu, T., Zou, M., Pan, Y., Guo, J., Xiahou, J.,
Fiecas, M. B., Coffman, C., Xu, M., Hendrickson, T. J., … Zhang, L. (2020). Multi-head self-attention
Mueller, B. A., Klimes-Dougan, B., & Cullen, K. R. model for classification of temporal lobe epilepsy
(2021). Approximate hidden semi-Markov models subtypes. Frontiers in Physiology, 11. doi: 10.3389/
for dynamic connectivity analysis in resting-state fphys.2020.604764
fMRI. bioRxiv. doi: 10.1101/2021.03.01.433385 Guan, H., Liu, Y., Yang, E., Yap, P.-T., Shen, D., &
Fortin, J. P., Cullen, N., Sheline, Y. I., Taylor, W. D., Liu, M. (2021). Multi-site MRI harmonization via
Aselcioglu, I., Cook, P. A., … McGrath, P. J. (2018). attention-guided deep domain adaptation for
Harmonization of cortical thickness measurements brain disorder identification. Medical Image Analy-
across scanners and sites. NeuroImage, 167, sis, 71. doi: 10.1016/j.media.2021.102076
104–120. Guerrout, E.-H., Mahiou, R., Melouk, A., & Harmali,
Fortin, J. P., Parker, D., Tunç, B., Watanabe, T., Elliott, I. (2020). 3D brain image segmentation model
M. A., Ruparel, K., … Gur, R. E. (2017). Harmoni- using deep learning and hidden Markov random
zation of multi-site diffusion tensor imaging data. fields. Paper presented at IEEE/ACS 17th Interna-
NeuroImage, 161, 149–170. tional Conference on Computer Systems and
Fox, P. T., Lancaster, J. L., Laird, A. R., & Eickhoff, S. B. Applications (AICCSA). doi: 10.1109/
(2014). Meta-analysis in human neuroimaging: AICCSA50499.2020.9316501
Computational modeling of large-scale databases. Guillaume, B., Hua, X., Thompson, P. M., Waldorp,
Annual Review of Neuroscience, 37, 409–434. L., Nichols, T. E., & Alzheimer’s Disease Neuroim-
Gao, S., Greene, A. S., Constable, R. T., & Scheinost, aging Initiative. (2014). Fast and accurate model-
D. (2019). Combining multiple connectomes ling of longitudinal and repeated measures
improves predictive modeling of phenotypic neuroimaging data. NeuroImage, 94, 287–302.
Novel Approaches to Large-Scale Data in Neuroimaging 563

Guo, H., Li, Y., Xu, Y., Jin, Y., Xiang, J., & Chen, J. Javed, F., Gilani, S. O., Latif, S., Waris, A., Jamil, M.,
(2018). Resting-state brain functional hyper-­ & Waqas, A. (2021). Predicting risk of antenatal
network construction based on elastic net and depression and anxiety using multi-layer percep-
group lasso methods. Frontiers in Neuroinformat- trons and support vector machines. Journal of
ics, 12. doi: 10.3389/fninf.2018.00025 Personalized Medicine, 11(3). doi: 10.3390/
Guo, Y., & Tang, L. (2013). A hierarchical model for jpm11030199
probabilistic independent component analysis of Jia, T., Chu, C., Liu, Y., van Dongen, J., Papastergios,
multi-subject fMRI studies. Biometrics, 69(4), E., Armstrong, N. J., … Harris, M. (2019).
970–981. ­Epigenome-wide meta-analysis of blood DNA
Hadj-Selem, F., Löfstedt, T., Dohmatob, E., Frouin, V., methylation and its association with subcortical
Dubois, M., Guillemot, V., & Duchesnay, E. (2018). volumes: Findings from the ENIGMA Epigenetics
Continuation of Nesterov’s smoothing for regres- Working Group. Molecular Psychiatry, 26,
sion with structured sparsity in high-dimensional 3884–3895.
neuroimaging. IEEE Transactions on Medical Imag- Jo, T., Nho, K., Risacher, S. L., & Saykin, A. J. (2020).
ing, 37(11), 2403–2413. Deep learning detection of informative features in
Hooker, J. M., & Carson, R. E. (2019). Human tau PET for Alzheimer’s disease classification. BMC
positron emission tomography neuroimaging. Bioinformatics, 21(S21), 1–13.
Annual Review of Biomedical Engineering, 21, Jo, T., Nho, K., & Saykin, A. J. (2019). Deep learning
551–581. in Alzheimer’s disease: Diagnostic classification
Hoshi, Y., & Michael, F. (2005). Functional near- and prognostic prediction using neuroimaging
infrared spectroscopy: potential and limitations in data. Frontiers in Aging Neuroscience, 11. doi:
neuroimaging studies. International Review of 10.3389/fnagi.2019.00220
Neurobiology, 66(5), 237–266. Johnson, W. E., Li, C., & Rabinovic, A. (2007). Adjust-
Huang, Z.-A., Zhu, Z., Yau, C. H., & Tan, K. C. (2020). ing batch effects in microarray expression data
Identifying autism spectrum disorder from resting- using empirical Bayes methods. Biostatistics, 8(1),
state fMRI using deep belief network. IEEE Trans- 118–127.
actions on Neural Networks and Learning Systems. Kamarajan, C., Ardekani, B. A., Pandey, A. K., Kinre-
32, 2487–2861. ich, S., Pandey, G., Chorlian, D. B., … Stimus, A. T.
Hupé, J.-M. (2015). Statistical inferences under the (2020). Random forest classification of alcohol use
Null hypothesis: Common mistakes and pitfalls in disorder using fMRI functional connectivity, neu-
neuroimaging studies. Frontiers in Neuroscience, ropsychological functioning, and impulsivity meas-
9. doi: 10.3389/fnins.2015.00018 ures. Brain Sciences, 10(2). doi: 10.3390/
Hutchison, R. M., Womelsdorf, T., Allen, E. A., Ban- brainsci10020115
dettini, P. A., Calhoun, V. D., Corbetta, M., … Kang, K., Cai, J., Song, X., & Zhu, H. (2019). Bayesian
Gonzalez-Castillo, J. (2013). Dynamic functional hidden Markov models for delineating the pathol-
connectivity: Promise, issues, and interpretations. ogy of Alzheimer’s disease. Statistical Methods in
NeuroImage, 80, 360–378. Medical Research, 28(7), 2112–2124.
Ithapu, V. K., Singh, V., Okonkwo, O., & Johnson, S. C. Kang, K., Song, X., Hu, X. J., & Zhu, H. (2019). Bayes-
(2014). Randomized denoising autoencoders for ian adaptive group lasso with semiparametric
smaller and efficient imaging-based AD clinical hidden Markov models. Statistics in Medicine,
trials. Paper presented at International Conference 38(9), 1634–1650.
on Medical Image Computing and Computer- Karami, V., Mahdavifar, R., Habibzadeh, A., & Nabavi,
Assisted Intervention (ICMICCAI). doi: S. M. (2020). Identification of Multiple Sclerosis
10.1007/978-3-319-10470-6_59 lesion subtypes and their quantitative assessments
Jalbrzikowski, M., Hayes, R. A., Wood, S. J., Nord- with EDSS using neuroimaging. Network Mode-
holm, D., Zhou, J. H., Fusar-Poli, P., … Kwak, Y. B. ling Analysis in Health Informatics and Bioinfor-
(2021). Association of structural magnetic reso- matics, 9. doi: 10.1007/s13721-020-00245-8
nance imaging measures with psychosis onset in Ke, Q., Zhang, J., Wei, W., Damaševičius, R., &
individuals at clinical high risk for developing psy- Woźniak, M. (2019). Adaptive independent sub-
chosis: An ENIGMA working group mega-analysis. space analysis of brain magnetic resonance imag-
JAMA Psychiatry. 78(7), 753–766. ing data. IEEE Access, 7, 12252–12261.
Jamaloo, F., Mikaeili, M., & Noroozian, M. (2020). Khan, M. A., Islam, M. R., & Molla, M. K. I. (2016).
Multi metric functional connectivity analysis based Artifact suppression from electroencephalography
on continuous hidden Markov model with applica- signals using stationary subspace analysis. Paper
tion in early diagnosis of Alzheimer’s disease. Bio- presented at 19th International Conference on
medical Signal Processing and Control, 61. doi: Computer and Information Technology (ICCIT).
10.1016/j.bspc.2020.102056 doi: 10.1109/ICCITECHN.2016.7860204
564 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Khosla, M., Jamison, K., Kuceyeski, A., & Sabuncu, Lin, L., Zhang, G., Wang, J., Tian, M., & Wu, S.
M. R. (2019). Ensemble learning with 3D convolu- (2021). Utilizing transfer learning of pre-trained
tional neural networks for functional connectome- AlexNet and relevance vector machine for regres-
based prediction. NeuroImage, 199, 651–662. sion for predicting healthy older adult’s brain age
Kim, J.-H., Zhang, Y., Han, K., Wen, Z., Choi, M., & Liu, Z. from structural MRI. Multimedia Tools and Appli-
(2021). Representation learning of resting state fMRI cations, 80(1), 24719–24735.
with variational autoencoder. NeuroImage, 241. doi: Lindquist, M. A., & Sobel, M. E. (2011). Graphical
10.1016/j.neuroimage.2021.118423 models, potential outcomes and causal inference:
Koshiyama, D., Fukunaga, M., Okada, N., Morita, K., Comment on Ramsey, Spirtes and Glymour. Neu-
Nemoto, K., Usui, K., … Kudo, N. (2020). White roImage, 57(2), 334–336.
matter microstructural alterations across four Liu, C., JaJa, J., & Pessoa, L. (2018). LEICA: Laplacian
major psychiatric disorders: Mega-analysis study in eigenmaps for group ICA decomposition of fMRI
2937 individuals. Molecular Psychiatry, 25(4), data. NeuroImage, 169, 363-373.
883–895. Liu, J., Ji, J., Xun, G., Yao, L., Huai, M., & Zhang, A.
Koush, Y., Masala, N., Scharnowski, F., & Van De (2020). EC-GAN: Inferring brain effective connec-
Ville, D. (2019). Data-driven tensor independent tivity via generative adversarial networks. Proceed-
component analysis for model-based connectivity ings of the AAAI Conference on Artificial
neurofeedback. NeuroImage, 184, 214–226. Intelligence. doi: 10.1609/aaai.v34i04.5921
Laforce Jr, R., Tosun, D., Ghosh, P., Lehmann, M., Liu, M., Li, F., Yan, H., Wang, K., Ma, Y., Shen, L., …
Madison, C. M., Weiner, M. W., … Rabinovici, Alzheimer’s Disease Neuroimaging Initiative. (2020).
G. D. (2014). Parallel ICA of FDG-PET and PiB-PET A multi-model deep convolutional neural network
in three conditions with underlying Alzheimer’s for automatic hippocampus segmentation and clas-
pathology. NeuroImage: Clinical, 4, 508–516. sification in Alzheimer’s disease. NeuroImage, 208.
Lahat, D., & Jutten, C. (2016). Joint independent doi: 10.1016/j.neuroimage.2019.116459
subspace analysis using second-order statistics. Liu, X., Goncalves, A. R., Cao, P., Zhao, D., Banerjee,
IEEE Transactions on Signal Processing, 64(18), A., & Alzheimer’s Disease Neuroimaging Initiative.
4891–4904. (2018). Modeling Alzheimer’s disease cognitive
Lariviere, S., Paquola, C., Park, B., Royer, J., Wang, Y., scores using multi-task sparse group lasso. Com-
Benkarim, O., … Kirschner, M. (2020). The puterized Medical Imaging and Graphics, 66,
ENIGMA Toolbox: Cross-disorder integration and 100–114.
multiscale neural contextualization of multisite Lukashenko, O., & Chernyaev, S. (2020). fMRI image
neuroimaging datasets. Nature Methods, 18, segmentation based on hidden Markov random
698–700. field with directional statistics observation model.
Latha, M., & Kavitha, G. (2019). Detection of Schizo- Paper presented at 12th International Conference
phrenia in brain MR images based on segmented on Machine Vision (ICMV 2019). doi:
ventricle region and deep belief networks. Neural 10.1117/12.2559545
Computing and Applications, 31(9), 5195–5206. Ma, S., Li, X.-L., Correa, N. M., Adali, T., & Calhoun,
Lee, M. B., Jahng, G.-H., Kim, H. J., & Kwon, O.-I. V. D. (2010). Independent subspace analysis with
(2021). High-frequency conductivity at Larmor-­ prior information for fMRI data. Paper presented
frequency in human brain using moving local at IEEE International Conference on Acoustics,
window multilayer perceptron neural network. PLoS Speech and Signal Processing. doi: 10.1109/
One, 16(5). doi: 10.1371/journal.pone.0251417 ICASSP.2010.5495320
Legarreta, J. H., Petit, L., Rheault, F., Theaud, G., Martin, P., Bender, B., & Focke, N. K. (2015). Post-
Lemaire, C., Descoteaux, M., & Jodoin, P.M. processing of structural MRI for individualized
(2021). Filtering in tractography using autoencod- diagnostics. Quantitative Imaging in Medicine and
ers (FINTA). Medical Image Analysis. doi: 10.1016/j. Surgery, 5(2), 188–203.
media.2021.102126 McKinley, R., Häni, L., Gralla, J., El-Koussy, M., Bauer,
Leuthardt, E. C., Guzman, G., Bandt, S. K., Hacker, S., Arnold, M., … Reyes, M. (2017). Fully auto-
C., Vellimana, A. K., Limbrick, D., … Roland, J. mated stroke tissue estimation using random
(2018). Integration of resting state functional MRI forest classifiers (FASTER). Journal of Cerebral
into clinical practice: A large single institution Blood Flow and Metabolism, 37(8), 2728–2741.
experience. PLoS One, 13(6). doi: 10.1371/journal. Moran, R. J., Pinotsis, D. A., & Friston, K. J. (2013).
pone.0198349 Neural masses and fields in dynamic causal mod-
Li, Y., Liu, J., Gao, X., Jie, B., Kim, M., Yap, P.-T., … eling. Frontiers in Computational Neuroscience, 7.
Shen, D. (2019). Multimodal hyper-connectivity of doi: 10.3389/fncom.2013.00057
functional networks using functionally-weighted Mourao-Miranda, J., Portual, L., Rondina, J., &
LASSO for MCI classification. Medical Image Anal- Shawe-Taylor, J. (2012). Elastic-net multiple kernel
ysis, 52, 80–96. learning for multi-region neuroimaging-based
Novel Approaches to Large-Scale Data in Neuroimaging 565

diagnosis. Proceedings of the 2nd NIPS Workshop Pinto, M. S., Paolella, R., Billiet, T., Van Dyck, P., Guns,
on Machine Learning and Interpretation in Neuro- P.-J., Jeurissen, B., … Sijbers, J. (2020). Harmoniza-
imaging. doi: 10.13140/RG.2.1.5137.7365 tion of brain diffusion MRI: Concepts and methods.
Nentwich, M., Ai, L., Madsen, J., Telesford, Q. K., Frontiers in Neuroscience, 14. doi: 10.3389/
Haufe, S., Milham, M. P., & Parra, L. C. (2020). Func- fnins.2020.00396
tional connectivity of EEG is subject-specific, associ- Pomponio, R., Erus, G., Habes, M., Doshi, J., Srinivasan,
ated with phenotype, and different from fMRI. D., Mamourian, E., … Fan, Y. (2020). Harmonization
NeuroImage, 218. doi: 10.1016/j.neuroimage. of large MRI datasets for the analysis of brain imag-
2020.117001 ing patterns throughout the lifespan. NeuroImage,
Nguyen, M., He, T., An, L., Alexander, D. C., Feng, J., 208. doi: 10.1016/j.neuroimage.2019.116450
Yeo, B. T., & Alzheimer’s Disease Neuroimaging Properzi, M. J., Buckley, R. F., Chhatwal, J. P., Dono-
Initiative. (2020). Predicting Alzheimer’s disease hue, M. C., Lois, C., Mormino, E. C., … Schultz, A.
progression using deep recurrent neural networks. P. (2019). Nonlinear distributional mapping
NeuroImage, 222. doi: 10.1016/j.neuroimage. (NoDiM) for harmonization across amyloid-PET
2020.117203 radiotracers. NeuroImage, 186, 446–454.
Ning, L., Bonet-Carne, E., Grussu, F., Sepehrband, F., Qi, S., Calhoun, V. D., Van Erp, T. G., Damaraju, E.,
Kaden, E., Veraart, J., … Kokkinos, I. (2020). Cross- Bustillo, J., Du, Y., … Voyvodic, J. (2016). Super-
scanner and cross-protocol multi-shell diffusion vised multimodal fusion and its application in
MRI data harmonization: Algorithms and results. searching joint neuromarkers of working memory
NeuroImage, 221. doi: 10.1016/j.neuroimage. deficits in schizophrenia. Paper presented at 38th
2020.117128 Annual International Conference of the IEEE Engi-
Nobakht, S., Schaeffer, M., Forkert, N. D., Nestor, S., neering in Medicine and Biology Society (EMBC).
E Black, S., Barber, P., & Alzheimer’s Disease Neu- doi: 10.1109/EMBC.2016.7591609
roimaging Initiative (2021). Combined atlas and Qiang, N., Dong, Q., Zhang, W., Ge, B., Ge, F., Liang,
convolutional neural network-based segmentation H., … Liu, T. (2020). Modeling task-based fMRI
of the hippocampus from MRI according to the data via deep belief network with neural architec-
ADNI harmonized protocol. Sensors, 21(7). doi: ture search. Computerized Medical Imaging and
10.3390/s21072427 Graphics, 83. doi: 10.1016/j.compmedimag.
Pecheva, D., Kelly, C., Kimpton, J., Bonthrone, A., 2020.101747
Batalle, D., Zhang, H., & Counsell, S. J. (2018). Rasero, J., Sentis, A. I., Yeh, F.-C., & Verstynen, T.
Recent advances in diffusion neuroimaging: (2021). Integrating across neuroimaging modali-
applications in the developing preterm brain. ties boosts prediction accuracy of cognitive ability.
F1000Research. doi: 10.12688/f1000research. PLoS Computational Biology, 17(3). doi: 10.1371/
15073.1 journal.pcbi.1008347
Phang, C.-R., Noman, F., Hussain, H., Ting, C.-M., & Reddan, M. C., Lindquist, M. A., & Wager, T. D.
Ombao, H. (2019). A multi-domain connectome (2017). Effect size estimation in neuroimaging.
convolutional neural network for identifying schiz- JAMA Psychiatry, 74(3), 207–208.
ophrenia from EEG connectivity patterns. IEEE Ren, Y., Tao, Z., Zhang, W., & Liu, T. (2021). Modeling
Journal of Biomedical and Health Informatics, hierarchical spatial and temporal patterns of natu-
24(5), 1333-1343. ralistic fMRI volume via volumetric deep belief
Pietrosanu, M., Zhang, L., Seres, P., Elkady, A., network with neural architecture search. Paper
Wilman, A. H., Kong, L., & Cobzas, D. (2021). presented at IEEE 18th International Symposium
Stable anatomy detection in multimodal imaging on Biomedical Imaging (ISBI). doi: 10.1109/
through sparse group regularization: A compara- ISBI48211.2021.9433811
tive study of iron accumulation in the aging brain. Richhariya, B., Tanveer, M., Rashid, A., & Alzheimer’s
Frontiers in Human Neuroscience, 15. doi: Disease Neuroimaging Initiative (2020). Diagnosis
10.3389/fnhum.2021.641616 of Alzheimer’s disease using universum support
Pinaya, W. H., Mechelli, A., & Sato, J. R. (2019). vector machine based recursive feature elimination
Using deep autoencoders to identify abnormal (USVM-RFE). Biomedical Signal Processing and
brain structural patterns in neuropsychiatric disor- Control, 59. doi: 10.1016/j.bspc.2020.101903
ders: A large-scale multi-sample study. Human Risk, B., & Gaynanova, I. (2020). Simultaneous non-
Brain Mapping, 40(3), 944–954. Gaussian component analysis (SING) for data
Pinti, P., Tachtsidis, I., Hamilton, A., Hirsch, J., Aichel- integration in neuroimaging. arXiv. doi: 10.1214/
burg, C., Gilbert, S., & Burgess, P. W. (2020). The 21-AOAS1466
present and future use of functional near-infrared Roine, T., Jeurissen, B., Perrone, D., Aelterman, J.,
spectroscopy (fNIRS) for cognitive neuroscience. Philips, W., Leemans, A., & Sijbers, J. (2015).
Annals of the New York Academy of Sciences, Informed constrained spherical deconvolution
1464(1), 5–29. (iCSD). Medical Image Analysis, 24(1), 269–281.
566 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Roudbar, M. A., Mousavi, S. F., Ardestani, S. S., placement prediction in deep brain stimulation
Lopes, F. B., Momen, M., Gianola, D., & Khatib, H. using support vector regression. Proceedings of
(2021). Prediction of biological age and evaluation Third International Symposium on Image Comput-
of genome-wide dynamic methylomic changes ing and Digital Medicine. doi: 10.1145/3364836.
throughout human aging. G3 Genes, Genomes, 3364876
Genetics. doi: 10.1093/g3journal/jkab112 Smit, D. J., Wright, M. J., Meyers, J. L., Martin, N. G.,
Rudie, J. D., Weiss, D. A., Saluja, R., Rauschecker, A. M., Ho, Y. Y., Malone, S. M., … de Geus, E. J. (2018).
Wang, J., Sugrue, L., … Colby, J. B. (2019). Multi- Genome-wide association analysis links multiple
disease segmentation of gliomas and white matter psychiatric liability genes to oscillatory brain activ-
hyperintensities in the BraTS data using a 3D con- ity. Human Brain Mapping, 39(11), 4183–4195.
volutional neural network. Frontiers in Computa- Smith, S. M., & Nichols, T. E. (2018). Statistical chal-
tional Neuroscience, 13. doi: 10.3389/fncom. lenges in “big data” human neuroimaging.
2019.00084 Neuron, 97(2), 263–268.
Ryali, S., Chen, T., Supekar, K., & Menon, V. (2012). Sørensen, L., Nielsen, M., & Alzheimer’s Disease Neu-
Estimation of functional connectivity in fMRI data roimaging Initiative (2018). Ensemble support
using stability selection-based sparse partial cor- vector machine classification of dementia using
relation with elastic net penalty. NeuroImage, structural MRI and mini-mental state examination.
59(4), 3852–3861. Journal of Neuroscience Methods, 302, 66–74.
Sarraf, S., Saverino, C., Ghaderi, H., & Anderson, J. Sperber, C., Wiesen, D., & Karnath, H. O. (2019). An
(2014). Brain network extraction from probabilistic empirical evaluation of multivariate lesion behav-
ICA using functional Magnetic Resonance Images iour mapping using support vector regression.
and advanced template matching techniques. Human Brain Mapping. 40(5), 1381–1390.
Paper presented at IEEE 27th Canadian Confer- Squarcina, L., Castellani, U., Bellani, M., Perlini, C.,
ence on Electrical and Computer Engineering Lasalvia, A., Dusi, N., … Rambaldelli, G. (2017).
(CCECE). doi: 10.1109/CCECE.2014.6901003 Classification of first-episode psychosis in a large
Sarwar, T., Ramamohanarao, K., & Zalesky, A. (2019). cohort of patients using support vector machine
Mapping connectomes with diffusion MRI: Deter- and multiple kernel learning techniques. NeuroIm-
ministic or probabilistic tractography? Magnetic age, 145, 238–245.
Resonance in Medicine, 81(2), 1368–1384. Steardo Jr, L., Carbone, E. A., de Filippis, R., Pisanu,
Seth, A. K., Barrett, A. B., & Barnett, L. (2015). C., Segura-Garcia, C., Squassina, A., … Steardo, L.
Granger causality analysis in neuroscience and (2020). Application of support vector machine on
neuroimaging. Journal of Neuroscience, 35(8), fMRI data as biomarkers in schizophrenia diagno-
3293–3297. sis: A systematic review. Frontiers in Psychiatry, 11.
Shen, T., Jiang, J., Lin, W., Ge, J., Wu, P., Zhou, Y., … doi: 10.3389/fpsyt.2020.00588
Shi, K. (2019). Use of overlapping group LASSO Su, J. H., Thomas, F. T., Kasoff, W. S., Tourdias, T.,
sparse deep belief network to discriminate Parkin- Choi, E. Y., Rutt, B. K., & Saranathan, M. (2019).
son’s disease and normal control. Frontiers in Thalamus Optimized Multi Atlas Segmentation
Neuroscience, 13. doi: 10.3389/fnins.2019.00396 (THOMAS): Fast, fully automated segmentation of
Shi, L., Zhao, L., Yeung, F. K., Wong, S. Y., Chan, thalamic nuclei from structural MRI. NeuroImage,
R. K., Tse, M. F., … Liu, K. (2018). Mapping the 194, 272–282.
contribution and strategic distribution patterns of Subramanian, I., Verma, S., Kumar, S., Jere, A., &
neuroimaging features of small vessel disease in Anamika, K. (2020). Multi-omics data integration,
poststroke cognitive impairment. Journal of Neu- interpretation, and its application. Bioinformatics
rology, Neurosurgery and Psychiatry, 89(9), and Biology Insights, 14. doi: 10.1177/
918–926. 1177932219899051
Silva, R. F., Plis, S. M., Adalı, T., Pattichis, M. S., & Sun, D., Rakesh, G., Clarke-Rubright, E. K., Haswell,
Calhoun, V. D. (2020). Multidataset Independent C. C., Logue, M., O’Leary, B. M., … Jahanshad, N.
Subspace Analysis with application to multimodal (2021). A comparison of methods to harmonize
fusion. IEEE Transactions on Image Processing, 30, cortical thickness measurements across scanners
588–602. and sites. bioRxiv. doi: 10.1101/2021.09.22.461242
Silva, R. F., Plis, S. M., Sui, J., Pattichis, M. S., Adali, Sun, H., Wang, A., Wang, W., & Liu, C. (2021). An
T., & Calhoun, V. D. (2016). Blind source separa- improved deep residual network prediction model
tion for unimodal and multimodal brain networks: for the early diagnosis of Alzheimer’s disease. Sen-
A unifying framework for subspace modeling. IEEE sors, 21(12). doi: 10.3390/s21124182
Journal of Selected Topics in Signal Processing, Symms, M., Jäger, H., Schmierer, K., & Yousry, T.
10(7), 1134–1149. (2004). A review of structural magnetic resonance
Singer, A., Zhang, C., Wang, T., Qiu, S., Li, D., Du, Y., neuroimaging. Journal of Neurology, Neurosur-
… Feng, Y. (2019). Post-operative electrode gery, and Psychiatry, 75(9), 1235–1244.
Novel Approaches to Large-Scale Data in Neuroimaging 567

Szucs, D., & Ioannidis, J. P. (2020). Sample size evolu- analyses using robust regression. NeuroImage,
tion in neuroimaging research: An evaluation of 26(1), 99–113.
highly-cited studies (1990–2012) and of latest Wang, X., Xie, H., Chen, T., Cotton, A. S., Salminen,
practices (2017–2018) in high-impact journals. L. E., Logue, M. W., … O’Leary, B. M. (2020).
NeuroImage, 221. doi: 10.1016/j.neuroimage. Cortical volume abnormalities in posttraumatic
2020.117164 stress disorder: An ENIGMA-psychiatric genomics
Tao, Q., Si, Y., Li, F., Li, P., Li, Y., Zhang, S., … Xu, P. consortium PTSD workgroup mega-analysis.
(2021). Decision-feedback stages revealed by Molecular Psychiatry, 26, 4331–4343.
hidden Markov modeling of EEG. International Wang, Y., & Guo, Y. (2019). A hierarchical independ-
Journal of Neural Systems, 31(7). doi: 10.1142/ ent component analysis model for longitudinal
S0129065721500313 neuroimaging studies. NeuroImage, 189,
Tessadori, J., Yamin, M. A., Valsasina, P., Filippi, M., 380–400.
Rocca, M. A., & Sona, D. (2021). Dynamic func- Wegmann, B., Eklund, A., & Villani, M. (2017). Bayes-
tional connectivity for the classification of multiple ian Rician regression for neuroimaging. Frontiers in
sclerosis phenotype: A hidden Markov model Neuroscience, 11, 586. doi: 10.3389/fnins.
approach. Paper presented at IEEE 18th Interna- 2017.00586
tional Symposium on Biomedical Imaging (ISBI). Wrobel, J., Martin, M., Bakshi, R., Calabresi, P. A.,
doi: 10.1109/ISBI48211.2021.9434160 Elliot, M., Roalf, D., … Nair, G. (2020). Intensity
Thomas, A. W., Heekeren, H. R., Müller, K.-R., & warping for multisite MRI harmonization. NeuroIm-
Samek, W. (2019). Analyzing neuroimaging data age, 223. doi: 10.1016/j.neuroimage.2020.117242
through recurrent deep learning models. Frontiers Yang, D., Huang, R., Yoo, S.-H., Shin, M.-J., Yoon, J.
in Neuroscience, 13. doi: 10.3389/fnins.2019. A., Shin, Y.-I., & Hong, K.-S. (2020). Detection of
01321 mild cognitive impairment using convolutional
Thompson, P. M., Jahanshad, N., Ching, C. R., Salm- neural network: Temporal-feature maps of func-
inen, L. E., Thomopoulos, S. I., Bright, J., … Bruin, tional near-infrared spectroscopy. Frontiers in
W. B. (2020). ENIGMA and global neuroscience: A Aging Neuroscience, 12. doi: 10.3389/fnagi.
decade of large-scale studies of the brain in health 2020.00141
and disease across more than 40 countries. Yang, J., Carl, B., Nimsky, C., & Bopp, M. H. (2020).
Translational Psychiatry, 10(1). doi: 10.1038/ The impact of position-orientation adaptive
s41398-020-0705-1 smoothing in diffusion weighted imaging: From
Tournier, J.-D., Yeh, C.-H., Calamante, F., Cho, K.-H., diffusion metrics to fiber tractography. PLoS One,
Connelly, A., & Lin, C.-P. (2008). Resolving cross- 15(5). doi: 10.1371/journal.pone.0233474
ing fibres using constrained spherical deconvolu- Yeo, B. T., Krienen, F. M., Sepulcre, J., Sabuncu, M. R.,
tion: Validation using diffusion-weighted imaging Lashkari, D., Hollinshead, M., … Buckner, R. L.
phantom data. NeuroImage, 42(2), 617–625. (2011). The organization of the human cerebral
Umapathy, L., Perez-Carrillo, G., Keerthivasan, M., cortex estimated by intrinsic functional connectivity.
Rosado-Toro, J., Altbach, M., Winegar, B., … Alz- Journal of Neurophysiology, 106(3), 1125–1165.
heimer’s Disease Neuroimaging Initiative. (2021). Yu, M., Linn, K. A., Cook, P. A., Phillips, M. L., McIn-
A stacked generalization of 3D orthogonal deep nis, M., Fava, M., … Sheline, Y. I. (2018). Statistical
learning convolutional neural networks for harmonization corrects site effects in functional
improved detection of white matter hyperintensi- connectivity measurements from multi-site fMRI
ties in 3D FLAIR images. American Journal of data. Human Brain Mapping, 39(11), 4213–4227.
Neuroradiology, 42(4), 639–647. Yun, J., Park, J. E., Lee, H., Ham, S., Kim, N., & Kim,
Van Der Meer, D., Sønderby, I. E., Kaufmann, T., H. S. (2019). Radiomic features and multilayer
Walters, G. B., Abdellaoui, A., Ames, D., … Ber- perceptron network classifier: A robust MRI classi-
nard, M. (2020). Association of copy number vari- fication strategy for distinguishing glioblastoma
ation of the 15q11.2BP1-BP2 region with cortical from primary central nervous system lymphoma.
and subcortical morphology and cognition. JAMA Scientific Reports, 9(1). doi: 10.1038/s41598-
Psychiatry, 77(4), 420–430. 019-42276-w
Wachinger, C., Becker, B. G., Rieckmann, A., & Pöl- Zeng, N., Li, H., & Peng, Y. (2021). A new deep belief
sterl, S. (2019). Quantifying confounding bias in network-based multi-task learning for diagnosis of
neuroimaging datasets with causal inference. Alzheimer’s disease. Neural Computing and Appli-
Paper presented at International Conference on cations. doi: 10.1007/s00521-021-06149-6
Medical Image Computing and Computer-Assisted Zhang, H., Schneider, T., Wheeler-Kingshott, C. A., &
Intervention (ICMICCAI). doi: 10.1007/ Alexander, D. C. (2012). NODDI: Practical in vivo
978-3-030-32251-9_53 neurite orientation dispersion and density imaging
Wager, T. D., Keller, M. C., Lacey, S. C., & Jonides, J. of the human brain. NeuroImage, 61(4),
(2005). Increased sensitivity in neuroimaging 1000–1016.
568 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Zhang, J., Zheng, B., Gao, A., Feng, X., Liang, D., & Zhao, Y., Lindquist, M. A., & Caffo, B. S. (2020). Sparse
Long, X. (2021). A 3D densely connected convolu- principal component based high-dimensional medi-
tion neural network with connection-wise attention ation analysis. Computational Statistics and Data
mechanism for Alzheimer’s disease classification. Analysis, 142. doi: 10.1016/j.csda.2019.106835
Magnetic Resonance Imaging, 78, 119–126. Zhao, Y., Ma, B., Jiang, P., Zeng, D., Wang, X., & Li, S.
Zhang, L., Lim, C. Y., Maiti, T., Li, Y., Choi, J., Bozoki, (2020). Prediction of Alzheimer’s Disease Progres-
A., … Alzheimer’s Disease Neuroimaging Initiative sion with Multi-Information Generative Adversar-
(2019). Analysis of conversion of Alzheimer’s dis- ial Network. IEEE Journal of Biomedical and Health
ease using a multi-state Markov model. Statistical Informatics, 25(3), 711–719.
Methods in Medical Research, 28(9), 2801–2819. Zheng-Lin, T., Hai-Xing, W., Shao-Xun, Y., Xiao, S., &
Zhang, X., Han, L., Zhu, W., Sun, L., & Zhang, D. Jian-Ming, X. (2018). Classification of Alzheimer’s
(2021). An explainable 3D residual self-attention Disease Based on Stacked Denoising Autoencoder.
deep neural network for joint atrophy localization Paper presented at 4th Annual International Confer-
and Alzheimer’s disease diagnosis using structural ence on Network and Information Systems for Com-
MRI. IEEE Journal of Biomedical and Health Infor- puters (ICNISC). doi: 10.1109/ICNISC.2018.00056
matics. doi: 10.1109/JBHI.2021.3066832 Zhong, J., Wang, Y., Li, J., Xue, X., Liu, S., Wang, M.,
Zhang, Z., Zhu, Q., Xie, G.-S., Chen, Y., Li, Z., & … Li, X. (2020). Inter-site harmonization based on
Wang, S. (2020). Discriminative margin-sensitive dual generative adversarial networks for diffusion
autoencoder for collective multi-view disease anal- tensor imaging: application to neonatal white
ysis. Neural Networks, 123, 94–107. matter development. Biomedical Engineering
Zhao, C., Carass, A., Lee, J., Jog, A., & Prince, J. L. Online, 19(1). doi: 10.1186/s12938-020-0748-9
(2017). A supervoxel based random forest synthe- Zhou, C., Cheng, Y., Ping, L., Xu, J., Shen, Z., Jiang, L., …
sis framework for bidirectional MR/CT synthesis. Xu, X. (2018). Support vector machine classification of
Paper presented at International Workshop on obsessive-compulsive disorder based on whole-brain
Simulation and Synthesis in Medical Imaging. doi: volumetry and diffusion tensor imaging. Frontiers
10.1007/978-3-319-68127-6_4 in Psychiatry, 9. doi: 10.3389/fpsyt.2018.00524
Zhao, Q., Adeli, E., & Pohl, K. M. (2020). Training Zhu, X., Du, X., Kerich, M., Lohoff, F. W., & Momenan,
confounder-free deep learning models for medical R. (2018). Random forest-based classification of alco-
applications. Nature Communications, 11(1). doi: hol dependence patients and healthy controls using
10.1038/s41467-020-19784-9 resting state MRI. Neuroscience Letters, 676, 27–33.
34
Advancing Cognitive Neuroscience
R o t e m B o t v i n i k - N e z e r a n d To r D . W a g e r

INTRODUCTION research areas, but very few attempts have been


made to develop theories and models that can be
tested and that potentially can explain and inter-
Since the establishment of cognitive neuroscience,
relate these distinct chunks of information.
researchers have mapped associations between
Over recent decades, there has been a rise in
brain topography and a variety of cognitive, affec-
synthesis-oriented thinking. Systems biology
tive, and social tasks. The field has grown rapidly,
involves creating models of interacting compo-
with rising numbers of laboratories, researchers,
nents such as proteins and molecules. Systems
data, tools, and publications. Exciting findings neuroscience has shifted in focus from isolated
have been uncovered and tremendous methodo- brain regions and single monosynaptic pathways
logical and conceptual developments have been to broader characterizations of large-scale brain
made. Overall, the field has laid a solid layer of dynamics, supported by advances such as wide-
foundation for the central goal of cognitive neuro- field optical imaging and two-photon microscopy.
science: Deciphering how the brain gives rise to Neuroimaging has undergone a similar sea change
mental states. Yet, the most prominent inroads when it comes to brain regions, gradually shifting
have been made into basic aspects of where differ- from focusing on isolated regions as meditators of
ent mental events occur. While this knowledge psychological functions to large-scale interactions,
constraints how we think of mental processes and characterized by networks and brain patterns.
their similarity, advances in understanding how A driver of recent scientific developments has
mental processes occur have been more modest. been prominent technological advancement in the
Cognitive neuroscience has followed a largely digital age. Quantity, complexity, and dimension-
analytic strategy, dissecting systems into small ality of data collected in the field have all grown
components and building deep knowledge about tremendously. Analyses that were impossible until
details within each area. Accordingly, deep valleys recently due to computational limitations are now
of research have been developed, accessible only becoming possible, and computations that used
to more specialized audiences, with area-specific to take weeks or months can now be performed
terms and relative separation from other closely within hours or days. As a consequence, new
related and highly relevant areas. We now have methods are gaining popularity, from Bayesian
an abundance of detailed findings within specific models to machine learning and deep learning.
570 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

This progress carries both opportunities and chal- effects are often smaller and less important for
lenges. First, with these new abilities, researchers real-world behavior than claimed in initial
can now ask research questions they could not publications.
address before. While this undoubtedly advances Cumulative knowledge is often not flashy or
the field and adds priceless knowledge, it also sub- surprising, and may require sacrificing short-term
stantially broadens the skills required in perform- interest value, including opportunities to publish
ing research and the knowledge required from or otherwise gain attention, in favor of long-term
peers to understand the methods and findings. accuracy. Prioritizing replicability of findings and
Second, as magnitude of data increases, automa- unbiased estimation of their effect sizes helps to
tion of research methods, from data collection and align incentives with the goal of developing a
quality control to data processing, statistical anal- shared understanding of how the world actually
ysis and visualizations, is needed more than ever. works.
Programming skills are becoming a necessity, but Various causes have been suggested for this
are not always taught in neuroscience programs. “replication crisis.” First, low statistical power is a
As the quantity and complexity of data increases, major problem (Button et al., 2013; Munafò et al.,,
so does the necessity of reproducible and trans- 2019; Yarkoni, 2009). The estimated median sam-
parent analyses, and tools and practices to identify ple size of fMRI studies in 2015 was merely 28.5
errors, since these are becoming harder to spot and (Poldrack et al., 2017), far from being sufficiently
more potent in changing results. powered to detect larger than typical effect sizes
Cognitive neuroscience is on the verge of a par- in fMRI studies, given the multiple comparison
adigm shift, at multiple levels, advancing toward a challenge inherent such studies. Underpowered
more systematic and integrative approach toward studies do not affect the likelihood of mistakenly
labs and studies, psychological domains, and brain rejecting the null hypothesis. However, they do
representations. We believe that the field must increase the false discovery rate. This problem
develop new approaches to manage the tension is inflated by publication bias (Ioannidis et al.,
between often-competing goals of rigor and repro- 2014). Significant findings are more likely to be
ducibility, innovation, and the practical constraints reported and accepted for publication in scientific
and limitations of human researchers and aca- journals, while reports with many null findings
demia more generally (e.g., incentive and funding often end up in researchers’ “file drawers,” allow-
structures). Below, we describe the recent devel- ing positive results (including a disproportionate
opment and our ideas for such novel approaches. number of false positive results) to dominate the
literature (Fanelli, 2012; Rosenthal, 1979). In the
context where most published studies report sig-
nificant findings, despite many being underpow-
ered, the likelihood of any reported finding being
ENHANCING REPRODUCIBILITY, true is alarmingly low (Ioannidis, 2005).
REPLICABILITY, AND ROBUSTNESS Second, some research practices that increase
the likelihood of false positives are quite common
Reproducibility and replicability of findings in (John et al., 2012). The most common questionable
many fields, from biomedical to social sciences, research practice is p-hacking, whereby researchers
have been called into question over the last decade. run different statistical tests and selectively report
Large studies have shown concerning rates of only ones that yield significant results (Simmons
unsuccessful replications and much lower effect et al., 2011; Simonsohn et al., 2014). Another exam-
sizes than in original studies (Camerer et al., 2016, ple is hypothesizing after the results are known
2018; Errington et al., 2021; Klein et al., 2014, (HARKing), when researchers present a post hoc
2018; Nosek and Errington, 2017; OSC, 2015). For hypothesis (informed by results) as an a priori
example, only 36 out of 100 impactful results in one (Kerr, 1998). These practices are incentivized,
Psychology were replicated with effect sizes half since researchers are measured and promoted based
that of the original on average (OSC, 2015), and in on their publication records, whereby statistically
preclinical cancer biology, only 23/50 results repli- significant results are almost always a prerequisite
cated (for another 143 results, it was not even pos- for publication (Nelson et al., 2018). Furthermore,
sible to conduct a replication) with effect sizes 15% such questionable research practices could happen
that of the original on average (Errington et al., unconsciously, since human inference is biased by
2021). While a failed replication does not necessar- prior beliefs (Nickerson, 1998) and desires (Kunda,
ily imply that the original finding was wrong 1990; Sharot and Garrett, 2016).
(Nosek and Errington, 2020a,b), consistently low Third, analytical flexibility within the field is
replication rates reveal that many reported findings very high (a thriving “garden of forking paths”;
are likely false positives, and that even replicable Gelman and Loken, 2013). Turning data into an
Advancing Cognitive Neuroscience 571

hypothesis test typically requires numerous ana- (Henrich et al., 2010). Hence, it is unclear whether
lytical choices, with multiple justifiable alter- findings within the field would in fact general-
natives at each step. “Optimal” choices often ize across the human population (Arnett, 2008;
do not exist or depend strongly on the specific Barrett, 2020; Chiao and Cheon, 2010; Rad et al.,
domain, hypothesis, dataset, sample size, effect 2018; Thalmayer et al., 2021).
sizes, and artifact patterns, among other features. Awareness of these problems is increasing, and
Furthermore, analysis is often performed by early there is a prominent “open science” or “science
career researchers who are learning the methods reform” movement rapidly growing, calling for
and tools while using them. Indeed, it is common improved scientific practices to enhance repro-
for researchers to conduct preliminary analy- ducibility, replicability, and generalizability, with
ses, which are later changed based on additional an emphasis on openness, transparency and inclu-
knowledge acquired about the methods (e.g., from sivity (Munafò et al., 2017; Nelson et al., 2018;
reading more papers or from discussions with Nosek, 2015; Poldrack et al., 2017). Cognitive
other researchers) or about the data (e.g., from neuroscientists have been taking an active role in
observed artifacts or covariates). Researchers are this movement, demonstrating self-reflection and
more likely to “improve” their analysis or test self-criticism (Gilmore et al., 2017). This sug-
additional ones if the original analysis did not gests an optimistic future, and indeed, tools and
yield a significant result in the expected direction. practices to mitigate current challenges are being
Thus, attempts to improve rigor and optimize anal- developed, thereby increasing the rigor of research
yses can easily manifest (perhaps unconsciously) into cognitive neuroscience.
as p-hacking. For example, sample sizes have been slowly
Analytical flexibility is a major challenge even if increasing in recent years. One recent study
it does not involve p-hacking and only one analysis showed that while high impact neuroimaging
is performed. A recent collaborative project includ- studies from 1990 to 2012 had a sample size of 12
ing almost 200 researchers made this abstract prob- participants on average, similar studies in 2017–
lem concrete by demonstrating startling analytical 2018 had sample sizes of about 23–24 partici-
variability with substantial effects on reported pants (Szucs and Ioannidis, 2020). One trend that
results in fMRI (Botvinik-Nezer et al., 2020). is substantially increasing statistical power is use
Seventy independent analysis teams received the of publicly shared data, particularly large datasets
same fMRI dataset and were instructed to analyze with thousands of participants, such as the Human
the data to test the same pre-defined nine hypothe- Connectome Project (HCP) (Elam et al., 2021;
ses. Notably, no two teams chose exactly the same Van Essen et al., 2013), the Adolescent Brain
analysis pipeline. This analytical variability led to Cognitive Development (ABCD) study (Casey
substantial variability in reported statistical maps et al. 2018), and UK BioBank (Alfaro-Almagro
and binary conclusions about hypothesis tests et al., 2018) (Table 34.1 and Figure 34.2). While
across teams (although statistical maps in earlier this indicates a trend toward larger sample sizes
stages of analysis were more similar across teams). and more powerful studies, the statistical power of
Other studies have also demonstrated the effects of most studies is still relatively small and needs to
analytical choices on results in fMRI (Carp, 2012; be improved considerably.
Li et al., 2021). A similar project is in progress in Similarly, questionable research practices such
EEG (EEGManyPipelines.org), while in PET it has as p-hacking and HARKing, which have been
been shown that choice of preprocessing strategy common in the past, are slowly becoming less
can have a major impact on outcomes (Nørgaard acceptable and consequently less prevalent. This
et al., 2020), and in diffusion-based tractography is a result of the surging focus on their implica-
high variability was found for tractography dissec- tions, in addition to new tools that are making such
tion (Schilling et al., 2021). Similar results have practices explicit and conscious, and prevent them
been obtained in the social sciences (Bastiaansen in some cases. For instance, it is becoming more
et al., 2020; Breznau et al., 2022; Silberzahn et al., common to pre-register studies prior to data col-
2018). Results are often not robust to arbitrary ana- lection (or at least prior to data analysis), although
lytical choices, in which case the value of a single it is still far from routine (Borghi and Van Gulick,
study using a single analysis pipeline becomes 2018; Button, 2019; Hardwicke et al., 2021; Paret
questionable, and testing the robustness of results et al., 2022). In a pre-registration, a frozen time-
to arbitrary analytical choices becomes essential. stamped description of the experimental design,
Finally, most cognitive neuroscience stud- sample size, hypotheses, analysis plan, and pre-
ies are conducted with participants (and by dictions is registered on a public platform, thus
researchers) from “WEIRD” (Western, Educated, constraining the researchers’ analytical flexibility
Industrialized, Rich, and Democratic) societies, and making explicit which hypotheses are tested
most commonly in North America or Europe and which choices are made, and whether they
572 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

are made prior to or after observing the data. Pre- While data and code sharing are becoming
registration distinguishes between confirmatory more prevalent, another type of sharing that is
(hypothesis testing) and exploratory (hypothesis just beginning to gain traction in cognitive neu-
generating) research (Nosek et al., 2018; Simmons roscience is model sharing (Poldrack et al., 2019).
et al., 2021). While both are necessary for scien- Models are powerful tools for both simulating and
tific progress, interpretation of results gener- understanding processes and for making quan-
ated by each approach is different. Registered titative, testable predictions. Machine learning
reports is a form of “extended” pre-registration, and deep learning-based models can provide new
where the study plan is submitted to a journal and measures, reducing complex data to patterns or
goes through peer review prior to data collection effects of interest. For example, they may reduce
(Chambers, 2019; Hardwicke and Ioannidis, 2018; people’s speech patterns and language during an
Nosek and Lakens, 2014). The review process can interview into a measure that predicts risk for sui-
result in an “in-principle acceptance,” meaning cide, reduce complex electrocardiogram patterns
that the study would be published as long as it to a measure of heart attack risk, and many more.
follows the accepted research plan, whatever the In neuroimaging, models can reduce images with
results show. This format helps mitigate p-hacking hundreds of thousands of voxels and billions of
as well as publication bias, since once a study pairwise connections–each of which provides a
has been accepted as a registered report, the pres- potential measure and hypothesis to test–into a sin-
sure to find a significant result for publication is gle, focused measure that can be tested as a target
removed, and there is less incentive to p-hack. for task manipulations or other interventions, rela-
Code and data sharing are also becoming more tionships with behavior or performance, and pre-
common, and studies are becoming more repro- diction of future behaviors, clinical diagnosis, or
ducible (Borghi and Van Gulick, 2018; Hardwicke clinical risk (Woo et al., 2017). Inspired by “Model
et al., 2021; Houtkoop et al., 2018; Paret et al., Zoos” in artificial intelligence, our Cognitive
2022; Poldrack and Gorgolewski, 2014). This pro- and Affective Neuorscience Lab (Tor Wager, PI)
cess is facilitated in part by recent methodological maintains a shared library of pre-trained predic-
and technological advancement such as guidelines tive models applicable to neuroimaging data from
for best practices (Knudsen et al., 2020; Nichols new individuals and scanners (https://github.com/
et al., 2017; Pernet et al., 2020), standards for canlab/Neuroimaging_Pattern_Masks). Testing a
data organization (e.g., the Brain Imaging Data priori brain models reduces the “garden of forking
Structure (Gorgolewski et al., 2016)), platforms paths” and potential for p-­hacking, eliminates need
for data analysis and sharing (e.g., brainlife.io and for multiple comparisons’ correction when testing
OpenNeuro (Hayashi et al., 2023; Markiewicz effects on a single model-based measure, and can
et al., 2021)), and tools for data and code man- dramatically increase effect sizes and replicability
agement from version control and data provenance (Kragel et al., 2018; Reddan et al., 2017). Model
tracking to reproducible analyses and data shar- sharing complements sharing of data and code,
ing (e.g., DataLad (Halchenko et al., 2021)). For making these measures available to all. Moreover,
example, our laboratory (Tor Wager, PI) maintains the more widely models are used and tested, the
a growing repository of general-use, object-ori- more extensively validated they become, and the
ented code for neuroimaging data analysis that is more is known about their generalizability and
publicly available at CANlab neuroimaging analy- other measurement properties such as sensitivity
sis tools (https://canlab.github.io). and specificity to particular task conditions and
Beyond ensuring more reproducible analy- populations.
ses and replicable findings, data sharing efforts While issues of low power and questionable
increase the value of each dataset, and therefore research practices are slowly improving with cur-
the (frequently public) resources invested in rent tools and approaches that are focused on
collecting it, as researchers outside the original reproducibility and transparency, there is still a
group can later further examine it and use it to long way to go, and other issues are even further
study additional directions. Data sharing further away from being solved. For example, the problem
facilitates projects that integrate data across mul- of analytical variability is drawing more attention
tiple studies and labs, studying research questions with recent demonstrations of its magnitude and
that frequently cannot be adequately addressed importance in the social sciences and neuroimag-
by a single study. Furthermore, it helps miti- ing (Botvinik-Nezer et al., 2020; Nørgaard et al.,
gate problems of p-hacking and irreproducibil- 2020; Silberzahn et al., 2018). Pre-registration
ity. When researchers share their data and code, and sharing of code, data, and models help ensure
others can test whether they can reproduce the that the analytical variability is not exploited to
claimed results using the same dataset or inde- find and report a single pipeline that yields sig-
pendent data. nificant results, and to allow other researchers to
Advancing Cognitive Neuroscience 573

test whether results are robust to other analytical Stroebe, 2019) and the Psychological Science
choices. However, it does not solve the problem Accelerator (PSA; Moshontz et al., 2018). PSA
completely, since the result of a single analysis is an ongoing, globally distributed, diverse, and
pipeline is still dependent on arbitrary choices inclusive network of researchers in psychology.
made, whether they were the only ones tested It includes over 1000 members from dozens of
or not. Clearly, a first crucial step is to fully and countries worldwide. Though it is only a few
transparently report all analytical choices. years old, it has already produced a few publica-
A more complete solution to this problem is a tions, preprints and in-principle-accepted regis-
“multiverse analysis,” in which the same data are tered reports, each reporting results from multiple
analyzed with multiple validated analysis pipe- and very diverse samples (Bago et al., 2019; Chen
lines, which are all reported, and the researcher et al., 2018; Forscher et al., 2019, 2020; Hall et al.,
tests for convergence (and divergence) across 2018; Jones et al., 2021). Resolving these issues
them (thus testing robustness of findings to arbi- and making cognitive neuroscience diverse, inclu-
trary analytical choices). While such approaches sive and representative is still in its infancy, and
have been suggested (Strother et al., 2002), mul- major attempts alongside creative adaptations of
tiple conceptual and practical developments are existing structures and ways of thought are needed
needed for full solutions to be available. First, to make s­ ubstantial progress. Also, current fund-
and perhaps most challenging, one needs to select ing structures and authorship assignment may be
which analysis pipelines to include. As mentioned problematic for large-scale collaborations (Coles
above, it is hard or even impossible to generalize et al., 2021, 2022).
optimal, best approaches across domains, hypoth- The last decade has marked the rise of more
eses, and datasets (e.g., fMRI data obtained with reproducible, replicable, and generalizable cog-
spiral vs. EPI acquisition protocols involve differ- nitive neuroscience. Growing emphasis on these
ent artifact patterns and edge-of-brain brightness, topics, together with an abundance of developed
which can interact with segmentation and normali- tools and practices to enhance them (Niso et al.,
zation algorithms). Hence, the default approach of 2022), are already “bearing fruit.” A prominent
using an analysis pipeline that works well on aver- challenge that is becoming apparent as “open
age with some specific customized adaptations science” becomes more prevalent is the tension
is sensible, but developing methods for choosing between theoretical ideals and practical con-
multiple analysis pipelines to test over is likely a straints. We turn to this next.
better approach. Second, once a “multiverse” of
analysis pipelines is defined, methods to test for
convergence across these dependent pipelines are
needed. Third, tools are needed to conduct such Balancing Theoretical Ideals and
analysis with reasonable computational resources Practical Constraints
at a single lab level. Forth, tools and guidelines
are needed for the reporting and visualization of In practice, research into cognitive neuroscience
the tested pipelines and the results. Such devel- often is carried out with limited resources – from
opments are currently in progress, but may take funding to participant pools to skilled and experi-
time to mature, mainly for complex and high-­ enced personnel – and within systems that often
dimensional datasets typical to cognitive neuro- incentivize novel, positive, publishable findings
science (Aczel et al., 2021; Clayson et al., 2021; (Munafò et al., 2017; Nosek et al., 2012). Putting
Dafflon et al., 2020; Li et al., 2021; Liu et al., things into perspective, Phase III clinical trials of
2021; Simonsohn et al., 2020). pharmaceutical agents are designed to test a single,
The diversity of researchers and participants well-specified hypothesis: that a drug produces a
populations has been gaining a lot of attention non-zero benefit over a placebo or other compara-
alongside societal movements toward inclusiv- tor. Measures, methods, and statistical techniques
ity, diversity, equality, and equity (Carroll et al., are well-established, as they follow blueprints laid
2021; Rad et al., 2018). The recent rise of “big out by regulatory agencies and many previous stud-
team science” may solve a fundamental prob- ies. Endpoints are standard clinical assessments,
lem of limited resources in the typical single- and complex biological assays are rare. Nonetheless,
lab studies, by allowing many research teams to such trials often cost tens to hundreds of millions of
pool together their resources (Coles et al., 2022; dollars each. This is due largely to the inherently
Forscher et al., 2020). It can also lead to global very challenging endeavors of establishing rigor-
collaborations, allowing more statistically pow- ous and well-documented protocols, ensuring com-
ered, diverse, and therefore generalizable stud- pliance with them, and establishing effects that
ies, as exemplified by the “Many Labs” projects generalize to a population. Can we expect scientists
(Ebersole et al., 2016; Klein et al., 2014, 2018; working on new, innovative ideas, developing, and
574 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

testing new complex measures (like fMRI, EEG, Phase A1: “Exploratory.” Exploratory studies (with
language, social media data) for which analyses not minimal constraints and requirements) are used
yet established and evolve rapidly from year to to discover preliminary findings for later valida-
year, to meet expected standards for documenta- tion. Practices described above, such as data and
tion, reproducibility, replicability, data sharing, and
code sharing are recommended, but not required.
generalizability when compromises between the
ideal and reality are inevitable? In addition, rigor of Many previous and current studies in cognitive
large-scale clinical trials comes at an extraordinary neuroscience are at this phase. However, it is
cost in innovation and development of new ideas desirable that specific findings are validated in
and methods. Below, we present our suggestions Phase B1 or B2 studies before being accepted by
for how to advance cognitive neuroscience with the scientific community.
innovative approaches and formalizing such Phase A2: “Shared Exploratory.” These are explor-
compromises. atory studies whose data and code are shared with
We suggest transitioning toward a “phase the research community. Publications from phase 2
model” (Figure 34.1), similar to models used in studies are accompanied by shared data and code.
biomedical research from preclinical through
Phase A3: “Reproducible Exploratory.” Studies
phase III clinical trials (Borsook et al., 2011),
and to the gradual approach we previously sug- with fully reproducible analyses, as validated by
gested for developing brain biomarkers (Woo external researchers. Shared code and data are
et al., 2017). The rationale is to lower barriers reviewed by independent parties prior to publica-
for entry and encourage exploration and devel- tion, to ensure results are analytically reproduc-
opment in Phase 1, but then impose increasingly ible (i.e., identical results are obtained by other
more constraints in later phases to ensure repro- researchers using the same data and code).
ducibility, generalizability, and robustness of find- Phase A4: “Robust Exploratory.” Exploratory stud-
ings. Specification of different phases requires a ies may be both fully reproducible and robust to
consensus group, but here we lay out a general analytical variability, as determined by a multi-
proposed architecture. The different phases are
verse analysis. Studies in this category specify
designed to have different criteria across two main
dimensions: First, each phase imposes additional and justify a systematic set of analysis pipelines
requirements, such as code and data sharing, ana- over which to generalize and report robustness of
lytical reproducibility, and analytical robustness, results across these analytical choices.
for establishing stronger findings. Second, studies Phase B1: “Confirmatory.” Confirmatory studies
in each phase could be either exploratory (group are used to test specific hypotheses, validate mea-
A) or confirmatory (i.e., pre-registered; group B). sures and biomarkers, and estimate effect sizes.

Figure 34.1 An illustration of the suggested “phase model” for cognitive neuroscience.
Advancing Cognitive Neuroscience 575

These activities require precisely defined targets. prior to data collection. An alternative is to use
In this phase, pre-registered hypotheses and pre- one dataset to optimize the analysis, and then pre-
dictions are tested with a pre-registered analysis register and apply the optimized analysis to a new
plan. dataset. This could be done either by dividing the
dataset to a “training” set and an independent “test”
Phase B2: “Shared Confirmatory.” Confirmatory
set or by collecting new data after the analysis of the
studies with publicly shared data and code (as original dataset (i.e., a pre-registered replication).
in A2). Second, as studies grow in size and complexity,
Phase B3: “Reproducible Confirmatory.” Confir- it becomes impractical to specify all hypotheses
matory studies that are fully reproducible (as in before data collection, and requiring this would
phase A3). either impose an uneven standard across stud-
Phase B4: “Robust Confirmatory.” Studies that are ies or preclude virtually any Phase B1 hypoth-
fully reproducible and tested for analytical robust- esis tests on large-scale studies. For example,
ness (as in phase A4), which were pre-registered the UK Biobank study began data collection in
(including plan for analytical robustness). about 2010, and neuroimaging data collection
is expected to be completed around 2025. Thus,
requiring pre-registration prior to data collection
Pre-registration is key to distinguishing between would preclude confirmatory studies on this data-
group A and group B studies. It is critical to deter- set and similar ones. A reasonable standard is to
mine when a study needs to be pre-registered in allow the formulation of hypotheses as long as the
order to be considered confirmatory. We suggest critical outcome data used to test them have not
that pre-registration be recommended prior to data been examined. For example, researchers could
collection, and required before critical outcome pre-register a hypothesis about effects of sleep on
data are observed. Critical outcome data are data brain connectivity, as long as they formulate the
required to test the hypothesis. For example, pri- hypothesis before any examination of the relation-
mary outcomes in a clinical trial, task condition ship. Examining or extracting brain connectivity
labels in a hypothesis test of task-evoked brain components in advance would be acceptable, as
responses, and group labels in between-groups long as the researchers did not examine their rela-
comparisons are critical outcome data. Analyses tionship with sleep (or, ideally, did not have access
can examine distribution of other variables prior to to the sleep variables) before pre-registration. This
hypothesis pre-registration (e.g., for the purposes standard is consistent with the practice of blinded
of checking data integrity, study procedures, and analysis, in which effects are analyzed without
statistical distributions, if outcome data are not awareness of the labels on the critical outcome
available to the analyst). Data or results available data (e.g., patient group or task conditions). Such a
prior to pre-registration should be described. phase model can facilitate gradual progress toward
Studies should be powered to detect the pre-­ more reproducible, replicable, and robust findings,
registered effects of interest, as is standard for while still encouraging innovation and risk taking
clinical trials. (but acknowledging practical barriers).
The concept of critical outcome data is important Another critical trade-off exists between statisti-
for maintaining appropriate controlled flexibility at cal power and replicability, which require large sam-
this stage. It is advantageous, and often critical, to ple sizes, and the need for innovation and diversity
examine data in order to know which modeling of approaches (Figure 34.2). This is a fundamental
choices should be made (e.g., how to identify and tradeoff, though there are other objectives that trade
correct invalid or missing values during data collec- off with these goals as well, such as generalizabil-
tion, deal with outliers, specify error distributions ity to diverse populations. There has been a strong
and variable transformations, test assumptions, trend toward larger and more diverse datasets, with
and determine the structure and nature of statisti- substantial public funds devoted to collecting and
cal models). If all hypotheses must be specified sharing ultra-large datasets like the HCP, ABCD, and
prior to data collection, their specification must UK BioBank. While these datasets are exceptionally
be imprecise with respect to all of these choices. useful for many research questions, they focus by
This leads to substantial analytical variability later, necessity on a few widely used measures and task
allowing thousands of variations on tests of a single paradigms. They are not designed for methodologi-
pre-­registered hypothesis. Indeed, almost all pub- cal innovation. One critical type of innovation is the
lications of pre-registered studies report deviations development of new tasks and computational models
from the pre-registered plan that are explained and that could provide novel measures with larger effect
justified. Pre-registering at a later stage, but before sizes and greater inferential precision. Another kind
outcome measures are observed, could thus con- of innovation is the development of new techniques
strain analytical flexibility better than pre-registering for collecting data (e.g., new imaging sequences and
576 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Figure 34.2 Tradeoff between sample size and task and context diversity. For description of
studies based on the acronym, see Table 34.1. Note that here we focus on diversity of task fMRI
(and its context) specifically, and thus studies that are diverse in other aspects (e.g., imaging
modalities) may still be low on the Y axis. There is a prominent trade-off between sample size
and task and context diversity that could be addressed by multi-task, multi-study consortia like
the Placebo Imaging Consortium and the Affective Neuroimaging Collaboratory (ANiC).

modalities). Thus, while large shared projects have progress requires integration of findings into test-
provided a tremendous step forward, they are limited able theories, and models, that organize and for-
to a very small portion of the space of cognitive and malize previous knowledge and predict new
affective processes that might be assessed and meth- findings. Psychological sciences may suffer from
ods for assessing them. a “theory crisis,” manifested in weak connections
One solution is to leverage the advantages of small, between theories and their empirical tests, as well
innovative studies and large, well-powered studies by as underappreciation of computational modeling
combining them. For example, predictive models and in theory building (Muthukrishna and Henrich,
measures from smaller studies can be correlated and 2019; Oberauer and Lewandowsky, 2019).
combined with those from large shared datasets (He Likewise, in the field of cognitive neuroscience,
et al., 2022). A broader, more ambitious solution is several developments are needed to shift the focus
to establish multi-lab, multi-study, multi-task consor- toward integration and theory building. Here, we
tia. In such consortia, researchers collaborate to share focus on two such developments: (1) Integrative
datasets collected within broad domains but with empirical analyses and ontologies; and
diverse designs and tasks and by different labs. Such (2) Cognitive architectures and unifying princi-
multi-study datasets are already yielding more robust, ples, particularly those based on predictive coding.
generalizable findings (Zunhammer et al., 2021).

Integrative Empirical Analyses


TOWARD UNIFYING THEORETICAL and Ontologies
FRAMEWORKS More systematic reviews and meta-analyses are
needed to better integrate existing knowledge in a
Achieving the goal of reproducible, replicable, specific research domain. Data sharing practices
and robust findings is a major cornerstone in the are already starting to improve meta-analyses by
advancement of cognitive neuroscience. However, providing researchers with richer information to
a collection of rigorous, true findings is not integrate over. Furthermore, as data sharing becomes
enough to lead us toward deciphering how the more common, advanced meta-analytic methods are
brain gives rise to human cognition. Scientific likely to be developed (Han and Park, 2019).
Table 34.1 Overview of large population-based studies, ordered based on sample size
Acronym Study / database Reference Sample Population

UKB United Kingdom Biobank (Alfaro-Almagro et al., 2018; 100,000 UK adults; many data types, six MRI modalities but only one fMRI task Population
Miller et al., 2016)
ENIGMA Enhancing Neuroimaging Genetics through (Adhikari et al., 2019; ∼60,000 from various studies with genetic and neuroimaging data Multiple
Meta Analysis Thompson et al., 2014)
ABCD Adolescent Brain Cognitive Development Study (Casey et al., 2018) 10,000 US adolescents, longitudinal, three fMRI tasks Development
FCP & INDI 1000 Functional Connectomes Project and the (Mennes et al., 2013) 1,300+ (FCP), thousands (INDI); focused on resting-state fMRI Multiple
International Neuroimaging Data-sharing
Initiative
ROS-MAP Religious Orders Study and Rush Memory and (Bennett et al., 2018) 3,500, with omics and postmortem, multi-modal MRI including resting-state Dementia
(RM) Aging Project fMRI
HCP & Human Connectome Project Somerville et al., 2018; ∼2500 Healthy US adults + children; different MRI modalities and six fMRI task Healthy,
dHCP Van Essen et al., 2013b) domains development
ABIDE I & II Autism Imaging Data Exchange (Di Martino et al., 2014, Multi-lab data sharing, ∼2200 with ASD and controls; focused on resting-state ASD
2017) fMRI
IMAGEN IMAGEN (Schumann et al., 2010) 2,000 adolescents, longitudinal; behavioral and neuropsychological measures, Development
structural and functional MRI (three tasks)
ADNI 1,2,3 The Alzheimer’s Disease Neuroimaging (Jack et al., 2008) Multi-lab data sharing, ∼1800 with Alzheimer’s, MCI, and controls; includes Dementia
Initiative structural MRI, PET, clinical, cognitive, biochemical, and genetic data
PPMI Parkinson’s Progression Markers Initiative (Marek et al., 2011) Aiming at ∼4000; including clinical data, biologic samples, genetic data, Parkinson’s
structural and diffusion MRI
OASIS Open Access Series of Imaging Studies (Marcus et al., 2007) Thousands, including cross-sectional and longitudinal data, multiple MRI modalities Dementia
Advancing Cognitive Neuroscience

(varies across sub projects, includes resting-state fMRI from OASIS 3 on)
GSP Brain Genomics Superstruct Project (Holmes et al., 2015) 1,570, some longitudinal; genetics, cognitive, personality, structural MRI and Healthy
resting state fMRI
IBC Individual Brain Charting (Pinho et al., 2018) 12 participants, multiple tasks Limited
MSC Midnight Scan Club (Gordon et al., 2017) 10 participants, 12 two-hours long sessions from each, including multiple modalities Limited
and five and a half hours of task fMRI data across three different tasks
NSD Natural Scenes Dataset (Allen et al., 2022) 8 participants, task data while viewing thousands of color natural scenes and Limited
performing a recognition task across 30–40 sessions
MC My Connectome (Poldrack et al., 2015) A single participant with data collected over 18 months including resting-state Single participant
fMRI, memory tasks, and many other types of data (physiological, structural,
577

physical health, gene expression, metabolism)


578 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

In neuroimaging, for example, coordination-based neuroimaging studies along with their methods
meta-analyses have traditionally been the norm, (sample size, modality, task, etc.) and the coordinate-
but image-based meta-analyses (Salimi-Khorshidi based results (the location of the peak activation
et al., 2009) are now becoming more accessible as in standard space) (Fox and Lancaster, 2002; Fox
more and more researchers share their statistical et al., 2005; Laird et al., 2005). It includes soft-
brain maps via platforms such as NeuroVault ware and tools for neuroimaging data sharing
(Gorgolewski et al., 2015). and has already yielded hundreds of publications.
In addition, cognitive neuroscientists should Importantly, it also developed a taxonomy for
be expected to (and therefore trained to) formal- classifying studies in a space of cognitive, affec-
ize their theories prior to testing (falsifying) them, tive, sensory, and motor processes to facilitate
preferably in the form of quantitative, testable aggregation and analysis. The Cognitive Paradigm
computational models (Guest and Martin, 2021). Ontology (CogPo; Turner and Laird, 2012) is built
This would require a higher level of formal sta- upon the BrainMap taxonomy to categorize task
tistical and computational training in the field paradigms based on the stimuli, instructions and
(Neuromatch Academy (NMA), 2021) as well as response rather than the cognitive function they
a better alignment between theories, or hypoth- putatively measure, avoiding constructed and
eses, and the empirical and statistical procedures potentially heterogeneous categories like “work-
they rely on (Yarkoni, 2020). Furthermore, even ing memory.”
when researchers are using formal models, these Despite the enormous contributions of
are frequently not shared in a way that allows BrainMap and other similar projects to the field,
other researchers to test them. It is crucial that they are limited because they depend on manual
researchers provide the full details of their mod- annotation of studies. NeuroSynth is a more
els, whether it is the formulations and algorithms, recent tool that took a very different approach.
the code, or ways of applying pre-trained models It exchanged quality with quantity: Instead of
to new data (Woo et al., 2017). Finally, a clearer high-quality but limited manual annotation of
distinction between confirmatory (theory-testing) single studies, it relies on automated extraction
and exploratory (discovery-oriented) research is of reported activations and semantic annota-
needed. This is already happening with the grow- tions from a large number of published studies
ing use of pre-registration (Nosek et al., 2018). (Yarkoni et al., 2011). Analyses can be performed
The framework for classifying studies outlined based on a flexible set of thousands of concepts
above is aligned with this need: The specification used in the papers and semantic topics based on
of exploratory and confirmatory goals is built into them (Poldrack and Yarkoni, 2016). Other recent
the classification system. approaches, like Neuroquery.org, build on this
Notably, to facilitate successful integration of concept (Dockès et al., 2020).
findings, it is crucial to have a common language The Cognitive Atlas is a more systematic
(or ontologies, which are explicit specifications attempt to formally represent mental processes,
of concepts (Gruber, 1993)) to base theories on, and the cognitive tasks used to manipulate and
as well as dedicated databases and tools based measure them, in order to later map these pro-
on such ontologies. Researchers need to under- cesses to brain functions (Poldrack et al., 2011). It
stand what other researchers mean when they is built collaboratively by any interested researcher
use a specific term, within and between differ- within cognitive neuroscience and related fields,
ent subdisciplines. Unfortunately, this is far from and facilitates a more precise use of terms across
being the reality in cognitive neuroscience. The the fields, better data mining of the published lit-
field’s reality is closer to the “Tower of Babel,” erature, metadata annotation, meta-analyses, and
with different researchers speaking different “lan- theory conceptualization and testing in general.
guages” and using different terms to describe These attempts to develop common terminol-
similar phenomena, thus compromising the ability ogy, data annotation, and literature aggregation
to integrate findings into a tower toward the truth. tools have advanced the field and increased the
Novel informatics-driven big data approaches to ability to achieve scientific consensus and dis-
brain-cognition mapping are particularly depend- seminate findings, as well as providing more rig-
ent on common annotations and integration tools orously defined hypotheses and targets for new
(Poldrack and Yarkoni, 2016). studies. Yet, cognitive neuroscience is still closer
Several attempts have been made to create a to the “Tower of Babel” than to the vision of
joint knowledge base or ontologies for cognitive these tools, and much work is needed for a fun-
neuroscience. For example, the BrainMap pro- damental change (Hastings et al., 2014). It is also
ject aims to facilitate data mining, aggregation, debated whether ontologies used to study the brain
and meta-analyses by providing annotated data should be based on psychological concepts or
of published human structural and functional neurobiological objects (Buzsáki, 2020; Poeppel
Advancing Cognitive Neuroscience 579

and Adolfi, 2020). Recent initiatives, such as the Another important direction is the development
Research Domain Criteria (RDoC) framework ini- of computational principles. Recently, the idea of
tiated by the National Institutes of Mental Health predictive coding subsumes several broad con-
(NIMH), aim to shift the classification of mental cepts that have great current appeal: that instead
disorders from symptom-based to neuroscience- of a stimulus–response machine, the brain has
based (Insel, 2014; Insel et al., 2010). evolved to predict future events and select actions
that minimize entropy (or, equivalently, surprise
or prediction error) with respect to the organism,
maximizing predictive control and thus future sur-
Cognitive Architectures and Unifying vival and wellbeing (Clark, 2013; Friston, 2010;
Principles Rao and Ballard, 1999). In essence, the theoreti-
cal framework is that the brain generates and opti-
Combination of highly specialized and isolated mizes internal mental models that predict sensory
research areas without shared, formalized termi- input and physiological states. These models are
nology creates a new, broader challenge: The need updated and optimized based on prediction errors–
for unifying, integrative theories that relate vari- the difference between the predicted and the actual
ous isolated research areas and findings together. input. Top-down signals generated by internal
Integrating different research areas into a unifying models convey the predictions and ­bottom-up
theory is extremely challenging, but it is arguably signals convey prediction errors (Bastos et al.,
the most pressing challenge of cognitive neurosci- 2012; Rao and Ballard, 1999). The minimization
ence. Some early attempts to construct integrative of prediction errors across hierarchical layers
cognitive architectures, for example the Soar occurs either by updating the internal model or by
architecture for general cognition (Newell, 1992, action. In this framework, perception, decision-
1994), the Adaptive Control of Thought—Rational making, emotion, and action are a matter of quasi-
(ACT-R) cognitive architecture (Anderson et al., optimal, quasi-Bayesian (or Bayesian) inference.
1998) and the executive-process interactive con- Information representation is efficient, so that
trol (EPIC) theoretical framework (Kieras, 2016; only deviations from predictions are passed for-
Meyer and Kieras, 1997), have not been used and ward to update internal mental models (Aitchison
developed nearly as widely as they might have and Lengyel, 2017; Knill and Pouget, 2004).
been, although they continued to be developed to The predictive coding theoretical framework
some extent (Anderson, 2009; Borst and Anderson, revolutionized many research areas in cognitive
2015; Laird, 2019). neuroscience and beyond, and has the potential to
One reason for this may be because they did integrate across areas. It already unifies percep-
not provide a level of description compatible with tion (for example vision (Rao and Ballard, 1999),
the field’s understanding at the neural circuit level. audition (Chennu et al., 2013), pain (Büchel et al.,
Early neural networks (Rumelhart et al., 1994) 2014; Song et al., 2021) and interoception (Barrett
were compatible with principles of neural func- and Simmons, 2015), motor action (active infer-
tion and information representation, but suffered ence (Adams et al., 2013; Friston et al., 2010)),
from a lack of computational power to implement attention (Hohwy, 2012), emotions (Barrett, 2017;
larger models and a lack of tools to interpret their Seth, 2013), theory of mind (Koster-Hale and
function. Recently, the former limitations have Saxe, 2013), mental health (Kaye and Krystal,
been partially overcome with technological, meth- 2020; Kube et al., 2020; Smith et al., 2021; Sterzer
odological, and statistical developments. Indeed, et al., 2018; Van de Cruys et al., 2014), and can
neural networks and deep learning have become also be related to structure and representational
immensely popular, substantially advancing both learning (or latent cause attribution (Niv, 2019;
basic cognitive neuroscience and applications Tenenbaum et al., 2011)) and other domains.
such as artificial intelligence (AI) (Hasson et al., Nonetheless, while predictive coding is compu-
2020; LeCun et al., 2015; Richards et al., 2019; tationally specific at the level of general informa-
Saxe et al., 2021). However, the second limita- tion processing principles, it is underconstrained
tion remains. Neural networks and deep learning in terms of the neural mechanisms: what is rep-
provide only a high-level description of how neu- resented where in the brain and how precisely it
ral information representation might occur, but is coded (Kogo and Trengove, 2015; Kwisthout
they are difficult to test, and to understand, and and van Rooij, 2020). It is also still not well inte-
therefore lack explanatory power (though some grated with learning theory, including reinforce-
researchers suggest that understanding how the ment learning (K. Friston, 2018) and other types
brain develops and learns is a better goal than of learning and memory, although the prediction
understanding the properties resulting from these error idea is central to reinforcement learning
processes (Lillicrap and Kording, 2019)). (Schultz et al., 1997; Tobler et al., 2005). Like
580 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

other integrative attempts, predictive coding has predictive mapping approach brain patterns are
strengths and weaknesses, and the future will tell used to predict (or decode) mental outcomes.
whether it will continue to grow as a unifying These analyses are largely based on computational
theoretical framework or be replaced with oth- approaches, particularly machine-learning tools,
ers. Whatever the future holds, it has undoubtedly and have been facilitated by recent technological
advanced cognitive neuroscience, both within and and computational developments, substantially
across research areas, demonstrating the value of reducing the required time and resources (Cohen
unifying, fundamental theoretical frameworks to et al., 2017). This paradigm shift in neuroimaging
the advancement of the field. is also grounded on the theoretical shift in cellular
neuroscience from the modular view of cognitive
processes as modular, separately-­implemented
processes, to a distributed view of neural popu-
TOWARD INTEGRATIVE BRAIN MODELS: lation coding and distributed representation
(Averbeck et al., 2006; Ni et al., 2018; Pouget
FROM SINGLE REGIONS TO PREDICTIVE et al., 2000; Rigotti et al., 2013).
MODELS The predictive modeling approach develops
brain models that predict specific mental functions
In addition to new, integrative theoretical principles, or health-related outcomes. Predictive models can
the future of cognitive neuroscience lies in develop- serve as biomarkers for these processes, and are
ing integrative, empirical models of brain organiza- much more likely to generalize to new popula-
tion and representation. Cognitive neuroscience tions compared to findings of the traditional
aims to describe the neural mechanisms of cogni- region or voxel-based methods, because they are
tion. The field has been predominantly focused on statistically measured on their ability to do so.
mapping specific cognitive processes to localized Such models have been advancing knowledge
brain regions. This approach has deep historical across cognitive neuroscience research areas, for
roots, from phrenology, the localizationist view and example object recognition (Haxby et al., 2011),
the parcellation of the brain to sub-regions by memory (Harrison and Tong, 2009), pain (Ceko
Broadman and others in the nineteenth century. et al., 2022; Wager et al., 2013; Woo and Wager,
Traditional approaches in neuroimaging in particu- 2015; Woo et al., 2017), emotions (Kragel and
lar are focused on localized regions and are based on LaBar, 2015), attention (Rosenberg et al., 2016),
analysis of isolated brain regions. In other words, reinforcement signals (Vickery et al., 2011), and
they describe which brain regions encode specific neurological and mental disorders (Arbabshirani
types of mental functions. Indeed, prominent dis- et al., 2017; Fan et al., 2008; Hahn et al., 2015;
coveries in cognitive neuroscience include the local- Koutsouleris et al., 2009; Woo et al., 2017). In
ization of specific mental processes, such as the addition to integration across brain regions, pre-
fusiform face area (FFA) for face perception dictive models can also integrate across measure-
(Kanwisher et al., 1997), the hippocampus for ments, such as functional and structural features,
memory (Squire, 1992), and the ventro-medial pre- diffusion-based measures, connectivity patterns,
frontal cortex for value-based decision making neurochemistry, electrical signals, and more.
(Chib et al., 2009; Levy and Glimcher, 2012). This approach is likely to continue to lead the
However, this approach has a fundamental problem: way for cognitive neuroscience. However, to real-
Individual brain regions are related to multiple cog- ize its promise, emphasis is needed on the ability
nitive and affective processes, and individual cogni- of the predictive brain models to accurately predict
tive/affective processes are related to multiple completely independent data. Many studies with
regions. The efforts to understand what processes predictive modeling make claims of prediction
and types of information the ventro-medial prefron- although statistically they only establish correla-
tal cortex (Delgado et al., 2016) or the insula (Chang tion (Poldrack et al., 2020). Importantly, in order
et al., 2013; Kurth et al., 2010) represent, for exam- to establish their predictive power, models that are
ple, nicely illustrate these issues. Therefore, it is being trained on a dataset should be tested on new,
problematic to reverse-infer mental functions from completely independent data. If cross-­validation
activations of single brain regions (Poldrack, 2011). is being performed, with the model tested on
Recently, the field has been shifting focus held-out data, all operations on the data includ-
toward integrated, multivariate, system-level pre- ing preprocessing and feature selection must be
dictive models of cognitive processes (Haynes, separately performed in each validation iteration,
2015; Kragel, Koban, et al., 2018; Norman et al., to ensure independence between the training and
2006; Poldrack and Farah, 2015). In neuroimag- test sets. Since data from the same dataset are
ing, instead of using mental measures to explain likely to be dependent, it is preferred that models
(statistically relate to) brain activations, in a will be trained and validated in a cross-validation
Advancing Cognitive Neuroscience 581

procedure, and once a model has been selected and stimulus modalities (pictures, sounds) (Chikazoe
optimized, it can be tested on completely new data et al., 2014; Wager et al., 2015) or integrating
to establish predictivity, without additional opti- across multiple studies with different contexts
mization procedures (Kragel, Koban, et al., 2018). when possible (Kragel et al., 2018a,b). Multi-lab
Brain signatures, or neuromarkers, are a specific consortia, as suggested above, lend themselves to
type of predictive model, aiming to use brain pat- this kind of generalization.
terns to predict mental processes across partici- Another important aspect is the interpretability
pants (i.e., population-level models) (Gabrieli et al., of predictive models. Interpable models can pro-
2015). It is crucial to evaluate the diagnostic value vide explanations for the outcome, beyond predict-
of such signatures: their ability to correctly identify ing it. Unlike activations in single brain regions,
true cases (sensitivity, or how robustly the measure distributed patterns across voxels might be harder
is induced by the outcome) and correctly identify to interpret, particularly (but not only) when
false cases (specificity, or how robustly the meas- non-linear models are used (Haufe et al., 2014).
ure only responds to its outcome and not to other A protocol for interpreting predictive models in
outcomes) (Kragel, Koban, et al., 2018; Woo et al., neuroimaging was recently published (Kohoutová
2017). This is because a signature that was devel- et al., 2020). One key idea is the ability to evalu-
oped to predict a particular outcome (i.e., behavior) ate models’ sensitivity, specificity, and generaliz-
based on a specific type of data does not necessarily ability, defining the “boundary conditions” of the
capture the behavior’s unique features. For exam- processes and tasks they relate to and those they
ple, signatures that aim to identify pain can in fact do not. This is the process of construct valida-
identify negative affect more generally, arousal, or tion (Cronbach and Meehl, 1955; Kragel et al.,
other processes that co-occur with pain. 2018a,b). A second key idea is to develop models
Predictive performance characteristics of brain whose parts are interpretable and can be related
signatures are already becoming central to their to mechanistic literature in nonhuman animals and
evaluation, and are likely to become more com- humans (pharmacology, brain stimulation, inva-
mon practice in the future as the methods develop. sive neuronal recordings, optogenetic and chemo-
Such evaluation of diagnostic value can be part genetic manipulation of circuits, etc.). Models that
of the original study, but could also be done pro- are sparse (have few predictors) can help if the few
spectively. New findings about which processes predictive variables make sense (Grosenick et al.,
are predicted by the signature can gradually 2013). Another approach is using structure coef-
refine the definition of the construct it assumes to ficients (or model encoding maps) to identify how
measure, just as science progresses gradually and individual brain voxels relate to a more integra-
theories are refined based on new observations. tive model (Čeko et al., 2022; Haufe et al., 2014).
Prospective tests of brain signatures, or predictive Finally, a third idea is to build integrative models
models more broadly, require researchers to share up hierarchically from combinations of individual
their published models in a way that allows other regions (Schrouff et al., 2013) or neural pathways
researchers to apply them on new data. This is (Kragel et al., 2021).
challenging in the current atmosphere in which, as Paradigm shift from single regions and single
was nicely stated more than a decade ago (Mischel, measurement types to integrative and distributed
2008), researchers in the field “treat other peoples’ predictive brain models holds promise for advanc-
theories [or in our case, models] like toothbrushes ing cognitive neuroscience by systematically
– no self-respecting person wants to use anyone developing models that can represent, predict, and
else’s.” The field will have to overcome this ten- explain cognitive outcomes. Continuous methodo-
dency in order to work together toward integrative logical and conceptual progress is likely to lead to
and useful models and theories. more fundamental contributions to the fields that
A fundamental challenge is to ensure that the can potentially change the way cognitive neuro-
predictive models are indeed reflecting brain scientists think about specific cognitive processes,
representations of mental processes, rather than and the brain.
confounding or contextual variables like head
movement, eye movement patterns, or physiologi-
cal noise. While such variables can contribute to
the knowledge base (for example, it is valuable
to learn that eye movement patterns predict the SUMMARY AND CONCLUSIONS
valence of visual information), they indicate that
the model does not capture the neural represen- Studying how the brain gives rise to cognition is
tation of the mental process (Woo et al., 2017). like solving an extremely challenging jigsaw
One approach to mitigate this challenge is to test puzzle. Until now, cognitive neuroscience has
models on varying contexts, for example varying largely focused on producing more and more
582 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

pieces of information, and putting them together in A massive 7T fMRI dataset to bridge cognitive
localized, specific, and frequently narrow research neuroscience and artificial intelligence. Nature
areas. Consequently, we constructed parts of the Neuroscience, 25(1), 116–126.
image, but are relatively far from connecting these Anderson, J. R. (2009). How can the human mind
isolated partial images together. Moreover, alarm- occur in the physical universe? Oxford, UK: Oxford
ing findings about the reproducibility, replicability, University Press.
reliability, generalizability, and validity of findings Anderson, J. R., Lebiere, C., Lovett, M., & Reder, L.
in cognitive neuroscience and related fields are sug- (1998). ACT-R: A higher-level account of process-
gesting that many of the puzzle pieces we are trying ing capacity. Behavioral and Brain Sciences, 21(6),
to put together do not belong in this puzzle. Since 831–832.
we have no idea what the final picture looks like, Arbabshirani, M. R., Plis, S., Sui, J., & Calhoun, V. D.
identifying such false positive findings is extremely (2017). Single subject prediction of brain disorders
hard. We suggest that to advance cognitive neuro- in neuroimaging: Promises and pitfalls. NeuroIm-
science toward integrative, robust, and valid knowl- age, 145(B), 137–165.
edge and theories, we need to (1) improve our Arnett, J. J. (2008). The neglected 95%: Why Ameri-
ability to identify true (reproducible, replicable, can psychology needs to become less American.
robust) pieces by balancing between theoretical American Psychologist, 63(7), 602–614.
ideals and practical constraints; (2) develop better Averbeck, B. B., Latham, P. E., & Pouget, A. (2006).
and testable models and theories for sub-areas of Neural correlations, population coding and computa-
the puzzle and how they all connect together to tion. Nature Reviews: Neuroscience, 7(5), 358–366.
represent the broader picture; and (3) integrate Bagó, B., Aczel, B., Kekecs, Z., Protzko, J., Kovacs,
across brain regions to build predictive models M., Nagy, T., … Dutra, N. (2019). Moral thinking
based on multivariate information that can accu- across the world: Exploring the influence of per-
rately predict cognitive outcome. By doing so, the sonal force and intention in moral dilemma judg-
picture of the neural basis of cognition, as repre- ments. doi: 10.31234/osf.io/9uaqm
sented by the puzzle picture, will start to emerge. Barrett, H. C. (2020). Towards a cognitive science of the
human: Cross-cultural approaches and their urgency.
Trends in Cognitive Sciences, 24(8), 620–638.
Barrett, L. F. (2017). The theory of constructed emo-
tion: an active inference account of interoception
REFERENCES and categorization. Social Cognitive and Affective
Neuroscience, 12(1), 1–23.
Aczel, B., Szaszi, B., Nilsonne, G., van den Akker, Barrett, L. F., & Simmons, W. K. (2015). Interoceptive
O. R., Albers, C. J., van Assen, M. A., … Wagen- predictions in the brain. Nature Reviews: Neurosci-
makers, E.-J. (2021). Consensus-based guidance ence, 16(7), 419–429.
for conducting and reporting multi-analyst stud- Bastiaansen, J. A., Kunkels, Y. K., Blaauw, F. J., Boker,
ies. eLife, 10. doi: 10.7554/eLife.72185 S. M., Ceulemans, E., Chen, M., … Bringmann, L. F.
Adams, R. A., Shipp, S., & Friston, K. J. (2013). Pre- (2020). Time to get personal? The impact of
dictions not commands: Active inference in the researchers’ choices on the selection of treatment
motor system. Brain Structure & Function, 218(3), targets using the experience sampling methodology.
611–643. Journal of Psychosomatic Research, 137, 110211.
Adhikari, B. M., Jahanshad, N., Shukla, D., Turner, J., Bastos, A. M., Usrey, W. M., Adams, R. A., Mangun,
Grotegerd, D., Dannlowski, U., … Kochunov, P. G. R., Fries, P., & Friston, K. J. (2012). Canonical
(2019). A resting state fMRI analysis pipeline for microcircuits for predictive coding. Neuron, 76(4),
pooling inference across diverse cohorts: An 695–711.
ENIGMA rs-fMRI protocol. Brain Imaging and Bennett, D. A., Buchman, A. S., Boyle, P. A., Barnes,
Behavior, 13(5), 1453–1467. L. L., Wilson, R. S., & Schneider, J. A. (2018). Reli-
Aitchison, L., & Lengyel, M. (2017). With or without gious orders study and rush memory and aging
you: Predictive coding and Bayesian inference in project. Journal of Alzheimer’s Disease, 64(S1),
the brain. Current Opinion in Neurobiology, 46, S161–S189.
219–227. Borghi, J. A., & Van Gulick, A. E. (2018). Data man-
Alfaro-Almagro, F., Jenkinson, M., Bangerter, N. K., agement and sharing in neuroimaging: Practices
Andersson, J. L. R., Griffanti, L., Douaud, G., … and perceptions of MRI researchers. PloS One,
Smith, S. M. (2018). Image processing and quality 13(7). doi: 10.1371/journal.pone.0200562
control for the first 10,000 brain imaging datasets Borsook, D., Becerra, L., & Hargreaves, R. (2011).
from UK Biobank. NeuroImage, 166, 400–424. Biomarkers for chronic pain and analgesia. Part 1:
Allen, E. J., St-Yves, G., Wu, Y., Breedlove, J. L., The need, reality, challenges, and solutions. Dis-
Prince, J. S., Dowdle, L. T., … Kay, K. (2022). covery Medicine, 11(58), 197–207.
Advancing Cognitive Neuroscience 583

Borst, J. P., & Anderson, J. R. (2015). Using the ACT-R Chambers, C. (2019). What’s next for registered
cognitive architecture in combination with fMRI reports? Nature, 573, 187–189.
data. In B. U. Forstmann & E.-J. Wagenmakers Chang, L. J., Yarkoni, T., Khaw, M. W., & Sanfey,
(Eds.), An introduction to model-based cognitive A. G. (2013). Decoding the role of the insula in
neuroscience (pp. 339–352). New York: Springer. human cognition: Functional parcellation and
Botvinik-Nezer, R., Holzmeister, F., Camerer, C. F., large-scale reverse inference. Cerebral Cortex,
Dreber, A., Huber, J., Johannesson, M., … Schon- 23(3), 739–749.
berg, T. (2020). Variability in the analysis of a Chennu, S., Noreika, V., Gueorguiev, D., Blenkmann,
single neuroimaging dataset by many teams. A., Kochen, S., Ibáñez, A., … Bekinschtein, T. A.
Nature, 582, 84–88. (2013). Expectation and attention in hierarchical
Breznau, N., Rinke, E. M., Wuttke, A., Nguyen, H. H. auditory prediction. Journal of Neuroscience,
V., Adem, M., Adriaans, J., … Żółtak, T. (2022). 33(27), 11194–11205.
Observing many researchers using the same data Chen, S.-C., Szabelska, A., Chartier, C. R., Kekecs, Z.,
and hypothesis reveals a hidden universe of uncer- Lynott, D., Bernabeu, P., … Schmidt, K. (2018).
tainty. Proceedings of the National Academy of Investigating object orientation effects across 14
Sciences of the United States of America, 119(44), languages. doi: 10.31234/osf.io/t2pjv
doi: 10.1073/pnas.2203150119 Chiao, J. Y., & Cheon, B. K. (2010). The weirdest
Büchel, C., Geuter, S., Sprenger, C., & Eippert, F. brains in the world. Behavioral and Brain Sciences,
(2014). Placebo analgesia: A predictive coding 33(2-3), 88–90.
perspective. Neuron, 81(6), 1223–1239. Chib, V. S., Rangel, A., Shimojo, S., & O’Doherty, J. P.
Button, K. S. (2019). Double-dipping revisited. Neu- (2009). Evidence for a common representation
roscience, 22, 688–690. of decision values for dissimilar goods in human
Button, K. S., Ioannidis, J. P. A., Mokrysz, C., Nosek, ventromedial prefrontal cortex. Journal of Neuro-
B. A., Flint, J., Robinson, E. S. J., & Munafò, M. R. science, 29(39), 12315–12320.
(2013). Power failure: Why small sample size Chikazoe, J., Lee, D. H., Kriegeskorte, N., & Ander-
undermines the reliability of neuroscience. Nature son, A. K. (2014). Population coding of affect
Reviews. Neuroscience, 14(5), 365–376. across stimuli, modalities and individuals. Nature
Buzsáki, G. (2020). The brain-cognitive behavior Neuroscience, 17(8), 1114–1122.
problem: A retrospective. eNeuro, 7(4). doi: Clark, A. (2013). Whatever next? Predictive brains,
10.1523/ENEURO.0069-20.2020 situated agents, and the future of cognitive science.
Camerer, C. F., Dreber, A., Forsell, E., Ho, T.-H., Behavioral and Brain Sciences, 36(3), 181–204.
Huber, J., Johannesson, M., … Wu, H. (2016). Clayson, P. E., Baldwin, S. A., Rocha, H. A., & Larson,
Evaluating replicability of laboratory experiments M. J. (2021). The data-processing multiverse of
in economics. Science, 351, 1433–1436. event-related potentials (ERPs): A roadmap for the
Camerer, C. F., Dreber, A., Holzmeister, F., Ho, T.-H., optimization and standardization of ERP process-
Huber, J., Johannesson, M., … Wu, H. (2018). ing and reduction pipelines. NeuroImage. doi:
Evaluating the replicability of social science experi- 10.1016/j.neuroimage.2021.118712
ments in Nature and Science between 2010 and Cohen, J. D., Daw, N., Engelhardt, B., Hasson, U., Li,
2015. Nature Human Behaviour, 2. doi: 10.1038/ K., Niv, Y., … Willke, T. L. (2017). Computational
s41562-018-0399-z approaches to fMRI analysis. Nature Neuroscience,
Carp, J. (2012). On the plurality of (methodological) 20(3), 304–313.
worlds: Estimating the analytic flexibility of fMRI Coles, N. A., Forscher, P. S., Flake, J. K., DeBruine, L.,
experiments. Frontiers in Neuroscience, 6. doi: & Jones, B. (2021). Promises and challenges of big
10.3389/fnins.2012.00149 team psychology. https://socialsciences.nature.
Carroll, S. R., Herczog, E., Hudson, M., Russell, K., & com/posts/promises-and-challenges-of-big-team-
Stall, S. (2021). Operationalizing the CARE and FAIR psychology (retrieved Sept. 29, 2022)
principles for indigenous data futures. Scientific Coles, N. A., Hamlin, J. K., Sullivan, L. L., Parker, T. H.,
Data, 8(1). doi: 10.1038/s41597-021-00892-0 & Altschul, D. (2022). Build up big-team science.
Casey, B. J., Cannonier, T., Conley, M. I., Cohen, Nature, 601, 505–507.
A. O., Barch, D. M., Heitzeg, M. M., … ABCD Cronbach, L. J., & Meehl, P. E. (1955). Construct
Imaging Acquisition Workgroup (2018). The Ado- validity in psychological tests. Psychological Bulle-
lescent Brain Cognitive Development (ABCD) tin, 52(4), 281–302.
study: Imaging acquisition across 21 sites. Devel- Dafflon, J., Da Costa, P. F., Váša, F., Monti, R. P.,
opmental Cognitive Neuroscience, 32, 43–54. Bzdok, D., Hellyer, P. J., … Leech, R. (2020). Neu-
Čeko, M., Kragel, P. A., Woo, C.-W., López-Solà, M., roimaging: Into the multiverse. bioRxiv. doi:
& Wager, T. D. (2022). Common and stimulus- 10.1101/2020.10.29.359778
type-specific brain representations of negative Delgado, M. R., Beer, J. S., Fellows, L. K., Huettel, S. A.,
affect. Nature Neuroscience, 25(6), 760–770. Platt, M. L., Quirk, G. J., & Schiller, D. (2016).
584 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Viewpoints: Dialogues on the functional role of evaluation. Human Brain Mapping, 25(1),
the ventromedial prefrontal cortex. Nature Neuro- 185–198.
science, 19(12), 1545–1552. Fox, P. T., & Lancaster, J. L. (2002). Opinion: Mapping
Di Martino, A., O’Connor, D., Chen, B., Alaerts, K., context and content: The BrainMap model. Nature
Anderson, J. S., Assaf, M., … Milham, M. P. (2017). Reviews: Neuroscience, 3(4), 319–321.
Enhancing studies of the connectome in autism Friston, K. (2010). The free-energy principle: a unified
using the autism brain imaging data exchange II. brain theory? Nature Reviews: Neuroscience. doi:
Scientific Data, 4. doi: 10.1038/sdata.2017.10 10.1038/nrn2787
Di Martino, A., Yan, C.-G., Li, Q., Denio, E., Castellanos, Friston, K. (2018). Does predictive coding have a
F. X., Alaerts, K., … Milham, M. P. (2014). The autism future? Nature Neuroscience, 21, 1019–1021.
brain imaging data exchange: Towards a large-scale Friston, K. J., Daunizeau, J., Kilner, J., & Kiebel, S. J.
evaluation of the intrinsic brain architecture in (2010). Action and behavior: A free-energy formu-
autism. Molecular Psychiatry, 19(6), 659–667. lation. Biological Cybernetics, 102(3), 227–260.
Dockès, J., Poldrack, R. A., Primet, R., Gözükan, H., Gabrieli, J. D. E., Ghosh, S. S., & Whitfield-Gabrieli, S.
Yarkoni, T., Suchanek, F., … Varoquaux, G. (2020). (2015). Prediction as a humanitarian and prag-
NeuroQuery, comprehensive meta-analysis of matic contribution from human cognitive neuro-
human brain mapping. eLife, 9. doi: 10.7554/ science. Neuron, 85(1), 11–26.
eLife.53385 Gelman, A., & Loken, E. (2013). The garden of fork-
Ebersole, C. R., Atherton, O. E., Belanger, A. L., Skul- ing paths: Why multiple comparisons can be a
borstad, H. M., Allen, J. M., Banks, J. B., … Nosek, problem, even when there is no “fishing expedi-
B. A. (2016). Many labs 3: Evaluating participant tion” or “p-hacking” and the research hypothesis
pool quality across the academic semester via rep- was posited ahead of time. http://stat.columbia.
lication. Journal of Experimental Social Psychology, edu/∼gelman/research/unpublished/forking.pdf
67, 68–82. (retrieved Sept. 29, 2022)
Elam, J. S., Glasser, M. F., Harms, M. P., Sotiropoulos, Gilmore, R. O., Diaz, M. T., Wyble, B. A., & Yarkoni,
S. N., Andersson, J. L. R., Burgess, G. C., … Van T. (2017). Progress toward openness, transpar-
Essen, D. C. (2021). The human connectome pro- ency, and reproducibility in cognitive neurosci-
ject: A retrospective. NeuroImage. doi: 10.1016/ ence. Annals of the New York Academy of
j.neuroimage.2021.118543 Sciences, 1396(1), 5–18.
Errington, T. M., Mathur, M., Soderberg, C. K., Denis, Gordon, E. M., Laumann, T. O., Gilmore, A. W., New-
A., Perfito, N., Iorns, E., & Nosek, B. A. (2021). bold, D. J., Greene, D. J., Berg, J. J., … Dosenbach,
Investigating the replicability of preclinical cancer N. U. F. (2017). Precision functional mapping of
biology. eLife, 10. doi: 10.7554/eLife.71601 individual human brains. Neuron, 95(4), 791–807.
Fanelli, D. (2012). Negative results are disappearing Gorgolewski, K. J., Auer, T., Calhoun, V. D., Crad-
from most disciplines and countries. Scientomet- dock, R. C., Das, S., Duff, E. P., … Poldrack, R. A.
rics, 90(3), 891–904. (2016). The brain imaging data structure, a format
Fan, Y., Batmanghelich, N., Clark, C. M., Davatzikos, for organizing and describing outputs of neuroim-
C., & Alzheimer’s disease neuroimaging initiative aging experiments. Scientific Data, 3. doi:
(2008). Spatial patterns of brain atrophy in MCI 10.1038/sdata.2016.44
patients, identified via high-dimensional pattern Gorgolewski, K. J., Varoquaux, G., Rivera, G., Schwarz,
classification, predict subsequent cognitive decline. Y., Ghosh, S. S., Maumet, C., … Margulies, D. S.
NeuroImage, 39(4), 1731–1743. (2015). NeuroVault.org: A web-based repository for
Forscher, P. S., Paris, B., Primbs, M., & Coles, N. A. collecting and sharing unthresholded statistical
(2020). PSACR: The psychological science accelera- maps of the human brain. Frontiers in Neuroinfor-
tor’s COVID-19 rapid-response project. doi: matics, 9. doi: 10.3389/fninf.2015.00008
10.31234/osf.io/x976j Grosenick, L., Klingenberg, B., Katovich, K., Knutson,
Forscher, P. S., Taylor, V. J., Cavagnaro, D., Lewis, N. A., B., & Taylor, J. E. (2013). Interpretable whole-brain
Buchanan, E. M., Moshontz, H., … Chartier, C. R. prediction analysis with GraphNet. NeuroImage,
(2019). Stereotype threat in black college students 72, 304–321.
across many operationalizations. doi: 10.31234/osf. Gruber, T. (1993). Ontolingua: A translation approach
io/6hju9 to providing portable ontology specifications.
Forscher, P. S., Wagenmakers, E.-J., Coles, N. A., Silan, Knowledge Acquisition, 5(2), 199–200.
M. A., Dutra, N. B., Basnight-Brown, D., & IJzer- Guest, O., & Martin, A. E. (2021). How computa-
man, H. (2020). The benefits, barriers, and risks of tional modeling can force theory building in psy-
big team science. doi: 10.31234/osf.io/2mdxh chological science. Perspectives on Psychological
Fox, P. T., Laird, A. R., Fox, S. P., Fox, P. M., Uecker, A. Science, 16(4), 789–802.
M., Crank, M., … Lancaster, J. L. (2005). BrainMap Hahn, T., Kircher, T., Straube, B., Wittchen, H.-U.,
taxonomy of experimental design: Description and Konrad, C., Ströhle, A., … Lueken, U. (2015).
Advancing Cognitive Neuroscience 585

Predicting treatment response to cognitive behav- Henrich, J., Heine, S. J., & Norenzayan, A. (2010).
ioral therapy in panic disorder with agoraphobia The weirdest people in the world? Behavioral and
by integrating local neural information. JAMA Brain Sciences, 33(2-3), 61–83.
Psychiatry, 72(1), 68–74. He, T., An, L., Chen, P., Chen, J., Feng, J., Bzdok, D.,
Halchenko, Y., Meyer, K., Poldrack, B., Solanky, D., … Yeo, B. T. T. (2022). Meta-matching as a simple
Wagner, A., Gors, J., … Hanke, M. (2021). Data- framework to translate phenotypic predictive
Lad: Distributed system for joint management of models from big to small data. Nature Neurosci-
code, data, and their relationship. Journal of ence, 25(6), 795–804.
Open Source Software , 6(63). doi: 10.21105/ Hohwy, J. (2012). Attention and conscious percep-
joss.03262 tion in the hypothesis testing brain. Frontiers in
Hall, B. F., Wagge, J. R., Pfuhl, G., Stieger, S., Ver- Psychology, 3. doi: 10.3389/fpsyg.2012.00096
gauwe, E., Lizerman, H., … Lewis, S. (2018). Holmes, A. J., Hollinshead, M. O., O’Keefe, T. M.,
Accelerated CREP—RRR: Turri, Buckwalter, & Petrov, V. I., Fariello, G. R., Wald, L. L., … Buckner,
Blouw (2015). doi: 10.31234/osf.io/zeux9 R. L. (2015). Brain genomics superstruct project
Han, H., & Park, J. (2019). Bayesian meta-analysis of initial data release with structural, functional, and
fMRI image data. Cognitive Neuroscience, 10(2), behavioral measures. Scientific Data, 2. doi:
66–76. 10.1038/sdata.2015.31
Hardwicke, T. E., & Ioannidis, J. P. A. (2018). Mapping Houtkoop, B. L., Chambers, C., Macleod, M., Bishop,
the universe of registered reports. Nature Human D. V. M., Nichols, T. E., & Wagenmakers, E.-J.
Behaviour, 2(11), 793–796. (2018). Data sharing in psychology: A survey on
Hardwicke, T. E., Thibault, R. T., Kosie, J. E., Wallach, barriers and preconditions. Advances in Methods
J. D., Kidwell, M. C., & Ioannidis, J. P. A. (2021). and Practices in Psychological Science, 1(1), 70–85.
Estimating the prevalence of transparency and Insel, T., Cuthbert, B., Garvey, M., Heinssen, R., Pine,
reproducibility-related research practices in psy- D. S., Quinn, K., … Wang, P. (2010). Research
chology (2014-2017). Perspectives on Psychologi- domain criteria (RDoC): Toward a new classifica-
cal Science, 17(1), 239–251. tion framework for research on mental disorders.
Harrison, S. A., & Tong, F. (2009). Decoding reveals American Journal of Psychiatry, 167(7), 748–751.
the contents of visual working memory in early Insel, T. R. (2014). The NIMH Research Domain Criteria
visual areas. Nature, 458, 632–635. (RDoC) project: Precision medicine for psychiatry.
Hasson, U., Nastase, S. A., & Goldstein, A. (2020). American Journal of Psychiatry, 171(4), 395–397.
Direct fit to nature: An evolutionary perspective on Ioannidis, J. P. A. (2005). Why most published
biological and artificial neural networks. Neuron, research findings are false. PLoS Medicine, 2(8).
105(3), 416–434. doi: 10.1371/journal.pmed.0020124
Hastings, J., Frishkoff, G. A., Smith, B., Jensen, M., Ioannidis, J. P. A., Munafò, M. R., Fusar-Poli, P.,
Poldrack, R. A., Lomax, J., … Martone, M. E. Nosek, B. A., & David, S. P. (2014). Publication
(2014). Interdisciplinary perspectives on the devel- and other reporting biases in cognitive sciences:
opment, integration, and application of cognitive Detection, prevalence, and prevention. Trends in
ontologies. Frontiers in Neuroinformatics, 8. doi: Cognitive Sciences, 18(5), 235–241.
10.3389/fninf.2014.00062 Jack, C. R., Jr, Bernstein, M. A., Fox, N. C., Thompson,
Haufe, S., Meinecke, F., Görgen, K., Dähne, S., P., Alexander, G., Harvey, D., … Weiner, M. W.
Haynes, J.-D., Blankertz, B., & Bießmann, F. (2014). (2008). The Alzheimer’s Disease Neuroimaging
On the interpretation of weight vectors of linear Initiative (ADNI): MRI methods. Journal of Mag-
models in multivariate neuroimaging. NeuroIm- netic Resonance Imaging, 27(4), 685–691.
age, 87, 96–110. John, L. K., Loewenstein, G., & Prelec, D. (2012).
Haxby, J. V., Guntupalli, J. S., Connolly, A. C., Measuring the prevalence of questionable research
Halchenko, Y. O., Conroy, B. R., Gobbini, M. I., … practices with incentives for truth telling. Psycho-
Ramadge, P. J. (2011). A common, high-­ logical Science, 23(5), 524–532.
dimensional model of the representational space Jones, B. C., DeBruine, L. M., Flake, J. K., Liuzza,
in human ventral temporal cortex. Neuron, 72(2), M. T., Antfolk, J., Arinze, N. C., … Coles, N. A.
404–416. (2021). To which world regions does the valence-
Hayashi, S., Caron, B., Heinsfeld, A. S., Vinci-Booher, dominance model of social perception apply?
S., McPherson, B. C., Bullock, D. N., … & Pestilli, F. Nature Human Behaviour, 5(1), 159–169.
(2023). brainlife. io: A decentralized and open Kanwisher, N., McDermott, J., & Chun, M. M. (1997).
source cloud platform to support neuroscience The fusiform face area: A module in human extras-
research. arXiv preprint arXiv:2306.02183. triate cortex specialized for face perception. Jour-
Haynes, J.-D. (2015). A primer on Ppttern-based nal of Neuroscience, 17(11), 4302–4311.
approaches to fMRI: Principles, pitfalls, and per- Kaye, A. P., & Krystal, J. H. (2020). Predictive process-
spectives. Neuron, 87(2), 257–270. ing in mental illness: Hierarchical circuitry for
586 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

perception and trauma. Journal of Abnormal Psy- Kragel, P. A., Koban, L., Barrett, L. F., & Wager, T. D.
chology, 129(6), 629–632. (2018b). Representation, pattern information, and
Kerr, N. L. (1998). HARKing: hypothesizing after the brain signatures: From neurons to neuroimaging.
results are known. Personality and Social Psychol- Neuron, 99(2), 257–273.
ogy Review, 2(3), 196–217. Kragel, P. A., & LaBar, K. S. (2015). Multivariate
Kieras, D. E. (2016). A summary of the EPIC cognitive neural biomarkers of emotional states are
architecture. In S. E. F. Chipman (Ed.), The Oxford ­categorically distinct. Social Cognitive and Affec-
handbook of cognitive science. Oxford, UK: tive Neuroscience, 10(11), 1437–1448.
Oxford University Press. Kube, T., Schwarting, R., Rozenkrantz, L., Glom-
Klein, R. A., Ratliff, K. A., Vianello, M., Adams, R. B., biewski, J. A., & Rief, W. (2020). Distorted cogni-
Bahník, Š., Bernstein, M. J., … Nosek, B. A. (2014). tive processes in major depression: A predictive
Investigating variation in replicability: A “many processing perspective. Biological Psychiatry,
labs” replication project. Social Psychology, 45(3), 87(5), 388–398.
142–152. Kunda, Z. (1990). The case for motivated reasoning.
Klein, R. A., Vianello, M., Hasselman, F., Adams, Psychological Bulletin, 108(3), 480–498.
B. G., Adams, R. B., Alper, S., … Nosek, B. A. Kurth, F., Zilles, K., Fox, P. T., Laird, A. R., & Eickhoff,
(2018). Many labs 2: Investigating variation in S. B. (2010). A link between the systems: Func-
replicability across samples and settings. Advances tional differentiation and integration within the
in Methods and Practices in Psychological Science, human insula revealed by meta-analysis. Brain
1(4), 443–490. Structure & Function, 214(5-6), 519–534.
Knill, D. C., & Pouget, A. (2004). The Bayesian brain: Kwisthout, J., & van Rooij, I. (2020). Computational
The role of uncertainty in neural coding and com- resource demands of a predictive Bayesian brain.
putation. Trends in Neurosciences, 27(12), Computational Brain & Behavior, 3(2), 174–188.
712–719. Laird, A. R., Lancaster, J. L., & Fox, P. T. (2005). Brain-
Knudsen, G. M., Ganz, M., Appelhoff, S., Boellaard, Map: The social evolution of a functional neuroim-
R., Bormans, G., Carson, R. E., … Innis, R. B. aging database. Neuroinformatics, 3, 65–78.
(2020). Guidelines for the content and format of Laird, J. E. (2019). The Soar cognitive architecture.
PET brain data in publications and archives: A Cambridge, MA: MIT Press.
consensus paper. Journal of Cerebral Blood Flow LeCun, Y., Bengio, Y., & Hinton, G. (2015). Deep
and Metabolism, 40(8), 1576–1585. learning. Nature, 521(7553), 436–444.
Kogo, N., & Trengove, C. (2015). Is predictive coding Levy, D. J., & Glimcher, P. W. (2012). The root of all
theory articulated enough to be testable? Frontiers value: A neural common currency for choice. Cur-
in Computational Neuroscience, 9. doi: 10.3389/ rent Opinion in Neurobiology, 22(6), 1027–1038.
fncom.2015.00111 Lillicrap, T. P., & Kording, K. P. (2019). What does it
Kohoutová, L., Heo, J., Cha, S., Lee, S., Moon, T., mean to understand a neural network? http://
Wager, T. D., & Woo, C.-W. (2020). Toward a uni- arxiv.org/abs/1907.06374 (retrieved Sept. 29,
fied framework for interpreting machine-learning 2022)
models in neuroimaging. Nature Protocols, 15(4), Liu, Y., Kale, A., Althoff, T., & Heer, J. (2021). Boba:
1399–1435. Authoring and visualizing multiverse analyses. IEEE
Koster-Hale, J., & Saxe, R. (2013). Theory of mind: A Transactions on Visualization and Computer
neural prediction problem. Neuron, 79(5), Graphics, 27(2), 1753–1763.
836–848. Li, X., Ai, L., Giavasis, S., Jin, H., Feczko, E., Xu, T., …
Koutsouleris, N., Meisenzahl, E. M., Davatzikos, C., Milham, M. P. (2021). Moving beyond processing
Bottlender, R., Frodl, T., Scheuerecker, J., … and analysis-related variation in neuroscience.
Gaser, C. (2009). Use of neuroanatomical pattern bioRxiv. doi: 10.1101/2021.12.01.470790
classification to identify subjects in at-risk mental Marcus, D. S., Wang, T. H., Parker, J., Csernansky,
states of psychosis and predict disease transition. J. G., Morris, J. C., & Buckner, R. L. (2007). Open
Archives of General Psychiatry, 66(7), 700–712. Access Series of Imaging Studies (OASIS): Cross-
Kragel, P. A., Čeko, M., Theriault, J., Chen, D., Satpute, sectional MRI data in young, middle aged, nonde-
A. B., Wald, L. W., … Wager, T. D. (2021). A human mented, and demented older adults. Journal of
colliculus-pulvinar-amygdala pathway encodes neg- Cognitive Neuroscience, 19(9), 1498–1507.
ative emotion. Neuron, 109(15), 2404–2412. Marek, K., Jennings, D., Lasch, S., Siderowf, A.,
Kragel, P. A., Kano, M., Van Oudenhove, L., Ly, H. G., Tanner, C., Simuni, T., … Taylor, P. (2011). The
Dupont, P., Rubio, A., … Wager, T. D. (2018a). Parkinson Progression Marker Initiative (PPMI).
Generalizable representations of pain, cognitive Progress in Neurobiology, 95(4), 629–635.
control, and negative emotion in medial frontal Markiewicz, C. J., Gorgolewski, K. J., Feingold, F.,
cortex. Nature Neuroscience, 21(2), 283–289. Blair, R., Halchenko, Y. O., Miller, E., … Poldrack,
R. (2021). The OpenNeuro resource for sharing of
Advancing Cognitive Neuroscience 587

neuroscience data. eLife, 10. doi: 10.7554/ neuroimaging using MRI. Nature Neuroscience,
eLife.71774 20(3), 299–303.
Mennes, M., Biswal, B. B., Castellanos, F. X., & Nickerson, R. S. (1998). Confirmation bias: A ubiqui-
Milham, M. P. (2013). Making data sharing work: tous phenomenon in many guises. Review of
The FCP/INDI experience. NeuroImage, 82, General Psychology, 2(2), 175–220.
683–691. Niso, G., Botvinik-Nezer, R., Appelhoff, S., Vega, A.
Meyer, D. E., & Kieras, D. E. (1997). A computational D. L., Esteban, O., Etzel, J. A., … Rieger, J. W.
theory of executive cognitive processes and multiple- (2022). Open and reproducible neuroimaging:
task performance: Part 1. Basic mechanisms. Psy- From study inception to publication. NeuroImage.
chological Review, 104(1), 3–65. doi: 10.1016/j.neuroimage.2022.119623
Miller, K. L., Alfaro-Almagro, F., Bangerter, N. K., Niv, Y. (2019). Learning task-state representations.
Thomas, D. L., Yacoub, E., Xu, J., … Smith, S. M. Nature Neuroscience, 22(10), 1544–1553.
(2016). Multimodal population brain imaging in Nørgaard, M., Ganz, M., Svarer, C., Frokjaer, V. G.,
the UK Biobank prospective epidemiological study. Greve, D. N., Strother, S. C., & Knudsen, G. M.
Nature Neuroscience, 19(11), 1523–1536. (2020). Different preprocessing strategies lead to
Mischel, W. (2008). The toothbrush problem. APS different conclusions: A [11C]DASB-PET reproduc-
Observer (Vol. 21). Washington, DC: Association ibility study. Journal of Cerebral Blood Flow and
for Psychological Science. Metabolism, 40(9), 1902–1911.
Moshontz, H., Campbell, L., Ebersole, C. R., IJzerman, Norman, K. a., Polyn, S. M., Detre, G. J., & Haxby, J. V.
H., Urry, H. L., Forscher, P. S., … Chartier, C. R. (2006). Beyond mind-reading: multi-voxel pattern
(2018). The psychological science accelerator: analysis of fMRI data. Trends in Cognitive Sciences,
Advancing psychology through a distributed col- 10(9), 424–430.
laborative network. Advances in Methods and Nosek, B. A. (2015). Promoting an open research
Practices in Psychological Science, 1(4), 501–515. culture. Science, 348, 1422–1425.
Munafò, M. R., Cremers, H. R., Wager, T. D., & Nosek, B. A., Ebersole, C. R., DeHaven, A. C., & Mellor,
Yarkoni, T. (2019). 12 - Power and design consid- D. T. (2018). The preregistration revolution. Proceed-
erations in imaging research. In A. Raz & R. T. Thiba- ings of the National Academy of Sciences of the
ult (Eds.), Casting light on the dark side of brain United States of America, 115(11), 2600–2606.
imaging (pp. 73–78). London: Elsevier/Academic. Nosek, B. A., & Errington, T. M. (2017). Making sense
Munafò, M. R., Nosek, B. A., Bishop, D. V. M., of replications. eLife, 6, 4–7.
Button, K. S., Chambers, C. D., du Sert, N. P., … Nosek, B. A., & Errington, T. M. (2020a). The best
Ioannidis, J. P. A. (2017). A manifesto for repro- time to argue about what a replication means?
ducible science. Nature Human Behaviour, 1. doi: Before you do it. Nature, 583, 518–520.
10.1038/s41562-016-0021 Nosek, B. A., & Errington, T. M. (2020b). What is
Muthukrishna, M., & Henrich, J. (2019). A problem in replication? PLoS Biology, 18(3). doi: 10.1371/
theory. Nature Human Behaviour, 3(3), 221–229. journal.pbio.3000691
Nelson, L. D., Simmons, J., & Simonsohn, U. (2018). Nosek, B. A., & Lakens, D. (2014). Registered reports:
Psychology’s renaissance. Annual Review of Psy- A method to increase the credibility of published
chology, 69, 511–534. results. Social Psychology, 45(3), 137–141.
Neuromatch Academy (NMA). (2021). Neuromatch Nosek, B. A., Spies, J. R., & Motyl, M. (2012). Scientific
academy: Teaching computational neuroscience utopia: II. Restructuring incentives and practices to
with global accessibility. Trends in Cognitive Sci- promote truth over publishability. Perspectives on
ences, 25(7), 535–538. Psychological Science, 7(6), 615–631.
Newell, A. (1992). Unified theories of cognition and Oberauer, K., & Lewandowsky, S. (2019). Addressing
the role of Soar. In J. A. Michon & A. Akyürek the theory crisis in psychology. Psychonomic Bul-
(Eds.), Soar: A cognitive architecture in perspective: letin & Review, 26(5), 1596–1618.
A tribute to Allen Newell (pp. 25–79). Dordrecht: OSC (2015). Estimating the reproducibility of psycho-
Springer. logical science. Science. (Open Science Collabora-
Newell, A. (1994). Unified theories of cognition. tion). doi: 10.1126/science.aac4716
Cambridge, MA: Harvard University Press. Paret, C., Unverhau, N., Feingold, F., Poldrack, R. A.,
Ni, A. M., Ruff, D. A., Alberts, J. J., Symmonds, J., & Stirner, M., Schmahl, C., & Sicorello, M. (2022).
Cohen, M. R. (2018). Learning and attention Survey on open science practices in functional neu-
reveal a general relationship between population roimaging, 257, 119306.
activity and behavior. Science, 359, 463–465. Pernet, C., Garrido, M. I., Gramfort, A., Maurits, N.,
Nichols, T. E., Das, S., Eickhoff, S. B., Evans, A. C., Michel, C. M., Pang, E., … Puce, A. (2020). Issues
Glatard, T., Hanke, M., … Yeo, B. T. T. (2017). Best and recommendations from the OHBM COBIDAS
practices in data analysis and sharing in MEEG committee for reproducible EEG and MEG
research. Nature Neuroscience, 23(12), 1473–1483.
588 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Pinho, A. L., Amadon, A., Ruest, T., Fabre, M., Reddan, M. C., Lindquist, M. A., & Wager, T. D.
Dohmatob, E., Denghien, I., … Thirion, B. (2018). (2017). Effect size estimation in neuroimaging.
Individual brain charting: A high-resolution fMRI JAMA Psychiatry, 74(3), 207–208.
dataset for cognitive mapping. Scientific data, 5. Richards, B. A., Lillicrap, T. P., Beaudoin, P., Bengio, Y.,
doi: 10.1038/sdata.2018.105. PMID: 29893753 Bogacz, R., Christensen, A., … Kording, K. P.
Poeppel, D., & Adolfi, F. (2020). Against the episte- (2019). A deep learning framework for neurosci-
mological primacy of the hardware: The brain ence. Nature Neuroscience, 22(11), 1761–1770.
from inside out, turned upside down. eNeuro. doi: Rigotti, M., Barak, O., Warden, M. R., Wang, X.-J.,
10.1523/ENEURO.0215-20.2020 Daw, N. D., Miller, E. K., & Fusi, S. (2013). The
Poldrack, R. A. (2011). Inferring mental states from importance of mixed selectivity in complex cogni-
neuroimaging data: From reverse inference to tive tasks. Nature, 497, 585–590.
large-scale decoding. Neuron, 72(5), 692–697. Rosenberg, M. D., Finn, E. S., Scheinost, D., Papad-
Poldrack, R. A., Baker, C. I., Durnez, J., Gorgolewski, emetris, X., Shen, X., Constable, R. T., & Chun, M.
K. J., Matthews, P. M., Munafò, M. R., … Yarkoni, M. (2016). A neuromarker of sustained attention
T. (2017). Scanning the horizon: Towards trans- from whole-brain functional connectivity. Nature
parent and reproducible neuroimaging research. Neuroscience, 19(1), 165–171.
Nature Reviews: Neuroscience, 18(2), 115–126. Rosenthal, R. (1979). The file drawer problem and
Poldrack, R. A., & Farah, M. J. (2015). Progress and tolerance for null results. Psychological Bulletin,
challenges in probing the human brain. Nature, 86(3), 638–641.
526, 371–379. Rumelhart, D. E., Widrow, B., & Lehr, M. A. (1994).
Poldrack, R. A., Feingold, F., Frank, M. J., Gleeson, P., The basic ideas in neural networks, Communica-
de Hollander, G., Huys, Q. J., … Cohen, J. D. tions of the AC M, 37(3), 87–92.
(2019). The importance of standards for sharing of Salimi-Khorshidi, G., Smith, S. M., Keltner, J. R.,
computational models and data. Computational Wager, T. D., & Nichols, T. E. (2009). Meta-analysis
Brain & Behavior, 2(3-4), 229–232. of neuroimaging data: A comparison of image-
Poldrack, R. A., & Gorgolewski, K. J. (2014). Making based and coordinate-based pooling of studies.
big data open: Data sharing in neuroimaging. NeuroImage, 45(3), 810–823.
Nature Neuroscience, 17(11), 1510–1517. Saxe, A., Nelli, S., & Summerfield, C. (2021). If deep
Poldrack, R. A., Huckins, G., & Varoquaux, G. (2020). learning is the answer, what is the question?
Establishment of best practices for evidence for pre- Nature Reviews: Neuroscience, 22(1), 55–67.
diction: A review. JAMA Psychiatry, 77(5), 534–540. Schilling, K. G., Rheault, F., Petit, L., Hansen, C. B.,
Poldrack, R. A., Kittur, A., Kalar, D., Miller, E., Seppa, Nath, V., Yeh, F.-C., … Descoteaux, M. (2021).
C., Gil, Y., … Bilder, R. M. (2011). The cognitive Tractography dissection variability: What happens
atlas: Toward a knowledge foundation for cogni- when 42 groups dissect 14 white matter bundles
tive neuroscience. Frontiers in Neuroinformatics, on the same dataset? NeuroImage, 243. doi:
5. doi: 10.3389/fninf.2011.00017 10.1016/j.neuroimage.2021.118502
Poldrack, R. A., Laumann, T. O., Koyejo, O., Gregory, Schrouff, J., Rosa, M. J., Rondina, J. M., Marquand, A.
B., Hover, A., Chen, M.-Y., … Mumford, J. A. F., Chu, C., Ashburner, J., … Mourão-Miranda, J.
(2015). Long-term neural and physiological phe- (2013). PRoNTo: Pattern recognition for neuroim-
notyping of a single human. Nature Communica- aging toolbox. Neuroinformatics, 11(3), 319–337.
tions, 6. doi: 10.1038/ncomms9885 Schultz, W., Dayan, P., & Montague, P. R. (1997). A
Poldrack, R. A., & Yarkoni, T. (2016). From brain neural substrate of prediction and reward. Sci-
maps to cognitive ontologies: Informatics and the ence, 275, 1593–1599.
search for mental structure. Annual Review of Schumann, G., Loth, E., Banaschewski, T., Barbot, A.,
Psychology, 67, 587–612. Barker, G., Büchel, C., … IMAGEN consortium.
Pouget, A., Dayan, P., & Zemel, R. (2000). Informa- (2010). The IMAGEN study: Reinforcement-
tion processing with population codes. Nature related behaviour in normal brain function and
Reviews: Neuroscience, 1(2), 125–132. psychopathology. Molecular Psychiatry, 15(12),
Rad, M. S., Martingano, A. J., & Ginges, J. (2018). 1128–1139.
Toward a psychology of homo sapiens: Making Seth, A. K. (2013). Interoceptive inference, emotion,
psychological science more representative of the and the embodied self. Trends in Cognitive Sci-
human population. Proceedings of the National ences, 17(11), 565–573.
Academy of Sciences of the United States of Sharot, T., & Garrett, N. (2016). Forming beliefs: Why
America, 115(45), 11401–11405. valence matters. Trends in Cognitive Sciences,
Rao, R. P., & Ballard, D. H. (1999). Predictive coding 20(1), 25–33.
in the visual cortex: A functional interpretation of Silberzahn, R., Uhlmann, E. L., Martin, D. P., Anselmi,
some extra-classical receptive-field effects. Nature P., Aust, F., Awtrey, E., … Nosek, B. A. (2018).
Neuroscience, 2(1), 79–87. Many analysts, one data set: Making transparent
Advancing Cognitive Neuroscience 589

how variations in analytic choices affect results. Thompson, P. M., Stein, J. L., Medland, S. E., Hibar,
Advances in Methods and Practices in Psychologi- D. P., Vasquez, A. A., Renteria, M. E., … Alzhei-
cal Science, 1(3), 337–356. mer’s disease neuroimaging initiative, EPIGEN
Simmons, J., Nelson, L., & Simonsohn, U. (2021). Pre- Consortium, IMAGEN Consortium, Saguenay
registration: Why and how. Journal of Consumer Youth Study (SYS) Group. (2014). The ENIGMA
Psychology, 31(1), 151–162. Consortium: Large-scale collaborative analyses of
Simmons, J. P., Nelson, L. D., & Simonsohn, U. neuroimaging and genetic data. Brain Imaging
(2011). False-positive psychology: Undisclosed and Behavior, 8(2), 153–182.
flexibility in data collection and analysis allows Tobler, P. N., Fiorillo, C. D., & Schultz, W. (2005).
presenting anything as significant. Psychological Adaptive coding of reward value by dopamine
Science, 22(11), 1359–1366. neurons. Science, 307, 1642–1645.
Simonsohn, U., Nelson, L. D., & Simmons, J. P. (2014). Turner, J. A., & Laird, A. R. (2012). The cognitive
P-curve: A key to the file-drawer. Journal of Experi- paradigm ontology: Design and application. Neu-
mental Psychology: General, 143(2), 534–547. roinformatics, 10(1), 57–66.
Simonsohn, U., Simmons, J. P., & Nelson, L. D. Van de Cruys, S., Evers, K., Van der Hallen, R., Van Eylen,
(2020). Specification curve analysis. Nature Human L., Boets, B., de-Wit, L., & Wagemans, J. (2014).
Behaviour, 4(11), 1208–1214. Precise minds in uncertain worlds: predictive coding
Smith, R., Badcock, P., & Friston, K. J. (2021). Recent in autism. Psychological Review, 121(4), 649–675.
advances in the application of predictive coding and Van Essen, D. C., Smith, S. M., Barch, D. M., Behrens, T.
active inference models within clinical neuroscience. E. J., Yacoub, E., Ugurbil, K., & WU-Minn HCP Con-
Psychiatry and Clinical Neurosciences, 75(1), 3–13. sortium. (2013). The WU-Minn human connectome
Somerville, L. H., Bookheimer, S. Y., Buckner, R. L., project: An overview. NeuroImage, 80, 62–79.
Burgess, G. C., Curtiss, S. W., Dapretto, M., … Vickery, T. J., Chun, M. M., & Lee, D. (2011). Ubiquity
Barch, D. M. (2018). The lifespan human connec- and specificity of reinforcement signals through-
tome project in development: A large-scale study out the human brain. Neuron, 72(1), 166–177.
of brain connectivity development in 5-21 year Wager, T. D., Atlas, L. Y., Lindquist, M. A., Roy, M.,
olds. NeuroImage, 183, 456–468. Woo, C.-W., & Kross, E. (2013). An fMRI-based
Song, Y., Yao, M., Kemprecos, H., Byrne, A., Xiao, Z., neurologic signature of physical pain. New Eng-
Zhang, Q., … Chen, Z. S. (2021). Predictive coding land Journal of Medicine, 368(15), 1388–1397.
models for pain perception. Journal of Computa- Wager, T. D., Kang, J., Johnson, T. D., Nichols, T. E.,
tional Neuroscience, 49(2), 107–127. Satpute, A. B., & Barrett, L. F. (2015). A Bayesian
Squire, L. R. (1992). Memory and the hippocampus: model of category-specific emotional brain
A synthesis from findings with rats, monkeys, and responses. PLoS Computational Biology, 11(4).
humans. Psychological Review, 99(2), 195–231. doi: 10.1371/journal.pcbi.1004066
Sterzer, P., Adams, R. A., Fletcher, P., Frith, C., Lawrie, Woo, C.-W., Chang, L. J., Lindquist, M. A., & Wager,
S. M., Muckli, L., … Corlett, P. R. (2018). The pre- T. D. (2017). Building better biomarkers: Brain
dictive coding account of psychosis. Biological models in translational neuroimaging. Nature
Psychiatry, 84(9), 634–643. Neuroscience, 20(3), 365–377.
Stroebe, W. (2019). What can we learn from many Woo, C.-W., & Wager, T. D. (2015). Neuroimaging-
lab replications? Basic and Applied Social Psychol- based biomarker discovery and validation. Pain,
ogy, 41(2), 91–103. 156(8), 1379–1381.
Strother, S. C., Anderson, J., Hansen, L. K., Kjems, U., Yarkoni, T. (2009). Big Correlations in Little Studies:
Kustra, R., Sidtis, J., … Rottenberg, D. (2002). The Inflated fMRI Correlations Reflect Low Statistical
quantitative evaluation of functional neuroimag- Power: Commentary on Vul et al. (2009). Perspec-
ing experiments: The NPAIRS data analysis frame- tives on Psychological Science, 4(3), 294–298.
work. NeuroImage, 15(4), 747–771. Yarkoni, T. (2020). The generalizability crisis. Behavio-
Szucs, D., & Ioannidis, J. P. (2020). Sample size evolu- ral and Brain Sciences, 45. doi: 10.1017/
tion in neuroimaging research: An evaluation of S0140525X20001685
highly-cited studies (1990-2012) and of latest prac- Yarkoni, T., Poldrack, R. A., Nichols, T. E., Van Essen,
tices (2017-2018) in high-impact journals. NeuroIm- D. C., & Wager, T. D. (2011). Large-scale auto-
age, 221. doi: 10.1016/j.neuroimage.2020.117164 mated synthesis of human functional neuroimag-
Tenenbaum, J. B., Kemp, C., Griffiths, T. L., & Goodman, ing data. Nature Methods, 8(8), 665–670.
N. D. (2011). How to grow a mind: Statistics, struc- Zunhammer, M., Spisák, T., Wager, T. D., Bingel, U.,
ture, and abstraction. Science, 331, 1279–1285. & Placebo Imaging Consortium (2021). Meta-
Thalmayer, A. G., Toscanelli, C., & Arnett, J. J. (2021). analysis of neural systems underlying placebo
The neglected 95% revisited: Is American psychol- analgesia from individual participant fMRI data.
ogy becoming less American? American Psycholo- Nature Communications, 12(1). doi: 10.1038/
gist, 76(1), 116–129. s41467-021-21179-3.
Index

Note: Page numbers followed by f or t indicate material in figures or tables respectively.

ABC transporters, 90 task-based fMRI, 36–37


Abdollahi, R. O., 289 white matter trajectory, 34–35
absolute pitch, 338 alertness–sleepiness rhythm, 318, 322, 329, 330
acetylcholine (ACh) signaling, 151–152 alpha rhythm, sleepiness, and alertness, 322–323
acquired MEG signal, 535 Altman, J., 496, 497
Adenosine A2A receptors Altschuler, E. L., 289
aged and neurodegenerative brains, 175–176 amygdala
behavioral inhibition, 175 aggression, 232–233
caffeine, 177 anxiety-related stimuli, 230f
cue-motivated behavior, 175 basolateral amygdala (BLA), 232
dopamine and glutamate signaling, 173 basolateral limbic circuit, 228f
memory impairment, 173–174 description of, 227
mood-related behaviors, 175 fear, 229–231
motivation, 175 functional neurological disorders, 233
No-Go pathway, 171–172 input–output relations, 229, 229t
overview of, 171 limbic system, 227–229
reverse cognitive impairments in AD and PD neurological disorders, 233
models, 176–177 neurons, 71, 71f
striatopallidal medium spiny neurons, 171–172 positive emotions, 231
synaptic plasticity, 172–173 prefrontal connectivity, 232
value sensitivity vs. action-outcome smell, 231
contingency, 175 Urbach-Wiethe disease, 233–234
Adolescent Brain Cognitive Development amygdala-precuneus connectivity, 230
(ABCD) study, 571 anatomical cerebro-cerebellar loops, 215–216
Adolfi, F., 18, 23 anterior middle cingulate cortex (aMCC), 295f, 296
Adolphs, R., 234 Apolipoprotein E (APOE), 33
adult neurogenesis Aquino, T. G., 231
acceptance of, 496 Aristotle, 360, 415
behavioral effects, 498 Arnolds, D. E., 186
degenerative diseases, 497 Aron, A. R., 2
evidence of, 496 artifact removal, 542–543
in humans, 497 astroglia
Aeschbach, D., 323 Ca2+ signal generation, 86
age-related alterations classification of, 82, 83f
cognitive control and working memory, 120 diversity, 81–83
episodic memory, 120–121 endfeet, 83
reward learning and decision making, extracellular K+ homeostasis, 88
121–122 functions of, 81–82, 82f, 88–90, 88t
aging brain changes, lifespan glycogen synthesize, 89
BOLD signal, 31 glymphatic system, 89, 89f
brain maintenance (BM) model, 39 homeostatic transporters, 90
changes in gray matter (GM), 31–33 ionic signalling, 87
changes in white matter (WM), 33 morphological appearance, 81–83
environmental effects and models, 38–39 secretory cells, 90–91
functional connectivity with rs-fMRI, 35–36 synaptic transmission, 90, 91f, 92f
future aspects, 39 types of, 84f
genetic effects, 37–38 ATP-dependent transporters, 90
resting state networks (RSN), 35 attention deficit-hyperactivity disorder (ADHD), 268
INDEX 591

auditory attention basolateral amygdala (BLA), 232


description of, 344–345 Bayesian network, 554t, 557
involuntary attention, 345–346 Bear, M. F., 499, 501
voluntary attention, 345 behaviorism, 2, 17
auditory information processing brain’s inner time–space shapes, 24
absolute pitch, 338 input–output relation vs. cognitive function,
auditory attention, 344–346 20–22
auditory processing pathway, 341–342 inside–out vs. outside–in approach, 23–24
auditory representations. see auditory outer time–space of cognition, 24
representations spontaneous activity vs. task-related activity, 22
cocktail-party problem, 337 spontaneous and stimulus induced activity,
high-pitched tone, 338 22–23
instantaneous auditory scene analysis, 343 Bellugi, U., 234
location, 339–340 Berger, H., 22
loudness, 339 Bernstein, M. A., 502
loud sound, 338 Besser, G., 401
neural encoding of sounds, 339–341 Betka, S., 422f
neuronal activation patterns, 340 Bickle, J., 495, 505
organ of Corti, 340 bidirectional activation, 72
pitch, 338 Bienenstock, E., 499
sequential auditory scene analysis, 343–344 Bishop, D. V. M., 22
sound source separation, 342–343 Bizley, J. K., 337
timbre, 339 blood-brain barrier (BBB), 185
tonotopy, 341 blood oxygen level dependent (BOLD) signals, 17, 36,
auditory representations 116, 121, 310, 533, 534
event representations and memory, 346 Bocchio, M., 232
neural representation and predictive coding, Bolognini, N., 2, 4, 5, 291, 438, 440, 444
343–344 Bouton, M. E., 159
autism spectrum disorder (ASD), 268 Boyle, G. J., 1–3, 102, 190, 242
autocorrelation window (ACW), 24 brain
autoencoders (AEs), 554t, 558 aging, 38–39
amygdala, 186
Barber, M., 502 cognitive functioning, 105, 186
Barbey, A. K., 2, 3 disorders, 268–269
Bard, P., 415 dynamic rhythms of, 306–310
Bares, M., 221 functional activity measurement, 533
Barrett, L. F., 415 functions of, 18
basal ganglia (BG) genetic and functional expression, 105–106
action selection, 204–205, 205f genetic effects, 37–38
anatomical structure vs. function, 197–198 hippocampus, 186
anatomy of, 198 imaging. see brain imaging
biophysical modeling, PD. see also Parkinson’s inner time-space, 24
disease (PD) prefrontal cortex (PFC), 186
computational models, 202–203, 206 structural neuroanatomy, 65
direct BG pathway, 200f, 202 structure of, 186
dorsal striatum, 198 brain asymmetries
globus pallidus, 198, 201 body asymmetries, 264, 265f
individual nuclei, 198–201 brain disorders, 268–269
isolated anatomical information, 202 cerebral lobes, 260
model description, 198, 200f ciliopathies, 264–266, 266t
post-mortem anatomical images, 199, 199f experiential and environmental factors, 267
reinforcement learning, 204–205 fetal brain, 261f
striatum (STR), 199–201 genetics and heritability, 266–267
substantia nigra (SN), 198, 201–202 left planum temporale, 260
subthalamic nucleus (STN), 198, 201 sex hormones, 267–268
synergistic approach, 206 sylvian/planum temporale asymmetry, 260
ventral striatum, 198 Yakovlevian counter-clockwise torque, 261
592 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

brain imaging case-controlled study, 268


functional magnetic resonance imaging (fMRI), Catani, M., 229
533–535 Cattaneo, G., 288
future aspects, 547–548 central nervous system (CNS)
machine learning, 548 astroglia, 88, 89
magnetic resonance spectroscopy, 547–548 functional principles, 66
magnetoencephalography (MEG), 535–536, 535f informational complexity, 67
multimodal brain imaging, 547 microglia, 81
positron emission tomography (PET), 536–538, multilevel processing, 66
537f secretory cells, astrocytes, 90–91
brain maintenance (BM) model, 38, 39 structural principles, 66
brain network activity cerebellar motor syndrome (CMS), 221
aversive learning tasks, 155–156 cerebellar reserve, 219
cholinergic activity, 151–152 cerebello-cortical circuits, 218–219
context complexity, 157–158 cerebellum
dopaminergic neurons, 153–154 amphibians, 213
emotional balance, 155 anatomical cerebro-cerebellar loops, 215–216
exteroceptive context, 158–159 cerebellar reserve, 219
glutamatergic activity, 154 cerebello-cortical circuits, 218–219
hedonic and/or reward processing, 157 cerebro-cerebellar connectivity, 215–218
hypothetical scheme, 156f climbing fibers, 213
interoceptive influences, 159 clinical syndromes, 221–222
learning and memory, 151 complex spikes, 214
noradrenergic activity, 152–153 description of, 212
peptides and endocannabinoid systems, 154–155 emotions assesment, 217–218
regulate reward and affective processing, 155 fish and amphibians, 213
reward learning, 156–157 functional anatomy, 216–217
brain rhythms in mammals, 213
connectivity mediated, 310–311 modulator of, 219–221
spatio-temporal dynamics, 306–310 modules, 215
Brain, W. R., 435 mossy fibers, 213
Braun, K. F., 502 motor and non-motor operations, 219–221
Bregman, A. S., 342 multiple climbing fibers, 213
Bremmer, F., 439 neurophysiological aspects, 218–219
Broca, P., 1, 2, 18, 227 physiological aspects, 214–215
Brock, L., 504 plasticity mechanisms, 214–215
Brodmann, K., 280, 374, 380, 522 posterolateral fissure, 213
Brownstone, R.M., 503 primary fissure, 213
Buccino, G., 288, 289 rebound depolarization, 214
Buchanan, T. W., 232 Schmahmann syndrome, 221–222
Buckner, R. L., 36 simple spikes, 214
build–measure–learn process, 50 structural anatomy, 213–214
Bullock, T. H., 504 vestibulospinal tract, 214
Burdach, K. F., 227 cerebral lateralization
brain asymmetries, 260–262
Calvo-Merino, B., 289 emotions and cognition, 262
Cambridge Neuropsychological Test Automated Battery fetal brain, 261f
(CANTAB), 3 handedness, 263–264
Canadian Study of Health and Aging (CSHA) study, hemispheric specialization, 260
177 language lateralization, 262–263
Candy Smell Test (CST), 401, 402 overview of, 259–260
Canli, T., 232 right hemisphere hypothesis (RHH), 262, 262t
Cannon, W. B., 415 valence hypothesis (VH), 262, 262t
canonical correlation analysis (CCA), 554t, 556 cerebro-cerebellar connectivity, 215–218
Cardiovascular risk factors, Aging and Dementia Cerkevich, C. M., 374
(CAIDE), 177 Chamberlain, S. R., 2
Carlson, J. M., 230 Chambers, R., 502
Caruana, F., 295 Chen, B., 403
INDEX 593

Cherry, E. C., 345 Cohen, Y. E., 337


cholinergic activity, 151–152 Cole, K. S., 503
cingulate-amygdala connectivity, 230 connectivity mediated, brain rhythms, 310–311
circuit-based mechanisms, 71, 72 connexins (Cx), 84
Cisek, P., 285 connexons, 83
Cochin, S., 289 continuity illusion, 343
cocktail-party problem, 337 convolutional neural network, 554t, 558
cognition, brain, and aging (COBRA) study, 122–123 Coombs, J., 504
cognitive-behavioral response, 67 Cooper, L. N., 499
cognitive control, 120 cortical responsivity, 72
cognitive neurogenetics Cox, S. R., 33
behavioral genetics, 104 Crawford, J., 358
cultural neurogenetics, 108 cultural neurogenetics, 108
functional pathways, 105 Curtis, H. J., 503
genes and brain function, 103t
genetic and functional expression, 105–107 Daan, S., 319
genetic and neural basis, 107–108, 109f Damasio, A., 415
genetic influences, 103 data harmonization, 554t, 556
genotype-to-phenotype model, 104, 104f Davis, S. W., 34
neurochemical signaling, 102 D1DR stimulation, 116
neurocognitive function, 106 D2DR stimulation, 116
neurotransmission, 104 deep belief network (DBN), 554t, 558
overview of, 102–103 default-mode network (DMN), 17
cognitive neuroscience Denenberg, V. H., 263
architectures and unifying principles, 579–580 Di Cesare, G., 297
BOLD fluctuations, 17 diffusion weighted imaging (DWI), 554
brain function, 18 Dimberg, U., 293
brain signatures/neuromarkers, 581 Ding, S.-L., 245
CANTAB, 3 Dolcos, S., 232
cognitive Atlas, 578 dopamine (DA)
cognitive ontology, 19–20 age-related alterations, 119–122
decade of the brain, 5 cognition, brain, and aging (COBRA) study,
development of, 2 122–123
emotions and nonconscious process, 3 cognitive control and working memory, 117–118
empirical analyses and ontologies, 576, 578–579 cytosolic DA, 115
fMRI research, 17, 18 depletion, 176
historical origin of, 2 dopaminergic pathways, 114–115, 115f
integrative theoretical principles, 580–581 episodic memory, 118–119
interpable models, 581 post-synaptic dopaminergic mechanisms,
large population-based studies, 577t 115–116, 115f
localizationism, 18–20 pre-synaptic dopaminergic mechanisms, 115–116,
methodologies and techniques, 4–5 115f
non-human primates, 6 reward learning and value-based decision making,
overview of, 1 116–117
phase model, 574–575, 574f dopaminergic activity, 153–154
practical constraints, 573–576 Dow, R., 213
predictive modeling approach, 580 Dudek, S. M., 501
from regions to modules, 18–19 dynamic causal modeling (DCM), 204, 554t, 557
replicability of findings, 570–573 dyslexia, 268
replication crisis, 570
reproducibility, 570–573 Ebner, T. J., 220
response inhibition, 2 Eccles, J., 504
robustness, 570–575 Edde, M., 35
scientific developments, 569 electroencephalography (EEG), 198
spatiotemporal cognitive neuroscience, 6 EMOTICOM battery, 3
stimulus–response (S–R) reflexes, 21 emotional memory, 187
cognitive revolution, 3 endogenous opioid system, 154–155
Cohen, D., 535 Engel, A. K., 310
594 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Enhancing Neuro-Imaging Genetics through Garcia, J., 21


Meta-Analysis (ENIGMA), 270 Gazzola, V., 289
epigenome-wide association study (EWAS), 37 Gee, D. G., 231
episodic- autobiographical memory, 232 generative adversarial network, 554t, 558
episodic memory, 120–121 genetic regulation, 107–108
equilibrium potential (EK), 85 genome-wide association study (GWAS), 37, 266, 267
Errante, A., 291 genotype modulation, 105, 106f
event-related brain potential (ERP), 343 genotype-to-phenotype model, 104, 104f
executive functioning (EF) Gerard, R., 502, 503
cognitive flexibility, 188 Gerard, R. W., 503, 504
experimental manipulation, 188–189 Ge, S., 291
inhibition, 188 Gestalt law of common fate, 344
natural hormone fluctuations, 189 Gibson, W. S., 294
pharmacological application, 188 globus pallidus, 201
explicit memory, 133 glutamate (GABA)-glutamine shuttle, 90, 92f
Eyzaguirre, C., 504 glutamatergic activity, 154
Golesorkhi, M., 24
Fadiga, L., 287 Gorbach, T., 33
familiarity-consolidation process Graham, J., 502, 503
catecholaminergic activity, 138 granularity mismatch problem (GMP), 24
cholinergic response, 139 Gray, C. M., 310
dopaminergic release, 138 Gross, C. G., 496
insular cortex, 139 Guell, X., 216
muscarinic administration, 139 gustatory dysfunction
norepinephrine administration, 140 clinical examinations, 405–406
Fechner, G., 472, 474–476, 478–480 description of, 405
Ferrari, P. F., 290 psychophysical tests, 401f
Ferri, J., 230 treatments, 405–406
Finland, Italy, Netherlands Elderly (FINE) study, 177 gustatory sensation
Florey, E., 502, 503 anatomy, 404–405
Flourens, M. J. P., 212, 351 description of, 398–399
Fluid Attenuation Inversion Recovery (FLAIR) gustatory dysfunction, 405–406
acquisitions, 33 physiology, 404–405
Frangou, S., 32
Frank, M. J., 204, 206 Hagiwara, S., 504
Frasnelli, J., 399 Haith, A. M., 387
Fregni, F., 3, 4 Haller, J., 232
French Three Cities study, 177 Hari, R., 289
functional magnetic resonance imaging (fMRI), 74, 116 Hartline, H. K., 504
blood oxygenation level-dependent (BOLD), 533, Hassler, R., 311
534 Hayes, N. L., 497
characteristics of, 534f head-related transfer function (HRTF), 340
independent components analysis (ICA), 540 Hebbian learning, 204
multiband acquisition, 538–539 He, B. J., 457
multi-echo acquisition, 539 hippocampal amnesia, 250
node based analysis, 540–541 hippocampal formation
slice-by-slice acquisition, 538, 539f afferents, 247
study, 2, 231, 232, 439 anatomy of, 243–246
temporal resolution, 534 body, 243, 244f, 246
ultra-high field fMRI, 534 cellular composition, 246–247, 246f
description of, 242–243
GABA neurotransmitter system, 67 efferent, 248
Gage, F. H., 496, 497 encoding, 252
Gallese, V., 293 entorhinal cortex inputs, 247
Galvani, L., 502 episodic memory, 250
Gangitano, M., 287 extrinsic connectivity, 247
gap junctional pathway, 86 head, 245–246
gap junction plaques, 83 intrinsic connectivity, 247
INDEX 595

location of, 244 immuno-competent receptors, 96


long-axis organization, 248–249 implicit memory, 133
long-term declarative memory, 252–253 independent component analysis (ICA), 216, 307,
macrostructural definitions, 243–245 540, 554t, 556
material specificity of, 252 instantaneous auditory scene analysis, 343
memory retrieval, 251 interaural time differences (ITDs), 339
microstructural definitions, 246–247 intercellular signalling system, 85
neurochemical composition, 246–247 interoception
parcellation scheme, 242 anatomy of, 418–419
post-mortem hippocampal tissue, 242, 243f aspects of, 415–416
ram’s horn, 243 central chemoreception, 417
relational memory, 250–251 chemoreception, 416
short- and long-term memory, 251–252 computational modeling, 424–425, 425f
spatial navigation, 252–253 detecting temperature, 417
structural and functional properties, 243 dimensions and clinical symptoms, 423
structure–function relationships, 249–250 dimensions of, 423t
tail, 244, 244f disorders of, 425–426, 427t
vascular supply, 248 emotion, 415
white matter structures, 246–247 exteroceptive senses, 414
Hodgkin, A., 503, 505 functions, 414
Holmes, G., 212 heartbeat detection tasks, 421, 422f
Ho, N. F., 232 humoral physiology, 419
Honolulu-Asia Aging study, 177 interoceptors, 416
hormones interregional functional connectivity, 420
bidirectionality, 190–191 levels of, 421–423
classes of, 185 mechanoreception, 416
cognitive function assesment, 189–190 NTS neurons, 418–419
description of, 185 pathways to brain, 418
effects of, 186 perception, 414–415
executive functioning (EF), 188–189 phasic interoceptive signals, 422
interactions, 186–189 psychological representations, 420–421
memory, 186–188 therapeutic targeting, 427–428
natural fluctuations, 186 thermal information, 419–420
peptide hormones, 185 intracellular recording
steroid hormones, 185 complex behaviors and cognition,
human astrocytes, 82 504–505
Human Brain Project (HBP) development of, 502
biological neurons communication, 517, 518f electrophysiology approach, 502
brain to robotics, 519–520 functional imaging and optogenetics, 505
cognition and perception, 515 investigations, 503
computational models, 514–515 mammalian muscle fibres, 503
EBRAINS’ computational resources, micropipette electrode, 503
511, 512 neurotransmission, 504
international context, 524–525, 526t revolution in, 505
mean-field models, 513, 514f vertebrate CNS neurons, 504
multidisciplinary approach, 511 intra-CNS pathways, 185
multi-level human brain Atlas, 522–524, 525f intrinsic connectivity, 72
neuroethical questions, 526 involuntary attention, 345–346
overview of, 511–512 Ishida, H., 290
psychological levels, 516 Ito, M., 220
representations and consciousness, 520–521 Ivry, R. B., 218
spiking neural network models, 516–519 Izmailov, C. A., 491
systems-level models, 512–513
testing contextual cortical processing, 516 Jahanshahi, M., 2, 4, 5, 386–389, 392
workflows, 521–522, 523f James–Lange therapy, 415
Hummel, T., 399 James, W., 351, 415, 421
Huntington’s disease, A2AR blockade, 176 Jansen, J., 213
Huxley, A., 503, 505 Jiang, R.-S., 152
596 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Kaas, J. H., 374 characteristics of, 544f


Kalaska, J. F., 285 network connectivity, 543–544
Kandel, E., 497 presurgical functional mapping, 536
Kant, I., 18, 19 sensors, 536
Kaplan, M., 496 source localization, 541–542, 542f
Karhu, J. J., 218 superconducting quantum interference device
Katsumi, Y., 232 (SQUID), 536
Kawato, M., 220 task-evoked cognitive process, 536
Keysers, C., 290, 293 major depressive disorder (MDD), 232
Koelling, R. A., 21 Malacarne, V., 212
Kohler, E., 283 Manan, H. A., 403
Kolliker, A., 496 many-to-one mapping, 68
Kraskov, A., 283 Mao, Y., 230
Krolak-Salmon, P., 293 Markov model, 554t, 557
Kuffler, S. W., 504 Marr-Albus-Ito theory, 215
Kuhn, T., 495, 497, 500 Marreiros, A. C., 206
Masssatricht Aging Study, 177
Lange, K. G., 415, 421 maternal aggressive behavior, 232
Langley, C., 3 Matsunaga, M., 294
Lanteaume, L., 294 Mattos, J. L., 400
Lanzilotto, M., 287 McDermott, J. H., 343
large-scale data, neuroimaging medium spiny projection neurons (MSN), 200
deep learning (DL) techniques, 558 Mehta, M. A., 3
machine learning (ML) methods, 557–558 Meltzoff, A. N., 290
modalities, 553–555 memory, hormone interactions
random forest (RF) model, 558 cortisol, 187
source separation, 556–557 experimental manipulation, 187–188
statistical challenges, 555–556 glucocorticoid cortisol, 187
Larsell, O., 213 natural hormone fluctuations, 188
Lashley, K., 2 pharmacological application, 187
lateral geniculate nucleus (LGN), 68 memory retrieval, 134
Laurent, V., 156 Merton, P. A., 385
L-DOPA administration, 121 Merzenich, M., 267
LeDoux, J. E., 229, 230 mesolimbic pathway, 118
Lew, C. H., 233 Mesulam, M.-M., 229
Ling, G., 502, 503, 504 Meyer, M. F., 20
Liu, X., 400 microglia
Li, X., 231 description of, 93–94
locus coeruleus (LC), 185 diversity of, 94t
locus coeruleus neurons, 118 ion signaling, 95–98
long-term episodic memory, 118–119 morphological properties, 94–95
long-term memory (LTM), 133, 134f physiological functions, 94, 95f
Lopes da Silva, F. H., 186 physiological microglial morphotypes, 95, 96f
loudness, 339 P2X7 receptors, 96
Luciani, L., 212 Midlife in the US (MIDUS) study, 190
Lui, F., 231 Mikhalevskaya, M. B., 474
Luria, A., 21 Mild Cognitive Impairment (MCI), 3, 31, 177
mirror mechanism
Mach, E., 351 anterior middle cingulate cortex (aMCC), 296
machine learning, 5, 39, 55, 56, 467, 516, 540, 548, cortical areas, 285
557, 569, 572, 580 cortical motor system and properties, 280–281,
MacLean, P. D., 227, 228f 280f
Maeda, K., 287 disgust and anterior insula, 293–294
magnetic resonance imaging (MRI), 198 electrophysiological studies, 289–290
magnetic resonance spectroscopy, 547–548 emotions, 293
magnetoencephalography (MEG) fear and amygdala, 294
acquired MEG signal, 535 frontal cortex, 285–286
artifact removal, 542–543 goal coding, 289
INDEX 597

historical premise, 279–280 overview of, 435


human mirror system, 287–288 RFs of visuo-tactile neuron, 436, 437f
in monkey, 282–284, 282f, 284f schematic representation, 436f
motor goals, 281 social side of, 443–444
neurons and intention, 292–293, 292f visuo-tactile stimulation, 444, 445
overview of, 279 neural communication, 65
parietal cortex, 286–287 neural encoding, 339–341
parieto-frontal mirror, 284–285 neural processes and activity
phylogenesis and development, 290 cognition and higher-level processes, 74–75
pregenual anterior cingulate cortex (pACC ), cognitive neural networks, 72–73
294–295, 295f cultural neuroscience, 72
somatosensory cortices, 290–291 facial recognition, 69, 70f
somatotopy of observed acts, 288–289 functional principles, 66
subcortical structures, 291 neural networks, 73–74
vitality forms, 296–297, 297f neurons types, 66–68
Mismatch Negativity (MMN) component, 344 sensation and perception, 68, 69f
molecular-imaging method, 116 social perception, 69–71
Moore, M. K., 290 structural principles, 66
Moran, R. J., 204, 206 type representataion, 68
motor control system neurite orientation dispersion and density imaging
afferent sensory information, 384 (NODDI), 34
building blocks, 380–382, 381f neurodegenerative disorders
internal forward and inverse models, 382–385, corticobasal degeneration, 269
383f Pick’s disease, 269
motor chauvinist’s perspective, 379 neurodevelopmental disorders
putative stages, 387 attention deficit-hyperactivity disorder (ADHD),
reaction time (RT) tasks, 386, 386f 268
response inhibition, 389–393, 391f autism spectrum disorder (ASD), 268
response preparation and initiation, 385–389 dyslexia, 268
roles, 379–380 neuroglia
transcranial magnetic stimulation, 385 astrocytic networks/syncytia, 83–85, 85f
MSN D1 receptors, 204 astroglia, 81–83, 88–90
MSN D2 receptors, 204 classification of, 80–81, 81f
Mukamel, R., 289 CNS neuroglia, 80
multiband acquisition, 538–539 defined, 79–80
Multidimensional Scale of Perceived Social Support, gliocrine system, 90–91
231 intracellular excitability, 86–88
multi-echo acquisition, 539 ion channels, 85–86
multi-layer perceptron (MLP), 554t, 558 ion distribution, 85–86
multimodal brain imaging, 547 mammalian nervous system, 80
multimodal neuroimaging study, 121, 122 membrane potential, 85–86
muscarinic receptors, 141 microglia, 93–95
myelination, 93 neurohormones, 86
neuromodulators, 86
Nastuk, W. L., 503 neurotransmitters, 86
natural sound waves, 339 numbers of, 81
nervous tissue oligodendrocyte precursors (NG2-glia), 92, 93
active milieu, 79, 80f overview of, 79
multicellular nature, 79 pathophysiology of, 97t–98t, 98–99
network connectivity, 543–544 PNS neuroglia, 80
neural bases, peripersonal space representation Neuroimaging Meta-Analysis Research Environment
appetitive function, 441–442 (NiMARE), 5
convergent evidence, 444 neuromodulators, 134
defensive function, 442–443 neuronal activation, 65
functional significance, 441 neuronal mechanisms
hand blink reflex (HBR), 442, 443f cortical specialization, 72
in humans, 438–441, 439f, 440f functional and emergent properties, 72
in monkey, 435–438, 436f, 437f response properties, 74
598 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

role of, 75 olfactory sensation


social perception, 71 age and gender, 398
neurotechnology startups anatomy, 399
advances in, 48 assessment, 399–400
application of, 59 description of, 398
brain–computer interfaces, 56 electrophysiology and functional brain
brain imaging, 54–55 imaging, 402–403
brain–machine interface technology, 53 neuroepithelium, 399
build–measure–learn process, 50 olfactory dysfunction, 403
cognitive and learning neurotechnologies, orthonasal olfaction, 399
58–59 physiology, 399
data ownership, 54 psychometric questionnaires, 399–400
development and innovation chain, 54f psychophysical tests, 400–402, 401f
electrophysiologic activity, 55–56 retronasal olfaction, 399
emphatic stimulation and investment, 53 structural brain imaging, 403
gene therapy, 55 testing in children, 402
innovation translation, 57–58 women, 398
innovative implementations, 48 oligodendroglia
investment, 52–53 functional properties of, 92–93
lean startup method, 50–51 myelination, 93
minimal viable product (MVB), 50 NG2-glia, 92, 93
neurostimulation devices, 57, 57t orientation dispersion index (ODI), 34
overview of, 48–49 orthonasal olfactory function, 400–401
pharmacological drugs, 55
pivoting, 51 Padlubnaya, D. B., 155
real-life neuroscience startups, 54 panglial syncytium, 84
remote EEG system, 56 Papez Circuit, 227
risk of failure, 49 Parker, D., 495
Stanford University’s Biodesign Model, Parkinson’s disease (PD)
49–50 beta oscillations, 204
user experience and innovation cycle, 49 biologically constrained computational
world’s brain research projects, 51–52 model, 205f
Nishitani, N., 289 cardinal symptoms, 203
nociception mechanism, 154 dopamine availability, 203
non-invasive neuromodulation techniques, 4–5 dynamical systems theory, 203
noradrenergic activity, 152–153 dynamic causal modelling (DCM), 204
norepinephrine system, 105 long feedback loops, 203
Northoff, G., 1, 3, 6, 17, 18, 20, 22–24 Pascual-Leone, A., 4, 5, 38, 306, 308
novel object recognition (NOR), 136 Patton, H., 504
novelty signaling Pavlov, I., 21
c-Fos expression, 136 Pearson, 490
dopamine reuptake, 137 Penfield, W., 358
dopaminergic response, 138 peptide hormones, 185
neurochemistry, 138–140 perfusion weighted imaging (PWI), 554
norepinephrine administration, 137 peripersonal space representation
Nowakowski, R. S., 497 appetitive function, 441–442
Nucleus of the Solitary Tract (NTS), 418 convergent evidence, 444
defensive function, 442–443
object location memory (OLM), 136–138 functional significance, 441
object-related negativity (ORN), 343, 344, 399 hand blink reflex (HBR), 442, 443f
Ogawa, S., 534 in humans, 438–441, 439f, 440f
Olbrich, S., 329 in monkey, 435–438, 436f, 437f
olfactory dysfunction overview of, 435
classification, 403 RFs of visuo-tactile neuron, 436, 437f
clinical examinations and treatments, 403–404 schematic representation, 436f
description of, 403 social side of, 443–444
epidemiology, 403 visuo-tactile stimulation, 444, 445
prevalence of, 403 Peterfi, T., 502
INDEX 599

Phillips, W. A., 293 psychophysical tests


Pick’s disease, 269 orthonasal olfactory function, 400–401
Pieniak, M., 402 retronasal olfactory function, 401–402
Pishnamazi, M., 234 Puccetti, N. A., 231
Poeppel, D., 18, 23 Purkinje cell layer, 213
positron emission tomography (PET) Purkinje, J. E., 351
advance methadology, 538 Purkyn, J. E., 212
characteristics of, 537f
kinetic modeling, 545–546 Raichle, M. E., 18, 22
metabolic connectivity, 546–547 Rakic, P., 496, 497
radiolabeled tracers, 536 Ramón y Cajal, S., 212, 496
radiopharmaceutical development, 537 rebound depolarization, 214
silicon photomultipliers (SiPMs), 537 recognition memory
time-of-flight (TOF) PET, 544–545 consolidation of, 138–139
Postema, M. C., 268 description of, 135
posterior inferior cerebellar artery (PICA), 221 familiarity, 135, 135f
pregenual anterior cingulate cortex (pACC ), hippocampal formation, 135
294–295, 295f learning and relearning, 140–141
presurgical functional mapping, 536
memory updating, 140–141
primary somatosensory cortex, 372–373
novel object recognition (NOR), 136
proprioceptive afferents, 367
novelty signaling, 136–138
protoplasmic astrocytes, 83, 86
object location memory (OLM), 136
psychiatric disorder
recollection, 135, 135f
bipolar affective disorder, 269
taste recognition memory, 135
major depressive disorder (MDD), 270
reconsolidation, 134
obsessive-compulsive disorder (OCD), 270
recurrent neural network (RNNs), 554t, 558
schizophrenia, 269
residual neural network, 554t, 558
psychophysical techniques
response inhibition
conception of two thresholds, 474
automatic mechanism, 390, 391, 393
confidence rating method (CRM), 484–485,
behavioral assessment, 389, 390, 391f
485t
conditional stop-signal task (CSST), 390
Fechner’s methods, 478–479
electrophysiological recordings, 392
forced choice method (FCM), 484
proactive inhibition, 390
High-Threshold Theory, 473–474
reactive inhibition, 389, 390
Low-Threshold Theory, 474
SMA impairments, 393
machine-like approach, 477
supplementary eye field (SEF), 393
MDS vs. factory analysis, 492
resting-state networks (RSN), 310, 311
method of adjustment, 480
restricted Boltzmann machines (RBMs), 558
method of constant stimuli, 480–482
Restuccia, D., 218
method of limits, 479–480
retronasal olfactory function, 401–402
metric multidimensional scaling (mMDS),
reward learning, 121–122
490–492
reward prediction error (RPE), 116
multidimensional scaling (MDS), 488–490
Rizzolatti, G., 293
neuro-quantum (NQ) theory, 473
Rolando, L., 212
non-metric multidimensional scaling (nMDS),
Rolls, E., 231
492
Rubin, J. E., 206
overview of, 472
Rutishauser, U., 232
psychology activity, 477–478
psychophysical law, 476–477
SDT sensory axis, 473–474 Sahakian, B. J., 1, 3, 5, 6, 138, 500
sensory range discreteness/continuity problem, Sanami, M., 58
472–473 sarco-endo-plasm reticulum (SERCA), 86
sensory sensitivity and decision making, 486 Sato, M., 231
signal detection theory, 473 Schepers, S. T., 159
Stevens’s scaling methods, 475–476, 486–487 Schmahmann, J. D., 212, 216
Theory of Two States, 474 Schmahmann syndrome, 221–222
Three States Theory, 474 Schwab, S., 310
Thurstone’s approach to scaling, 474–475 Schwann cells, 80
600 THE SAGE HANDBOOK OF COGNITIVE AND SYSTEMS NEUROSCIENCE

Sejnowski, T. J., 499 sound pressure level (SPL), 281, 339


Sejnowski, T.J., 501 sound source separation, 342–343
Self-Reported Mini Olfactory Questionnaire source localization, 541–542, 542f
(Self-MOQ), 400 spatial navigation, 252–253
Seligman, M., 21 spatiotemporal cognitive neuroscience, 6
sensory stimulus, 68, 72, 73, 340, 341, 438 spatio-temporal dynamics, brain rhythms
sequential auditory scene analysis, 343–344 advantages, 308
Sherrington, C. S., 415, 416 decomposition of EEG signal, 308, 309f
short-term memory (STM), 133, 139 EEG oscillations, 305, 306, 308
silicon photomultipliers (SiPMs), 537 generalized eigenvalue decompositions (GED),
Simoncelli, E. P., 343 307, 308
Sinek, S., 49 ongoing debate, 308
Singer, T., 296 overview, 305–306
single nucleotide polymorphism (SNP), 266, 267 quantitative EEG (qEEG) approach, 306
Sjogard, M., 312 spatial non-stationarity of Alpha-EEG, 306, 307f
Skeldon, A. C., 319 time-frequency approach, 310
Skinner, B. F., 20, 21 Specter, M., 496
sleep, alertness, and circadian rhythms spinothalamic pathways, 368–369
alertness–sleepiness rhythm, 322 Sprengelmeyer, R., 293
alpha rhythm, sleepiness, and alertness, 322–323 Stanford University’s Biodesign Model
EEG slow wave activity, 319 identification phase, 49–50
overview of, 318–319 implementation phase, 50
sleep times and sleepiness levels, 321t invention phase, 50
sleep–wake regulation (SWA), 319–321, 320f in neurotech field, 50
sleepiness, alpha waves Stanton, P.K., 501
artifact-free epochs, 324 Stent, G. S., 499
diurnal variation, 329 Stern, D., 296
EEG recording session, 324, 325 steroid hormones, 185
experimental deprivation studies, 325–326 Stevens, S., 475–476, 487
independent factors, 328t Stevens’s scaling methods, 475–476, 486–487
methods of deprivation studies, 324–325 striatal neurogenesis, 497
objective and subjective measures, 328t striatal neurons, 202, 204
power spectra and time courses, 326, 327f striatum (STR)
repeated measure ANOVAs (rANOVAs), 325 anatomo-functional specificity, 200
self-ratings of sleepiness, 326 dorsal STR, 199
subjective sensation, 323 microscopic appearance, 200
sleep–wake regulation (SWA), 319–321, 320f topographical zonation, 201
slice-by-slice acquisition, 538, 539f Strick, P. L., 216
solute carrier transporters (SLCs), 90 Stuart, D.G., 503
somatosensory cortex, 370–372, 371f substantia nigra (SN), 114, 115, 201–202
somatosensory midbrain, 369 subthalamic nucleus, 201
somatosensory nuclei, 370f superconducting quantum interference device (SQUID),
somatosensory processing 536
afferent pathways and target nuclei, 367–368 superior colliculus (SC), 369
contralateral body surface, 373 superior temporal sulcus (STS), 69
glabrous skin and hairy skin, 366–367 cellular mechanisms, 70
primary somatosensory cortex, 372–373 electrophysiological signaling, 71
processing levels, 368f response selectivity, 70
proprioceptive inputs, 373–374 support vector machine, 464, 554t, 557
somatosensory cortex, 370–372, 371f sympathetic nervous system (SNS), 190
somatosensory midbrain, 369 synaptic plasticity, 204
somatosensory nuclei, 370f anti-Hebbian process, 499
somatosensory thalamus, 369–370 BCM model, 499, 500
spinothalamic pathways, 368–369 long-term depression (LTD), 172, 173
tactile receptors and afferents, 366–367 long-term potentiation (LTP), 172, 173, 498, 501
upper bank of lateral sulcus, 374 LTD-LTP shift, 173
somatosensory thalamus, 369–370 NMDAR activity, 173
Sorrells, S. F., 497 wet neurobiology, 498–502
INDEX 601

tactile afferents, 367 EEG signature, 358


Takahashi, R. N., 231 endolymph, 351
Talairach, J., 296 galvanic vestibular stimulation (GVS) effect,
Tang, Y., 232 356, 358
Tasaki, I., 504 hair cells, 352–353, 353f
taste powders, 402 head movement and sensation, 351, 352f
Taylor, C. V., 503 human subcortical regions, 353–354
Tesche, C. D., 218 multimodal representation of verticality, 357
thermoreception, 418–419 multisensory guidance of action, 356–357
Thurstone, L., 474, 487 nucleus tractus solitarius (NTS), 359
timbre, 339 otolith organs, 352
time-of-flight (TOF) PET, 544–545 sensory organs, 351
time series data spatial resolution and compatibility, 355–356
cross-frequency coupling, 458f, 463f thalamic neuronal responses, 355
data analysis, 456 vestibulo-ocular reflexes (VORs), 354
electrophysiological data analysis, 456 visually induced motion sickness (VIMS), 359
event-related analysis, 456–457, 457f visually induced vestibular subjective
intracranial human electrophysiology, 455f experiences, 358–359
multivariate representations and analyses, vestibulo-cerebellar syndrome (VCS), 221
462–465, 464f, 465f vitality forms, 296–297, 297f
neural recordings, 454–455 voluntary attention, 345
oscillatory and broadband activity, 459–460, 459f Von der Heide, R. J., 233
oscillatory and waveform shape features, Von der Malsburg, C., 498, 499
460–461, 461f von Holst, E., 21
overview of, 453–454 von Monakov, C., 2
peri-stimulus time histograms (PTSH), 457 Voogd, J., 213
scale-free phenomena, 465–466, 466f
spatial and temporal scales, 461–462 Watson, J. B., 20
spectral analysis, 457–459, 459f, 460f Wernicke, C., 2
statistical considerations, 466 wet neurobiology
theoretical and decoding approaches, 464, 465f adult neurogenesis, 495–498
Tkach, D., 285 intracellular recording, 502–505
Tolman, E., 21 overview of, 495
tonotopy, 341 synaptic plasticity, 498–502
Torgerson, W., 490 Whitaker, D. M., 503
transcranial electrical stimulation, 218 Wolff, A., 23
transcranial magnetic stimulation (TMS), 51, 218 Woodbury, J. W., 503, 504
transient receptor potential (TRP), 87 working memory, 120, 188
Trier Social Stress Test (TSST), 190 World’s brain research projects
two-day and one-night sleep deprivation study, 325, 326 China Brain Project (CBP), 51
two-day deprivation study, 326 Human Brain Project (HBP), 51
two-day sleep deprivation study, 324 International Brain Initiative (IBI), 52
Neurotechnology Industry Organization
UK Biobank study, 575 (NIO), 52
ultra-high field fMRI, 534 US BRAIN initiative project, 51
Urbach-Wiethe disease, 233–234
Yoshino, A., 285, 286, 402
valence hypothesis (VH), 262, 262t Yuval-Greenberg, S., 311
Van Hoesen, G. W., 245
ventral tegmental area (VTA), 114, 115, 185 Zabrodin, Yu. M., 477
ventroposterior nucleus (VP), 369 Zeiss, C., 502
verbal fluency, 188 zero-phase synchronization
vestibular processing EEG/MEG recording, 311
benign paroxysmal positioning vertigo (BPPV), meaning phase-lagged connectivity, 312
359 simultaneous synchronous network, 312
body representation in space, 357–358 volume conduction, 311, 312, 313f
cortical processing, 354–355 Zou, L. Q., 400

You might also like